Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 56(3): 516-530.e9, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36738738

RESUMEN

In vitro studies have associated oxidative phosphorylation (OXPHOS) with anti-inflammatory macrophages, whereas pro-inflammatory macrophages rely on glycolysis. However, the metabolic needs of macrophages in tissues (TMFs) to fulfill their homeostatic activities are incompletely understood. Here, we identified OXPHOS as the highest discriminating process among TMFs from different organs in homeostasis by analysis of RNA-seq data in both humans and mice. Impairing OXPHOS in TMFs via Tfam deletion differentially affected TMF populations. Tfam deletion resulted in reduction of alveolar macrophages (AMs) due to impaired lipid-handling capacity, leading to increased cholesterol content and cellular stress, causing cell-cycle arrest in vivo. In obesity, Tfam depletion selectively ablated pro-inflammatory lipid-handling white adipose tissue macrophages (WAT-MFs), thus preventing insulin resistance and hepatosteatosis. Hence, OXPHOS, rather than glycolysis, distinguishes TMF populations and is critical for the maintenance of TMFs with a high lipid-handling activity, including pro-inflammatory WAT-MFs. This could provide a selective therapeutic targeting tool.


Asunto(s)
Inflamación , Fosforilación Oxidativa , Humanos , Ratones , Animales , Inflamación/metabolismo , Macrófagos/metabolismo , Homeostasis , Lípidos , Tejido Adiposo/metabolismo
2.
Nature ; 618(7964): 365-373, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225978

RESUMEN

Birth presents a metabolic challenge to cardiomyocytes as they reshape fuel preference from glucose to fatty acids for postnatal energy production1,2. This adaptation is triggered in part by post-partum environmental changes3, but the molecules orchestrating cardiomyocyte maturation remain unknown. Here we show that this transition is coordinated by maternally supplied γ-linolenic acid (GLA), an 18:3 omega-6 fatty acid enriched in the maternal milk. GLA binds and activates retinoid X receptors4 (RXRs), ligand-regulated transcription factors that are expressed in cardiomyocytes from embryonic stages. Multifaceted genome-wide analysis revealed that the lack of RXR in embryonic cardiomyocytes caused an aberrant chromatin landscape that prevented the induction of an RXR-dependent gene expression signature controlling mitochondrial fatty acid homeostasis. The ensuing defective metabolic transition featured blunted mitochondrial lipid-derived energy production and enhanced glucose consumption, leading to perinatal cardiac dysfunction and death. Finally, GLA supplementation induced RXR-dependent expression of the mitochondrial fatty acid homeostasis signature in cardiomyocytes, both in vitro and in vivo. Thus, our study identifies the GLA-RXR axis as a key transcriptional regulatory mechanism underlying the maternal control of perinatal cardiac metabolism.


Asunto(s)
Ácidos Grasos , Glucosa , Corazón , Leche Humana , Ácido gammalinolénico , Femenino , Humanos , Recién Nacido , Embarazo , Cromatina/genética , Ácidos Grasos/metabolismo , Ácido gammalinolénico/metabolismo , Ácido gammalinolénico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Corazón/efectos de los fármacos , Corazón/embriología , Corazón/crecimiento & desarrollo , Homeostasis , Técnicas In Vitro , Leche Humana/química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Receptores X Retinoide/metabolismo , Factores de Transcripción/metabolismo
3.
Mol Cell ; 74(5): 877-890.e6, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31023583

RESUMEN

Endoplasmic reticulum (ER) stress and unfolded protein response are energetically challenging under nutrient stress conditions. However, the regulatory mechanisms that control the energetic demand under nutrient and ER stress are largely unknown. Here we show that ER stress and glucose deprivation stimulate mitochondrial bioenergetics and formation of respiratory supercomplexes (SCs) through protein kinase R-like ER kinase (PERK). Genetic ablation or pharmacological inhibition of PERK suppresses nutrient and ER stress-mediated increases in SC levels and reduces oxidative phosphorylation-dependent ATP production. Conversely, PERK activation augments respiratory SCs. The PERK-eIF2α-ATF4 axis increases supercomplex assembly factor 1 (SCAF1 or COX7A2L), promoting SCs and enhanced mitochondrial respiration. PERK activation is sufficient to rescue bioenergetic defects caused by complex I missense mutations derived from mitochondrial disease patients. These studies have identified an energetic communication between ER and mitochondria, with implications in cell survival and diseases associated with mitochondrial failures.


Asunto(s)
Factor de Transcripción Activador 4/genética , Metabolismo Energético/genética , Factor 2 Eucariótico de Iniciación/genética , Mitocondrias/genética , eIF-2 Quinasa/genética , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Línea Celular , Supervivencia Celular/genética , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/genética , Glucosa/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Mutación Missense/genética , Nutrientes/metabolismo , Fosforilación , Factores de Empalme Serina-Arginina/genética , Transducción de Señal
4.
Nature ; 586(7828): 287-291, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32728214

RESUMEN

All metazoans depend on the consumption of O2 by the mitochondrial oxidative phosphorylation system (OXPHOS) to produce energy. In addition, the OXPHOS uses O2 to produce reactive oxygen species that can drive cell adaptations1-4, a phenomenon that occurs in hypoxia4-8 and whose precise mechanism remains unknown. Ca2+ is the best known ion that acts as a second messenger9, yet the role ascribed to Na+ is to serve as a mere mediator of membrane potential10. Here we show that Na+ acts as a second messenger that regulates OXPHOS function and the production of reactive oxygen species by modulating the fluidity of the inner mitochondrial membrane. A conformational shift in mitochondrial complex I during acute hypoxia11 drives acidification of the matrix and the release of free Ca2+ from calcium phosphate (CaP) precipitates. The concomitant activation of the mitochondrial Na+/Ca2+ exchanger promotes the import of Na+ into the matrix. Na+ interacts with phospholipids, reducing inner mitochondrial membrane fluidity and the mobility of free ubiquinone between complex II and complex III, but not inside supercomplexes. As a consequence, superoxide is produced at complex III. The inhibition of Na+ import through the Na+/Ca2+ exchanger is sufficient to block this pathway, preventing adaptation to hypoxia. These results reveal that Na+ controls OXPHOS function and redox signalling through an unexpected interaction with phospholipids, with profound consequences for cellular metabolism.


Asunto(s)
Transporte de Electrón , Hipoxia/metabolismo , Mitocondrias/metabolismo , Sistemas de Mensajero Secundario , Sodio/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Fosfatos de Calcio/metabolismo , Línea Celular Tumoral , Precipitación Química , Humanos , Masculino , Fluidez de la Membrana , Ratones Endogámicos C57BL , Membranas Mitocondriales/química , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Fosforilación Oxidativa , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Intercambiador de Sodio-Calcio/metabolismo
5.
J Physiol ; 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38630964

RESUMEN

In eukaryotic cells, aerobic energy is produced by mitochondria through oxygen uptake. However, little is known about the early mitochondrial responses to moderate hypobaric hypoxia (MHH) in highly metabolic active tissues. Here, we describe the mitochondrial responses to acute MHH in the heart and skeletal muscle. Rats were randomly allocated into a normoxia control group (n = 10) and a hypoxia group (n = 30), divided into three groups (0, 6, and 24 h post-MHH). The normoxia situation was recapitulated at the University of Granada, at 662 m above sea level. The MHH situation was performed at the High-Performance Altitude Training Centre of Sierra Nevada located in Granada at 2320 m above sea level. We found a significant increase in mitochondrial supercomplex assembly in the heart as soon as the animals reached 2320 m above sea level and their levels are maintained 24 h post-exposure, but not in skeletal muscle. Furthermore, in skeletal muscle, at 0 and 6 h, there was increased dynamin-related protein 1 (Drp1) expression and a significant reduction in Mitofusin 2. In conclusion, mitochondria from the muscle and heart respond differently to MHH: mitochondrial supercomplexes increase in the heart, whereas, in skeletal muscle, the mitochondrial pro-fission response is trigged. Considering that skeletal muscle was not actively involved in the ascent when the heart was beating faster to compensate for the hypobaric, hypoxic conditions, we speculate that the different responses to MHH are a result of the different energetic requirements of the tissues upon MHH. KEY POINTS: The heart and the skeletal muscle showed different mitochondrial responses to moderate hypobaric hypoxia. Moderate hypobaric hypoxia increases the assembly of the electron transport chain complexes into supercomplexes in the heart. Skeletal muscle shows an early mitochondrial pro-fission response following exposure to moderate hypobaric hypoxia.

6.
PLoS Biol ; 19(11): e3001447, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34758018

RESUMEN

During the first weeks of postnatal heart development, cardiomyocytes undergo a major adaptive metabolic shift from glycolytic energy production to fatty acid oxidation. This metabolic change is contemporaneous to the up-regulation and activation of the p38γ and p38δ stress-activated protein kinases in the heart. We demonstrate that p38γ/δ contribute to the early postnatal cardiac metabolic switch through inhibitory phosphorylation of glycogen synthase 1 (GYS1) and glycogen metabolism inactivation. Premature induction of p38γ/δ activation in cardiomyocytes of newborn mice results in an early GYS1 phosphorylation and inhibition of cardiac glycogen production, triggering an early metabolic shift that induces a deficit in cardiomyocyte fuel supply, leading to whole-body metabolic deregulation and maladaptive cardiac pathogenesis. Notably, the adverse effects of forced premature cardiac p38γ/δ activation in neonate mice are prevented by maternal diet supplementation of fatty acids during pregnancy and lactation. These results suggest that diet interventions have a potential for treating human cardiac genetic diseases that affect heart metabolism.


Asunto(s)
Glucógeno Sintasa/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Miocardio/enzimología , Animales , Animales Recién Nacidos , Cardiomegalia/enzimología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Dieta Alta en Grasa , Activación Enzimática , Conducta Alimentaria , Femenino , Eliminación de Gen , Intolerancia a la Glucosa/enzimología , Glucógeno/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Miocitos Cardíacos/enzimología , Especificidad de Órganos , Fosforilación
8.
Circulation ; 145(14): 1084-1101, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35236094

RESUMEN

BACKGROUND: In most eukaryotic cells, the mitochondrial DNA (mtDNA) is transmitted uniparentally and present in multiple copies derived from the clonal expansion of maternally inherited mtDNA. All copies are therefore near-identical, or homoplasmic. The presence of >1 mtDNA variant in the same cytoplasm can arise naturally or result from new medical technologies aimed at preventing mitochondrial genetic diseases and improving fertility. The latter is called divergent nonpathologic mtDNA heteroplasmy (DNPH). We hypothesized that DNPH is maladaptive and usually prevented by the cell. METHODS: We engineered and characterized DNPH mice throughout their lifespan using transcriptomic, metabolomic, biochemical, physiologic, and phenotyping techniques. We focused on in vivo imaging techniques for noninvasive assessment of cardiac and pulmonary energy metabolism. RESULTS: We show that DNPH impairs mitochondrial function, with profound consequences in critical tissues that cannot resolve heteroplasmy, particularly cardiac and skeletal muscle. Progressive metabolic stress in these tissues leads to severe pathology in adulthood, including pulmonary hypertension and heart failure, skeletal muscle wasting, frailty, and premature death. Symptom severity is strongly modulated by the nuclear context. CONCLUSIONS: Medical interventions that may generate DNPH should address potential incompatibilities between donor and recipient mtDNA.


Asunto(s)
Fragilidad , Cardiopatías , Hipertensión Pulmonar , Adulto , Animales , ADN Mitocondrial/genética , Fragilidad/patología , Cardiopatías/patología , Heteroplasmia , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Ratones , Mitocondrias/genética
9.
Biol Chem ; 404(5): 399-415, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36952351

RESUMEN

The orchestrated activity of the mitochondrial respiratory or electron transport chain (ETC) and ATP synthase convert reduction power (NADH, FADH2) into ATP, the cell's energy currency in a process named oxidative phosphorylation (OXPHOS). Three out of the four ETC complexes are found in supramolecular assemblies: complex I, III, and IV form the respiratory supercomplexes (SC). The plasticity model suggests that SC formation is a form of adaptation to changing conditions such as energy supply, redox state, and stress. Complex I, the NADH-dehydrogenase, is part of the largest supercomplex (CI + CIII2 + CIVn). Here, we demonstrate the role of NDUFB10, a subunit of the membrane arm of complex I, in complex I and supercomplex assembly on the one hand and bioenergetics function on the other. NDUFB10 knockout was correlated with a decrease of SCAF1, a supercomplex assembly factor, and a reduction of respiration and mitochondrial membrane potential. This likely is due to loss of proton pumping since the CI P P -module is downregulated and the P D -module is completely abolished in NDUFB10 knock outs.


Asunto(s)
Complejo I de Transporte de Electrón , NADH Deshidrogenasa , Adenosina Trifosfato/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Mitocondrias/metabolismo , NAD/metabolismo , Fosforilación Oxidativa , NADH Deshidrogenasa/metabolismo
11.
Int J Mol Sci ; 24(2)2023 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36674816

RESUMEN

As the last step of the OXPHOS system, mitochondrial ATP synthase (or complex V) is responsible for ATP production by using the generated proton gradient, but also has an impact on other important functions linked to this system. Mutations either in complex V structural subunits, especially in mtDNA-encoded ATP6 gene, or in its assembly factors, are the molecular cause of a wide variety of human diseases, most of them classified as neurodegenerative disorders. The role of ATP synthase alterations in cancer development or metastasis has also been postulated. In this work, we reported the generation and characterization of the first mt-Atp6 pathological mutation in mouse cells, an m.8414A>G transition that promotes an amino acid change from Asn to Ser at a highly conserved residue of the protein (p.N163S), located near the path followed by protons from the intermembrane space to the mitochondrial matrix. The phenotypic consequences of the p.N163S change reproduce the effects of MT-ATP6 mutations in human diseases, such as dependence on glycolysis, defective OXPHOS activity, ATP synthesis impairment, increased ROS generation or mitochondrial membrane potential alteration. These observations demonstrate that this mutant cell line could be of great interest for the generation of mouse models with the aim of studying human diseases caused by alterations in ATP synthase. On the other hand, mutant cells showed lower migration capacity, higher expression of MHC-I and slightly lower levels of HIF-1α, indicating a possible reduction of their tumorigenic potential. These results could suggest a protective role of ATP synthase inhibition against tumor transformation that could open the door to new therapeutic strategies in those cancer types relying on OXPHOS metabolism.


Asunto(s)
Mitocondrias , ATPasas de Translocación de Protón Mitocondriales , Animales , Humanos , Ratones , Adenosina Trifosfato/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , ADN Mitocondrial/genética , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Mutación , Fenotipo , Respiración
12.
Small ; 18(16): e2106570, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35263020

RESUMEN

Manganese ferrite nanoparticles display interesting features in bioimaging and catalytic therapies. They have been recently used in theranostics as contrast agents in magnetic resonance imaging (MRI), and as catalase-mimicking nanozymes for hypoxia alleviation. These promising applications encourage the development of novel synthetic procedures to enhance the bioimaging and catalytic properties of these nanomaterials simultaneously. Herein, a cost-efficient synthetic microwave method is developed to manufacture ultrasmall manganese ferrite nanoparticles as advanced multimodal contrast agents in MRI and positron emission tomography (PET), and improved nanozymes. Such a synthetic method allows doping ferrites with Mn in a wide stoichiometric range (Mnx Fe3-x O4 , 0.1 ≤ x ≤ 2.4), affording a library of nanoparticles with different magnetic relaxivities and catalytic properties. These tuned magnetic properties give rise to either positive or dual-mode MRI contrast agents. On the other hand, higher levels of Mn doping enhance the catalytic efficiency of the resulting nanozymes. Finally, through their intracellular catalase-mimicking activity, these ultrasmall manganese ferrite nanoparticles induce an unprecedented tumor growth inhibition in a breast cancer murine model. All of these results show the robust characteristics of these nanoparticles for nanobiotechnological applications.


Asunto(s)
Medios de Contraste , Nanopartículas , Animales , Catalasa , Compuestos Férricos , Imagen por Resonancia Magnética/métodos , Compuestos de Manganeso , Ratones
13.
IUBMB Life ; 74(7): 629-644, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35166025

RESUMEN

The flavin mononucleotide (FMN) cofactor of respiratory complex I occupies a key position in the electron transport chain. Here, the electrons coming from NADH start the sequence of oxidoreduction reactions, which drives the generation of the proton-motive force necessary for ATP synthesis. The overall architecture and the general catalytic proprieties of the FMN site are mostly well established. However, several aspects regarding the complex I flavin cofactor are still unknown. For example, the flavin binding to the N-module, the NADH-oxidizing portion of complex I, lacks a molecular description. The dissociation of FMN from the enzyme is beginning to emerge as an important regulatory mechanism of complex I activity and ROS production. Finally, how mitochondria import and metabolize FMN is still uncertain. This review summarizes the current knowledge on complex I flavin cofactor and discusses the open questions for future research.


Asunto(s)
Complejo I de Transporte de Electrón , Mononucleótido de Flavina , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Mononucleótido de Flavina/química , Mononucleótido de Flavina/metabolismo , Flavinas/química , Flavinas/metabolismo , NAD/química , NAD/metabolismo , Oxidación-Reducción
14.
EMBO Rep ; 21(7): e50287, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32496654

RESUMEN

The oxidative phosphorylation (OXPHOS) system is a dynamic system in which the respiratory complexes coexist with super-assembled quaternary structures called supercomplexes (SCs). The physiological role of SCs is still disputed. Here, we used zebrafish to study the relevance of respiratory SCs. We combined immunodetection analysis and deep data-independent proteomics to characterize these structures and found similar SCs to those described in mice, as well as novel SCs including III2  + IV2 , I + IV, and I + III2  + IV2 . To study the physiological role of SCs, we generated two null allele zebrafish lines for supercomplex assembly factor 1 (scaf1). scaf1-/- fish displayed altered OXPHOS activity due to the disrupted interaction of complexes III and IV. scaf1-/- fish were smaller in size and showed abnormal fat deposition and decreased female fertility. These physiological phenotypes were rescued by doubling the food supply, which correlated with improved bioenergetics and alterations in the metabolic gene expression program. These results reveal that SC assembly by Scaf1 modulates OXPHOS efficiency and allows the optimization of metabolic resources.


Asunto(s)
Complejo IV de Transporte de Electrones , Factores de Empalme Serina-Arginina/metabolismo , Pez Cebra , Animales , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético/genética , Femenino , Ratones , Membranas Mitocondriales/metabolismo , Fosforilación Oxidativa , Pez Cebra/genética , Pez Cebra/metabolismo
15.
Nature ; 539(7630): 579-582, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27775717

RESUMEN

Respiratory chain complexes can super-assemble into quaternary structures called supercomplexes that optimize cellular metabolism. The interaction between complexes III (CIII) and IV (CIV) is modulated by supercomplex assembly factor 1 (SCAF1, also known as COX7A2L). The discovery of SCAF1 represented strong genetic evidence that supercomplexes exist in vivo. SCAF1 is present as a long isoform (113 amino acids) or a short isoform (111 amino acids) in different mouse strains. Only the long isoform can induce the super-assembly of CIII and CIV, but it is not clear whether SCAF1 is required for the formation of the respirasome (a supercomplex of CI, CIII2 and CIV). Here we show, by combining deep proteomics and immunodetection analysis, that SCAF1 is always required for the interaction between CIII and CIV and that the respirasome is absent from most tissues of animals containing the short isoform of SCAF1, with the exception of heart and skeletal muscle. We used directed mutagenesis to characterize SCAF1 regions that interact with CIII and CIV and discovered that this interaction requires the correct orientation of a histidine residue at position 73 that is altered in the short isoform of SCAF1, explaining its inability to interact with CIV. Furthermore, we find that the CIV subunit COX7A2 is replaced by SCAF1 in supercomplexes containing CIII and CIV and by COX7A1 in CIV dimers, and that dimers seem to be more stable when they include COX6A2 rather than the COX6A1 isoform.


Asunto(s)
Membranas Mitocondriales/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Complejo IV de Transporte de Electrones/química
16.
Nature ; 535(7613): 561-5, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27383793

RESUMEN

Human mitochondrial DNA (mtDNA) shows extensive within population sequence variability. Many studies suggest that mtDNA variants may be associated with ageing or diseases, although mechanistic evidence at the molecular level is lacking. Mitochondrial replacement has the potential to prevent transmission of disease-causing oocyte mtDNA. However, extension of this technology requires a comprehensive understanding of the physiological relevance of mtDNA sequence variability and its match with the nuclear-encoded mitochondrial genes. Studies in conplastic animals allow comparison of individuals with the same nuclear genome but different mtDNA variants, and have provided both supporting and refuting evidence that mtDNA variation influences organismal physiology. However, most of these studies did not confirm the conplastic status, focused on younger animals, and did not investigate the full range of physiological and phenotypic variability likely to be influenced by mitochondria. Here we systematically characterized conplastic mice throughout their lifespan using transcriptomic, proteomic,metabolomic, biochemical, physiological and phenotyping studies. We show that mtDNA haplotype profoundly influences mitochondrial proteostasis and reactive oxygen species generation,insulin signalling, obesity, and ageing parameters including telomere shortening and mitochondrial dysfunction, resulting in profound differences in health longevity between conplastic strains.


Asunto(s)
Envejecimiento/genética , Núcleo Celular/genética , ADN Mitocondrial/genética , Variación Genética/genética , Metabolismo/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Envejecimiento/fisiología , Animales , Femenino , Genoma Mitocondrial/genética , Haplotipos , Insulina/metabolismo , Longevidad/genética , Masculino , Metabolismo/fisiología , Metabolómica , Ratones , Ratones Congénicos , Mitocondrias/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Obesidad/genética , Obesidad/metabolismo , Fenotipo , Proteómica , Especies Reactivas de Oxígeno/metabolismo , Acortamiento del Telómero , Transcriptoma , Respuesta de Proteína Desplegada
17.
EMBO J ; 36(22): 3356-3371, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29018038

RESUMEN

In the amyloidogenic pathway associated with Alzheimer disease (AD), the amyloid precursor protein (APP) is cleaved by ß-secretase to generate a 99-aa C-terminal fragment (C99) that is then cleaved by γ-secretase to generate the ß-amyloid (Aß) found in senile plaques. In previous reports, we and others have shown that γ-secretase activity is enriched in mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) and that ER-mitochondrial connectivity and MAM function are upregulated in AD We now show that C99, in addition to its localization in endosomes, can also be found in MAM, where it is normally processed rapidly by γ-secretase. In cell models of AD, however, the concentration of unprocessed C99 increases in MAM regions, resulting in elevated sphingolipid turnover and an altered lipid composition of both MAM and mitochondrial membranes. In turn, this change in mitochondrial membrane composition interferes with the proper assembly and activity of mitochondrial respiratory supercomplexes, thereby likely contributing to the bioenergetic defects characteristic of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Mitocondrias/metabolismo , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular , Respiración de la Célula , Retículo Endoplásmico/ultraestructura , Humanos , Membranas Intracelulares/ultraestructura , Ratones , Mitocondrias/ultraestructura , Mutación/genética , Consumo de Oxígeno , Presenilinas/genética , Transporte de Proteínas , Esfingolípidos/metabolismo , Regulación hacia Arriba
18.
IUBMB Life ; 73(3): 511-529, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33369015

RESUMEN

The oxidative phosphorylation (OXPHOS) system is the only structure in animal cells with components encoded by two genomes, maternally transmitted mitochondrial DNA (mtDNA), and biparentally transmitted nuclear DNA (nDNA). MtDNA-encoded genes have to physically assemble with their counterparts encoded in the nucleus to build together the functional respiratory complexes. Therefore, structural and functional matching requirements between the protein subunits of these molecular complexes are rigorous. The crosstalk between nDNA and mtDNA needs to overcome some challenges, as the nuclear-encoded factors have to be imported into the mitochondria in a correct quantity and match the high number of organelles and genomes per mitochondria that encode and synthesize their own components locally. The cell is able to sense the mito-nuclear match through changes in the activity of the OXPHOS system, modulation of the mitochondrial biogenesis, or reactive oxygen species production. This implies that a complex signaling cascade should optimize OXPHOS performance to the cellular-specific requirements, which will depend on cell type, environmental conditions, and life stage. Therefore, the mitochondria would function as a cellular metabolic information hub integrating critical information that would feedback the nucleus for it to respond accordingly. Here, we review the current understanding of the complex interaction between mtDNA and nDNA.


Asunto(s)
Núcleo Celular/genética , ADN Mitocondrial , Fosforilación Oxidativa , Animales , Citoplasma/genética , Citoplasma/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Evolución Molecular , Regulación de la Expresión Génica , Genoma , Haplotipos , Humanos , Mitocondrias/genética
19.
Biochem Soc Trans ; 49(6): 2655-2668, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34747989

RESUMEN

Mitochondria are one of the most exhaustively investigated organelles in the cell and most attention has been paid to the components of the mitochondrial electron transport chain (ETC) in the last 100 years. The ETC collects electrons from NADH or FADH2 and transfers them through a series of electron carriers within multiprotein respiratory complexes (complex I to IV) to oxygen, therefore generating an electrochemical gradient that can be used by the F1-F0-ATP synthase (also named complex V) in the mitochondrial inner membrane to synthesize ATP. The organization and function of the ETC is a continuous source of surprises. One of the latest is the discovery that the respiratory complexes can assemble to form a variety of larger structures called super-complexes (SCs). This opened an unexpected level of complexity in this well-known and fundamental biological process. This review will focus on the current evidence for the formation of different SCs and will explore how they modulate the ETC organization according to the metabolic state. Since the field is rapidly growing, we also comment on the experimental techniques used to describe these SC and hope that this overview may inspire new technologies that will help to advance the field.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejos Multienzimáticos/metabolismo , Animales , Mitocondrias/metabolismo , Fosforilación Oxidativa
20.
PLoS Biol ; 16(7): e2004455, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29979672

RESUMEN

Adipose tissue has emerged as an important regulator of whole-body metabolism, and its capacity to dissipate energy in the form of heat has acquired a special relevance in recent years as potential treatment for obesity. In this context, the p38MAPK pathway has arisen as a key player in the thermogenic program because it is required for the activation of brown adipose tissue (BAT) thermogenesis and participates also in the transformation of white adipose tissue (WAT) into BAT-like depot called beige/brite tissue. Here, using mice that are deficient in p38α specifically in adipose tissue (p38αFab-KO), we unexpectedly found that lack of p38α protected against high-fat diet (HFD)-induced obesity. We also showed that p38αFab-KO mice presented higher energy expenditure due to increased BAT thermogenesis. Mechanistically, we found that lack of p38α resulted in the activation of the related protein kinase family member p38δ. Our results showed that p38δ is activated in BAT by cold exposure, and lack of this kinase specifically in adipose tissue (p38δ Fab-KO) resulted in overweight together with reduced energy expenditure and lower body and skin surface temperature in the BAT region. These observations indicate that p38α probably blocks BAT thermogenesis through p38δ inhibition. Consistent with the results obtained in animals, p38α was reduced in visceral and subcutaneous adipose tissue of subjects with obesity and was inversely correlated with body mass index (BMI). Altogether, we have elucidated a mechanism implicated in physiological BAT activation that has potential clinical implications for the treatment of obesity and related diseases such as diabetes.


Asunto(s)
Tejido Adiposo Pardo/enzimología , Tejido Adiposo Pardo/fisiología , Proteína Quinasa 13 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Termogénesis , Adipocitos Marrones/enzimología , Adulto , Animales , Índice de Masa Corporal , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/prevención & control , Dieta , Metabolismo Energético , Activación Enzimática , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Modelos Biológicos , Obesidad/enzimología , Obesidad/prevención & control , Proteína Desacopladora 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA