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











Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 41(16): e110476, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35912435

RESUMO

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.


Assuntos
Proteostase , Prótons , Proteases Dependentes de ATP/genética , Proteases Dependentes de ATP/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteoma/metabolismo
2.
Life Sci Alliance ; 3(6)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32321733

RESUMO

Lipid droplets (LDs) are metabolic organelles that store neutral lipids and dynamically respond to changes in energy availability by accumulating or mobilizing triacylglycerols (TAGs). How the plastic behavior of LDs is regulated is poorly understood. Hereditary spastic paraplegia is a central motor axonopathy predominantly caused by mutations in SPAST, encoding the microtubule-severing protein spastin. The spastin-M1 isoform localizes to nascent LDs in mammalian cells; however, the mechanistic significance of this targeting is not fully explained. Here, we show that tightly controlled levels of spastin-M1 are required to inhibit LD biogenesis and TAG accumulation. Spastin-M1 maintains the morphogenesis of the ER when TAG synthesis is prevented, independent from microtubule binding. Moreover, spastin plays a microtubule-dependent role in mediating the dispersion of LDs from the ER upon glucose starvation. Our results reveal a dual role of spastin to shape ER tubules and to regulate LD movement along microtubules, opening new perspectives for the pathogenesis of hereditary spastic paraplegia.


Assuntos
Retículo Endoplasmático/metabolismo , Gotículas Lipídicas/metabolismo , Microtúbulos/metabolismo , Transdução de Sinais/genética , Paraplegia Espástica Hereditária/metabolismo , Espastina/deficiência , Animais , Linhagem Celular Tumoral , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Isoenzimas , Camundongos , Neurônios Motores/metabolismo , Mutação , Paraplegia Espástica Hereditária/genética , Espastina/genética , Transfecção , Triglicerídeos/metabolismo
3.
Glia ; 67(8): 1526-1541, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30989755

RESUMO

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.


Assuntos
Proteases Dependentes de ATP/deficiência , ATPases Associadas a Diversas Atividades Celulares/deficiência , Astrócitos/enzimologia , Cerebelo/enzimologia , Metaloendopeptidases/deficiência , Mitocôndrias/enzimologia , Doenças Neurodegenerativas/enzimologia , Proteases Dependentes de ATP/genética , ATPases Associadas a Diversas Atividades Celulares/genética , Animais , Astrócitos/patologia , Cerebelo/patologia , Modelos Animais de Doenças , Feminino , Inflamação/enzimologia , Inflamação/patologia , Masculino , Metaloendopeptidases/genética , Camundongos Transgênicos , Mitocôndrias/patologia , Doenças Neurodegenerativas/patologia , Células de Purkinje/enzimologia , Células de Purkinje/patologia
4.
Nat Commun ; 10(1): 1566, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30952952

RESUMO

The class 3 phosphoinositide 3-kinase (PI3K) is required for lysosomal degradation by autophagy and vesicular trafficking, assuring nutrient availability. Mitochondrial lipid catabolism is another energy source. Autophagy and mitochondrial metabolism are transcriptionally controlled by nutrient sensing nuclear receptors. However, the class 3 PI3K contribution to this regulation is unknown. We show that liver-specific inactivation of Vps15, the essential regulatory subunit of the class 3 PI3K, elicits mitochondrial depletion and failure to oxidize fatty acids. Mechanistically, transcriptional activity of Peroxisome Proliferator Activated Receptor alpha (PPARα), a nuclear receptor orchestrating lipid catabolism, is blunted in Vps15-deficient livers. We find PPARα repressors Histone Deacetylase 3 (Hdac3) and Nuclear receptor co-repressor 1 (NCoR1) accumulated in Vps15-deficient livers due to defective autophagy. Activation of PPARα or inhibition of Hdac3 restored mitochondrial biogenesis and lipid oxidation in Vps15-deficient hepatocytes. These findings reveal roles for the class 3 PI3K and autophagy in transcriptional coordination of mitochondrial metabolism.


Assuntos
Autofagia/fisiologia , Metabolismo dos Lipídeos , Mitocôndrias/metabolismo , PPAR alfa/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Fenofibrato/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histona Desacetilases/fisiologia , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Correpressor 1 de Receptor Nuclear/genética , Correpressor 1 de Receptor Nuclear/metabolismo , Correpressor 1 de Receptor Nuclear/fisiologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Transcrição Gênica/efeitos dos fármacos , Proteína VPS15 de Distribuição Vacuolar/genética , Proteína VPS15 de Distribuição Vacuolar/metabolismo , Proteína VPS15 de Distribuição Vacuolar/fisiologia
5.
J Cell Biol ; 216(3): 675-693, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28188211

RESUMO

Mitochondria are essential organelles that host crucial metabolic pathways and produce adenosine triphosphate. The mitochondrial proteome is heterogeneous among tissues and can dynamically change in response to different metabolic conditions. Although the transcriptional programs that govern mitochondrial biogenesis and respiratory function are well known, posttranscriptional regulatory mechanisms remain unclear. In this study, we show that the cytosolic RNA-binding protein clustered mitochondria homologue (CLUH) regulates the expression of a mitochondrial protein network supporting key metabolic programs required under nutrient deprivation. CLUH exerts its function by controlling the stability and translation of target messenger RNAs. In the absence of Cluh, mitochondria are severely depleted of crucial enzymes involved in catabolic energy-converting pathways. CLUH preserves oxidative mitochondrial function and glucose homeostasis, thus preventing death at the fetal-neonatal transition. In the adult liver, CLUH ensures maximal respiration capacity and the metabolic response to starvation. Our results shed new light on the posttranscriptional mechanisms controlling the expression of mitochondrial proteins and suggest novel strategies to tailor mitochondrial function to physiological and pathological conditions.


Assuntos
Mitocôndrias/metabolismo , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Citosol/metabolismo , Citosol/fisiologia , Metabolismo Energético/fisiologia , Regulação da Expressão Gênica/fisiologia , Homeostase/fisiologia , Metabolismo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Interferência de RNA/fisiologia
6.
PLoS Genet ; 12(12): e1006463, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27911893

RESUMO

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.


Assuntos
Proteases Dependentes de ATP/genética , Doenças Desmielinizantes/genética , Cabelo/metabolismo , Metaloendopeptidases/genética , Mitocôndrias/genética , Proteases Dependentes de ATP/biossíntese , ATPases Associadas a Diversas Atividades Celulares , Animais , Morte Celular/genética , Sobrevivência Celular/genética , Cabelo/crescimento & desenvolvimento , Humanos , Metaloendopeptidases/biossíntese , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mutação , Bainha de Mielina/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Oligodendroglia/metabolismo , Células de Schwann/metabolismo
7.
Mol Cell ; 64(1): 148-162, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27642048

RESUMO

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.


Assuntos
Canais de Cálcio/metabolismo , Cerebelo/metabolismo , Corpo Estriado/metabolismo , Hipocampo/metabolismo , Metaloendopeptidases/genética , Mitocôndrias/metabolismo , Neurônios/metabolismo , Proteases Dependentes de ATP/genética , Proteases Dependentes de ATP/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Animais , Cálcio/metabolismo , Canais de Cálcio/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Morte Celular , Cerebelo/patologia , Corpo Estriado/patologia , Regulação da Expressão Gênica , Células HEK293 , Hipocampo/patologia , Homeostase/genética , Humanos , Transporte de Íons , Metaloendopeptidases/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/patologia , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Neurônios/patologia , Mapeamento de Interação de Proteínas , Transdução de Sinais
8.
PLoS Genet ; 11(4): e1005149, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25875445

RESUMO

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.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Drosophila/metabolismo , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Motivos de Aminoácidos , Animais , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Retículo Endoplasmático/metabolismo , Mucosa Intestinal/metabolismo , Músculo Esquelético/metabolismo , Neurônios/metabolismo , Transporte Proteico , Triglicerídeos/metabolismo
9.
EMBO J ; 33(19): 2171-87, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25056906

RESUMO

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.


Assuntos
Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/fisiologia , Proteínas de Transporte/metabolismo , Disenteria Bacilar/imunologia , Mitocôndrias/imunologia , Proteínas Mitocondriais/metabolismo , Shigella/imunologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/fisiologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Western Blotting , Caspases/metabolismo , Proliferação de Células , Células Cultivadas , Disenteria Bacilar/microbiologia , Disenteria Bacilar/patologia , Feminino , Hepatócitos/imunologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Técnicas Imunoenzimáticas , Integrases/metabolismo , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/imunologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Shigella/patogenicidade , Transdução de Sinais , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
10.
EMBO J ; 33(9): 1011-26, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24681487

RESUMO

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.


Assuntos
Proteases Dependentes de ATP/genética , Mitocôndrias/metabolismo , Proteínas tau/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Acetilcisteína/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Embrião de Mamíferos , Sistema de Sinalização das MAP Quinases/genética , Metaloproteases/genética , Metaloproteases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/genética , Espécies Reativas de Oxigênio/farmacologia
11.
J Clin Invest ; 122(11): 4048-58, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23041622

RESUMO

Mutations in the AFG3L2 gene have been linked to spinocerebellar ataxia type 28 and spastic ataxia-neuropathy syndrome in humans; however, the pathogenic mechanism is still unclear. AFG3L2 encodes a subunit of the mitochondrial m-AAA protease, previously implicated in quality control of misfolded inner mitochondrial membrane proteins and in regulatory functions via processing of specific substrates. Here, we used a conditional Afg3l2 mouse model that allows restricted deletion of the gene in Purkinje cells (PCs) to shed light on the pathogenic cascade in the neurons mainly affected in the human diseases. We demonstrate a cell-autonomous requirement of AFG3L2 for survival of PCs. Examination of PCs prior to neurodegeneration revealed fragmentation and altered distribution of mitochondria in the dendritic tree, indicating that abnormal mitochondrial dynamics is an early event in the pathogenic process. Moreover, PCs displayed features pointing to defects in mitochondrially encoded respiratory chain subunits at early stages. To unravel the underlying mechanism, we examined a constitutive knockout of Afg3l2, which revealed a decreased rate of mitochondrial protein synthesis associated with impaired mitochondrial ribosome assembly. We therefore propose that defective mitochondrial protein synthesis, leading to early-onset fragmentation of the mitochondrial network, is a central causative factor in AFG3L2-related neurodegeneration.


Assuntos
Proteases Dependentes de ATP/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/biossíntese , Proteínas do Tecido Nervoso/metabolismo , Biossíntese de Proteínas/fisiologia , Proteases Dependentes de ATP/genética , ATPases Associadas a Diversas Atividades Celulares , Animais , Sobrevivência Celular , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Espasticidade Muscular/genética , Espasticidade Muscular/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Atrofia Óptica/genética , Atrofia Óptica/metabolismo , Células de Purkinje , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo , Degenerações Espinocerebelares/genética , Degenerações Espinocerebelares/metabolismo
12.
PLoS One ; 7(5): e36337, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563492

RESUMO

BACKGROUND: Hereditary spastic paraplegia defines a group of genetically heterogeneous diseases characterized by weakness and spasticity of the lower limbs owing to retrograde degeneration of corticospinal axons. One autosomal recessive form of the disease is caused by mutation in the SPG7 gene. Paraplegin, the product of SPG7, is a component of the m-AAA protease, a high molecular weight complex that resides in the mitochondrial inner membrane, and performs crucial quality control and biogenesis functions in mitochondria. PRINCIPAL FINDINGS: Here we show the existence in the mouse of a novel isoform of paraplegin, which we name paraplegin-2, encoded by alternative splicing of Spg7 through usage of an alternative first exon. Paraplegin-2 lacks the mitochondrial targeting sequence, and is identical to the mature mitochondrial protein. Remarkably, paraplegin-2 is targeted to the endoplasmic reticulum. We find that paraplegin-2 exposes the catalytic domains to the lumen of the endoplasmic reticulum. Moreover, endogenous paraplegin-2 accumulates in microsomal fractions prepared from mouse brain and retina. Finally, we show that the previously generated mouse model of Spg7-linked hereditary spastic paraplegia is an isoform-specific knock-out, in which mitochondrial paraplegin is specifically ablated, while expression of paraplegin-2 is retained. CONCLUSIONS/SIGNIFICANCE: These data suggest a possible additional role of AAA proteases outside mitochondria and open the question of their implication in neurodegeneration.


Assuntos
Processamento Alternativo , Retículo Endoplasmático/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Proteases Dependentes de ATP/genética , Proteases Dependentes de ATP/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Animais , Western Blotting , Encéfalo/metabolismo , Células Cultivadas , Éxons/genética , Etiquetas de Sequências Expressas , Humanos , Células Híbridas , Camundongos , Camundongos Knockout , Microscopia Confocal , Microssomos/metabolismo , Mitocôndrias/metabolismo , Neurônios Motores/citologia , Neuroblastoma/patologia , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/metabolismo
13.
PLoS Genet ; 7(10): e1002325, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22022284

RESUMO

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.


Assuntos
Proteases Dependentes de ATP/genética , Encéfalo/anormalidades , Metaloendopeptidases/genética , Mitocôndrias/enzimologia , Paraplegia Espástica Hereditária/genética , Degenerações Espinocerebelares/genética , ATPases Associadas a Diversas Atividades Celulares , Adolescente , Sequência de Aminoácidos , Animais , Encéfalo/patologia , Criança , Diagnóstico Diferencial , Exoma/genética , Genótipo , Células HeLa , Homozigoto , Humanos , Masculino , Metaloendopeptidases/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Paraplegia , Dobramento de Proteína , Irmãos , Paraplegia Espástica Hereditária/patologia , Ataxias Espinocerebelares/congênito , Degenerações Espinocerebelares/patologia , Leveduras/genética
14.
Brain ; 134(Pt 6): 1808-28, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21576111

RESUMO

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.


Assuntos
DNA Helicases/metabolismo , Fator 8 de Crescimento de Fibroblasto/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/citologia , RNA Helicases/metabolismo , Transdução de Sinais/fisiologia , Animais , Caspase 3/metabolismo , Morte Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , DNA Helicases/genética , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Humanos , Camundongos , Enzimas Multifuncionais , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , RNA Helicases/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção/métodos , Tretinoína/farmacologia
15.
Biochim Biophys Acta ; 1797(1): 1-10, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19664590

RESUMO

Fine tuning of integrated mitochondrial functions is essential in neurons and rationalizes why mitochondrial dysfunction plays an important pathogenic role in neurodegeneration. Mitochondria can contribute to neuronal cell death and axonal dysfunction through a plethora of mechanisms, including low ATP levels, increased reactive oxygen species, defective calcium regulation, and impairment of dynamics and transport. Recently, mitochondrial proteases in the inner mitochondrial membrane have emerged as culprits in several human neurodegenerative diseases. Mitochondrial proteases degrade misfolded and non-assembled polypeptides, thus performing quality control surveillance in the organelle. Moreover, they regulate the activity of specific substrates by mediating essential processing steps. Mitochondrial proteases may be directly involved in neurodegenerative diseases, as recently shown for the m-AAA protease, or may regulate crucial mitochondrial molecules, such as OPA1, which in turn is implicated in human disease. The mitochondrial proteases HTRA2 and PARL increase the susceptibility of neurons to apoptotic cell death. Here we review our current knowledge on how disturbances of the mitochondrial proteolytic system affect neuronal maintenance and axonal function.


Assuntos
Metaloendopeptidases/metabolismo , Mitocôndrias/fisiologia , Degeneração Neural/fisiopatologia , Adenosina Trifosfatases/metabolismo , Animais , Apoptose/fisiologia , Axônios/enzimologia , Axônios/fisiologia , Morte Celular , Cerebelo/enzimologia , Cerebelo/fisiopatologia , Proteínas Fúngicas/metabolismo , Humanos , Metaloendopeptidases/genética , Camundongos , Mutação de Sentido Incorreto , Degeneração Neural/genética , Atrofia Óptica/genética , Saccharomyces cerevisiae/enzimologia
16.
J Cell Biol ; 187(7): 1023-36, 2009 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-20038678

RESUMO

Mitochondrial fusion depends on the dynamin-like guanosine triphosphatase OPA1, whose activity is controlled by proteolytic cleavage. Dysfunction of mitochondria induces OPA1 processing and results in mitochondrial fragmentation, allowing the selective removal of damaged mitochondria. In this study, we demonstrate that two classes of metallopeptidases regulate OPA1 cleavage in the mitochondrial inner membrane: isoenzymes of the adenosine triphosphate (ATP)-dependent matrix AAA (ATPase associated with diverse cellular activities [m-AAA]) protease, variable assemblies of the conserved subunits paraplegin, AFG3L1 and -2, and the ATP-independent peptidase OMA1. Functionally redundant isoenzymes of the m-AAA protease ensure the balanced accumulation of long and short isoforms of OPA1 required for mitochondrial fusion. The loss of AFG3L2 in mouse tissues, down-regulation of AFG3L1 and -2 in mouse embryonic fibroblasts, or the expression of a dominant-negative AFG3L2 variant in human cells decreases the stability of long OPA1 isoforms and induces OPA1 processing by OMA1. Moreover, cleavage by OMA1 causes the accumulation of short OPA1 variants if mitochondrial DNA is depleted or mitochondrial activities are impaired. Our findings link distinct peptidases to constitutive and induced OPA1 processing and shed new light on the pathogenesis of neurodegenerative disorders associated with mutations in m-AAA protease subunits.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Metaloendopeptidases/fisiologia , Metaloproteases/fisiologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/fisiologia , Proteases Dependentes de ATP , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/metabolismo , Animais , Células Cultivadas , Estabilidade Enzimática , GTP Fosfo-Hidrolases/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Isoenzimas/fisiologia , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Metaloproteases/genética , Metaloproteases/metabolismo , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Interferência de RNA
17.
Hum Mol Genet ; 18(11): 2001-13, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19289403

RESUMO

The mitochondrial m-AAA protease has a crucial role in axonal development and maintenance. Human mitochondria possess two m-AAA protease isoenzymes: a hetero-oligomeric complex, composed of paraplegin and AFG3L2 (Afg3 like 2), and a homo-oligomeric AFG3L2 complex. Loss of function of paraplegin (encoded by the SPG7 gene) causes hereditary spastic paraplegia, a disease characterized by retrograde degeneration of cortical motor axons. Spg7(-/-) mice show a late-onset degeneration of long spinal and peripheral axons with accumulation of abnormal mitochondria. In contrast, Afg3l2(Emv66/Emv66) mutant mice, lacking the AFG3L2 protein, are affected by a severe neuromuscular phenotype, due to defects in motor axon development. The role of the homo-oligomeric m-AAA protease and the extent of cooperation and redundancy between the two isoenzymes in adult neurons are still unclear. Here we report an early-onset severe neurological phenotype in Spg7(-/-) Afg3l2(Emv66/+) mice, characterized by loss of balance, tremor and ataxia. Spg7(-/-) Afg3l2(Emv66/+) mice display acceleration and worsening of the axonopathy observed in paraplegin-deficient mice. In addition, they show prominent cerebellar degeneration with loss of Purkinje cells and parallel fibers, and reactive astrogliosis. Mitochondria from affected tissues are prone to lose mt-DNA and have unstable respiratory complexes. At late stages, neurons contain structural abnormal mitochondria defective in COX-SDH reaction. Our data demonstrate genetic interaction between the m-AAA isoenzymes and suggest that different neuronal populations have variable thresholds of susceptibility to reduced levels of the m-AAA protease. Moreover, they implicate impaired mitochondrial proteolysis as a novel pathway in cerebellar degeneration.


Assuntos
Adenosina Trifosfatases/metabolismo , Doenças Cerebelares/enzimologia , Metaloendopeptidases/metabolismo , Paraplegia Espástica Hereditária/enzimologia , Proteases Dependentes de ATP , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Animais , Axônios/enzimologia , Axônios/fisiologia , Doenças Cerebelares/genética , Doenças Cerebelares/fisiopatologia , Modelos Animais de Doenças , Feminino , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/enzimologia , Mitocôndrias/genética , Ligação Proteica , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/fisiopatologia
18.
J Neurochem ; 108(5): 1277-88, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19141076

RESUMO

Hereditary spastic paraplegia (HSP) is characterized by weakness and spasticity of the lower limbs, owing to degeneration of corticospinal axons. The most common form is due to heterozygous mutations in the SPG4 gene, encoding spastin, a microtubule (MT)-severing protein. Here, we show that neurite growth in immortalized and primary neurons responds in pleiotropic ways to changes in spastin levels. Spastin depletion alters the development of primary hippocampal neurons leading to abnormal neuron morphology, dystrophic neurites, and axonal growth defects. By live imaging with End-Binding Protein 3-Fluorescent Green Protein (EB3-GFP), a MT plus-end tracking protein, we ascertained that the assembly rate of MTs is reduced when spastin is down-regulated. Spastin over-expression at high levels strongly suppresses neurite maintenance, while slight spastin up-regulation using an endogenous promoter enhances neurite branching and elongation. Spastin severing activity is exerted preferentially on stable acetylated and detyrosinated MTs. We further show that SPG4 nonsense or splice site mutations found in hereditary spastic paraplegia patients result in reduced spastin levels, supporting haploinsufficiency as the molecular cause of the disease. Our study reveals that SPG4 is a dosage-sensitive gene, and broadens the understanding of the role of spastin in neurite growth and MT dynamics.


Assuntos
Adenosina Trifosfatases/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mutação/genética , Neuritos/fisiologia , Paraplegia Espástica Hereditária/genética , Adenosina Trifosfatases/metabolismo , Animais , Encéfalo/citologia , Células Cultivadas , Embrião de Mamíferos , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Neuritos/efeitos dos fármacos , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Espastina
20.
BMC Biol ; 6: 31, 2008 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-18613979

RESUMO

BACKGROUND: Mutations in SPG4 cause the most common form of autosomal dominant hereditary spastic paraplegia, a neurodegenerative disease characterized by weakness and spasticity of the lower limbs due to degeneration of the corticospinal tract. SPG4 encodes spastin, a microtubule-severing ATPase belonging to the AAA family. Two isoforms of spastin, 68 and 60 kDa, respectively, are variably abundant in tissues, show different subcellular localizations and interact with distinct molecules. The isoforms arise through alternative initiation of translation from two AUG codons in exon 1; however, it is unclear how regulation of their expression may be achieved. RESULTS: We present data that rule out the hypothesis that a cap-independent mechanism may be involved in the translation of the 60-kDa spastin isoform. Instead, we provide evidence for a complex transcriptional regulation of SPG4 that involves both a TATA-less ubiquitous promoter and a cryptic promoter in exon 1. The cryptic promoter covers the 5'-UTR and overlaps with the coding region of the gene. By using promoter-less constructs in various experimental settings, we found that the cryptic promoter is active in HeLa, HEK293 and motoneuronal NSC34 cells but not in SH-SY-5Y neuroblastoma cells. We showed that the cryptic promoter directs the synthesis of a SPG4 transcript that contains a shorter 5'-UTR and translates the 60-kDa spastin isoform selectively. Two polymorphisms (S44L and P45Q), leading to an early onset severe form of hereditary spastic paraplegia when present in heterozygosity with a mutant allele, fall a few nucleotides downstream of the novel transcriptional start site, opening up the possibility that they may exert their modifier effect at the transcriptional level. We provide evidence that at least one of them decreases the activity of the cryptic promoter in luciferase assays. CONCLUSION: We identified a cryptic promoter in exon 1 of the SPG4 gene that selectively drives the expression of the 60-kDa spastin isoform in a tissue-regulated manner. These data may have implications for the understanding of the biology of spastin and the pathogenic basis of hereditary spastic paraplegia.


Assuntos
Adenosina Trifosfatases/genética , Éxons/genética , Regulação Enzimológica da Expressão Gênica , Regiões Promotoras Genéticas/genética , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Células HeLa , Humanos , Isoenzimas/genética , Camundongos , Fenótipo , Polimorfismo Genético , Espastina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA