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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(4)2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38396915

RESUMEN

Mitochondrial ATP synthase (Complex V) catalyzes the last step of oxidative phosphorylation and provides most of the energy (ATP) required by human cells. The mitochondrial genes MT-ATP6 and MT-ATP8 encode two subunits of the multi-subunit Complex V. Since the discovery of the first MT-ATP6 variant in the year 1990 as the cause of Neuropathy, Ataxia, and Retinitis Pigmentosa (NARP) syndrome, a large and continuously increasing number of inborn variants in the MT-ATP6 and MT-ATP8 genes have been identified as pathogenic. Variants in these genes correlate with various clinical phenotypes, which include several neurodegenerative and multisystemic disorders. In the present review, we report the pathogenic variants in mitochondrial ATP synthase genes and highlight the molecular mechanisms underlying ATP synthase deficiency that promote biochemical dysfunctions. We discuss the possible structural changes induced by the most common variants found in patients by considering the recent cryo-electron microscopy structure of human ATP synthase. Finally, we provide the state-of-the-art of all therapeutic proposals reported in the literature, including drug interventions targeting mitochondrial dysfunctions, allotopic gene expression- and nuclease-based strategies, and discuss their potential translation into clinical trials.


Asunto(s)
Enfermedades Mitocondriales , ATPasas de Translocación de Protón Mitocondriales , Humanos , Adenosina Trifosfato , Microscopía por Crioelectrón , ADN Mitocondrial/genética , Genes Mitocondriales , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/terapia , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Mutación
2.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731874

RESUMEN

The mitochondrial protein IF1 is upregulated in many tumors and acts as a pro-oncogenic protein through its interaction with the ATP synthase and the inhibition of apoptosis. We have recently characterized the molecular nature of the IF1-Oligomycin Sensitivity Conferring Protein (OSCP) subunit interaction; however, it remains to be determined whether this interaction could be targeted for novel anti-cancer therapeutic intervention. We generated mitochondria-targeting peptides to displace IF1 from the OSCP interaction. The use of one selective peptide led to displacement of the inhibitor IF1 from ATP synthase, as shown by immunoprecipitation. NMR spectroscopy analysis, aimed at clarifying whether these peptides were able to directly bind to the OSCP protein, identified a second peptide which showed affinity for the N-terminal region of this subunit overlapping the IF1 binding region. In situ treatment with the membrane-permeable derivatives of these peptides in HeLa cells, that are silenced for the IF1 inhibitor protein, showed significant inhibition in mitochondrial permeability transition and no effects on mitochondrial respiration. These peptides mimic the effects of the IF1 inhibitor protein in cancer HeLa cells and confirm that the IF1-OSCP interaction inhibits apoptosis. A third peptide was identified which counteracts the anti-apoptotic role of IF1, showing that OSCP is a promising target for anti-cancer therapies.


Asunto(s)
Mitocondrias , ATPasas de Translocación de Protón Mitocondriales , Péptidos , Humanos , Apoptosis/efectos de los fármacos , Proteína Inhibidora ATPasa/efectos de los fármacos , Proteína Inhibidora ATPasa/metabolismo , Células HeLa , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , Péptidos/farmacología , Péptidos/química , Péptidos/metabolismo , Unión Proteica
3.
Int J Mol Sci ; 25(6)2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38542395

RESUMEN

Mitochondria are involved in multiple aspects of neurodevelopmental processes and play a major role in the pathogenetic mechanisms leading to neuro-degenerative diseases. Fragile-X-related disorders (FXDs) are genetic conditions that occur due to the dynamic expansion of CGG repeats of the FMR1 gene encoding for the RNA-binding protein FMRP, particularly expressed in the brain. This gene expansion can lead to premutation (PM, 56-200 CGGs), full mutation (FM, >200 CGGs), or unmethylated FM (UFM), resulting in neurodegeneration, neurodevelopmental disorders, or no apparent intellectual disability, respectively. To investigate the mitochondrial mechanisms that are involved in the FXD patients, we analyzed mitochondrial morphology and bioenergetics in fibroblasts derived from patients. Donut-shaped mitochondrial morphology and excessive synthesis of critical mitochondrial proteins were detected in FM, PM, and UFM cells. Analysis of mitochondrial oxidative phosphorylation in situ reveals lower respiration in PM fibroblasts. Importantly, mitochondrial permeability transition-dependent apoptosis is sensitized to reactive oxygen species in FM, PM, and UFM models. This study elucidated the mitochondrial mechanisms that are involved in the FXD phenotypes, and indicated altered mitochondrial function and morphology. Importantly, a sensitization to permeability transition and apoptosis was revealed in FXD cells. Overall, our data suggest that mitochondria are novel drug targets to relieve the FXD symptoms.


Asunto(s)
Síndrome del Cromosoma X Frágil , Discapacidad Intelectual , Enfermedades Mitocondriales , Humanos , Síndrome del Cromosoma X Frágil/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Discapacidad Intelectual/genética , Muerte Celular/genética , Enfermedades Mitocondriales/genética , Mutación , Expansión de Repetición de Trinucleótido
4.
Int J Mol Sci ; 24(19)2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37834071

RESUMEN

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.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Mitocondrias/metabolismo , Hipoxia/metabolismo , Osteosarcoma/metabolismo , Neoplasias Óseas/metabolismo , Isquemia/metabolismo , Proliferación Celular , Adenosina Trifosfato/metabolismo
5.
Int J Mol Sci ; 24(15)2023 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-37569836

RESUMEN

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.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Productos de Tabaco , Embarazo , Ratas , Animales , Masculino , Femenino , Humanos , Humo , Ratas Sprague-Dawley , Productos de Tabaco/efectos adversos , Hígado
6.
Biochem Soc Trans ; 49(2): 815-827, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33929490

RESUMEN

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.


Asunto(s)
Metabolismo Energético/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Neoplasias/genética , Proteínas/genética , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Modelos Moleculares , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Proteínas/química , Proteínas/metabolismo , Homología de Secuencia de Aminoácido , Proteína Inhibidora ATPasa
7.
Circ Res ; 122(10): e75-e83, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29483093

RESUMEN

RATIONALE: Disrupted proteostasis is one major pathological trait that heart failure (HF) shares with other organ proteinopathies, such as Alzheimer and Parkinson diseases. Yet, differently from the latter, whether and how cardiac preamyloid oligomers (PAOs) develop in acquired forms of HF is unclear. OBJECTIVE: We previously reported a rise in monophosphorylated, aggregate-prone desmin in canine and human HF. We now tested whether monophosphorylated desmin acts as the seed nucleating PAOs formation and determined whether positron emission tomography is able to detect myocardial PAOs in nongenetic HF. METHODS AND RESULTS: Here, we first show that toxic cardiac PAOs accumulate in the myocardium of mice subjected to transverse aortic constriction and that PAOs comigrate with the cytoskeletal protein desmin in this well-established model of acquired HF. We confirm this evidence in cardiac extracts from human ischemic and nonischemic HF. We also demonstrate that Ser31 phosphorylated desmin aggregates extensively in cultured cardiomyocytes. Lastly, we were able to detect the in vivo accumulation of cardiac PAOs using positron emission tomography for the first time in acquired HF. CONCLUSIONS: Ser31 phosphorylated desmin is a likely candidate seed for the nucleation process leading to cardiac PAOs deposition. Desmin post-translational processing and misfolding constitute a new, attractive avenue for the diagnosis and treatment of the cardiac accumulation of toxic PAOs that can now be measured by positron emission tomography in acquired HF.


Asunto(s)
Amiloide/metabolismo , Desmina/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional , Amiloide/análisis , Amiloide/efectos de los fármacos , Animales , Catequina/análogos & derivados , Catequina/farmacología , Células Cultivadas , Desmina/genética , Femenino , Vectores Genéticos , Insuficiencia Cardíaca/etiología , Humanos , Masculino , Espectrometría de Masas/métodos , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Isquemia Miocárdica/complicaciones , Fosforilación , Polimorfismo de Nucleótido Simple , Tomografía de Emisión de Positrones/métodos , Presión , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína , Ratas , Proteínas Recombinantes/metabolismo , alfa-Cristalinas/deficiencia , beta-Cristalinas/deficiencia
8.
Cell Physiol Biochem ; 47(3): 1230-1243, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29913456

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Señalización del Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Metabolismo Energético/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Polifenoles/farmacología , Té/química , Administración Oral , Animales , Masculino , Miocitos Cardíacos/citología , Fosforilación/efectos de los fármacos , Polifenoles/química , Ratas , Ratas Wistar , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
9.
J Biol Chem ; 290(10): 6338-48, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25605724

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/metabolismo , ATPasas de Translocación de Protón Mitocondriales/genética , Osteosarcoma/metabolismo , Proteínas/genética , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Metabolismo Energético/genética , Silenciador del Gen , Humanos , Potencial de la Membrana Mitocondrial/genética , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , Osteosarcoma/genética , Osteosarcoma/patología , Fosforilación Oxidativa , Proteínas/antagonistas & inhibidores , ARN Interferente Pequeño , Proteína Inhibidora ATPasa
10.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166879, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37689158

RESUMEN

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.


Asunto(s)
NAD , Neoplasias , Humanos , Adenosina Trifosfato/metabolismo , Mitocondrias/metabolismo , NAD/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación , Proteína Inhibidora ATPasa
11.
Cell Death Dis ; 14(1): 54, 2023 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-36690622

RESUMEN

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.


Asunto(s)
ATPasas de Translocación de Protón Mitocondriales , Neoplasias , Humanos , Células HeLa , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Mitocondrias/metabolismo , Óxido Nítrico Sintasa/metabolismo , Apoptosis , Adenosina Trifosfato/metabolismo , Neoplasias/patología
12.
Biochim Biophys Acta ; 1807(6): 534-42, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20849810

RESUMEN

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.


Asunto(s)
Neoplasias/metabolismo , Fosforilación Oxidativa , Animales , Humanos , Redes y Vías Metabólicas/fisiología , Mitocondrias/metabolismo , Mitocondrias/patología , Mitocondrias/fisiología , Modelos Biológicos , Neoplasias/patología , Neoplasias/fisiopatología , Consumo de Oxígeno/fisiología
13.
ScientificWorldJournal ; 2012: 573410, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22593698

RESUMEN

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.


Asunto(s)
Oxigenoterapia Hiperbárica/métodos , Hipotermia Inducida/métodos , Hígado , Preservación de Órganos/métodos , Animales , Disacáridos/farmacología , Electrólitos/farmacología , Regulación Enzimológica de la Expresión Génica , Glutamatos/farmacología , Glutatión/metabolismo , Glutatión/farmacología , Disulfuro de Glutatión/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Histidina/farmacología , Hipotermia Inducida/instrumentación , Glucógeno Hepático/metabolismo , Manitol/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Soluciones Preservantes de Órganos/farmacología , Estrés Oxidativo/efectos de los fármacos , Perfusión , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Compuestos de Sulfhidrilo/metabolismo
14.
Front Physiol ; 13: 917203, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35991181

RESUMEN

The endogenous inhibitor of ATP synthase is a protein of about 10 kDa, known as IF1 which binds to the catalytic domain of the enzyme during ATP hydrolysis. The main role of IF1 consists of limiting ATP dissipation under condition of severe oxygen deprivation or in the presence of dysfunctions of mitochondrial respiratory complexes, causing a collapse in mitochondrial membrane potential and therefore ATP hydrolysis. New roles of IF1 are emerging in the fields of cancer and neurodegeneration. Its high expression levels in tumor tissues have been associated with different roles favouring tumor formation, progression and evasion. Since discordant mechanisms of action have been proposed for IF1 in tumors, it is of the utmost importance to clarify them in the prospective of defining novel approaches for cancer therapy. Other IF1 functions, including its involvement in mitophagy, may be protective for neurodegenerative and aging-related diseases. In the present review we aim to clarify and discuss the emerging mechanisms in which IF1 is involved, providing a critical view of the discordant findings in the literature.

15.
J Comp Physiol B ; 192(2): 349-360, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35001173

RESUMEN

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.


Asunto(s)
Hipotermia , Letargo , Animales , Respiración de la Célula , Ratones , Mitocondrias/metabolismo , Ratas , Receptor de Adenosina A1/fisiología , Letargo/fisiología
16.
Front Aging Neurosci ; 14: 1058665, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36698863

RESUMEN

Introduction: Growth Differentiation Factor 15 (GDF15) is a mitochondrial-stress-responsive molecule whose expression strongly increases with aging and age-related diseases. However, its role in neurodegenerative diseases, including Alzheimer's disease (AD), is still debated. Methods: We have characterized the expression of GDF15 in brain samples from AD patients and non-demented subjects (controls) of different ages. Results: Although no difference in CSF levels of GDF15 was found between AD patients and controls, GDF15 was expressed in different brain areas and seems to be predominantly localized in neurons. The ratio between its mature and precursor form was higher in the frontal cortex of AD patients compared to age-matched controls (p < 0.05). Moreover, this ratio was even higher for centenarians (p < 0.01), indicating that aging also affects GDF15 expression and maturation. A lower expression of OXPHOS complexes I, III, and V in AD patients compared to controls was also noticed, and a positive correlation between GDF15 and IL-6 mRNA levels was observed. Finally, when GDF15 was silenced in vitro in dermal fibroblasts, a decrease in OXPHOS complexes transcript levels and an increase in IL-6 levels were observed. Discussion: Although GDF15 seems not to be a reliable CSF marker for AD, it is highly expressed in aging and AD brains, likely as a part of stress response aimed at counteracting mitochondrial dysfunction and neuroinflammation.

17.
Biochim Biophys Acta ; 1797(6-7): 1171-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20153717

RESUMEN

It is now clear that mitochondrial defects are associated with a large variety of clinical phenotypes. This is the result of the mitochondria's central role in energy production, reactive oxygen species homeostasis, and cell death. These processes are interdependent and may occur under various stressing conditions, among which low oxygen levels (hypoxia) are certainly prominent. Cells exposed to hypoxia respond acutely with endogenous metabolites and proteins promptly regulating metabolic pathways, but if low oxygen levels are prolonged, cells activate adapting mechanisms, the master switch being the hypoxia-inducible factor 1 (HIF-1). Activation of this factor is strictly bound to the mitochondrial function, which in turn is related with the oxygen level. Therefore in hypoxia, mitochondria act as [O2] sensors, convey signals to HIF-1 directly or indirectly, and contribute to the cell redox potential, ion homeostasis, and energy production. Although over the last two decades cellular responses to low oxygen tension have been studied extensively, mechanisms underlying these functions are still indefinite. Here we review current knowledge of the mitochondrial role in hypoxia, focusing mainly on their role in cellular energy and reactive oxygen species homeostasis in relation with HIF-1 stabilization. In addition, we address the involvement of HIF-1 and the inhibitor protein of F1F0 ATPase in the hypoxia-induced mitochondrial autophagy.


Asunto(s)
Hipoxia de la Célula/fisiología , Mitocondrias/metabolismo , Oxígeno/metabolismo , Animales , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Metabolismo Energético , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Modelos Biológicos , Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Inhibidora ATPasa
18.
Biochim Biophys Acta ; 1797(2): 314-23, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19931505

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Metabolismo Energético , Fibroblastos/metabolismo , Genes ras , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular , Respiración de la Célula/fisiología , Células Cultivadas , Perfilación de la Expresión Génica , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidación-Reducción , Fosforilación Oxidativa , Oxígeno/metabolismo
19.
Biochim Biophys Acta ; 1797(6-7): 633-40, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20116362

RESUMEN

Recent investigations by native gel electrophoresis showed the existence of supramolecular associations of the respiratory complexes, confirmed by electron microscopy analysis and single particle image processing. Flux control analysis demonstrated that Complex I and Complex III in mammalian mitochondria kinetically behave as a single unit with control coefficients approaching unity for each component, suggesting the existence of substrate channeling within the super-complex. The formation of this supramolecular unit largely depends on the lipid content and composition of the inner mitochondrial membrane. The function of the super-complexes appears not to be restricted to kinetic advantages in electron transfer: we discuss evidence on their role in the stability and assembly of the individual complexes, particularly Complex I, and in preventing excess oxygen radical formation. There is increasing evidence that disruption of the super-complex organization leads to functional derangements responsible for pathological changes, as we have found in K-ras-transformed fibroblasts.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/fisiología , Mitocondrias/patología , Mitocondrias/fisiología , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/fisiología , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/fisiología , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/fisiología , Humanos , Cinética , Enfermedades Mitocondriales/patología , Enfermedades Mitocondriales/fisiopatología , Modelos Biológicos , Multimerización de Proteína , Estabilidad Proteica , Especies Reactivas de Oxígeno/metabolismo
20.
Life (Basel) ; 11(4)2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33917760

RESUMEN

Human diseases range from gene-associated to gene-non-associated disorders, including age-related diseases, neurodegenerative, neuromuscular, cardiovascular, diabetic diseases, neurocognitive disorders and cancer. Mitochondria participate to the cascades of pathogenic events leading to the onset and progression of these diseases independently of their association to mutations of genes encoding mitochondrial protein. Under physiological conditions, the mitochondrial ATP synthase provides the most energy of the cell via the oxidative phosphorylation. Alterations of oxidative phosphorylation mainly affect the tissues characterized by a high-energy metabolism, such as nervous, cardiac and skeletal muscle tissues. In this review, we focus on human diseases caused by altered expressions of ATP synthase genes of both mitochondrial and nuclear origin. Moreover, we describe the contribution of ATP synthase to the pathophysiological mechanisms of other human diseases such as cardiovascular, neurodegenerative diseases or neurocognitive disorders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA