Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Rev Physiol Biochem Pharmacol ; 185: 153-193, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-32789789

RESUMEN

Endoplasmic reticulum (ER)-mitochondria regions are specialized subdomains called also mitochondria-associated membranes (MAMs). MAMs allow regulation of lipid synthesis and represent hubs for ion and metabolite signaling. As these two organelles can module both the amplitude and the spatiotemporal patterns of calcium (Ca2+) signals, this particular interaction controls several Ca2+-dependent pathways well known for their contribution to tumorigenesis, such as metabolism, survival, sensitivity to cell death, and metastasis. Mitochondria-mediated apoptosis arises from mitochondrial Ca2+ overload, permeabilization of the mitochondrial outer membrane, and the release of mitochondrial apoptotic factors into the cytosol. Decreases in Ca2+ signaling at the ER-mitochondria interface are being studied in depth as failure of apoptotic-dependent cell death is one of the predominant characteristics of cancer cells. However, some recent papers that linked MAMs Ca2+ crosstalk-related upregulation to tumor onset and progression have aroused the interest of the scientific community.In this review, we will describe how different MAMs-localized proteins modulate the effectiveness of Ca2+-dependent apoptotic stimuli by causing both increases and decreases in the ER-mitochondria interplay and, specifically, by modulating Ca2+ signaling.


Asunto(s)
Señalización del Calcio , Neoplasias , Humanos , Señalización del Calcio/fisiología , Mitocondrias , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Muerte Celular , Proteínas de la Membrana/metabolismo , Calcio/metabolismo , Neoplasias/metabolismo
2.
FASEB J ; 38(3): e23466, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38318780

RESUMEN

Despite decades of research, the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) is still not completely understood. Based on the evidence from preclinical models, one of the factors proposed as a main driver of disease development is oxidative stress. This study aimed to search for the resemblance between the profiles of oxidative stress and antioxidant defense in the animal model of MASLD and the group of MASLD patients. C57BL/6J mice were fed with the Western diet for up to 24 weeks and served as the animal model of MASLD. The antioxidant profile of mice hepatic tissue was determined by liquid chromatography-MS3 spectrometry (LC-MS/MS). The human cohort consisted of 20 patients, who underwent bariatric surgery, and 6 controls. Based on histological analysis, 4 bariatric patients did not have liver steatosis and as such were also classified as controls. Total antioxidant activity was measured in sera and liver biopsy samples. The hepatic levels of antioxidant enzymes and oxidative damage were determined by Western Blot. The levels of antioxidant enzymes were significantly altered in the hepatic tissue of mice with MASLD. In contrast, there were no significant changes in the antioxidant profile of hepatic tissue of MASLD patients, except for the decreased level of carbonylated proteins. Decreased protein carbonylation together with significant correlations between the thioredoxin system and parameters describing metabolic health suggest alterations in the thiol-redox signaling. Altogether, these data show that even though the phenotype of mice closely resembles human MASLD, the animal-to-human translation of cellular and molecular processes such as oxidative stress may be more challenging.


Asunto(s)
Hígado Graso , Enfermedades Metabólicas , Humanos , Animales , Ratones , Ratones Endogámicos C57BL , Antioxidantes , Cromatografía Liquida , Espectrometría de Masas en Tándem , Estrés Oxidativo , Modelos Animales
3.
Eur J Clin Invest ; 54(7): e14217, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38644687

RESUMEN

OBJECTIVES AND SCOPE: Primary mitochondrial diseases (PMDs) are rare genetic disorders resulting from mutations in genes crucial for effective oxidative phosphorylation (OXPHOS) that can affect mitochondrial function. In this review, we examine the bioenergetic alterations and oxidative stress observed in cellular models of primary mitochondrial diseases (PMDs), shedding light on the intricate complexity between mitochondrial dysfunction and cellular pathology. We explore the diverse cellular models utilized to study PMDs, including patient-derived fibroblasts, induced pluripotent stem cells (iPSCs) and cybrids. Moreover, we also emphasize the connection between oxidative stress and neuroinflammation. INSIGHTS: The central nervous system (CNS) is particularly vulnerable to mitochondrial dysfunction due to its dependence on aerobic metabolism and the correct functioning of OXPHOS. Similar to other neurodegenerative diseases affecting the CNS, individuals with PMDs exhibit several neuroinflammatory hallmarks alongside neurodegeneration, a pattern also extensively observed in mouse models of mitochondrial diseases. Based on histopathological analysis of postmortem human brain tissue and findings in mouse models of PMDs, we posit that neuroinflammation is not merely a consequence of neurodegeneration but a potential pathogenic mechanism for disease progression that deserves further investigation. This recognition may pave the way for novel therapeutic strategies for this group of devastating diseases that currently lack effective treatments. SUMMARY: In summary, this review provides a comprehensive overview of bioenergetic alterations and redox imbalance in cellular models of PMDs while underscoring the significance of neuroinflammation as a potential driver in disease progression.


Asunto(s)
Metabolismo Energético , Enfermedades Mitocondriales , Enfermedades Neuroinflamatorias , Estrés Oxidativo , Humanos , Estrés Oxidativo/fisiología , Enfermedades Mitocondriales/fisiopatología , Enfermedades Mitocondriales/metabolismo , Enfermedades Neuroinflamatorias/fisiopatología , Enfermedades Neuroinflamatorias/metabolismo , Animales , Metabolismo Energético/fisiología , Fosforilación Oxidativa , Ratones , Mitocondrias/metabolismo , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/genética , Enfermedad de Leigh/fisiopatología , Síndrome MELAS/metabolismo , Síndrome MELAS/fisiopatología , Síndrome MELAS/genética , Modelos Animales de Enfermedad
4.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34099564

RESUMEN

Multiple sclerosis (MS) is a neuroinflammatory and neurodegenerative disease characterized by myelin damage followed by axonal and ultimately neuronal loss. The etiology and physiopathology of MS are still elusive, and no fully effective therapy is yet available. We investigated the role in MS of autophagy (physiologically, a controlled intracellular pathway regulating the degradation of cellular components) and of mitophagy (a specific form of autophagy that removes dysfunctional mitochondria). We found that the levels of autophagy and mitophagy markers are significantly increased in the biofluids of MS patients during the active phase of the disease, indicating activation of these processes. In keeping with this idea, in vitro and in vivo MS models (induced by proinflammatory cytokines, lysolecithin, and cuprizone) are associated with strongly impaired mitochondrial activity, inducing a lactic acid metabolism and prompting an increase in the autophagic flux and in mitophagy. Multiple structurally and mechanistically unrelated inhibitors of autophagy improved myelin production and normalized axonal myelination, and two such inhibitors, the widely used antipsychotic drugs haloperidol and clozapine, also significantly improved cuprizone-induced motor impairment. These data suggest that autophagy has a causal role in MS; its inhibition strongly attenuates behavioral signs in an experimental model of the disease. Therefore, haloperidol and clozapine may represent additional therapeutic tools against MS.


Asunto(s)
Antipsicóticos/uso terapéutico , Autofagia , Mitofagia , Esclerosis Múltiple/tratamiento farmacológico , Animales , Antipsicóticos/farmacología , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/sangre , Proteínas Relacionadas con la Autofagia/líquido cefalorraquídeo , Axones/efectos de los fármacos , Axones/metabolismo , Biomarcadores/metabolismo , Clozapina/farmacología , Citocinas/metabolismo , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Glucosa/metabolismo , Haloperidol/farmacología , Inflamación/patología , Interleucina-1beta/metabolismo , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos , Modelos Biológicos , Actividad Motora/efectos de los fármacos , Esclerosis Múltiple/sangre , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/fisiopatología , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/metabolismo , Estrés Fisiológico/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
5.
Int J Mol Sci ; 25(14)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39063068

RESUMEN

Oxidative stress has been known about in biological sciences for several decades; however, the understanding of this concept has evolved greatly since its foundation. Over the past years, reactive oxygen species, once viewed as solely deleterious, have become recognized as intrinsic components of life. In contrast, antioxidants, initially believed to be cure-all remedies, have failed to prove their efficacy in clinical trials. Fortunately, research on the health-promoting properties of antioxidants has been ongoing. Subsequent years showed that the former assumption that all antioxidants acted similarly was greatly oversimplified. Redox-active compounds differ in their chemical structures, electrochemical properties, mechanisms of action, and bioavailability; therefore, their efficacy in protecting against oxidative stress also varies. In this review, we discuss the changing perception of oxidative stress and its sources, emphasizing everyday-life exposures, particularly those of dietary origin. Finally, we posit that a better understanding of the physicochemical properties and biological outcomes of antioxidants is crucial to fully utilize their beneficial impact on health.


Asunto(s)
Antioxidantes , Homeostasis , Oxidación-Reducción , Estrés Oxidativo , Especies Reactivas de Oxígeno , Antioxidantes/metabolismo , Antioxidantes/química , Humanos , Especies Reactivas de Oxígeno/metabolismo , Animales , Oxidantes/metabolismo , Oxidantes/química
6.
Brain ; 145(1): 45-63, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-34849584

RESUMEN

Mitochondria are small cellular constituents that generate cellular energy (ATP) by oxidative phosphorylation (OXPHOS). Dysfunction of these organelles is linked to a heterogeneous group of multisystemic disorders, including diabetes, cancer, ageing-related pathologies and rare mitochondrial diseases. With respect to the latter, mutations in subunit-encoding genes and assembly factors of the first OXPHOS complex (complex I) induce isolated complex I deficiency and Leigh syndrome. This syndrome is an early-onset, often fatal, encephalopathy with a variable clinical presentation and poor prognosis due to the lack of effective intervention strategies. Mutations in the nuclear DNA-encoded NDUFS4 gene, encoding the NADH:ubiquinone oxidoreductase subunit S4 (NDUFS4) of complex I, induce 'mitochondrial complex I deficiency, nuclear type 1' (MC1DN1) and Leigh syndrome in paediatric patients. A variety of (tissue-specific) Ndufs4 knockout mouse models were developed to study the Leigh syndrome pathomechanism and intervention testing. Here, we review and discuss the role of complex I and NDUFS4 mutations in human mitochondrial disease, and review how the analysis of Ndufs4 knockout mouse models has generated new insights into the MC1ND1/Leigh syndrome pathomechanism and its therapeutic targeting.


Asunto(s)
Complejo I de Transporte de Electrón , Enfermedad de Leigh , Enfermedades Mitocondriales , Animales , Complejo I de Transporte de Electrón/genética , Humanos , Enfermedad de Leigh/genética , Ratones , Ratones Noqueados , Enfermedades Mitocondriales/genética , Fosforilación Oxidativa
7.
Semin Cell Dev Biol ; 98: 167-180, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31108186

RESUMEN

Organelles were originally considered to be individual cellular compartments with a defined organization and function. However, recent studies revealed that organelles deeply communicate within each other via Ca2+ exchange. This communication, mediated by specialized membrane regions in close apposition between two organelles, regulate cellular functions, including metabolism and cell fate decisions. Advances in microscopy techniques, molecular biology and biochemistry have increased our understanding of these interorganelle platforms. Research findings suggest that interorganellar Ca2+ signaling, which is altered in cancer, influences tumorigenesis and tumor progression by controlling cell death programs and metabolism. Here, we summarize the available data on the existence and composition of interorganelle platforms connecting the endoplasmic reticulum with mitochondria, the plasma membrane, or endolysosomes. Finally, we provide a timely overview of the potential function of interorganellar Ca2+ signaling in maintaining cellular homeostasis.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Orgánulos/metabolismo , Animales , Homeostasis , Humanos
8.
Eur J Clin Invest ; 52(3): e13622, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34050922

RESUMEN

According to the 'multiple-hit' hypothesis, several factors can act simultaneously in nonalcoholic fatty liver disease (NAFLD) progression. Increased nitro-oxidative (nitroso-oxidative) stress may be considered one of the main contributors involved in the development and risk of NAFLD progression to nonalcoholic steatohepatitis (NASH) characterized by inflammation and fibrosis. Moreover, it has been repeatedly postulated that mitochondrial abnormalities are closely related to the development and progression of liver steatosis and NAFLD pathogenesis. However, it is difficult to determine with certainty whether mitochondrial dysfunction or oxidative stress are primary events or a simple consequence of NAFLD development. On the one hand, increasing lipid accumulation in hepatocytes could cause a wide range of effects from mild to severe mitochondrial damage with a negative impact on cell fate. This can start the cascade of events, including an increase of cellular reactive nitrogen species (RNS) and reactive oxygen species (ROS) production that promotes disease progression from simple steatosis to more severe NAFLD stages. On the other hand, progressing mitochondrial bioenergetic catastrophe and oxidative stress manifestation could be considered accompanying events in the vast spectrum of abnormalities observed during the transition from NAFL to NASH and cirrhosis. This review updates our current understanding of NAFLD pathogenesis and clarifies whether mitochondrial dysfunction and ROS/RNS are culprits or bystanders of NAFLD progression.


Asunto(s)
Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Estrés Oxidativo , Humanos
9.
Pharmacol Res ; 177: 106119, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35131483

RESUMEN

Intracellular calcium signaling is a universal language source shared by the most part of biological entities inside cells that, all together, give rise to physiological and functional anatomical units, the organ. Although preferentially recognized as signaling between cell life and death processes, in the heart it assumes additional relevance considered the importance of calcium cycling coupled to ATP consumption in excitation-contraction coupling. The concerted action of a plethora of exchangers, channels and pumps inward and outward calcium fluxes where needed, to convert energy and electric impulses in muscle contraction. All this without realizing it, thousands of times, every day. An improper function of those proteins (i.e., variation in expression, mutations onset, dysregulated channeling, differential protein-protein interactions) being part of this signaling network triggers a short circuit with severe acute and chronic pathological consequences reported as arrhythmias, cardiac remodeling, heart failure, reperfusion injury and cardiomyopathies. By acting with chemical, peptide-based and pharmacological modulators of these players, a correction of calcium homeostasis can be achieved accompanied by an amelioration of clinical symptoms. This review will focus on all those defects in calcium homeostasis which occur in the most common cardiac diseases, including myocardial infarction, arrhythmia, hypertrophy, heart failure and cardiomyopathies. This part will be introduced by the state of the art on the proteins involved in calcium homeostasis in cardiomyocytes and followed by the therapeutic treatments that to date, are able to target them and to revert the pathological phenotype.


Asunto(s)
Cardiomiopatías , Insuficiencia Cardíaca , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Cardiomiopatías/metabolismo , Homeostasis , Humanos , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Retículo Sarcoplasmático
10.
Cell Commun Signal ; 19(1): 116, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34801048

RESUMEN

BACKGROUND: Wolfram syndrome (WFS) is a rare autosomal recessive syndrome in which diabetes mellitus and neurodegenerative disorders occur as a result of Wolframin deficiency and increased ER stress. In addition, WFS1 deficiency leads to calcium homeostasis disturbances and can change mitochondrial dynamics. The aim of this study was to evaluate protein levels and changes in gene transcription on human WFS cell model under experimental ER stress. METHODS: We performed transcriptomic and proteomic analysis on WFS human cell model-skin fibroblasts reprogrammed into induced pluripotent stem (iPS) cells and then into neural stem cells (NSC) with subsequent ER stress induction using tunicamycin (TM). Results were cross-referenced with publicly available RNA sequencing data in hippocampi and hypothalami of mice with WFS1 deficiency. RESULTS: Proteomic analysis identified specific signal pathways that differ in NSC WFS cells from healthy ones. Next, detailed analysis of the proteins involved in the mitochondrial function showed the down-regulation of subunits of the respiratory chain complexes in NSC WFS cells, as well as the up-regulation of proteins involved in Krebs cycle and glycolysis when compared to the control cells. Based on pathway enrichment analysis we concluded that in samples from mice hippocampi the mitochondrial protein import machinery and OXPHOS were significantly down-regulated. CONCLUSIONS: Our results show the functional and morphological secondary mitochondrial damage in patients with WFS. Video Abstract.


Asunto(s)
Síndrome de Wolfram
11.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202179

RESUMEN

The progression of non-alcoholic fatty liver (NAFL) into non-alcoholic steatohepatitis implicates multiple mechanisms, chief of which is mitochondrial dysfunction. However, the sequence of events underlying mitochondrial failure are still poorly clarified. In this work, male C57BL/6J mice were fed with a high-fat plus high-sucrose diet for 16, 20, 22, and 24 weeks to induce NAFL. Up to the 20th week, an early mitochondrial remodeling with increased OXPHOS subunits levels and higher mitochondrial respiration occurred. Interestingly, a progressive loss of mitochondrial respiration along "Western diet" feeding was identified, accompanied by higher susceptibility to mitochondrial permeability transition pore opening. Importantly, our findings prove that mitochondrial alterations and subsequent impairment are independent of an excessive mitochondrial reactive oxygen species (ROS) generation, which was found to be progressively diminished along with disease progression. Instead, increased peroxisomal abundance and peroxisomal fatty acid oxidation-related pathway suggest that peroxisomes may contribute to hepatic ROS generation and oxidative damage, which may accelerate hepatic injury and disease progression. We show here for the first time the sequential events of mitochondrial alterations involved in non-alcoholic fatty liver disease (NAFLD) progression and demonstrate that mitochondrial ROS are not one of the first hits that cause NAFLD progression.


Asunto(s)
Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/metabolismo , Autofagia , Ésteres del Colesterol/metabolismo , Biología Computacional/métodos , Susceptibilidad a Enfermedades , Fibrosis , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Mitocondrias/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Oxidación-Reducción , Estrés Oxidativo , Triglicéridos/metabolismo
12.
Cell Physiol Biochem ; 54(2): 230-251, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32153152

RESUMEN

BACKGROUND/AIMS: Adverse effects of cigarette smoke on health are widely known. Heating rather than combusting tobacco is one of strategies to reduce the formation of toxicants. The sensitive nature of mitochondrial dynamics makes the mitochondria an early indicator of cellular stress. For this reason, we studied the morphology and dynamics of the mitochondrial network in human bronchial epithelial cells (BEAS-2B) exposed to total particulate matter (TPM) generated from 3R4F reference cigarette smoke and from aerosol from a new candidate modified risk tobacco product, the Tobacco Heating System (THS 2.2). METHODS: Cells were subjected to short (1 week) and chronic (12 weeks) exposure to a low (7.5 µg/mL) concentration of 3R4F TPM and low (7.5 µg/mL), medium (37.5 µg/mL), and high (150 µg/mL) concentrations of TPM from THS 2.2. Confocal microscopy was applied to assess cellular and mitochondrial morphology. Cytosolic Ca2+ levels, mitochondrial membrane potential and mitochondrial mass were measured with appropriate fluorescent probes on laser scanning cytometer. The levels of proteins regulating mitochondrial dynamics and biogenesis were determined by Western blot. RESULTS: In BEAS-2B cells exposed for one week to the low concentration of 3R4F TPM and the high concentration of THS 2.2 TPM we observed clear changes in cell morphology, mitochondrial network fragmentation, altered levels of mitochondrial fusion and fission proteins and decreased biogenesis markers. Also cellular proliferation was slowed down. Upon chronic exposure (12 weeks) many parameters were affected in the opposite way comparing to short exposure. We observed strong increase of NRF2 protein level, reorganization of mitochondrial network and activation of the mitochondrial biogenesis process. CONCLUSION: Comparison of the effects of TPMs from 3R4F and from THS 2.2 revealed, that similar extent of alterations in mitochondrial dynamics and biogenesis is observed at 7.5 µg/mL of 3R4F TPM and 150 µg/mL of THS 2.2 TPM. 7 days exposure to the investigated components of cigarette smoke evoke mitochondrial stress, while upon chronic, 12 weeks exposure the hallmarks of cellular adaptation to the stressor were visible. The results also suggest that mitochondrial stress signaling is involved in the process of cellular adaptation under conditions of chronic stress caused by 3R4F and high concentration of THS 2.2.


Asunto(s)
Aerosoles/química , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Material Particulado/toxicidad , Calcio/metabolismo , Línea Celular , Colorantes Fluorescentes/química , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microscopía Confocal , Mitocondrias/efectos de los fármacos , Material Particulado/química , Humo/efectos adversos , Factores de Tiempo , Productos de Tabaco/análisis
13.
Int J Mol Sci ; 21(14)2020 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-32664529

RESUMEN

Calcific aortic stenosis is a disorder that impacts the physiology of heart valves. Fibrocalcific events progress in conjunction with thickening of the valve leaflets. Over the years, these events promote stenosis and obstruction of blood flow. Known and common risk factors are congenital defects, aging and metabolic syndromes linked to high plasma levels of lipoproteins. Inflammation and oxidative stress are the main molecular mediators of the evolution of aortic stenosis in patients and these mediators regulate both the degradation and remodeling processes. Mitochondrial dysfunction and dysregulation of autophagy also contribute to the disease. A better understanding of these cellular impairments might help to develop new ways to treat patients since, at the moment, there is no effective medical treatment to diminish neither the advancement of valve stenosis nor the left ventricular function impairments, and the current approaches are surgical treatment or transcatheter aortic valve replacement with prosthesis.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Calcinosis/metabolismo , Mitocondrias Cardíacas/fisiología , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/ultraestructura , Estenosis de la Válvula Aórtica/diagnóstico , Estenosis de la Válvula Aórtica/epidemiología , Estenosis de la Válvula Aórtica/cirugía , Autofagia , Membrana Basal/ultraestructura , Progresión de la Enfermedad , Células Endoteliales/patología , Humanos , Inflamación , Lípidos/análisis , Óxido Nítrico Sintasa de Tipo III/fisiología , Estrés Oxidativo , Terapias en Investigación , Respuesta de Proteína Desplegada
14.
Int J Obes (Lond) ; 43(5): 963-973, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30082750

RESUMEN

BACKGROUND/OBJECTIVES: Obesity is a complex disease characterized by the accumulation of excess body fat, which is caused by an increase in adipose cell size and number. The major source of adipocytes comes from mesenchymal stem cells (MSCs), although their roles in obesity remain unclear. An understanding of the mechanisms, regulation, and outcomes of adipogenesis is crucial for the development of new treatments for obesity-related diseases. Recently an unexpected role for the tumor suppressor promyelocytic leukemia protein (PML) in hematopoietic stem cell biology and metabolism regulation has come to light, but its role in MSC biology remains unknown. Here, we investigated the molecular pathway underlying the role of PML in the control of adipogenic MSC differentiation. SUBJECTS/METHODS: Muscle-derived stem cells (MDSCs) and adipose-derived stem cells (ADSCs) obtained from mice and voluntary patients (as a source of MSCs) were cultured in the presence of high glucose (HG) concentration, a nutrient stress condition known to promote MSCs differentiation into mature adipocytes and the adipogenic potential of PML was assessed. RESULTS: PML is essential for a correct HG-dependent adipogenic differentiation, and the enhancement of PML levels is fundamental during adipogenesis. Increased PML expression enables the upregulation of protein kinase Cß (PKCß), which, in turn, by controlling autophagy levels permits an increase in peroxisome proliferator-activated receptor γ (PPARγ) that leads the adipogenic differentiation. Therefore, genetic and pharmacological depletion of PML prevents PKCß expression, and by increasing autophagy levels, impairs the MSCs adipogenic differentiation. Human ADSCs isolated from overweight patients displayed increased PML and PKCß levels compared to those found in normal weight individuals, indicating that the PML-PKCß pathway is directly involved in the enhancement of adipogenesis and human metabolism. CONCLUSIONS: The new link found among PML, PKCß, and autophagy opens new therapeutic avenues for diseases characterized by an imbalance in the MSCs differentiation process, such as metabolic syndromes and cancer.


Asunto(s)
Adipogénesis/fisiología , Autofagia , Diabetes Mellitus Tipo 2/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Obesidad/metabolismo , PPAR gamma/metabolismo , Adipocitos , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glucosa/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Noqueados
15.
J Bioenerg Biomembr ; 51(4): 259-276, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31197632

RESUMEN

Mitochondria are multifunctional and dynamic organelles deeply integrated into cellular physiology and metabolism. Disturbances in mitochondrial function are involved in several disorders such as neurodegeneration, cardiovascular diseases, metabolic diseases, and also in the aging process. Nicotine is a natural alkaloid present in the tobacco plant which has been well studied as a constituent of cigarette smoke. It has also been reported to influence mitochondrial function both in vitro and in vivo. This review presents a comprehensive overview of the present knowledge of nicotine action on mitochondrial function. Observed effects of nicotine exposure on the mitochondrial respiratory chain, oxidative stress, calcium homeostasis, mitochondrial dynamics, biogenesis, and mitophagy are discussed, considering the context of the experimental design. The potential action of nicotine on cellular adaptation and cell survival is also examined through its interaction with mitochondria. Although a large number of studies have demonstrated the impact of nicotine on various mitochondrial activities, elucidating its mechanism of action requires further investigation.


Asunto(s)
Fumar Cigarrillos/metabolismo , Mitocondrias/metabolismo , Nicotina , Animales , Calcio/metabolismo , Fumar Cigarrillos/patología , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Mitocondrias/patología , Mitofagia/efectos de los fármacos , Nicotina/efectos adversos , Nicotina/farmacocinética , Estrés Oxidativo/efectos de los fármacos
16.
EMBO Rep ; 18(7): 1077-1089, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28566520

RESUMEN

The impact of the mitochondrial permeability transition (MPT) on cellular physiology is well characterized. In contrast, the composition and mode of action of the permeability transition pore complex (PTPC), the supramolecular entity that initiates MPT, remain to be elucidated. Specifically, the precise contribution of the mitochondrial F1FO ATP synthase (or subunits thereof) to MPT is a matter of debate. We demonstrate that F1FO ATP synthase dimers dissociate as the PTPC opens upon MPT induction. Stabilizing F1FO ATP synthase dimers by genetic approaches inhibits PTPC opening and MPT Specific mutations in the F1FO ATP synthase c subunit that alter C-ring conformation sensitize cells to MPT induction, which can be reverted by stabilizing F1FO ATP synthase dimers. Destabilizing F1FO ATP synthase dimers fails to trigger PTPC opening in the presence of mutants of the c subunit that inhibit MPT The current study does not provide direct evidence that the C-ring is the long-sought pore-forming subunit of the PTPC, but reveals that PTPC opening requires the dissociation of F1FO ATP synthase dimers and involves the C-ring.


Asunto(s)
Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico , Ciclosporina/farmacología , Células HEK293 , Humanos , Ratones , Proteínas de Transporte de Membrana Mitocondrial/química , Membranas Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/genética , Necrosis , Permeabilidad , Conformación Proteica , Multimerización de Proteína , Ratas
17.
Metab Brain Dis ; 33(6): 2005-2017, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30120672

RESUMEN

Huntington disease (HD) is an autosomal dominant neurodegenerative disorder manifesting as progressive impairment of motor function and different neuropsychiatric symptoms caused by an expansion of CAG repeats in huntingtin gene (HTT). Mitochondrial dysfunction and bioenergetic defects can contribute to the course of the disease, however, the molecular mechanism underlying this process is still largely unknown. In this study, we aimed to determine several mitochondrial parameters in HD fibroblasts and assess their relevance to the disease progression as well as to value mitochondrial pathology in peripheral cells as disease potential biomarker. We showed that HD fibroblasts demonstrate significantly lower growth rate compared to control fibroblasts despite the lack of cell cycle perturbations. In order to investigate mitochondrial contribution to cell growth differences between HD and healthy cells, we provided insight into various mitochondrial parameters. Conducted experiments have revealed a significant reduction of the ATP level in HD fibroblasts accompanied by a decrease in mitochondrial metabolic activity in relation to the cells from healthy donors. Importantly, there were no differences in the mitochondrial membrane potential (mtΔΨ) and OXPHOS complexes' levels. Slightly increased level of mitochondrial superoxide (mt. O2•-), but not cytosolic reactive oxygen species (cyt. ROS), has been demonstrated. We have also observed significantly elevated levels of some antioxidant enzymes (SOD2 and GR) which may serve as an indicator of antioxidant defense system in HD patients. Thus, we suggest that mitochondrial alterations in skin fibroblasts of Huntington's disease patients might be helpful in searching for novel disease biomarkers.


Asunto(s)
Fibroblastos/metabolismo , Enfermedad de Huntington/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Adulto , Anciano , Proliferación Celular/fisiología , Células Cultivadas , Femenino , Fibroblastos/patología , Humanos , Enfermedad de Huntington/patología , Masculino , Persona de Mediana Edad , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo
18.
Proc Natl Acad Sci U S A ; 112(6): 1779-84, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25624484

RESUMEN

The tumor suppressor p53 is a key protein in preventing cell transformation and tumor progression. Activated by a variety of stimuli, p53 regulates cell-cycle arrest and apoptosis. Along with its well-documented transcriptional control over cell-death programs within the nucleus, p53 exerts crucial although still poorly understood functions in the cytoplasm, directly modulating the apoptotic response at the mitochondrial level. Calcium (Ca(2+)) transfer between the endoplasmic reticulum (ER) and mitochondria represents a critical signal in the induction of apoptosis. However, the mechanism controlling this flux in response to stress stimuli remains largely unknown. Here we show that, in the cytoplasm, WT p53 localizes at the ER and at specialized contact domains between the ER and mitochondria (mitochondria-associated membranes). We demonstrate that, upon stress stimuli, WT p53 accumulates at these sites and modulates Ca(2+) homeostasis. Mechanistically, upon activation, WT p53 directly binds to the sarco/ER Ca(2+)-ATPase (SERCA) pump at the ER, changing its oxidative state and thus leading to an increased Ca(2+) load, followed by an enhanced transfer to mitochondria. The consequent mitochondrial Ca(2+) overload causes in turn alterations in the morphology of this organelle and induction of apoptosis. Pharmacological inactivation of WT p53 or naturally occurring p53 missense mutants inhibits SERCA pump activity at the ER, leading to a reduction of the Ca(2+) signaling from the ER to mitochondria. These findings define a critical nonnuclear function of p53 in regulating Ca(2+) signal-dependent apoptosis.


Asunto(s)
Apoptosis/fisiología , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Aequorina/metabolismo , Animales , Western Blotting , Línea Celular , Citosol/metabolismo , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Fura-2 , Técnicas de Silenciamiento del Gen , Humanos , Inmunoprecipitación , Ratones , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Proteína p53 Supresora de Tumor/genética
19.
Biochim Biophys Acta Bioenerg ; 1858(8): 615-627, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28087257

RESUMEN

Until 1972, the term 'apoptosis' was used to differentiate the programmed cell death that naturally occurs in organismal development from the acute tissue death referred to as necrosis. Many studies on cell death and programmed cell death have been published and most are, at least to some degree, related to cancer. Some key proteins and molecular pathways implicated in cell death have been analyzed, whereas others are still being actively researched; therefore, an increasing number of cellular compartments and organelles are being implicated in cell death and cancer. Here, we discuss the mitochondria and subdomains of the endoplasmic reticulum (ER) that interact with mitochondria, the mitochondria-associated membranes (MAMs), which have been identified as critical hubs in the regulation of cell death and tumor growth. MAMs-dependent calcium (Ca2+) release from the ER allows selective Ca2+ uptake by the mitochondria. The perturbation of Ca2+ homeostasis in cancer cells is correlated with sustained cell proliferation and the inhibition of cell death through the modulation of Ca2+ signaling. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Asunto(s)
Calcio/fisiología , Mitocondrias/fisiología , Membranas Mitocondriales/fisiología , Animales , Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Muerte Celular , División Celular , Transformación Celular Neoplásica , Progresión de la Enfermedad , Retículo Endoplásmico/metabolismo , Homeostasis , Humanos , Proteínas de la Membrana/fisiología , Membranas Mitocondriales/ultraestructura , Proteínas Mitocondriales/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Oncogénicas/fisiología , Transducción de Señal
20.
Eur J Clin Invest ; 47(1): 19-29, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27805735

RESUMEN

BACKGROUND: Patients with diabetes are at a high risk of developing both micro- and macrovascular disease. Hyperglycaemia seems to be the main factor in the pathogenesis of diabetic cardiomyopathy, often based on increased oxidative stress. Carvedilol, a ß-adrenergic blocker, has intrinsic antioxidant properties and was previously described to be effective in the protection of cardiac mitochondria against oxidative stress. The objective of this study was to evaluate the effect of carvedilol on hyperglycaemia-induced oxidative damage and mitochondrial abnormalities in cardiac and skeletal muscle in streptozotocin-treated rats. MATERIALS AND METHODS: Body mass, blood glucose, the level of protein carbonylation, caspase-9- and caspase-3-like activities, mitochondrial proteins, the status of antioxidant defence system and stress-related proteins were evaluated in streptozotocin vs streptozotocin + carvedilol (1 mg/kg/day)-treated rats. RESULTS: The results showed that carvedilol decreased blood glucose in streptozotocin-treated animals. Content of catalase in the heart and SOD2, SOD1 and catalase in skeletal muscle were increased by carvedilol treatment in streptozotocin-treated animals. At this particular time point, streptozotocin-induced hyperglycaemia did not cause caspase activation or increase in protein carbonylation status. The data showed that carvedilol increased the level of antioxidant enzymes, what may contribute to preserve cell redox balance during hyperglycaemia. We also showed here for the first time that carvedilol effects on streptozotocin-treated rats are tissue dependent, with a more predominant effect on skeletal muscle. CONCLUSIONS: Based on data showing modulation of the antioxidant network in the heart, carvedilol may be beneficial in diabetic patients without advanced disease complications, delaying their progression.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Carbazoles/farmacología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Musculares/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Propanolaminas/farmacología , Animales , Antioxidantes , Glucemia/metabolismo , Carvedilol , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Catalasa/efectos de los fármacos , Catalasa/metabolismo , Modelos Animales de Enfermedad , Masculino , Mitocondrias Cardíacas/metabolismo , Mitocondrias Musculares/metabolismo , Ratas , Ratas Wistar , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/efectos de los fármacos , Superóxido Dismutasa-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA