Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell ; 64(1): 148-162, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27642048

ABSTRACT

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.


Subject(s)
Calcium Channels/metabolism , Cerebellum/metabolism , Corpus Striatum/metabolism , Hippocampus/metabolism , Metalloendopeptidases/genetics , Mitochondria/metabolism , Neurons/metabolism , ATP-Dependent Proteases/genetics , ATP-Dependent Proteases/metabolism , ATPases Associated with Diverse Cellular Activities , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Death , Cerebellum/pathology , Corpus Striatum/pathology , Gene Expression Regulation , HEK293 Cells , Hippocampus/pathology , Homeostasis/genetics , Humans , Ion Transport , Metalloendopeptidases/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Neurons/pathology , Protein Interaction Mapping , Signal Transduction
2.
J Cell Biol ; 212(2): 157-66, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26783299

ABSTRACT

Proteolytic cleavage of the dynamin-like guanosine triphosphatase OPA1 in mitochondria is emerging as a central regulatory hub that determines mitochondrial morphology under stress and in disease. Stress-induced OPA1 processing by OMA1 triggersmitochondrial fragmentation, which is associated with mitophagy and apoptosis in vitro. Here, we identify OMA1 as a critical regulator of neuronal survival in vivo and demonstrate that stress-induced OPA1 processing by OMA1 promotes neuronal death and neuroinflammatory responses. Using mice lacking prohibitin membrane scaffolds as a model of neurodegeneration, we demonstrate that additional ablation of Oma1 delays neuronal loss and prolongs lifespan. This is accompanied by the accumulation of fusion-active, long OPA1 forms, which stabilize the mitochondrial genome but do not preserve mitochondrial cristae or respiratory chain supercomplex assembly in prohibitin-depleted neurons. Thus, long OPA1 forms can promote neuronal survival independently of cristae shape, whereas stress-induced OMA1 activation and OPA1 cleavage limit mitochondrial fusion and promote neuronal death.


Subject(s)
GTP Phosphohydrolases/metabolism , Metalloproteases/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Nerve Degeneration , Animals , Apoptosis , Brain/metabolism , Brain/pathology , Cell Respiration , Cell Survival/genetics , Cells, Cultured , DNA, Mitochondrial/metabolism , Gene Deletion , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Nerve Degeneration/genetics , Neurons/metabolism , Neurons/pathology , Prohibitins , Repressor Proteins/metabolism
3.
Oncotarget ; 6(39): 41750-65, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26497683

ABSTRACT

We previously described diaryl trifluorothiazoline compound 1a (hereafter referred to as fluorizoline) as a first-in-class small molecule that induces p53-independent apoptosis in a wide range of tumor cell lines. Fluorizoline directly binds to prohibitin 1 and 2 (PHBs), two proteins involved in the regulation of several cellular processes, including apoptosis. Here we demonstrate that fluorizoline-induced apoptosis is mediated by PHBs, as cells depleted of these proteins are highly resistant to fluorizoline treatment. In addition, BAX and BAK are necessary for fluorizoline-induced cytotoxic effects, thereby proving that apoptosis occurs through the intrinsic pathway. Expression analysis revealed that fluorizoline induced the upregulation of Noxa and Bim mRNA levels, which was not observed in PHB-depleted MEFs. Finally, Noxa(-/-)/Bim(-/-) MEFs and NOXA-downregulated HeLa cells were resistant to fluorizoline-induced apoptosis. All together, these findings show that fluorizoline requires PHBs to execute the mitochondrial apoptotic pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Fibroblasts/drug effects , Membrane Proteins/metabolism , Mitochondria/drug effects , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Thiazoles/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Fibroblasts/metabolism , Fibroblasts/pathology , HT29 Cells , HeLa Cells , Humans , Jurkat Cells , Membrane Proteins/genetics , Mice , Mitochondria/metabolism , Mitochondria/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Prohibitins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Cells, Cultured , Up-Regulation
4.
Cell Metab ; 20(1): 158-71, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24856930

ABSTRACT

Prohibitins form large protein and lipid scaffolds in the inner membrane of mitochondria that are required for mitochondrial morphogenesis, neuronal survival, and normal lifespan. Here, we have defined the interactome of PHB2 in mitochondria and identified DNAJC19, mutated in dilated cardiomyopathy with ataxia, as binding partner of PHB complexes. We observed impaired cell growth, defective cristae morphogenesis, and similar transcriptional responses in the absence of either DNAJC19 or PHB2. The loss of PHB/DNAJC19 complexes affects cardiolipin acylation and leads to the accumulation of cardiolipin species with altered acyl chains. Similar defects occur in cells lacking the transacylase tafazzin, which is mutated in Barth syndrome. Our experiments suggest that PHB/DNAJC19 membrane domains regulate cardiolipin remodeling by tafazzin and explain similar clinical symptoms in two inherited cardiomyopathies by an impaired cardiolipin metabolism in mitochondrial membranes.


Subject(s)
Cardiolipins/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Acyltransferases , Amino Acid Sequence , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Line , HEK293 Cells , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Metalloendopeptidases/metabolism , Mice , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Molecular Sequence Data , Prohibitins , RNA Interference , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
5.
EMBO J ; 33(9): 1011-26, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24681487

ABSTRACT

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.


Subject(s)
ATP-Dependent Proteases/genetics , Mitochondria/metabolism , tau Proteins/metabolism , ATPases Associated with Diverse Cellular Activities , Acetylcysteine/pharmacology , Animals , Biological Transport/drug effects , Biological Transport/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryo, Mammalian , MAP Kinase Signaling System/genetics , Metalloproteases/genetics , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphorylation/genetics , Reactive Oxygen Species/pharmacology
6.
EMBO J ; 33(6): 578-93, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24550258

ABSTRACT

The dynamic network of mitochondria fragments under stress allowing the segregation of damaged mitochondria and, in case of persistent damage, their selective removal by mitophagy. Mitochondrial fragmentation upon depolarisation of mitochondria is brought about by the degradation of central components of the mitochondrial fusion machinery. The OMA1 peptidase mediates the degradation of long isoforms of the dynamin-like GTPase OPA1 in the inner membrane. Here, we demonstrate that OMA1-mediated degradation of OPA1 is a general cellular stress response. OMA1 is constitutively active but displays strongly enhanced activity in response to various stress insults. We identify an amino terminal stress-sensor domain of OMA1, which is only present in homologues of higher eukaryotes and which modulates OMA1 proteolysis and activation. OMA1 activation is associated with its autocatalyic degradation, which initiates from both termini of OMA1 and results in complete OMA1 turnover. Autocatalytic proteolysis of OMA1 ensures the reversibility of the response and allows OPA1-mediated mitochondrial fusion to resume upon alleviation of stress. This differentiated stress response maintains the functional integrity of mitochondria and contributes to cell survival.


Subject(s)
Enzyme Activation/physiology , GTP Phosphohydrolases/metabolism , Metalloproteases/metabolism , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Models, Biological , Stress, Physiological/physiology , Animals , Centrifugation, Density Gradient , Electrophoresis, Polyacrylamide Gel , Fibroblasts , Immunoblotting , Metalloproteases/genetics , Mice , Mice, Knockout , Microscopy, Fluorescence , Mitochondrial Proteins/genetics
7.
J Clin Invest ; 124(4): 1552-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569455

ABSTRACT

Familial Alzheimer's disease (FAD) is characterized by autosomal dominant heritability and early disease onset. Mutations in the gene encoding presenilin-1 (PS1) are found in approximately 80% of cases of FAD, with some of these patients presenting cerebellar damage with amyloid plaques and ataxia with unclear pathophysiology. A Colombian kindred carrying the PS1-E280A mutation is the largest known cohort of PS1-FAD patients. Here, we investigated PS1-E280A-associated cerebellar dysfunction and found that it occurs early in PS1-E208A carriers, while cerebellar signs are highly prevalent in patients with dementia. Postmortem analysis of cerebella of PS1-E280A carrier revealed greater Purkinje cell (PC) loss and more abnormal mitochondria compared with controls. In PS1-E280A tissue, ER/mitochondria tethering was impaired, Ca2+ channels IP3Rs and CACNA1A were downregulated, and Ca2+-dependent mitochondrial transport proteins MIRO1 and KIF5C were reduced. Accordingly, expression of PS1-E280A in a neuronal cell line altered ER/mitochondria tethering and transport compared with that in cells expressing wild-type PS1. In a murine model of PS1-FAD, animals exhibited mild ataxia and reduced PC simple spike activity prior to cerebellar ß-amyloid deposition. Our data suggest that impaired calcium homeostasis and mitochondrial dysfunction in PS1-FAD PCs reduces their activity and contributes to motor coordination deficits prior to Aß aggregation and dementia. We propose that PS1-E280A affects both Ca2+ homeostasis and Aß precursor processing, leading to FAD and neurodegeneration.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Calcium/metabolism , Cerebellum/metabolism , Mutation, Missense , Presenilin-1/genetics , Presenilin-1/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amino Acid Substitution , Amyloid beta-Protein Precursor/metabolism , Animals , Case-Control Studies , Cell Line , Cerebellum/pathology , Disease Models, Animal , Endoplasmic Reticulum/pathology , Female , Genes, Dominant , Heterozygote , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Models, Neurological , Purkinje Cells/metabolism
8.
PLoS Genet ; 8(11): e1003021, 2012.
Article in English | MEDLINE | ID: mdl-23144624

ABSTRACT

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.


Subject(s)
Mitochondria , Neurodegenerative Diseases , Neurons , Optic Atrophy, Autosomal Dominant , Repressor Proteins , Animals , Apoptosis , Caenorhabditis elegans Proteins , Genome, Mitochondrial , Membrane Fusion , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membranes/metabolism , Morphogenesis , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Optic Atrophy, Autosomal Dominant/genetics , Optic Atrophy, Autosomal Dominant/metabolism , Phosphorylation , Prohibitins , Repressor Proteins/genetics , Repressor Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...