Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167169, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631408

RESUMEN

Mitochondrial dysregulation is pivotal in Alzheimer's disease (AD) pathogenesis. Calcium governs vital mitochondrial processes impacting energy conversion, oxidative stress, and cell death signaling. Disruptions in mitochondrial calcium (mCa2+) handling induce calcium overload and trigger the opening of mitochondrial permeability transition pore, ensuing energy deprivation and resulting in AD-related neuronal cell death. However, the role of mCa2+ in non-neuronal cells (microglia, astrocytes, oligodendrocytes, endothelial cells, and pericytes) remains elusive. This review provides a comprehensive exploration of mitochondrial heterogeneity and calcium signaling, offering insights into specific differences among various brain cell types in AD.


Asunto(s)
Enfermedad de Alzheimer , Señalización del Calcio , Calcio , Mitocondrias , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , Señalización del Calcio/fisiología , Animales , Calcio/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Pericitos/metabolismo , Pericitos/patología , Microglía/metabolismo , Microglía/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Estrés Oxidativo , Oligodendroglía/metabolismo , Oligodendroglía/patología , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Neuronas/metabolismo , Neuronas/patología
2.
bioRxiv ; 2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37904949

RESUMEN

Alzheimer's disease (AD) is characterized by the extracellular deposition of amyloid beta, intracellular neurofibrillary tangles, synaptic dysfunction, and neuronal cell death. These phenotypes correlate with and are linked to elevated neuronal intracellular calcium ( i Ca 2+ ) levels. Recently, our group reported that mitochondrial calcium ( m Ca 2+ ) overload, due to loss of m Ca 2+ efflux capacity, contributes to AD development and progression. We also noted proteomic remodeling of the mitochondrial calcium uniporter channel (mtCU) in sporadic AD brain samples, suggestive of altered m Ca 2+ uptake in AD. Since the mtCU is the primary mechanism for Ca 2+ uptake into the mitochondrial matrix, inhibition of the mtCU has the potential to reduce or prevent m Ca 2+ overload in AD. Here, we report that neuronal-specific loss of mtCU-dependent m Ca 2+ uptake in the 3xTg-AD mouse model of AD reduced Aß and tau-pathology, synaptic dysfunction, and cognitive decline. Knockdown of Mcu in a cellular model of AD significantly decreased matrix Ca 2+ content, oxidative stress, and cell death. These results suggest that inhibition of neuronal m Ca 2+ uptake is a novel therapeutic target to impede AD progression.

3.
Front Cell Dev Biol ; 11: 1196466, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37601094

RESUMEN

Mitochondria play a critical role in energy metabolism and signal transduction, which is tightly regulated by proteins, metabolites, and ion fluxes. Metabolites and ion homeostasis are mainly mediated by channels and transporters present on mitochondrial membranes. Mitochondria comprise two distinct compartments, the outer mitochondrial membrane (OMM) and the inner mitochondrial membrane (IMM), which have differing permeabilities to ions and metabolites. The OMM is semipermeable due to the presence of non-selective molecular pores, while the IMM is highly selective and impermeable due to the presence of specialized channels and transporters which regulate ion and metabolite fluxes. These channels and transporters are modulated by various post-translational modifications (PTMs), including phosphorylation, oxidative modifications, ions, and metabolites binding, glycosylation, acetylation, and others. Additionally, the mitochondrial protein quality control (MPQC) system plays a crucial role in ensuring efficient molecular flux through the mitochondrial membranes by selectively removing mistargeted or defective proteins. Inefficient functioning of the transporters and channels in mitochondria can disrupt cellular homeostasis, leading to the onset of various pathological conditions. In this review, we provide a comprehensive overview of the current understanding of mitochondrial channels and transporters in terms of their functions, PTMs, and quality control mechanisms.

4.
bioRxiv ; 2023 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-37131819

RESUMEN

Background: Mitochondrial calcium (mCa2+) uptake through the mitochondrial calcium uniporter channel (mtCU) stimulates metabolism to meet acute increases in cardiac energy demand. However, excessive mCa2+ uptake during stress, as in ischemia-reperfusion, initiates permeability transition and cell death. Despite these often-reported acute physiological and pathological effects, a major unresolved controversy is whether mtCU-dependent mCa2+ uptake and long-term elevation of cardiomyocyte mCa2+ contributes to the heart's adaptation during sustained increases in workload. Objective: We tested the hypothesis that mtCU-dependent mCa2+ uptake contributes to cardiac adaptation and ventricular remodeling during sustained catecholaminergic stress. Methods: Mice with tamoxifen-inducible, cardiomyocyte-specific gain (αMHC-MCM × flox-stop-MCU; MCU-Tg) or loss (αMHC-MCM × Mcufl/fl; Mcu-cKO) of mtCU function received 2-wk catecholamine infusion. Results: Cardiac contractility increased after 2d of isoproterenol in control, but not Mcu-cKO mice. Contractility declined and cardiac hypertrophy increased after 1-2-wk of isoproterenol in MCU-Tg mice. MCU-Tg cardiomyocytes displayed increased sensitivity to Ca2+- and isoproterenol-induced necrosis. However, loss of the mitochondrial permeability transition pore (mPTP) regulator cyclophilin D failed to attenuate contractile dysfunction and hypertrophic remodeling, and increased isoproterenol-induced cardiomyocyte death in MCU-Tg mice. Conclusions: mtCU mCa2+ uptake is required for early contractile responses to adrenergic signaling, even those occurring over several days. Under sustained adrenergic load excessive MCU-dependent mCa2+ uptake drives cardiomyocyte dropout, perhaps independent of classical mitochondrial permeability transition pore opening, and compromises contractile function. These findings suggest divergent consequences for acute versus sustained mCa2+ loading, and support distinct functional roles for the mPTP in settings of acute mCa2+ overload versus persistent mCa2+ stress.

5.
Sci Signal ; 16(782): eabi8948, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37098122

RESUMEN

MICU1 is a calcium (Ca2+)-binding protein that regulates the mitochondrial Ca2+ uniporter channel complex (mtCU) and mitochondrial Ca2+ uptake. MICU1 knockout mice display disorganized mitochondrial architecture, a phenotype that is distinct from that of mice with deficiencies in other mtCU subunits and, thus, is likely not explained by changes in mitochondrial matrix Ca2+ content. Using proteomic and cellular imaging techniques, we found that MICU1 localized to the mitochondrial contact site and cristae organizing system (MICOS) and directly interacted with the MICOS components MIC60 and CHCHD2 independently of the mtCU. We demonstrated that MICU1 was essential for MICOS complex formation and that MICU1 ablation resulted in altered cristae organization, mitochondrial ultrastructure, mitochondrial membrane dynamics, and cell death signaling. Together, our results suggest that MICU1 is an intermembrane space Ca2+ sensor that modulates mitochondrial membrane dynamics independently of matrix Ca2+ uptake. This system enables distinct Ca2+ signaling in the mitochondrial matrix and at the intermembrane space to modulate cellular energetics and cell death in a concerted manner.


Asunto(s)
Membranas Mitocondriales , Proteómica , Ratones , Animales , Membranas Mitocondriales/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Ratones Noqueados , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas Mitocondriales/metabolismo
6.
iScience ; 26(3): 106296, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36936788

RESUMEN

Mitochondrial calcium overload contributes to neurodegenerative disease development and progression. We recently reported that loss of the mitochondrial sodium/calcium exchanger (NCLX), the primary mechanism of mCa2+ efflux, promotes mCa2+ overload, metabolic derangement, redox stress, and cognitive decline in models of Alzheimer's disease (AD). However, whether disrupted mCa2+ signaling contributes to neuronal pathology and cognitive decline independent of pre-existing amyloid or tau pathology remains unknown. Here, we generated mice with neuronal deletion of the mitochondrial sodium/calcium exchanger (NCLX, Slc8b1 gene), and evaluated age-associated changes in cognitive function and neuropathology. Neuronal loss of NCLX resulted in an age-dependent decline in spatial and cued recall memory, moderate amyloid deposition, mild tau pathology, synaptic remodeling, and indications of cell death. These results demonstrate that loss of NCLX-dependent mCa2+ efflux alone is sufficient to induce an Alzheimer's disease-like pathology and highlights the promise of therapies targeting mCa2+ exchange.

7.
J Mol Cell Cardiol ; 167: 52-66, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35358843

RESUMEN

Mitochondrial calcium (mCa2+) uptake couples changes in cardiomyocyte energetic demand to mitochondrial ATP production. However, excessive mCa2+ uptake triggers permeability transition and necrosis. Despite these established roles during acute stress, the involvement of mCa2+ signaling in cardiac adaptations to chronic stress remains poorly defined. Changes in NCLX expression are reported in heart failure (HF) patients and models of cardiac hypertrophy. Therefore, we hypothesized that altered mCa2+ homeostasis contributes to the hypertrophic remodeling of the myocardium that occurs upon a sustained increase in cardiac workload. The impact of mCa2+ flux on cardiac function and remodeling was examined by subjecting mice with cardiomyocyte-specific overexpression (OE) of the mitochondrial Na+/Ca2+ exchanger (NCLX), the primary mediator of mCa2+ efflux, to several well-established models of hypertrophic and non-ischemic HF. Cardiomyocyte NCLX-OE preserved contractile function, prevented hypertrophy and fibrosis, and attenuated maladaptive gene programs in mice subjected to chronic pressure overload. Hypertrophy was attenuated in NCLX-OE mice, prior to any decline in cardiac contractility. NCLX-OE similarly attenuated deleterious cardiac remodeling in mice subjected to chronic neurohormonal stimulation. However, cardiomyocyte NCLX-OE unexpectedly reduced overall survival in mice subjected to severe neurohormonal stress with angiotensin II + phenylephrine. Adenoviral NCLX expression limited mCa2+ accumulation, oxidative metabolism, and de novo protein synthesis during hypertrophic stimulation of cardiomyocytes in vitro. Our findings provide genetic evidence for the contribution of mCa2+ to early pathological remodeling in non-ischemic heart disease, but also highlight a deleterious consequence of increasing mCa2+ efflux when the heart is subjected to extreme, sustained neurohormonal stress.


Asunto(s)
Insuficiencia Cardíaca , Intercambiador de Sodio-Calcio , Animales , Calcio/metabolismo , Señalización del Calcio , Cardiomegalia/metabolismo , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo , Remodelación Ventricular
8.
Acta Neuropathol Commun ; 9(1): 124, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34233766

RESUMEN

The cellular and molecular mechanisms that drive neurodegeneration remain poorly defined. Recent clinical trial failures, difficult diagnosis, uncertain etiology, and lack of curative therapies prompted us to re-examine other hypotheses of neurodegenerative pathogenesis. Recent reports establish that mitochondrial and calcium dysregulation occur early in many neurodegenerative diseases (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, and others. However, causal molecular evidence of mitochondrial and metabolic contributions to pathogenesis remains insufficient. Here we summarize the data supporting the hypothesis that mitochondrial and metabolic dysfunction result from diverse etiologies of neuropathology. We provide a current and comprehensive review of the literature and interpret that defective mitochondrial metabolism is upstream and primary to protein aggregation and other dogmatic hypotheses of NDDs. Finally, we identify gaps in knowledge and propose therapeutic modulation of mCa2+ exchange and mitochondrial function to alleviate metabolic impairments and treat NDDs.


Asunto(s)
Señalización del Calcio/fisiología , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Animales , Humanos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología
9.
Virology ; 553: 135-153, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33278736

RESUMEN

JC virus encodes an important regulatory protein, known as Agnoprotein (Agno). We have recently reported Agno's first protein-interactome with its cellular partners revealing that it targets various cellular networks and organelles, including mitochondria. Here, we report further characterization of the functional consequences of its mitochondrial targeting and demonstrated its co-localization with the mitochondrial networks and with the mitochondrial outer membrane. The mitochondrial targeting sequence (MTS) of Agno and its dimerization domain together play major roles in this targeting. Data also showed alterations in various mitochondrial functions in Agno-positive cells; including a significant reduction in mitochondrial membrane potential, respiration rates and ATP production. In contrast, a substantial increase in ROS production and Ca2+ uptake by the mitochondria were also observed. Finally, findings also revealed a significant decrease in viral replication when Agno MTS was deleted, highlighting a role for MTS in the function of Agno during the viral life cycle.


Asunto(s)
Virus JC/metabolismo , Mitocondrias/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Viroporinas/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Respiración de la Célula , Dimerización , Humanos , Potencial de la Membrana Mitocondrial , Membranas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Viroporinas/química , Replicación Viral
10.
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
11.
Genes (Basel) ; 11(5)2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32443488

RESUMEN

Mitochondria serve as a hub for many cellular processes, including bioenergetics, metabolism, cellular signaling, redox balance, calcium homeostasis, and cell death. The mitochondrial proteome includes over a thousand proteins, encoded by both the mitochondrial and nuclear genomes. The majority (~99%) of proteins are nuclear encoded that are synthesized in the cytosol and subsequently imported into the mitochondria. Within the mitochondria, polypeptides fold and assemble into their native functional form. Mitochondria health and integrity depend on correct protein import, folding, and regulated turnover termed as mitochondrial protein quality control (MPQC). Failure to maintain these processes can cause mitochondrial dysfunction that leads to various pathophysiological outcomes and the commencement of diseases. Here, we summarize the current knowledge about the role of different MPQC regulatory systems such as mitochondrial chaperones, proteases, the ubiquitin-proteasome system, mitochondrial unfolded protein response, mitophagy, and mitochondria-derived vesicles in the maintenance of mitochondrial proteome and health. The proper understanding of mitochondrial protein quality control mechanisms will provide relevant insights to treat multiple human diseases.


Asunto(s)
Núcleo Celular/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Biosíntesis de Proteínas/genética , Humanos , Mitofagia/genética , Complejo de la Endopetidasa Proteasomal/genética , Pliegue de Proteína , Ubiquitina/genética , Respuesta de Proteína Desplegada/genética
12.
Oncotarget ; 11(6): 634-649, 2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-32110282

RESUMEN

Insulin-signalling is an important pathway in multiple cellular functions and organismal ageing across the taxa. A strong association of insulin-signalling with Parkinson's disease (PD) has been proposed but the exact nature of molecular events and genetic associations are yet to be understood. We employed transgenic C. elegans strain harboring human α-synuclein::YFP transgene, towards studying the aggregation pattern of α-synuclein, a PD-associated endpoint, under human insulin (Huminsulin®) treatment and DAF-16/DAF-2 knockdown conditions, independently and in combination. The aggregation was increased when DAF-16 was knocked-down independently or alongwith a co-treatment of Human insulin (HumINS) and decreased when DAF-2 was knocked-down independently or alongwith a co-treatment of HumINS; whereas HumINS treatment per se, reduced the aggregation. Our results depicted that HumINS decreases α-synuclein aggregation via DAF-2/DAF-16 pathway by acting as an antagonist for DAF-2 receptor. Knockdown of reported DAF-2 agonist (INS-6) and antagonists (INS-17 and INS-18) also resulted in a similar effect on α-synuclein aggregation. Further by utilizing bioinformatics tools, we compared the differences between the binding sites of probable agonists and antagonists on DAF-2 including HumINS. Our results suggest that HumINS treatment and DAF-16 expression play a protective role against α-synuclein aggregation and its associated effects.

13.
Nat Commun ; 10(1): 4509, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586055

RESUMEN

Fibroblast to myofibroblast differentiation is crucial for the initial healing response but excessive myofibroblast activation leads to pathological fibrosis. Therefore, it is imperative to understand the mechanisms underlying myofibroblast formation. Here we report that mitochondrial calcium (mCa2+) signaling is a regulatory mechanism in myofibroblast differentiation and fibrosis. We demonstrate that fibrotic signaling alters gating of the mitochondrial calcium uniporter (mtCU) in a MICU1-dependent fashion to reduce mCa2+ uptake and induce coordinated changes in metabolism, i.e., increased glycolysis feeding anabolic pathways and glutaminolysis yielding increased α-ketoglutarate (αKG) bioavailability. mCa2+-dependent metabolic reprogramming leads to the activation of αKG-dependent histone demethylases, enhancing chromatin accessibility in loci specific to the myofibroblast gene program, resulting in differentiation. Our results uncover an important role for the mtCU beyond metabolic regulation and cell death and demonstrate that mCa2+ signaling regulates the epigenome to influence cellular differentiation.


Asunto(s)
Señalización del Calcio/fisiología , Diferenciación Celular/genética , Epigénesis Genética/fisiología , Infarto del Miocardio/patología , Miofibroblastos/fisiología , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Metilación de ADN/fisiología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Epigenoma , Femenino , Fibrosis , Glucólisis/fisiología , Humanos , Ácidos Cetoglutáricos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/etiología , Miocardio/citología , Miocardio/patología , Cultivo Primario de Células
14.
Circulation ; 140(21): 1720-1733, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31533452

RESUMEN

BACKGROUND: The mitochondrial calcium uniporter (mtCU) is an ≈700-kD multisubunit channel residing in the inner mitochondrial membrane required for mitochondrial Ca2+ (mCa2+) uptake. Here, we detail the contribution of MCUB, a paralog of the pore-forming subunit MCU, in mtCU regulation and function and for the first time investigate the relevance of MCUB to cardiac physiology. METHODS: We created a stable MCUB knockout cell line (MCUB-/-) using CRISPR-Cas9n technology and generated a cardiac-specific, tamoxifen-inducible MCUB mutant mouse (CAG-CAT-MCUB x MCM; MCUB-Tg) for in vivo assessment of cardiac physiology and response to ischemia/reperfusion injury. Live-cell imaging and high-resolution spectrofluorometery were used to determine intracellular Ca2+ exchange and size-exclusion chromatography; blue native page and immunoprecipitation studies were used to determine the molecular function and impact of MCUB on the high-molecular-weight mtCU complex. RESULTS: Using genetic gain- and loss-of-function approaches, we show that MCUB expression displaces MCU from the functional mtCU complex and thereby decreases the association of mitochondrial calcium uptake 1 and 2 (MICU1/2) to alter channel gating. These molecular changes decrease MICU1/2-dependent cooperative activation of the mtCU, thereby decreasing mCa2+ uptake. Furthermore, we show that MCUB incorporation into the mtCU is a stress-responsive mechanism to limit mCa2+ overload during cardiac injury. Indeed, overexpression of MCUB is sufficient to decrease infarct size after ischemia/reperfusion injury. However, MCUB incorporation into the mtCU does come at a cost; acute decreases in mCa2+ uptake impair mitochondrial energetics and contractile function. CONCLUSIONS: We detail a new regulatory mechanism to modulate mtCU function and mCa2+ uptake. Our results suggest that MCUB-dependent changes in mtCU stoichiometry are a prominent regulatory mechanism to modulate mCa2+ uptake and cellular physiology.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Sistemas CRISPR-Cas , Canales de Calcio/deficiencia , Canales de Calcio/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/patología , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Contracción Miocárdica , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/patología , Función Ventricular Izquierda
15.
Nat Commun ; 10(1): 3885, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467276

RESUMEN

Impairments in neuronal intracellular calcium (iCa2+) handling may contribute to Alzheimer's disease (AD) development. Metabolic dysfunction and progressive neuronal loss are associated with AD progression, and mitochondrial calcium (mCa2+) signaling is a key regulator of both of these processes. Here, we report remodeling of the mCa2+ exchange machinery in the prefrontal cortex of individuals with AD. In the 3xTg-AD mouse model impaired mCa2+ efflux capacity precedes neuropathology. Neuronal deletion of the mitochondrial Na+/Ca2+ exchanger (NCLX, Slc8b1 gene) accelerated memory decline and increased amyloidosis and tau pathology. Further, genetic rescue of neuronal NCLX in 3xTg-AD mice is sufficient to impede AD-associated pathology and memory loss. We show that mCa2+ overload contributes to AD progression by promoting superoxide generation, metabolic dysfunction and neuronal cell death. These results provide a link between the calcium dysregulation and metabolic dysfunction hypotheses of AD and suggest mCa2+ exchange as potential therapeutic target in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Calcio/metabolismo , Progresión de la Enfermedad , Mitocondrias/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Neuroblastoma/patología , Neuronas/metabolismo , Neuronas/patología , Agregado de Proteínas , Intercambiador de Sodio-Calcio/genética
16.
Cell Rep ; 26(13): 3709-3725.e7, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917323

RESUMEN

Mitochondrial Ca2+ uniporter (MCU)-mediated Ca2+ uptake promotes the buildup of reducing equivalents that fuel oxidative phosphorylation for cellular metabolism. Although MCU modulates mitochondrial bioenergetics, its function in energy homeostasis in vivo remains elusive. Here we demonstrate that deletion of the Mcu gene in mouse liver (MCUΔhep) and in Danio rerio by CRISPR/Cas9 inhibits mitochondrial Ca2+ (mCa2+) uptake, delays cytosolic Ca2+ (cCa2+) clearance, reduces oxidative phosphorylation, and leads to increased lipid accumulation. Elevated hepatic lipids in MCUΔhep were a direct result of extramitochondrial Ca2+-dependent protein phosphatase-4 (PP4) activity, which dephosphorylates AMPK. Loss of AMPK recapitulates hepatic lipid accumulation without changes in MCU-mediated Ca2+ uptake. Furthermore, reconstitution of active AMPK, or PP4 knockdown, enhances lipid clearance in MCUΔhep hepatocytes. Conversely, gain-of-function MCU promotes rapid mCa2+ uptake, decreases PP4 levels, and reduces hepatic lipid accumulation. Thus, our work uncovers an MCU/PP4/AMPK molecular cascade that links Ca2+ dynamics to hepatic lipid metabolism.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Proteínas Mitocondriales/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Canales de Calcio/genética , Células Cultivadas , Femenino , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Quinasas/metabolismo , Pez Cebra
17.
Cell Rep ; 23(13): 3710-3720.e8, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949756

RESUMEN

Mitochondria are composed of many small proteins that control protein synthesis, complex assembly, metabolism, and ion and reactive oxygen species (ROS) handling. We show that a skeletal muscle- and heart-enriched long non-coding RNA, LINC00116, encodes a highly conserved 56-amino-acid microprotein that we named mitoregulin (Mtln). Mtln localizes to the inner mitochondrial membrane, where it binds cardiolipin and influences protein complex assembly. In cultured cells, Mtln overexpression increases mitochondrial membrane potential, respiration rates, and Ca2+ retention capacity while decreasing mitochondrial ROS and matrix-free Ca2+. Mtln-knockout mice display perturbations in mitochondrial respiratory (super)complex formation and activity, fatty acid oxidation, tricarboxylic acid (TCA) cycle enzymes, and Ca2+ retention capacity. Blue-native gel electrophoresis revealed that Mtln co-migrates alongside several complexes, including the complex I assembly module, complex V, and supercomplexes. Under denaturing conditions, Mtln remains in high-molecular-weight complexes, supporting its role as a sticky molecular tether that enhances respiratory efficiency by bolstering protein complex assembly and/or stability.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , ARN Largo no Codificante/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Cardiolipinas/química , Cardiolipinas/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Células HeLa , Humanos , Ratones , Ratones Noqueados , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Oxidación-Reducción , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Alineación de Secuencia
18.
Cell Signal ; 45: 71-80, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29410282

RESUMEN

The accumulation of aggregate-prone proteins is a major representative of many neurological disorders, including Parkinson's disease (PD) wherein the cellular clearance mechanisms, such as the ubiquitin-proteasome and autophagy pathways are impaired. PD, known to be associated with multiple genetic and environmental factors, is characterized by the aggregation of α-synuclein protein and loss of dopaminergic neurons in midbrain. This disease is also associated with other cardiovascular ailments. Herein, we report our findings from studies on the effect of hyper and hypo-osmotic induced toxicity representing hyper and hypotensive condition as an extrinsic epigenetic factor towards modulation of Parkinsonism, using a genetic model Caenorhabditis elegans (C. elegans). Our studies showed that osmotic toxicity had an adverse effect on α-synuclein aggregation, autophagic puncta, lipid content and oxidative stress. Further, we figure that reduced autophagic activity may cause the inefficient clearance of α-synuclein aggregates in osmotic stress toxicity, thereby promoting α-synuclein deposition. Pharmacological induction of autophagy by spermidine proved to be a useful mechanism for protecting cells against the toxic effects of these proteins in such stress conditions. Our studies provide evidence that autophagy is required for the removal of aggregated proteins in these conditions. Studying specific autophagy pathways, we observe that the osmotic stress induced toxicity was largely associated with atg-7 and lgg-1 dependent autophagy pathway, brought together by involvement of mTOR pathway. This represents a unifying pathway to disease in hyper- and hypo-osmotic conditions within PD model of C. elegans.


Asunto(s)
Autofagia , Caenorhabditis elegans , Modelos Animales de Enfermedad , Presión Osmótica , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Caenorhabditis elegans/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Metabolismo de los Lípidos , Proteínas Asociadas a Microtúbulos/metabolismo , Estrés Oxidativo , Espermidina/farmacología , Ubiquitina/metabolismo , alfa-Sinucleína/genética
19.
Mol Neurobiol ; 55(8): 6914-6926, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29363043

RESUMEN

Circular RNAs (circRNAs) are peculiar non-coding RNA molecules which are known to be present across taxa. Considering the body of evidence that establishes critical functions of non-coding RNA molecules, we endeavored to study circRNAs in the context of Parkinson's disease (PD). Employing transgenic C. elegans model of PD, we used RNase R-mediated cleavage of linear RNA followed by divergent primer-based amplifications towards identifying circzip-2, a novel circRNA molecule. We went on to sequence circzip-2 which is synthesized from functionally important gene zip-2. Studying RNAi-induced knockdown conditions of zip-2, we observed a reduced aggregation of α-synuclein protein along with an enhanced lifespan of the worms. We further carried out transcriptome analysis of zip-2 silenced worms, which suggested that zip-2 might be functioning via Daf-16 pathway. Further interaction studies revealed that circzip-2 possibly sponges microRNA molecule miR-60 towards asserting an important role in various processes associated with PD.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Genes de Helminto , Enfermedad de Parkinson/genética , ARN/genética , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Amplificación de Genes , Técnicas de Silenciamiento del Gen , Longevidad , ARN/metabolismo , ARN Circular , Especies Reactivas de Oxígeno/metabolismo , Reproducibilidad de los Resultados , Regulación hacia Arriba/genética , alfa-Sinucleína/metabolismo
20.
J Pharmacol Exp Ther ; 358(3): 441-9, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27353074

RESUMEN

Current therapies are less effective for treating sustained/permanent versus paroxysmal atrial fibrillation (AF). We and others have previously shown that histone deacetylase (HDAC) inhibition reverses structural and electrical atrial remodeling in mice with inducible, paroxysmal-like AF. Here, we hypothesize an important, specific role for class I HDACs in determining structural atrial alterations during sustained AF. The class I HDAC inhibitor N-acetyldinaline [4-(acetylamino)-N-(2-amino-phenyl) benzamide] (CI-994) was administered for 2 weeks (1 mg/kg/day) to Hopx transgenic mice with atrial remodeling and inducible AF and to dogs with atrial tachypacing-induced sustained AF. Class I HDAC inhibition prevented atrial fibrosis and arrhythmia inducibility in mice. Dogs were divided into three groups: 1) sinus rhythm, 2) sustained AF plus vehicle, and 3) sustained AF plus CI-994. In group 3, the time in AF over 2 weeks was reduced by 30% compared with group 2, along with attenuated atrial fibrosis and intra-atrial adipocyte infiltration. Moreover, group 2 dogs had higher atrial and serum inflammatory cytokines, adipokines, and atrial immune cells and adipocytes compared with groups 1 and 3. On the other hand, groups 2 and 3 displayed similar left atrial size, ventricular function, and mitral regurgitation. Importantly, the same histologic alterations found in dogs with sustained AF and reversed by CI-994 were also present in atrial tissue from transplanted patients with chronic AF. This is the first evidence that, in sustained AF, class I HDAC inhibition can reduce the total time of fibrillation, atrial fibrosis, intra-atrial adipocytes, and immune cell infiltration without significant effects on cardiac function.


Asunto(s)
Fibrilación Atrial/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Fenilendiaminas/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/patología , Animales , Fibrilación Atrial/inmunología , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Remodelación Atrial/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Benzamidas , Biomarcadores/metabolismo , Citocinas/sangre , Citocinas/metabolismo , Perros , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Inhibidores de Histona Desacetilasas/uso terapéutico , Ratones , Fenilendiaminas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...