Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
EMBO J ; 39(14): e103812, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32488939

RESUMEN

It is controversial whether mitochondrial dysfunction in skeletal muscle is the cause or consequence of metabolic disorders. Herein, we demonstrate that in vivo inhibition of mitochondrial ATP synthase in muscle alters whole-body lipid homeostasis. Mice with restrained mitochondrial ATP synthase activity presented intrafiber lipid droplets, dysregulation of acyl-glycerides, and higher visceral adipose tissue deposits, poising these animals to insulin resistance. This mitochondrial energy crisis increases lactate production, prevents fatty acid ß-oxidation, and forces the catabolism of branched-chain amino acids (BCAA) to provide acetyl-CoA for de novo lipid synthesis. In turn, muscle accumulation of acetyl-CoA leads to acetylation-dependent inhibition of mitochondrial respiratory complex II enhancing oxidative phosphorylation dysfunction which results in augmented ROS production. By screening 702 FDA-approved drugs, we identified edaravone as a potent mitochondrial antioxidant and enhancer. Edaravone administration restored ROS and lipid homeostasis in skeletal muscle and reinstated insulin sensitivity. Our results suggest that muscular mitochondrial perturbations are causative of metabolic disorders and that edaravone is a potential treatment for these diseases.


Asunto(s)
Aminoácidos de Cadena Ramificada/metabolismo , Lipogénesis , Músculo Esquelético/metabolismo , Fosforilación Oxidativa , Animales , Ratones , Ratones Transgénicos
2.
Glia ; 69(3): 619-637, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33010069

RESUMEN

Fast synaptic transmission in vertebrates is critically dependent on myelin for insulation and metabolic support. Myelin is produced by oligodendrocytes (OLs) that maintain multilayered membrane compartments that wrap around axonal fibers. Alterations in myelination can therefore lead to severe pathologies such as multiple sclerosis. Given that hypomyelination disorders have complex etiologies, reproducing clinical symptoms of myelin diseases from a neurological perspective in animal models has been difficult. We recently reported that R-Ras1-/- and/or R-Ras2-/- mice, which lack GTPases essential for OL survival and differentiation processes, present different degrees of hypomyelination in the central nervous system with a compounded hypomyelination in double knockout (DKO) mice. Here, we discovered that the loss of R-Ras1 and/or R-Ras2 function is associated with aberrant myelinated axons with increased numbers of mitochondria, and a disrupted mitochondrial respiration that leads to increased reactive oxygen species levels. Consequently, aberrant myelinated axons are thinner with cytoskeletal phosphorylation patterns typical of axonal degeneration processes, characteristic of myelin diseases. Although we observed different levels of hypomyelination in a single mutant mouse, the combined loss of function in DKO mice lead to a compromised axonal integrity, triggering the loss of visual function. Our findings demonstrate that the loss of R-Ras function reproduces several characteristics of hypomyelinating diseases, and we therefore propose that R-Ras1-/- and R-Ras2-/- neurological models are valuable approaches for the study of these myelin pathologies.


Asunto(s)
Axones , Vaina de Mielina , Animales , Diferenciación Celular , Sistema Nervioso Central , Ratones , Oligodendroglía
3.
Mol Cell ; 45(6): 731-42, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22342343

RESUMEN

Recent findings indicate that prevalent human carcinomas overexpress the mitochondrial ATPase Inhibitory Factor 1 (IF1). Overexpression of IF1 inhibits the synthase activity of the mitochondrial H(+)-ATP synthase and plays a crucial role in metabolic adaptation of cancer cells to enhanced aerobic glycolysis. Herein, we demonstrate that IF1 overexpression in colon cancer cells triggers mitochondrial hyperpolarization and the subsequent production of superoxide radical, a reactive oxygen species (ROS). ROS are required to promote the transcriptional activation of the NFκB pathway via phosphorylation-dependent IκBα degradation. Activation of NFκB results in a cellular adaptive response that includes proliferation and Bcl-xL mediated resistance to drug-induced cell death. Quenching the mitochondrial production of ROS prevents the activation of NFκB and abolishes the IF1-mediated cellular adaptive response. Overall, our findings provide evidence linking the activity of a mitochondrial protein with retrograde signaling to the nucleus to promote cellular proliferation and survival.


Asunto(s)
Proliferación Celular , Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Metabolismo Energético , Fluorouracilo/farmacología , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Proteínas/genética , Transducción de Señal , Estaurosporina/farmacología , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Proteína Inhibidora ATPasa
4.
Diabetologia ; 61(12): 2674, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30324489

RESUMEN

Owing to an oversight, the authors omitted to note that Dr Taub is a co-founder of and equity holder in Cardero Therapeutics.

5.
EMBO J ; 33(7): 762-78, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24521670

RESUMEN

A key transducer in energy conservation and signaling cell death is the mitochondrial H(+)-ATP synthase. The expression of the ATPase inhibitory factor 1 (IF1) is a strategy used by cancer cells to inhibit the activity of the H(+)-ATP synthase to generate a ROS signal that switches on cellular programs of survival. We have generated a mouse model expressing a mutant of human IF1 in brain neurons to assess the role of the H(+)-ATP synthase in cell death in vivo. The expression of hIF1 inhibits the activity of oxidative phosphorylation and mediates the shift of neurons to an enhanced aerobic glycolysis. Metabolic reprogramming induces brain preconditioning affording protection against quinolinic acid-induced excitotoxicity. Mechanistically, preconditioning involves the activation of the Akt/p70S6K and PARP repair pathways and Bcl-xL protection from cell death. Overall, our findings provide the first in vivo evidence highlighting the H(+)-ATP synthase as a target to prevent neuronal cell death.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , Proteínas/genética , Transducción de Señal , Animales , Apoptosis , Conducta Animal , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Glucólisis/efectos de los fármacos , Humanos , Masculino , Redes y Vías Metabólicas , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Modelos Animales , Mutación Missense , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neurotoxinas/farmacología , Fosforilación Oxidativa , Regiones Promotoras Genéticas/genética , Proteínas/metabolismo , Ácido Quinolínico/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteína Inhibidora ATPasa
6.
Diabetologia ; 60(10): 2052-2065, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28770317

RESUMEN

AIMS/HYPOTHESIS: Mitochondria are important regulators of the metabolic phenotype in type 2 diabetes. A key factor in mitochondrial physiology is the H+-ATP synthase. The expression and activity of its physiological inhibitor, ATPase inhibitory factor 1 (IF1), controls tissue homeostasis, metabolic reprogramming and signalling. We aimed to characterise the putative role of IF1 in mediating skeletal muscle metabolism in obesity and diabetes. METHODS: We examined the 'mitochondrial signature' of obesity and type 2 diabetes in a cohort of 100 metabolically characterised human skeletal muscle biopsy samples. The expression and activity of H+-ATP synthase, IF1 and key mitochondrial proteins were characterised, including their association with BMI, fasting plasma insulin, fasting plasma glucose and HOMA-IR. IF1 was also overexpressed in primary cultures of human myotubes derived from the same biopsies to unveil the possible role played by the pathological inhibition of the H+-ATP synthase in skeletal muscle. RESULTS: The results indicate that type 2 diabetes and obesity act via different mechanisms to impair H+-ATP synthase activity in human skeletal muscle (76% reduction in its catalytic subunit vs 280% increase in IF1 expression, respectively) and unveil a new pathway by which IF1 influences lipid metabolism. Mechanistically, IF1 altered cellular levels of α-ketoglutarate and L-carnitine metabolism in the myotubes of obese (84% of control) and diabetic (76% of control) individuals, leading to limited ß-oxidation of fatty acids (60% of control) and their cytosolic accumulation (164% of control). These events led to enhanced release of TNF-α (10 ± 2 pg/ml, 27 ± 5 pg/ml and 35 ± 4 pg/ml in control, obese and type 2 diabetic participants, respectively), which probably contributes to an insulin resistant phenotype. CONCLUSIONS/INTERPRETATION: Overall, our data highlight IF1 as a novel regulator of lipid metabolism and metabolic disorders, and a possible target for therapeutic intervention.


Asunto(s)
Dislipidemias/metabolismo , Resistencia a la Insulina/fisiología , Mitocondrias Musculares/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Músculo Esquelético/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Masculino , Obesidad/metabolismo , Proteómica
7.
J Biol Chem ; 288(51): 36530-7, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24194524

RESUMEN

Massive poly(ADP-ribose) formation by poly(ADP-ribose) polymerase-1 (PARP-1) triggers NAD depletion and cell death. These events have been invariantly related to cellular energy failure due to ATP shortage. The latter occurs because of both ATP consumption for NAD resynthesis and impairment of mitochondrial ATP formation caused by an increase of the AMP/ADP ratio. ATP depletion is therefore thought to be an inevitable consequence of NAD loss and a hallmark of PARP-1 activation. Here, we challenge this scenario by showing that PARP-1 hyperactivation in cells cultured in the absence of glucose (Glu(-) cells) is followed by NAD depletion and an unexpected PARP-1 activity-dependent ATP increase. We found increased ADP content in resting Glu(-) cells, a condition that counteracts the increase of the AMP/ADP ratio during hyperpoly(ADP-ribosyl)ation and preserves mitochondrial coupling. We also show that the increase of ATP in Glu(-) cells is due to adenylate kinase activity, transforming AMP into ADP which, in turn, is converted into ATP by coupled mitochondria. Interestingly, PARP-1-dependent mitochondrial release of apoptosis-inducing factor (AIF) and cytochrome complex (Cyt c) is reduced in Glu(-) cells, even though cell death eventually occurs. Overall, the present study identifies basal ADP content and adenylate kinase as key determinants of bioenergetics during PARP-1 hyperactivation and unequivocally demonstrates that ATP loss is not metabolically related to NAD depletion.


Asunto(s)
Metabolismo Energético , Glucosa/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Células 3T3 , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Adenilato Quinasa/metabolismo , Animales , Factor Inductor de la Apoptosis/metabolismo , Citocromos c/metabolismo , Activación Enzimática , Células HeLa , Humanos , Ratones , Mitocondrias/metabolismo , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Poli Adenosina Difosfato Ribosa/metabolismo
8.
Nat Metab ; 6(2): 209-225, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38243131

RESUMEN

Coenzyme Q (Q) is a key lipid electron transporter, but several aspects of its biosynthesis and redox homeostasis remain undefined. Various flavoproteins reduce ubiquinone (oxidized form of Q) to ubiquinol (QH2); however, in eukaryotes, only oxidative phosphorylation (OXPHOS) complex III (CIII) oxidizes QH2 to Q. The mechanism of action of CIII is still debated. Herein, we show that the Q reductase electron-transfer flavoprotein dehydrogenase (ETFDH) is essential for CIII activity in skeletal muscle. We identify a complex (comprising ETFDH, CIII and the Q-biosynthesis regulator COQ2) that directs electrons from lipid substrates to the respiratory chain, thereby reducing electron leaks and reactive oxygen species production. This metabolon maintains total Q levels, minimizes QH2-reductive stress and improves OXPHOS efficiency. Muscle-specific Etfdh-/- mice develop myopathy due to CIII dysfunction, indicating that ETFDH is a required OXPHOS component and a potential therapeutic target for mitochondrial redox medicine.


Asunto(s)
Flavoproteínas Transportadoras de Electrones , Fosforilación Oxidativa , Ubiquinona , Animales , Ratones , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Homeostasis , Lípidos , Músculo Esquelético/metabolismo , Ubiquinona/metabolismo
9.
Eur J Neurosci ; 36(1): 1993-2005, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22639866

RESUMEN

Ischemic tolerance is a phenomenon in which exposure to a mild preconditioning stress results in resistance to a subsequent lethal ischemic insult. Here we investigated the role of poly(ADP-ribose) polymerase (PARP) in the development of ischemic tolerance by using organotypic rat hippocampal slices exposed to 30 min oxygen-glucose deprivation (OGD), which leads to selective injury of the CA1 subregion 24 h later. We developed models of pharmacological preconditioning by exposing slices to subtoxic concentrations of either N-methyl-D-aspartate (NMDA) or (S)-3,5-dihydroxyphenylglycine (DHPG) and then, 24 h later, to 30 min OGD. Under these conditions, we observed a significant reduction in OGD-induced CA1 damage. Exposure of slices to the PARP-1 and -2 inhibitors TIQ-A, PJ-34 and UPF 1069 during preconditioning prevented the development of OGD tolerance in a concentration-dependent manner. NMDA and DHPG preconditioning increased the activity of PARP, as detected by immunoblots using antibodies against the poly(ADP-ribose) polymer product, but was not associated with consumption of cellular NAD(+) or ATP. Neuroprotection induced by preconditioning was also prevented by the caspase inhibitor Z-VAD-FMK. The modest but significant increase in caspase-3/7 induced by preconditioning, however, was not associated with PARP-1 cleavage, as occurred with staurosporine. Finally, TIQ-A prevented the activation of ERK1/2 and Akt induced by NMDA preconditioning, suggesting that the protective mechanism evoked by PARP requires activation of these prosurvival mediators. Our results suggest that preconditioning with appropriate pharmacological stimuli may promote neuroprotective mechanisms triggered by the sublethal activation of two otherwise deleterious executioners such as PARP and caspase-3/7.


Asunto(s)
Glicina/análogos & derivados , Hipocampo/enzimología , N-Metilaspartato/farmacología , Fármacos Neuroprotectores/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Resorcinoles/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Inhibidores de Caspasas , Muerte Celular , Hipoxia de la Célula , Células Cultivadas , Glucosa/metabolismo , Glucosa/fisiología , Glicina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Precondicionamiento Isquémico , Isoquinolinas/farmacología , Sistema de Señalización de MAP Quinasas , Neuronas/metabolismo , Fenantrenos/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Ratas , Ratas Wistar , Tiofenos/farmacología
10.
Cell Death Dis ; 13(6): 561, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35732639

RESUMEN

Tubular aggregates (TA) are honeycomb-like arrays of sarcoplasmic-reticulum (SR) tubules affecting aged glycolytic fibers of male individuals and inducing severe sarcomere disorganization and muscular pain. TA develop in skeletal muscle from Tubular Aggregate Myopathy (TAM) patients as well as in other disorders including endocrine syndromes, diabetes, and ageing, being their primary cause unknown. Nowadays, there is no cure for TA. Intriguingly, both hypoxia and calcium dyshomeostasis prompt TA formation, pointing to a possible role for mitochondria in their setting. However, a functional link between mitochondrial dysfunctions and TA remains unknown. Herein, we investigate the alteration in muscle-proteome of TAM patients, the molecular mechanism of TA onset and a potential therapy in a preclinical mouse model of the disease. We show that in vivo chronic inhibition of the mitochondrial ATP synthase in muscle causes TA. Upon long-term restrained oxidative phosphorylation (OXPHOS), oxidative soleus experiments a metabolic and structural switch towards glycolytic fibers, increases mitochondrial fission, and activates mitophagy to recycle damaged mitochondria. TA result from the overresponse of the fission controller DRP1, that upregulates the Store-Operate-Calcium-Entry and increases the mitochondria-SR interaction in a futile attempt to buffer calcium overloads upon prolonged OXPHOS inhibition. Accordingly, hypoxic muscles cultured ex vivo show an increase in mitochondria/SR contact sites and autophagic/mitophagic zones, where TA clusters grow around defective mitochondria. Moreover, hypoxia triggered a stronger TA formation upon ATP synthase inhibition, and this effect was reduced by the DRP1 inhibitor mDIVI. Remarkably, the muscle proteome of TAM patients displays similar alterations in mitochondrial dynamics and in ATP synthase contents. In vivo edaravone treatment in mice with restrained OXPHOS restored a healthy phenotype by prompting mitogenesis and mitochondrial fusion. Altogether, our data provide a functional link between the ATP synthase/DRP1 axis and the setting of TA, and repurpose edaravone as a possible treatment for TA-associated disorders.


Asunto(s)
ATPasas de Translocación de Protón Mitocondriales , Retículo Sarcoplasmático , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Edaravona/metabolismo , Humanos , Hipoxia/metabolismo , Masculino , Ratones , Dinámicas Mitocondriales/fisiología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Músculo Esquelético/metabolismo , Proteoma/metabolismo , Retículo Sarcoplasmático/metabolismo
11.
J Biol Chem ; 285(44): 34106-14, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20724478

RESUMEN

The NAD rescue pathway consists of two enzymatic steps operated by nicotinamide phosphoribosyltransferase (Nampt) and nicotinamide mononucleotide adenylyltransferases. Recently, the potent Nampt inhibitor FK866 has been identified and evaluated in clinical trials against cancer. Yet, how Nampt inhibition affects NAD contents and bioenergetics is in part obscure. It is also unknown whether NAD rescue takes place in mitochondria, and FK866 alters NAD homeostasis within the organelle. Here, we show that FK866-dependent reduction of the NAD contents is paralleled by a concomitant increase of ATP in various cell types, in keeping with ATP utilization for NAD resynthesis. We also show that poly- and mono(ADP-ribose) transferases rather than Sirt-1 are responsible for NAD depletion in HeLa cells exposed to FK866. Mass spectrometry reveals that the drug distributes in the cytosolic and mitochondrial compartment. However, the cytoplasmic but not the mitochondrial NAD pool is reduced upon acute or chronic exposure to the drug. Accordingly, Nampt does not localize within the organelles and their bioenergetics is not affected by the drug. In the mouse, FK866-dependent reduction of NAD contents in various organs is prevented by inhibitors of poly(ADP-ribose) polymerases or the NAD precursor kynurenine. For the first time, our data indicate that mitochondria lack the canonical NAD rescue pathway, broadening current understanding of cellular bioenergetics.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Mitocondrias/metabolismo , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Acrilamidas/farmacología , Adenosina Trifosfato/química , Animales , Fibroblastos/metabolismo , Células HeLa , Humanos , Quinurenina/química , Masculino , Ratones , NAD/química , Piperidinas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo
12.
J Biol Chem ; 285(33): 25308-13, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20538613

RESUMEN

The H(+)-ATP synthase is a reversible engine of mitochondria that synthesizes or hydrolyzes ATP upon changes in cell physiology. ATP synthase dysfunction is involved in the onset and progression of diverse human pathologies. During ischemia, the ATP hydrolytic activity of the enzyme is inhibited by the ATPase inhibitory factor 1 (IF1). The expression of IF1 in human tissues and its participation in the development of human pathology are unknown. Here, we have developed monoclonal antibodies against human IF1 and determined its expression in paired normal and tumor biopsies of human carcinomas. We show that the relative mitochondrial content of IF1 increases significantly in carcinomas, suggesting the participation of IF1 in oncogenesis. The expression of IF1 varies significantly in cancer cell lines. To investigate the functional activity of IF1 in cancer, we have manipulated its cellular content. Overexpression of IF1 or of its pH-insensitive H49K mutant in cells that express low levels of IF1 triggers the up-regulation of aerobic glycolysis and the inhibition of oxidative phosphorylation with concurrent mitochondrial hyperpolarization. Treatment of the cells with the H(+)-ATP synthase inhibitor oligomycin mimicked the effects of IF1 overexpression. Conversely, small interfering RNA-mediated silencing of IF1 in cells that express high levels of IF1 promotes the down-regulation of aerobic glycolysis and the increase in oxidative phosphorylation. Overall, these findings support that the mitochondrial content of IF1 controls the activity of oxidative phosphorylation mediating the shift of cancer cells to an enhanced aerobic glycolysis, thus supporting an oncogenic role for the de-regulated expression of IF1 in cancer.


Asunto(s)
ATPasas de Translocación de Protón Mitocondriales/metabolismo , Neoplasias/metabolismo , Proteínas/metabolismo , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Glucólisis/efectos de los fármacos , Glucólisis/genética , Células HeLa , Células Hep G2 , Humanos , Técnicas In Vitro , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Microscopía Fluorescente , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , Mutación , Oligomicinas/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Proteínas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Ratas , Proteína Inhibidora ATPasa
13.
STAR Protoc ; 2(3): 100735, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34430910

RESUMEN

Depending on metabolic requirements, skeletal muscle mitochondria integrate O2 consumption and ATP production with lipid, glucose, or amino acid metabolism. Free fatty acids (FFAs) are the main source of energy during rest and mild-intensity exercise. We present a detailed protocol for measuring FFA-ß-oxidation coupled with O2 respiration by a Clark-type electrode in isolated mitochondria from mouse soleus oxidative muscle. We optimized the procedure, including buffer composition, protease treatment, and quantifiable parameters (P/O, Phosphate/Oxygen Ratio; OCR, Oxygen Consumption Rate; RCR,Respiration Control Rate; OSR, Oligomycin Sensitive Respiration). For complete details on the use and execution of this protocol, please refer to Sanchez-Gonzalez et al. (2020).


Asunto(s)
Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Consumo de Oxígeno/fisiología , Oxígeno/análisis , Animales , Células Cultivadas , Centrifugación , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Musculares/química , Músculo Esquelético/citología , Oxidación-Reducción , Oxígeno/metabolismo , Espectrofotometría
14.
IUBMB Life ; 62(7): 554-60, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20552634

RESUMEN

Metabolic reprogramming of cancer cells is a phenotypic trait necessary to promote proliferation and survival. Despite past controversies, recent transcriptomic, proteomic, functional and structural studies of mitochondria of the cancer cell indicate that an impaired biogenesis and activity of the organelle is required for the development of some tumors. Cancer aggressiveness can be estimated by its bioenergetic signature, a protein ratio that correlates the expression of b-F1-ATPase of oxidative phosphorylation relative to the glycolytic GAPDH. The bioenergetic signature also provides a gauge that informs of the metabolic activity of tumors and cancer cells as well as of the response to chemotherapy. The convergence of different epithelial tumors on the same bioenergetic signature supports that it provides an important tool and common target for cancer therapy. We stress that targeting the energetic metabolism of tumors affords a valuable strategy to combat the disease.


Asunto(s)
Carcinoma/metabolismo , Metabolismo Energético/fisiología , Mitocondrias/fisiología , Animales , Carcinoma/fisiopatología , Glucólisis , Humanos , Fosforilación Oxidativa , ATPasas de Translocación de Protón/metabolismo , Proteína p53 Supresora de Tumor/fisiología
15.
Nat Commun ; 11(1): 3606, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32681016

RESUMEN

Mitochondrial metabolism has emerged as a promising target against the mechanisms of tumor growth. Herein, we have screened an FDA-approved library to identify drugs that inhibit mitochondrial respiration. The ß1-blocker nebivolol specifically hinders oxidative phosphorylation in cancer cells by concertedly inhibiting Complex I and ATP synthase activities. Complex I inhibition is mediated by interfering the phosphorylation of NDUFS7. Inhibition of the ATP synthase is exerted by the overexpression and binding of the ATPase Inhibitory Factor 1 (IF1) to the enzyme. Remarkably, nebivolol also arrests tumor angiogenesis by arresting endothelial cell proliferation. Altogether, targeting mitochondria and angiogenesis triggers a metabolic and oxidative stress crisis that restricts the growth of colon and breast carcinomas. Nebivolol holds great promise to be repurposed for the treatment of cancer patients.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Inductores de la Angiogénesis/farmacología , Neoplasias de la Mama/fisiopatología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/fisiopatología , Mitocondrias/efectos de los fármacos , Nebivolol/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Femenino , Humanos , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Proteínas/genética , Proteínas/metabolismo , Proteína Inhibidora ATPasa
16.
J Forensic Sci ; 65(5): 1424-1431, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32453457

RESUMEN

Forensic DNA phenotyping refers to an emerging field of forensic sciences aimed at the prediction of externally visible characteristics of unknown sample donors directly from biological materials. The aging process significantly affects most of the above characteristics making the development of a reliable method of age prediction very important. Today, the so-called "epigenetic clocks" represent the most accurate models for age prediction. Since they are technically not achievable in a typical forensic laboratory, forensic DNA technology has triggered efforts toward the simplification of these models. The present study aimed to build an epigenetic clock using a set of methylation markers of five different genes in a sample of the Italian population of different ages covering the whole span of adult life. In a sample of 330 subjects, 42 selected markers were analyzed with a machine learning approach for building a prediction model for age prediction. A ridge linear regression model including eight of the proposed markers was identified as the best performing model across a plethora of candidates. This model was tested on an independent sample of 83 subjects providing a median error of 4.5 years. In the present study, an epigenetic model for age prediction was validated in a sample of the Italian population. However, its applicability to advanced ages still represents the main limitation in forensic caseworks.


Asunto(s)
Envejecimiento/genética , Epigénesis Genética , Genética Forense/métodos , Adulto , Anciano , Anciano de 80 o más Años , Islas de CpG , Metilación de ADN , Elongasas de Ácidos Grasos/genética , Femenino , Marcadores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Homeodominio LIM/genética , Modelos Lineales , Aprendizaje Automático , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética , Reacción en Cadena de la Polimerasa , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Adulto Joven
17.
Nutrients ; 11(12)2019 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-31817660

RESUMEN

Alu hypomethylation promotes genomic instability and is associated with aging and age-related diseases. Dietary factors affect global DNA methylation, leading to changes in genomic stability and gene expression with an impact on longevity and the risk of disease. This preliminary study aims to investigate the relationship between nutritional factors, such as circulating trace elements, lipids and antioxidants, and Alu methylation in elderly subjects and offspring of healthy nonagenarians. Alu DNA methylation was analyzed in sixty RASIG (randomly recruited age-stratified individuals from the general population) and thirty-two GO (GeHA offspring) enrolled in Italy in the framework of the MARK-AGE project. Factor analysis revealed a different clustering between Alu CpG1 and the other CpG sites. RASIG over 65 years showed lower Alu CpG1 methylation than those of GO subjects in the same age class. Moreover, Alu CpG1 methylation was associated with fruit and whole-grain bread consumption, LDL2-Cholesterol and plasma copper. The preserved Alu methylation status in GO, suggests Alu epigenetic changes as a potential marker of aging. Our preliminary investigation shows that Alu methylation may be affected by food rich in fibers and antioxidants, or circulating LDL subfractions and plasma copper.


Asunto(s)
Envejecimiento/genética , Elementos Alu , Metilación de ADN , Nutrientes/sangre , Adulto , Anciano , Envejecimiento/sangre , Antioxidantes/análisis , Islas de CpG , Dieta , Femenino , Voluntarios Sanos , Humanos , Italia , Lipoproteínas/sangre , Lipoproteínas/genética , Longevidad/genética , Masculino , Persona de Mediana Edad , Estado Nutricional , Oligoelementos/sangre
18.
Br J Pharmacol ; 175(2): 272-283, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28320070

RESUMEN

BACKGROUND AND PURPOSE: Dexpramipexole, a drug recently tested in patients with amyotrophic lateral sclerosis (ALS,) is able to bind F1Fo ATP synthase and increase mitochondrial ATP production. Here, we have investigated its effects on experimental ischaemic brain injury. EXPERIMENTAL APPROACH: The effects of dexpramipexole on bioenergetics, Ca2+ fluxes, electrophysiological functions and death were evaluated in primary neural cultures and hippocampal slices exposed to oxygen-glucose deprivation (OGD). Effects on infarct volumes and neurological functions were also evaluated in mice following proximal or distal middle cerebral artery occlusion (MCAo). Distribution of dexpramipexole within the ischaemic brain was evaluated by means of mass spectrometry imaging. KEY RESULTS: Dexpramipexole increased mitochondrial ATP production in cultured neurons or glia and reduces energy failure, prevents intracellular Ca2+ overload and affords cytoprotection when cultures are exposed to OGD. This compound also counteracted ATP depletion, mitochondrial swelling, anoxic depolarization, loss of synaptic activity and neuronal death in hippocampal slices subjected to OGD. Post-ischaemic treatment with dexpramipexole, at doses consistent with those already used in ALS patients, reduced brain infarct size and ameliorated neuroscore in mice subjected to transient or permanent MCAo. Notably, the concentrations of dexpramipexole reached within the ischaemic penumbra equalled those found neuroprotective in vitro. CONCLUSION AND IMPLICATIONS: Dexpramipexole, a compound able to increase mitochondrial F1Fo ATP-synthase activity reduced ischaemic brain injury. These findings, together with the excellent brain penetration and favourable safety profile in humans, make dexpramipexole a drug with realistic translational potential for the treatment of stroke. LINKED ARTICLES: This article is part of a themed section on Inventing New Therapies Without Reinventing the Wheel: The Power of Drug Repurposing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.2/issuetoc.


Asunto(s)
Benzotiazoles/farmacología , Benzotiazoles/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Adenosina Trifosfato/metabolismo , Animales , Benzotiazoles/farmacocinética , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Potenciales Evocados/fisiología , Hipocampo/metabolismo , Hipocampo/fisiología , Hipocampo/ultraestructura , Infarto de la Arteria Cerebral Media , Masculino , Ratones , Mitocondrias/metabolismo , Neuronas/fisiología , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/farmacología , Pramipexol , Cultivo Primario de Células , Ratas , Accidente Cerebrovascular/metabolismo
19.
Biochim Biophys Acta Mol Basis Dis ; 1864(12): 3655-3667, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30261289

RESUMEN

Insulin resistance (IR) and obesity are important risk factors for non-alcoholic fatty liver disease (NAFLD). G protein-coupled receptor kinase 2 (GRK2) is involved in the development of IR and obesity in vivo. However, its possible contribution to NAFLD and/or non-alcoholic steatohepatitis (NASH) independently of its role on IR or fat mass accretion has not been explored. Here, we used wild-type (WT) or GRK2 hemizygous (GRK2±) mice fed a high-fat diet (HFD) or a methionine and choline-deficient diet (MCD) as a model of NASH independent of adiposity and IR. GRK2± mice were protected from HFD-induced NAFLD. Moreover, MCD feeding caused an increased in triglyceride content and liver-to-body weight ratio in WT mice, features that were attenuated in GRK2± mice. According to their NAFLD activity score, MCD-fed GRK2± mice were diagnosed with simple steatosis and not overt NASH. They also showed reduced expression of lipogenic and lipid-uptake markers and less signs of inflammation in the liver. GRK2± mice preserved hepatic protective mechanisms as enhanced autophagy and mitochondrial fusion and biogenesis, together with reduced endoplasmic reticulum stress. GRK2 protein was increased in MCD-fed WT but not in GRK2± mice, and enhanced GRK2 expression potentiated palmitic acid-triggered lipid accumulation in human hepatocytes directly relating GRK2 levels to steatosis. GRK2 protein and mRNA levels were increased in human liver biopsies from simple steatosis or NASH patients in two different human cohorts. Our results describe a functional relationship between GRK2 levels and hepatic lipid accumulation and implicate GRK2 in the establishment and/or development of NASH.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Línea Celular , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Quinasa 2 del Receptor Acoplado a Proteína-G/análisis , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Humanos , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , ARN Mensajero/genética , Regulación hacia Arriba
20.
Cornea ; 26(1): 73-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17198017

RESUMEN

PURPOSE: Polysaccharides are frequently used as viscoelastic agents to improve pharmacokinetics of ophthalmic preparations. Recently, polysaccharides from yeast cell walls such as beta-glucans have emerged as bioactive molecules endowed with immunomodulatory and cytoprotective properties. In this study, we investigated the effects of carboxymethyl beta-glucan (CMG), a water-soluble derivative of yeast beta-glucan, on cultured rabbit corneal epithelial cells. METHODS: We developed a fluorescein-labeled CMG to visualize its binding to corneal cells by means of digital microscopy and image deconvolution. The effects of CMG on adhesion and survival of corneal epithelial cells exposed to noxious stimuli were also studied. RESULTS: CMG binds defined regions scattered throughout the body of corneal cells, suggesting binding specificity. Tridimensional reconstruction of fluorescence shows that binding is localized mainly at the plasma and nuclear membranes. Interestingly, CMG binding is highly represented at the level of focal adhesion of cells spreading onto laminin. Accordingly, CMG promotes adhesion of corneal epithelial cells to laminin without affecting their proliferation rate. CMG also protects cells from oxidative stress-dependent cell death, being ineffective in preventing ultraviolet B cytotoxicity. CONCLUSIONS: Data show that CMG dynamically binds to corneal epithelial cells, promoting cell adhesion and resistance to oxidative stress.


Asunto(s)
Adhesión Celular/fisiología , Epitelio Corneal/metabolismo , Laminina/metabolismo , Estrés Oxidativo , beta-Glucanos/metabolismo , Animales , Membrana Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Epitelio Corneal/efectos de los fármacos , Epitelio Corneal/efectos de la radiación , Colorantes Fluorescentes , Peróxido de Hidrógeno/toxicidad , Conejos , Saccharomyces cerevisiae/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA