Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
EMBO J ; 41(16): e110476, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35912435

RESUMEN

Mitochondria adapt to different energetic demands reshaping their proteome. Mitochondrial proteases are emerging as key regulators of these adaptive processes. Here, we use a multiproteomic approach to demonstrate the regulation of the m-AAA protease AFG3L2 by the mitochondrial proton gradient, coupling mitochondrial protein turnover to the energetic status of mitochondria. We identify TMBIM5 (previously also known as GHITM or MICS1) as a Ca2+ /H+ exchanger in the mitochondrial inner membrane, which binds to and inhibits the m-AAA protease. TMBIM5 ensures cell survival and respiration, allowing Ca2+ efflux from mitochondria and limiting mitochondrial hyperpolarization. Persistent hyperpolarization, however, triggers degradation of TMBIM5 and activation of the m-AAA protease. The m-AAA protease broadly remodels the mitochondrial proteome and mediates the proteolytic breakdown of respiratory complex I to confine ROS production and oxidative damage in hyperpolarized mitochondria. TMBIM5 thus integrates mitochondrial Ca2+ signaling and the energetic status of mitochondria with protein turnover rates to reshape the mitochondrial proteome and adjust the cellular metabolism.


Asunto(s)
Proteostasis , Protones , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteoma/metabolismo
2.
EMBO J ; 39(9): e102731, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32149416

RESUMEN

Mitochondria house anabolic and catabolic processes that must be balanced and adjusted to meet cellular demands. The RNA-binding protein CLUH (clustered mitochondria homolog) binds mRNAs of nuclear-encoded mitochondrial proteins and is highly expressed in the liver, where it regulates metabolic plasticity. Here, we show that in primary hepatocytes, CLUH coalesces in specific ribonucleoprotein particles that define the translational fate of target mRNAs, such as Pcx, Hadha, and Hmgcs2, to match nutrient availability. Moreover, CLUH granules play signaling roles, by recruiting mTOR kinase and the RNA-binding proteins G3BP1 and G3BP2. Upon starvation, CLUH regulates translation of Hmgcs2, involved in ketogenesis, inhibits mTORC1 activation and mitochondrial anabolic pathways, and promotes mitochondrial turnover, thus allowing efficient reprograming of metabolic function. In the absence of CLUH, a mitophagy block causes mitochondrial clustering that is rescued by rapamycin treatment or depletion of G3BP1 and G3BP2. Our data demonstrate that metabolic adaptation of liver mitochondria to nutrient availability depends on a compartmentalized CLUH-dependent post-transcriptional mechanism that controls both mTORC1 and G3BP signaling and ensures survival.


Asunto(s)
Mitocondrias Hepáticas/fisiología , Proteínas Mitocondriales/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Animales , Células COS , Chlorocebus aethiops , Gránulos Citoplasmáticos/genética , Gránulos Citoplasmáticos/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Mitofagia , Proteínas de Unión al ARN/genética
3.
Chemistry ; 30(30): e202400808, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38506349

RESUMEN

Lipid droplet (LD) degradation provides metabolic energy and important building blocks for various cellular processes. The two major LD degradation pathways include autophagy (lipophagy), which involves delivery of LDs to autolysosomes, and lipolysis, which is mediated by lipases. While abnormalities in LD degradation are associated with various pathological disorders, our understanding of lipophagy is still rudimentary. In this study, we describe the development of a lipophilic dye containing two fluorophores, one of which is pH-sensitive and the other pH-stable. We further demonstrate that this "Lipo-Fluddy" can be used to visualize and quantify lipophagy in living cells, in an easily applicable and protein label-free approach. After estimating the ability of compound candidates to penetrate LDs, we synthesized several BODIPY and (pH-switchable) rhodol dyes, whose fluorescence properties (incl. their photophysical compatibility) were analyzed. Of three Lipo-Fluddy dyes synthesized, one exhibited the desired properties and allowed observation of lipophagy by fluorescence microscopy. Also, this dye proved to be non-toxic and suitable for the examination of various cell lines. Moreover, a method was developed to quantify the lipophagy process using flow cytometry, which could be applied in the future in the identification of lipophagy-related genes or in the screening of potential drugs against lipophagy-related diseases.


Asunto(s)
Autofagia , Compuestos de Boro , Colorantes Fluorescentes , Gotas Lipídicas , Colorantes Fluorescentes/química , Concentración de Iones de Hidrógeno , Humanos , Gotas Lipídicas/química , Gotas Lipídicas/metabolismo , Compuestos de Boro/química , Microscopía Fluorescente , Células HeLa , Lipólisis
4.
Brain ; 146(10): 4117-4131, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37086482

RESUMEN

Hereditary spastic paraplegia is a neurological condition characterized by predominant axonal degeneration in long spinal tracts, leading to weakness and spasticity in the lower limbs. The nicotinamide adenine dinucleotide (NAD+)-consuming enzyme SARM1 has emerged as a key executioner of axonal degeneration upon nerve transection and in some neuropathies. An increase in the nicotinamide mononucleotide/NAD+ ratio activates SARM1, causing catastrophic NAD+ depletion and axonal degeneration. However, the role of SARM1 in the pathogenesis of hereditary spastic paraplegia has not been investigated. Here, we report an enhanced mouse model for hereditary spastic paraplegia caused by mutations in SPG7. The eSpg7 knockout mouse carries a deletion in both Spg7 and Afg3l1, a redundant homologue expressed in mice but not in humans. The eSpg7 knockout mice recapitulate the phenotypic features of human patients, showing progressive symptoms of spastic-ataxia and degeneration of axons in the spinal cord as well as the cerebellum. We show that the lack of SPG7 rewires the mitochondrial proteome in both tissues, leading to an early onset decrease in mito-ribosomal subunits and a remodelling of mitochondrial solute carriers and transporters. To interrogate mechanisms leading to axonal degeneration in this mouse model, we explored the involvement of SARM1. Deletion of SARM1 delays the appearance of ataxic signs, rescues mitochondrial swelling and axonal degeneration of cerebellar granule cells and dampens neuroinflammation in the cerebellum. The loss of SARM1 also prevents endoplasmic reticulum abnormalities in long spinal cord axons, but does not halt the degeneration of these axons. Our data thus reveal a neuron-specific interplay between SARM1 and mitochondrial dysfunction caused by lack of SPG7 in hereditary spastic paraplegia.


Asunto(s)
Paraplejía Espástica Hereditaria , Animales , Humanos , Ratones , Proteínas del Dominio Armadillo/genética , ATPasas Asociadas con Actividades Celulares Diversas , Axones/patología , Cerebelo , Proteínas del Citoesqueleto/genética , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , NAD , Paraplejía Espástica Hereditaria/genética
5.
Mol Cell ; 64(1): 148-162, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27642048

RESUMEN

Mutations in subunits of mitochondrial m-AAA proteases in the inner membrane cause neurodegeneration in spinocerebellar ataxia (SCA28) and hereditary spastic paraplegia (HSP7). m-AAA proteases preserve mitochondrial proteostasis, mitochondrial morphology, and efficient OXPHOS activity, but the cause for neuronal loss in disease is unknown. We have determined the neuronal interactome of m-AAA proteases in mice and identified a complex with C2ORF47 (termed MAIP1), which counteracts cell death by regulating the assembly of the mitochondrial Ca2+ uniporter MCU. While MAIP1 assists biogenesis of the MCU subunit EMRE, the m-AAA protease degrades non-assembled EMRE and ensures efficient assembly of gatekeeper subunits with MCU. Loss of the m-AAA protease results in accumulation of constitutively active MCU-EMRE channels lacking gatekeeper subunits in neuronal mitochondria and facilitates mitochondrial Ca2+ overload, mitochondrial permeability transition pore opening, and neuronal death. Together, our results explain neuronal loss in m-AAA protease deficiency by deregulated mitochondrial Ca2+ homeostasis.


Asunto(s)
Canales de Calcio/metabolismo , Cerebelo/metabolismo , Cuerpo Estriado/metabolismo , Hipocampo/metabolismo , Metaloendopeptidasas/genética , Mitocondrias/metabolismo , Neuronas/metabolismo , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Calcio/metabolismo , Canales de Calcio/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Muerte Celular , Cerebelo/patología , Cuerpo Estriado/patología , Regulación de la Expresión Génica , Células HEK293 , Hipocampo/patología , Homeostasis/genética , Humanos , Transporte Iónico , Metaloendopeptidasas/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Neuronas/patología , Mapeo de Interacción de Proteínas , Transducción de Señal
6.
Cell Mol Life Sci ; 79(6): 334, 2022 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-35652974

RESUMEN

Mitochondria in animals are associated with development, as well as physiological and pathological behaviors. Several conserved mitochondrial genes exist between plants and higher eukaryotes. Yet, the similarities in mitochondrial function between plant and animal species is poorly understood. Here, we show that FMT (FRIENDLY MITOCHONDRIA) from Arabidopsis thaliana, a highly conserved homolog of the mammalian CLUH (CLUSTERED MITOCHONDRIA) gene family encoding mitochondrial proteins associated with developmental alterations and adult physiological and pathological behaviors, affects whole plant morphology and development under both stressed and normal growth conditions. FMT was found to regulate mitochondrial morphology and dynamics, germination, and flowering time. It also affects leaf expansion growth, salt stress responses and hyponastic behavior, including changes in speed of hyponastic movements. Strikingly, Cluh± heterozygous knockout mice also displayed altered locomotive movements, traveling for shorter distances and had slower average and maximum speeds in the open field test. These observations indicate that homologous mitochondrial genes may play similar roles and affect homologous functions in both plants and animals.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Animales , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Locomoción , Mamíferos/metabolismo , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo
7.
Crit Rev Biochem Mol Biol ; 53(6): 652-666, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30741581

RESUMEN

Mitochondria are dynamic and plastic organelles, which flexibly adapt morphology, ATP production, and metabolic function to meet extrinsic challenges and demands. Regulation of mitochondrial biogenesis is essential during development and in adult life to survive stress and pathological insults, and is achieved not only by increasing mitochondrial mass, but also by remodeling the organellar proteome, metabolome, and lipidome. In the last decade, the post-transcriptional regulation of the expression of nuclear-encoded mitochondrial proteins has emerged as a fast, flexible, and powerful mechanism to shape mitochondrial function and coordinate it with other cellular processes. At the heart of post-transcriptional responses are a number of RNA-binding proteins that specifically bind mRNAs encoding mitochondrial proteins and define their fate, by influencing transcript maturation, stability, translation, and localization. Thus, RNA-binding proteins provide a uniquely complex regulatory code that orchestrates mitochondrial function during physiological and pathological conditions.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , ARN Mensajero/metabolismo , ARN Mitocondrial/metabolismo , Proteínas de Unión al ARN/farmacocinética , Animales , Humanos , Mitocondrias/genética , Proteínas Mitocondriales/genética , ARN Mensajero/genética , ARN Mitocondrial/genética , Proteínas de Unión al ARN/genética
8.
Glia ; 67(8): 1526-1541, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30989755

RESUMEN

Mitochondrial dysfunction causes neurodegeneration but whether impairment of mitochondrial homeostasis in astrocytes contributes to this pathological process remains largely unknown. The m-AAA protease exerts quality control and regulatory functions crucial for mitochondrial homeostasis. AFG3L2, which encodes one of the subunits of the m-AAA protease, is mutated in spinocerebellar ataxia SCA28 and in infantile syndromes characterized by spastic-ataxia, epilepsy and premature death. Here, we investigate the role of Afg3l2 and its redundant homologue Afg3l1 in the Bergmann glia (BG), radial astrocytes of the cerebellum that have functional connections with Purkinje cells (PC) and regulate glutamate homeostasis. We show that astrocyte-specific deletion of Afg3l2 in the mouse leads to late-onset motor impairment and to degeneration of BG, which display aberrant morphology, altered expression of the glutamate transporter EAAT2, and a reactive inflammatory signature. The neurological and glial phenotypes are drastically exacerbated when astrocytes lack both Afg31l and Afg3l2, and therefore, are totally depleted of the m-AAA protease. Moreover, mitochondrial stress responses and necroptotic markers are induced in the cerebellum. In both mouse models, targeted BG show a fragmented mitochondrial network and loss of mitochondrial cristae, but no signs of respiratory dysfunction. Importantly, astrocyte-specific deficiency of Afg3l1 and Afg3l2 triggers secondary morphological degeneration and electrophysiological changes in PCs, thus demonstrating a non-cell-autonomous role of glia in neurodegeneration. We propose that astrocyte dysfunction amplifies both neuroinflammation and glutamate excitotoxicity in patients carrying mutations in AFG3L2, leading to a vicious circle that contributes to neuronal death.


Asunto(s)
Proteasas ATP-Dependientes/deficiencia , ATPasas Asociadas con Actividades Celulares Diversas/deficiencia , Astrocitos/enzimología , Cerebelo/enzimología , Metaloendopeptidasas/deficiencia , Mitocondrias/enzimología , Enfermedades Neurodegenerativas/enzimología , Proteasas ATP-Dependientes/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , Animales , Astrocitos/patología , Cerebelo/patología , Modelos Animales de Enfermedad , Femenino , Inflamación/enzimología , Inflamación/patología , Masculino , Metaloendopeptidasas/genética , Ratones Transgénicos , Mitocondrias/patología , Enfermedades Neurodegenerativas/patología , Células de Purkinje/enzimología , Células de Purkinje/patología
9.
PLoS Genet ; 12(12): e1006463, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27911893

RESUMEN

The m-AAA protease preserves proteostasis of the inner mitochondrial membrane. It ensures a functional respiratory chain, by controlling the turnover of respiratory complex subunits and allowing mitochondrial translation, but other functions in mitochondria are conceivable. Mutations in genes encoding subunits of the m-AAA protease have been linked to various neurodegenerative diseases in humans, such as hereditary spastic paraplegia and spinocerebellar ataxia. While essential functions of the m-AAA protease for neuronal survival have been established, its role in adult glial cells remains enigmatic. Here, we show that deletion of the highly expressed subunit AFG3L2 in mature mouse oligodendrocytes provokes early-on mitochondrial fragmentation and swelling, as previously shown in neurons, but causes only late-onset motor defects and myelin abnormalities. In contrast, total ablation of the m-AAA protease, by deleting both Afg3l2 and its paralogue Afg3l1, triggers progressive motor dysfunction and demyelination, owing to rapid oligodendrocyte cell death. Surprisingly, the mice showed premature hair greying, caused by progressive loss of melanoblasts that share a common developmental origin with Schwann cells and are targeted in our experiments. Thus, while both neurons and glial cells are dependant on the m-AAA protease for survival in vivo, complete ablation of the complex is necessary to trigger death of oligodendrocytes, hinting to cell-autonomous thresholds of vulnerability to m-AAA protease deficiency.


Asunto(s)
Proteasas ATP-Dependientes/genética , Enfermedades Desmielinizantes/genética , Cabello/metabolismo , Metaloendopeptidasas/genética , Mitocondrias/genética , Proteasas ATP-Dependientes/biosíntesis , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Muerte Celular/genética , Supervivencia Celular/genética , Cabello/crecimiento & desarrollo , Humanos , Metaloendopeptidasas/biosíntesis , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Mutación , Vaina de Mielina/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Células de Schwann/metabolismo
10.
EMBO J ; 33(9): 1011-26, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24681487

RESUMEN

The m-AAA protease subunit AFG3L2 is involved in degradation and processing of substrates in the inner mitochondrial membrane. Mutations in AFG3L2 are associated with spinocerebellar ataxia SCA28 in humans and impair axonal development and neuronal survival in mice. The loss of AFG3L2 causes fragmentation of the mitochondrial network. However, the pathogenic mechanism of neurodegeneration in the absence of AFG3L2 is still unclear. Here, we show that depletion of AFG3L2 leads to a specific defect of anterograde transport of mitochondria in murine cortical neurons. We observe similar transport deficiencies upon loss of AFG3L2 in OMA1-deficient neurons, indicating that they are not caused by OMA1-mediated degradation of the dynamin-like GTPase OPA1 and inhibition of mitochondrial fusion. Treatment of neurons with antioxidants, such as N-acetylcysteine or vitamin E, or decreasing tau levels in axons restored mitochondrial transport in AFG3L2-depleted neurons. Consistently, tau hyperphosphorylation and activation of ERK kinases are detected in mouse neurons postnatally deleted for Afg3l2. We propose that reactive oxygen species signaling leads to cytoskeletal modifications that impair mitochondrial transport in neurons lacking AFG3L2.


Asunto(s)
Proteasas ATP-Dependientes/genética , Mitocondrias/metabolismo , Proteínas tau/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Acetilcisteína/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Embrión de Mamíferos , Sistema de Señalización de MAP Quinasas/genética , Metaloproteasas/genética , Metaloproteasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/genética , Especies Reactivas de Oxígeno/farmacología
11.
EMBO J ; 33(19): 2171-87, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25056906

RESUMEN

The X-linked inhibitor of apoptosis protein (XIAP) is a potent caspase inhibitor, best known for its anti-apoptotic function in cancer. During apoptosis, XIAP is antagonized by SMAC, which is released from the mitochondria upon caspase-mediated activation of BID. Recent studies suggest that XIAP is involved in immune signaling. Here, we explore XIAP as an important mediator of an immune response against the enteroinvasive bacterium Shigella flexneri, both in vitro and in vivo. Our data demonstrate for the first time that Shigella evades the XIAP-mediated immune response by inducing the BID-dependent release of SMAC from the mitochondria. Unlike apoptotic stimuli, Shigella activates the calpain-dependent cleavage of BID to trigger the release of SMAC, which antagonizes the inflammatory action of XIAP without inducing apoptosis. Our results demonstrate how the cellular death machinery can be subverted by an invasive pathogen to ensure bacterial colonization.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Proteínas Portadoras/metabolismo , Disentería Bacilar/inmunología , Mitocondrias/inmunología , Proteínas Mitocondriales/metabolismo , Shigella/inmunología , Proteína Inhibidora de la Apoptosis Ligada a X/fisiología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Western Blotting , Caspasas/metabolismo , Proliferación Celular , Células Cultivadas , Disentería Bacilar/microbiología , Disentería Bacilar/patología , Femenino , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/inmunología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Shigella/patogenicidad , Transducción de Señal , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
12.
PLoS Genet ; 11(4): e1005149, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25875445

RESUMEN

Mutations in SPAST, encoding spastin, are the most common cause of autosomal dominant hereditary spastic paraplegia (HSP). HSP is characterized by weakness and spasticity of the lower limbs, owing to progressive retrograde degeneration of the long corticospinal axons. Spastin is a conserved microtubule (MT)-severing protein, involved in processes requiring rearrangement of the cytoskeleton in concert to membrane remodeling, such as neurite branching, axonal growth, midbody abscission, and endosome tubulation. Two isoforms of spastin are synthesized from alternative initiation codons (M1 and M87). We now show that spastin-M1 can sort from the endoplasmic reticulum (ER) to pre- and mature lipid droplets (LDs). A hydrophobic motif comprised of amino acids 57 through 86 of spastin was sufficient to direct a reporter protein to LDs, while mutation of arginine 65 to glycine abolished LD targeting. Increased levels of spastin-M1 expression reduced the number but increased the size of LDs. Expression of a mutant unable to bind and sever MTs caused clustering of LDs. Consistent with these findings, ubiquitous overexpression of Dspastin in Drosophila led to bigger and less numerous LDs in the fat bodies and increased triacylglycerol levels. In contrast, Dspastin overexpression increased LD number when expressed specifically in skeletal muscles or nerves. Downregulation of Dspastin and expression of a dominant-negative variant decreased LD number in Drosophila nerves, skeletal muscle and fat bodies, and reduced triacylglycerol levels in the larvae. Moreover, we found reduced amount of fat stores in intestinal cells of worms in which the spas-1 homologue was either depleted by RNA interference or deleted. Taken together, our data uncovers an evolutionarily conserved role of spastin as a positive regulator of LD metabolism and open up the possibility that dysfunction of LDs in axons may contribute to the pathogenesis of HSP.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Drosophila/metabolismo , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Secuencias de Aminoácidos , Animales , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Retículo Endoplásmico/metabolismo , Mucosa Intestinal/metabolismo , Músculo Esquelético/metabolismo , Neuronas/metabolismo , Transporte de Proteínas , Triglicéridos/metabolismo
13.
EMBO J ; 31(6): 1336-49, 2012 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22354038

RESUMEN

Neuronal survival critically depends on the integrity and functionality of mitochondria. A hierarchical system of cellular surveillance mechanisms protects mitochondria against stress, monitors mitochondrial damage and ensures the selective removal of dysfunctional mitochondrial proteins or organelles. Mitochondrial proteases emerge as central regulators that coordinate different quality control (QC) pathways within an interconnected network of mechanisms. A failure of this system causes neuronal loss in a steadily increasing number of neurodegenerative disorders, which include Parkinson's disease, spinocerebellar ataxia, spastic paraplegia and peripheral neuropathies. Here, we will discuss the role of the mitochondrial QC network for neuronal survival and neurodegeneration.


Asunto(s)
Apoptosis/fisiología , Supervivencia Celular/fisiología , Mitocondrias/metabolismo , Neuronas/citología , Animales , Humanos , Proteínas Mitocondriales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Péptido Hidrolasas/metabolismo
14.
PLoS Genet ; 8(11): e1003021, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144624

RESUMEN

Fusion and fission of mitochondria maintain the functional integrity of mitochondria and protect against neurodegeneration, but how mitochondrial dysfunctions trigger neuronal loss remains ill-defined. Prohibitins form large ring complexes in the inner membrane that are composed of PHB1 and PHB2 subunits and are thought to function as membrane scaffolds. In Caenorhabditis elegans, prohibitin genes affect aging by moderating fat metabolism and energy production. Knockdown experiments in mammalian cells link the function of prohibitins to membrane fusion, as they were found to stabilize the dynamin-like GTPase OPA1 (optic atrophy 1), which mediates mitochondrial inner membrane fusion and cristae morphogenesis. Mutations in OPA1 are associated with dominant optic atrophy characterized by the progressive loss of retinal ganglion cells, highlighting the importance of OPA1 function in neurons. Here, we show that neuron-specific inactivation of Phb2 in the mouse forebrain causes extensive neurodegeneration associated with behavioral impairments and cognitive deficiencies. We observe early onset tau hyperphosphorylation and filament formation in the hippocampus, demonstrating a direct link between mitochondrial defects and tau pathology. Loss of PHB2 impairs the stability of OPA1, affects mitochondrial ultrastructure, and induces the perinuclear clustering of mitochondria in hippocampal neurons. A destabilization of the mitochondrial genome and respiratory deficiencies manifest in aged neurons only, while the appearance of mitochondrial morphology defects correlates with tau hyperphosphorylation in the absence of PHB2. These results establish an essential role of prohibitin complexes for neuronal survival in vivo and demonstrate that OPA1 stability, mitochondrial fusion, and the maintenance of the mitochondrial genome in neurons depend on these scaffolding proteins. Moreover, our findings establish prohibitin-deficient mice as a novel genetic model for tau pathologies caused by a dysfunction of mitochondria and raise the possibility that tau pathologies are associated with other neurodegenerative disorders caused by deficiencies in mitochondrial dynamics.


Asunto(s)
Mitocondrias , Enfermedades Neurodegenerativas , Neuronas , Atrofia Óptica Autosómica Dominante , Proteínas Represoras , Animales , Apoptosis , Proteínas de Caenorhabditis elegans , Genoma Mitocondrial , Fusión de Membrana , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Membranas Mitocondriales/metabolismo , Morfogénesis , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Atrofia Óptica Autosómica Dominante/genética , Atrofia Óptica Autosómica Dominante/metabolismo , Fosforilación , Prohibitinas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
15.
PLoS Genet ; 7(10): e1002325, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22022284

RESUMEN

We report an early onset spastic ataxia-neuropathy syndrome in two brothers of a consanguineous family characterized clinically by lower extremity spasticity, peripheral neuropathy, ptosis, oculomotor apraxia, dystonia, cerebellar atrophy, and progressive myoclonic epilepsy. Whole-exome sequencing identified a homozygous missense mutation (c.1847G>A; p.Y616C) in AFG3L2, encoding a subunit of an m-AAA protease. m-AAA proteases reside in the mitochondrial inner membrane and are responsible for removal of damaged or misfolded proteins and proteolytic activation of essential mitochondrial proteins. AFG3L2 forms either a homo-oligomeric isoenzyme or a hetero-oligomeric complex with paraplegin, a homologous protein mutated in hereditary spastic paraplegia type 7 (SPG7). Heterozygous loss-of-function mutations in AFG3L2 cause autosomal-dominant spinocerebellar ataxia type 28 (SCA28), a disorder whose phenotype is strikingly different from that of our patients. As defined in yeast complementation assays, the AFG3L2(Y616C) gene product is a hypomorphic variant that exhibited oligomerization defects in yeast as well as in patient fibroblasts. Specifically, the formation of AFG3L2(Y616C) complexes was impaired, both with itself and to a greater extent with paraplegin. This produced an early-onset clinical syndrome that combines the severe phenotypes of SPG7 and SCA28, in additional to other "mitochondrial" features such as oculomotor apraxia, extrapyramidal dysfunction, and myoclonic epilepsy. These findings expand the phenotype associated with AFG3L2 mutations and suggest that AFG3L2-related disease should be considered in the differential diagnosis of spastic ataxias.


Asunto(s)
Proteasas ATP-Dependientes/genética , Encéfalo/anomalías , Metaloendopeptidasas/genética , Mitocondrias/enzimología , Paraplejía Espástica Hereditaria/genética , Degeneraciones Espinocerebelosas/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adolescente , Secuencia de Aminoácidos , Animales , Encéfalo/patología , Niño , Diagnóstico Diferencial , Exoma/genética , Genotipo , Células HeLa , Homocigoto , Humanos , Masculino , Metaloendopeptidasas/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación Missense , Paraplejía , Pliegue de Proteína , Hermanos , Paraplejía Espástica Hereditaria/patología , Ataxias Espinocerebelosas/congénito , Degeneraciones Espinocerebelosas/patología , Levaduras/genética
16.
Life Sci Alliance ; 7(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38782601

RESUMEN

Complexes of ERLIN1 and ERLIN2 (ER lipid raft-associated 1 and 2) form large ring-like cup-shaped structures on the endoplasmic reticulum (ER) membrane and serve as platforms to bind cholesterol and E3 ubiquitin ligases, potentially defining functional nanodomains. Here, we show that ERLIN scaffolds mediate the interaction between the full-length isoform of TMUB1 (transmembrane and ubiquitin-like domain-containing 1) and RNF170 (RING finger protein 170). We identify a luminal N-terminal conserved region in TMUB1 and RNF170, which is required for this interaction. Three-dimensional modelling shows that this conserved motif binds the stomatin/prohibitin/flotillin/HflKC domain of two adjacent ERLIN subunits at different interfaces. Protein variants that preclude these interactions have been previously linked to hereditary spastic paraplegia. Using omics-based approaches in combination with phenotypic characterization of HeLa cells lacking both ERLINs, we demonstrate a role of ERLIN scaffolds in limiting cholesterol esterification, thereby favouring cholesterol transport from the ER to the Golgi apparatus and regulating Golgi morphology and the secretory pathway.


Asunto(s)
Colesterol , Retículo Endoplásmico , Aparato de Golgi , Proteínas de la Membrana , Vías Secretoras , Ubiquitina-Proteína Ligasas , Humanos , Proteínas de la Membrana/metabolismo , Colesterol/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Retículo Endoplásmico/metabolismo , Células HeLa , Aparato de Golgi/metabolismo , Unión Proteica , Proteínas del Tejido Nervioso
17.
Sci Adv ; 10(22): eadn2050, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38809982

RESUMEN

Transporting and translating mRNAs in axons is crucial for neuronal viability. Local synthesis of nuclear-encoded mitochondrial proteins protects long-lived axonal mitochondria from damage; however, the regulatory factors involved are largely unknown. We show that CLUH, which binds mRNAs encoding mitochondrial proteins, prevents peripheral neuropathy and motor deficits in the mouse. CLUH is enriched in the growth cone of developing spinal motoneurons and is required for their growth. The lack of CLUH affects the abundance of target mRNAs and the corresponding mitochondrial proteins more prominently in axons, leading to ATP deficits in the growth cone. CLUH interacts with ribosomal subunits, translation initiation, and ribosome recycling components and preserves axonal translation. Overexpression of the ribosome recycling factor ABCE1 rescues the mRNA and translation defects, as well as the growth cone size, in CLUH-deficient motoneurons. Thus, we demonstrate a role for CLUH in mitochondrial quality control and translational regulation in axons, which is essential for their development and long-term integrity and function.


Asunto(s)
Axones , Mitocondrias , Neuronas Motoras , Enfermedades del Sistema Nervioso Periférico , Biosíntesis de Proteínas , Animales , Neuronas Motoras/metabolismo , Mitocondrias/metabolismo , Axones/metabolismo , Ratones , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Conos de Crecimiento/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Ratones Noqueados
18.
Brain ; 134(Pt 6): 1808-28, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21576111

RESUMEN

Senataxin is encoded by the SETX gene and is mainly involved in two different neurodegenerative diseases, the dominant juvenile form of amyotrophic lateral sclerosis type 4 and a recessive form of ataxia with oculomotor apraxia type 2. Based on protein homology, senataxin is predicted to be a putative DNA/RNA helicase, while senataxin interactors from patients' lymphoblast cell lines suggest a possible involvement of the protein in different aspects of RNA metabolism. Except for an increased sensitivity to oxidative DNA damaging agents shown by some ataxia with neuropathy patients' cell lines, no data are available about possible functional consequences of dominant SETX mutations and no studies address the function of senataxin in neurons. To start elucidating the physiological role of senataxin in neurons and how disease-causing mutations in this protein lead to neurodegeneration, we analysed the effect of senataxin on neuronal differentiation in primary hippocampal neurons and retinoic acid-treated P19 cells by modulating the expression levels of wild-type senataxin and three different dominant mutant forms of the protein. Wild-type senataxin overexpression was required and sufficient to trigger neuritogenesis and protect cells from apoptosis during differentiation. These actions were reversed by silencing of senataxin. In contrast, overexpression of the dominant mutant forms did not affect the regular differentiation process in primary hippocampal neurons. Analysis of the cellular pathways leading to neuritogenesis and cytoprotection revealed a role of senataxin in modulating the expression levels and signalling activity of fibroblast growth factor 8. Silencing of senataxin reduced, while overexpression enhanced, fibroblast growth factor 8 expression levels and the phosphorylation of related target kinases and effector proteins. The effects of senataxin overexpression were prevented when fibroblast growth factor 8 signalling was inhibited, while exogenous fibroblast growth factor 8 reversed the effects of senataxin silencing. Overall, these results reveal a key role of senataxin in neuronal differentiation through the fibroblast growth factor 8 signalling and provide initial molecular bases to explain the neurodegeneration associated with loss-of-function mutations in senataxin found in recessive ataxia. The lack of effect on neuritogenesis observed with the overexpression of the dominant mutant forms of senataxin apparently excludes a dominant negative effect of these mutants while favouring haploinsufficiency as the pathogenic mechanism implicated in the amyotrophic lateral sclerosis 4-related degenerative condition. Alternatively, a different protein function, other than the one involved in neuritogenesis, may be implicated in these dominant degenerative processes.


Asunto(s)
ADN Helicasas/metabolismo , Factor 8 de Crecimiento de Fibroblastos/farmacología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/citología , ARN Helicasas/metabolismo , Transducción de Señal/fisiología , Animales , Caspasa 3/metabolismo , Muerte Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , ADN Helicasas/genética , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Humanos , Ratones , Enzimas Multifuncionales , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/patología , Neuronas/efectos de los fármacos , ARN Helicasas/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección/métodos , Tretinoina/farmacología
19.
Elife ; 112022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35559794

RESUMEN

Proliferating cells undergo metabolic changes in synchrony with cell cycle progression and cell division. Mitochondria provide fuel, metabolites, and ATP during different phases of the cell cycle, however it is not completely understood how mitochondrial function and the cell cycle are coordinated. CLUH (clustered mitochondria homolog) is a post-transcriptional regulator of mRNAs encoding mitochondrial proteins involved in oxidative phosphorylation and several metabolic pathways. Here, we show a role of CLUH in regulating the expression of astrin, which is involved in metaphase to anaphase progression, centrosome integrity, and mTORC1 inhibition. We find that CLUH binds both the SPAG5 mRNA and its product astrin, and controls the synthesis and the stability of the full-length astrin-1 isoform. We show that CLUH interacts with astrin-1 specifically during interphase. Astrin-depleted cells show mTORC1 hyperactivation and enhanced anabolism. On the other hand, cells lacking CLUH show decreased astrin levels and increased mTORC1 signaling, but cannot sustain anaplerotic and anabolic pathways. In absence of CLUH, cells fail to grow during G1, and progress faster through the cell cycle, indicating dysregulated matching of growth, metabolism, and cell cycling. Our data reveal a role of CLUH in coupling growth signaling pathways and mitochondrial metabolism with cell cycle progression.


Asunto(s)
Mitocondrias , Proteínas Mitocondriales , Azul Alcián , Ciclo Celular , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Metafase , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Fenazinas , Fenotiazinas , ARN Mensajero/metabolismo , Resorcinoles
20.
Cell Rep ; 38(7): 110370, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35172139

RESUMEN

The transition between quiescence and activation in neural stem and progenitor cells (NSPCs) is coupled with reversible changes in energy metabolism with key implications for lifelong NSPC self-renewal and neurogenesis. How this metabolic plasticity is ensured between NSPC activity states is unclear. We find that a state-specific rewiring of the mitochondrial proteome by the i-AAA peptidase YME1L is required to preserve NSPC self-renewal. YME1L controls the abundance of numerous mitochondrial substrates in quiescent NSPCs, and its deletion activates a differentiation program characterized by broad metabolic changes causing the irreversible shift away from a fatty-acid-oxidation-dependent state. Conditional Yme1l deletion in adult NSPCs in vivo results in defective self-renewal and premature differentiation, ultimately leading to NSPC pool depletion. Our results disclose an important role for YME1L in coordinating the switch between metabolic states of NSPCs and suggest that NSPC fate is regulated by compartmentalized changes in protein network dynamics.


Asunto(s)
Células Madre Adultas/metabolismo , Autorrenovación de las Células , Metaloendopeptidasas/metabolismo , Mitocondrias/enzimología , Células-Madre Neurales/metabolismo , Células Madre Adultas/citología , Animales , Proliferación Celular , Ciclo del Ácido Cítrico , Ácidos Grasos/metabolismo , Eliminación de Gen , Metaloendopeptidasas/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/ultraestructura , Células-Madre Neurales/citología , Nucleótidos/metabolismo , Oxidación-Reducción , Proteolisis , Proteoma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA