Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
Cell ; 174(4): 982-998.e20, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29909982

RESUMEN

The diversity of cell types and regulatory states in the brain, and how these change during aging, remains largely unknown. We present a single-cell transcriptome atlas of the entire adult Drosophila melanogaster brain sampled across its lifespan. Cell clustering identified 87 initial cell clusters that are further subclustered and validated by targeted cell-sorting. Our data show high granularity and identify a wide range of cell types. Gene network analyses using SCENIC revealed regulatory heterogeneity linked to energy consumption. During aging, RNA content declines exponentially without affecting neuronal identity in old brains. This single-cell brain atlas covers nearly all cells in the normal brain and provides the tools to study cellular diversity alongside other Drosophila and mammalian single-cell datasets in our unique single-cell analysis platform: SCope (http://scope.aertslab.org). These results, together with SCope, allow comprehensive exploration of all transcriptional states of an entire aging brain.


Asunto(s)
Envejecimiento , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Redes Reguladoras de Genes , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Drosophila melanogaster/fisiología , Femenino , Perfilación de la Expresión Génica , Masculino
2.
EMBO J ; 41(6): e110002, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35199384

RESUMEN

The use of animals in neuroscience and biomedical research remains controversial. Policy is built around the "3R" principle of "Refining, Reducing and Replacing" animal experiments, and across the globe, different initiatives stimulate the use of animal-free methods. Based on an extensive literature screen to map the development and adoption of animal-free methods in Alzheimer's and Parkinson's disease research, we find that at least two in three examined studies rely on animals or on animal-derived models. Among the animal-free studies, the relative contribution of innovative models that may replace animal experiments is limited. We argue that the distinction between animal research and alternative models presents a false dichotomy, as the role and scientific value of both animal and animal-free approaches are intertwined. Calls to halt all animal experiments appear premature, as insufficient non-animal-based alternatives are available and their development lags behind. In light of this, we highlight the need for objective, unprejudiced monitoring, and more robust performance indicators of animal-free approaches.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Parkinson , Animales , Modelos Animales
3.
Cell ; 145(1): 117-32, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21458671

RESUMEN

Exchange of proteins at sorting endosomes is not only critical to numerous signaling pathways but also to receptor-mediated signaling and to pathogen entry into cells; however, how this process is regulated in synaptic vesicle cycling remains unexplored. In this work, we present evidence that loss of function of a single neuronally expressed GTPase activating protein (GAP), Skywalker (Sky) facilitates endosomal trafficking of synaptic vesicles at Drosophila neuromuscular junction boutons, chiefly by controlling Rab35 GTPase activity. Analyses of genetic interactions with the ESCRT machinery as well as chimeric ubiquitinated synaptic vesicle proteins indicate that endosomal trafficking facilitates the replacement of dysfunctional synaptic vesicle components. Consequently, sky mutants harbor a larger readily releasable pool of synaptic vesicles and show a dramatic increase in basal neurotransmitter release. Thus, the trafficking of vesicles via endosomes uncovered using sky mutants provides an elegant mechanism by which neurons may regulate synaptic vesicle rejuvenation and neurotransmitter release.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Endosomas/metabolismo , Membranas Intracelulares/metabolismo , Mutación , Sistema Nervioso/metabolismo , Unión Neuromuscular/metabolismo , Neuronas/citología , Neuronas/metabolismo , Neurotransmisores/metabolismo , Proteínas de Unión al GTP rab/genética
4.
Mol Cell ; 71(5): 689-702.e9, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30193096

RESUMEN

Hsp90 is an essential chaperone that guards proteome integrity and amounts to 2% of cellular protein. We now find that Hsp90 also has the ability to directly interact with and deform membranes via an evolutionarily conserved amphipathic helix. Using a new cell-free system and in vivo measurements, we show this amphipathic helix allows exosome release by promoting the fusion of multivesicular bodies (MVBs) with the plasma membrane. We dissect the relationship between Hsp90 conformation and membrane-deforming function and show that mutations and drugs that stabilize the open Hsp90 dimer expose the helix and allow MVB fusion, while these effects are blocked by the closed state. Hence, we structurally separated the Hsp90 membrane-deforming function from its well-characterized chaperone activity, and we show that this previously unrecognized function is required for exosome release.


Asunto(s)
Membrana Celular/metabolismo , Exosomas/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Animales , Sistema Libre de Células/metabolismo , Drosophila/metabolismo , Femenino , Masculino , Chaperonas Moleculares/metabolismo , Cuerpos Multivesiculares/metabolismo , Unión Proteica/fisiología , Conformación Proteica
5.
EMBO J ; 40(17): e106914, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34313336

RESUMEN

The interphase nuclear envelope (NE) is extensively remodeled during nuclear pore complex (NPC) insertion. How this remodeling occurs and why it requires Torsin ATPases, which also regulate lipid metabolism, remains poorly understood. Here, we show that Drosophila Torsin (dTorsin) affects lipid metabolism via the NEP1R1-CTDNEP1 phosphatase and the Lipin phosphatidic acid (PA) phosphatase. This includes that Torsins remove NEP1R1-CTDNEP1 from the NE in fly and mouse cells, leading to subsequent Lipin exclusion from the nucleus. NEP1R1-CTDNEP1 downregulation also restores nuclear pore membrane fusion in post-mitotic dTorsinKO fat body cells. However, dTorsin-associated nuclear pore defects do not correlate with lipidomic abnormalities and are not resolved by silencing of Lipin. Further testing confirmed that membrane fusion continues in cells with hyperactivated Lipin. It also led to the surprising finding that excessive PA metabolism inhibits recruitment of the inner ring complex Nup35 subunit, resulting in elongated channel-like structures in place of mature nuclear pores. We conclude that the NEP1R1-CTDNEP1 phosphatase affects interphase NPC biogenesis by lipid-dependent and lipid-independent mechanisms, explaining some of the pleiotropic effects of Torsins.


Asunto(s)
Proteínas de Drosophila/metabolismo , Poro Nuclear/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Cuerpo Adiposo/citología , Cuerpo Adiposo/metabolismo , Metabolismo de los Lípidos , Fusión de Membrana , Fosfoproteínas Fosfatasas/genética
6.
Proc Natl Acad Sci U S A ; 119(45): e2203499119, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36322761

RESUMEN

Correct spatiotemporal distribution of organelles and vesicles is crucial for healthy cell functioning and is regulated by intracellular transport mechanisms. Controlled transport of bulky mitochondria is especially important in polarized cells such as neurons that rely on these organelles to locally produce energy and buffer calcium. Mitochondrial transport requires and depends on microtubules that fill much of the available axonal space. How mitochondrial transport is affected by their position within the microtubule bundles is not known. Here, we found that anterograde transport, driven by kinesin motors, is susceptible to the molecular conformation of tubulin in neurons both in vitro and in vivo. Anterograde velocities negatively correlate with the density of elongated tubulin dimers like guanosine triphosphate (GTP)-tubulin. The impact of the tubulin conformation depends primarily on where a mitochondrion is positioned, either within or at the rim of microtubule bundle. Increasing elongated tubulin levels lowers the number of motile anterograde mitochondria within the microtubule bundle and increases anterograde transport speed at the microtubule bundle rim. We demonstrate that the increased kinesin velocity and density on microtubules consisting of elongated dimers add to the increased mitochondrial dynamics. Our work indicates that the molecular conformation of tubulin contributes to the regulation of mitochondrial motility and as such to the local distribution of mitochondria along axons.


Asunto(s)
Transporte Axonal , Tubulina (Proteína) , Tubulina (Proteína)/metabolismo , Cinesinas , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Axones/metabolismo , Conformación Molecular
7.
Neurobiol Dis ; 163: 105595, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34933093

RESUMEN

Synapses are critical for neuronal communication and brain function. To maintain neuronal homeostasis, synapses rely on autophagy. Autophagic alterations cause neurodegeneration and synaptic dysfunction is a feature in neurodegenerative diseases. In Parkinson's disease (PD), where the loss of synapses precedes dopaminergic neuron loss, various PD-causative proteins are involved in the regulation of autophagy. So far only a few factors regulating autophagy at the synapse have been identified and the molecular mechanisms underlying autophagy at the synapse is only partially understood. Here, we describe Endophilin-B (EndoB) as a novel player in the regulation of synaptic autophagy in health and disease. We demonstrate that EndoB is required for autophagosome biogenesis at the synapse, whereas the loss of EndoB blocks the autophagy induction promoted by the PD mutation LRRK2G2019S. We show that EndoB is required to prevent neuronal loss. Moreover, loss of EndoB in the Drosophila visual system leads to an increase in synaptic contacts between photoreceptor terminals and their post-synaptic synapses. These data confirm the role of autophagy in synaptic contact formation and neuronal survival.


Asunto(s)
Aciltransferasas/metabolismo , Autofagia/genética , Neuronas Dopaminérgicas/metabolismo , Proteínas de Drosophila/metabolismo , Degeneración Nerviosa/metabolismo , Sinapsis/metabolismo , Aciltransferasas/genética , Animales , Animales Modificados Genéticamente , Neuronas Dopaminérgicas/patología , Drosophila , Proteínas de Drosophila/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Sinapsis/genética
8.
EMBO J ; 37(18)2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30065071

RESUMEN

Parkinson's disease, the second most common neurodegenerative disorder, affects millions of people globally. There is no cure, and its prevalence will double by 2030. In recent years, numerous causative genes and risk factors for Parkinson's disease have been identified and more than half appear to function at the synapse. Subtle synaptic defects are thought to precede blunt neuronal death, but the mechanisms that are dysfunctional at synapses are only now being unraveled. Here, we review recent work and propose a model where different Parkinson proteins interact in a cell compartment-specific manner at the synapse where these proteins regulate endocytosis and autophagy. While this field is only recently emerging, the work suggests that the loss of synaptic homeostasis may contribute to neurodegeneration and is a key player in Parkinson's disease.


Asunto(s)
Autofagia , Endocitosis , Homeostasis , Enfermedad de Parkinson/metabolismo , Sinapsis/metabolismo , Humanos , Enfermedad de Parkinson/patología , Sinapsis/patología
9.
EMBO J ; 36(10): 1392-1411, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28331029

RESUMEN

Presynaptic terminals are metabolically active and accrue damage through continuous vesicle cycling. How synapses locally regulate protein homeostasis is poorly understood. We show that the presynaptic lipid phosphatase synaptojanin is required for macroautophagy, and this role is inhibited by the Parkinson's disease mutation R258Q. Synaptojanin drives synaptic endocytosis by dephosphorylating PI(4,5)P2, but this function appears normal in SynaptojaninRQ knock-in flies. Instead, R258Q affects the synaptojanin SAC1 domain that dephosphorylates PI(3)P and PI(3,5)P2, two lipids found in autophagosomal membranes. Using advanced imaging, we show that SynaptojaninRQ mutants accumulate the PI(3)P/PI(3,5)P2-binding protein Atg18a on nascent synaptic autophagosomes, blocking autophagosome maturation at fly synapses and in neurites of human patient induced pluripotent stem cell-derived neurons. Additionally, we observe neurodegeneration, including dopaminergic neuron loss, in SynaptojaninRQ flies. Thus, synaptojanin is essential for macroautophagy within presynaptic terminals, coupling protein turnover with synaptic vesicle cycling and linking presynaptic-specific autophagy defects to Parkinson's disease.


Asunto(s)
Autofagosomas/metabolismo , Autofagia , Proteínas del Tejido Nervioso/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Terminales Presinápticos/enzimología , Terminales Presinápticos/metabolismo , Sustitución de Aminoácidos , Animales , Proteínas Relacionadas con la Autofagia/análisis , Células Cultivadas , Drosophila , Humanos , Proteínas de la Membrana/análisis , Mutación Missense , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/patología , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/genética
10.
Acta Neuropathol ; 141(2): 173-192, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33427938

RESUMEN

In Alzheimer's disease (AD), tau-protein undergoes a multi-step process involving the transition from a natively unfolded monomer to large, aggregated structures such as neurofibrillary tangles (NFTs). However, it is not yet clear which events initiate the early preclinical phase of AD tauopathy and whether they have impact on the propagation of tau pathology in later disease stages. To address this question, we analyzed the distribution of tau species phosphorylated at T231, S396/S404 and S202/T205, conformationally modified at the MC1 epitope and fibrillary tau detected by the Gallyas method (Gallyas-tau), in the brains of 15 symptomatic and 20 asymptomatic cases with AD pathology as well as of 19 nonAD cases. As initial tau lesions, we identified phosphorylated-T231-tau diffusely distributed within the somatodendritic compartment (IC-tau) and phosphorylated-S396/pS404-tau in axonal lesions of the white matter and in the neuropil (IN-tau). The subcellular localization of pT231-tau in the cell body and pS396/pS404-tau in the presynapse was confirmed in hP301L mutant Drosophila larvae. Phosphorylated-S202/T205-tau, MC1-tau and Gallyas-tau were negative for these lesions. IC- and IN-tau were observed in all analyzed regions of the human brain, including early affected regions in nonAD cases (entorhinal cortex) and late affected regions in symptomatic AD cases (cerebellum), indicating that tau pathology initiation follows similar processes when propagating into previously unaffected regions. Furthermore, a sequence of AD-related maturation of tau-aggregates was observed, initiated by the appearance of IC- and IN-tau, followed by the formation of pretangles exhibiting pT231-tau, pS396/pS404-tau and pS202/pT205-tau, then by MC1-conformational tau, and, finally, by the formation of Gallyas-positive NFTs. Since cases classified as nonAD [Braak NFT stages < I (including a-1b)] already showed IC- and IN-tau, our findings suggest that these lesions are a prerequisite for the development of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Citoplasma/patología , Ovillos Neurofibrilares/patología , Sinapsis/patología , Tauopatías/patología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Autopsia , Cerebelo/química , Cerebelo/patología , Citoplasma/química , Drosophila , Corteza Entorrinal/química , Corteza Entorrinal/patología , Femenino , Humanos , Inmunohistoquímica , Larva , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/química , Fosforilación , Conformación Proteica , Sinapsis/química
11.
Brain ; 143(6): 1746-1765, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32516804

RESUMEN

TOR1A/TorsinA mutations cause two incurable diseases: a recessive congenital syndrome that can be lethal, and a dominantly-inherited childhood-onset dystonia (DYT-TOR1A). TorsinA has been linked to phosphatidic acid lipid metabolism in Drosophila melanogaster. Here we evaluate the role of phosphatidic acid phosphatase (PAP) enzymes in TOR1A diseases using induced pluripotent stem cell-derived neurons from patients, and mouse models of recessive Tor1a disease. We find that Lipin PAP enzyme activity is abnormally elevated in human DYT-TOR1A dystonia patient cells and in the brains of four different Tor1a mouse models. Its severity also correlated with the dosage of Tor1a/TOR1A mutation. We assessed the role of excess Lipin activity in the neurological dysfunction of Tor1a disease mouse models by interbreeding these with Lpin1 knock-out mice. Genetic reduction of Lpin1 improved the survival of recessive Tor1a disease-model mice, alongside suppressing neurodegeneration, motor dysfunction, and nuclear membrane pathology. These data establish that TOR1A disease mutations cause abnormal phosphatidic acid metabolism, and suggest that approaches that suppress Lipin PAP enzyme activity could be therapeutically useful for TOR1A diseases.


Asunto(s)
Chaperonas Moleculares/metabolismo , Fosfatidato Fosfatasa/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Distonía/genética , Distonía/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Mutación , Neuronas/metabolismo , Fosfatidato Fosfatasa/genética , Fosfatidato Fosfatasa/fisiología
12.
Mol Cell ; 50(6): 831-43, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23685073

RESUMEN

The prevalence of intellectual disability is around 3%; however, the etiology of the disease remains unclear in most cases. We identified a series of patients with X-linked intellectual disability presenting mutations in the Rad6a (Ube2a) gene, which encodes for an E2 ubiquitin-conjugating enzyme. Drosophila deficient for dRad6 display defective synaptic function as a consequence of mitochondrial failure. Similarly, mouse mRad6a (Ube2a) knockout and patient-derived hRad6a (Ube2a) mutant cells show defective mitochondria. Using in vitro and in vivo ubiquitination assays, we show that RAD6A acts as an E2 ubiquitin-conjugating enzyme that, in combination with an E3 ubiquitin ligase such as Parkin, ubiquitinates mitochondrial proteins to facilitate the clearance of dysfunctional mitochondria in cells. Hence, we identify RAD6A as a regulator of Parkin-dependent mitophagy and establish a critical role for RAD6A in maintaining neuronal function.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/genética , Mitofagia , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitina-Proteína Ligasas/metabolismo , Adolescente , Adulto , Animales , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Estudios de Casos y Controles , Línea Celular , Niño , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Exoma , Estudios de Asociación Genética , Humanos , Cinética , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/fisiología , Mutación Missense , Unión Neuromuscular/metabolismo , Linaje , Análisis de Secuencia de ADN , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación , Desacopladores/farmacología
13.
Brain ; 142(8): 2319-2335, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31257402

RESUMEN

Genetic mutations in TBC1D24 have been associated with multiple phenotypes, with epilepsy being the main clinical manifestation. The TBC1D24 protein consists of the unique association of a Tre2/Bub2/Cdc16 (TBC) domain and a TBC/lysin motif domain/catalytic (TLDc) domain. More than 50 missense and loss-of-function mutations have been described and are spread over the entire protein. Through whole genome/exome sequencing we identified compound heterozygous mutations, R360H and G501R, within the TLDc domain, in an index family with a Rolandic epilepsy exercise-induced dystonia phenotype (http://omim.org/entry/608105). A 20-year long clinical follow-up revealed that epilepsy was self-limited in all three affected patients, but exercise-induced dystonia persisted into adulthood in two. Furthermore, we identified three additional sporadic paediatric patients with a remarkably similar phenotype, two of whom had compound heterozygous mutations consisting of an in-frame deletion I81_K84 and an A500V mutation, and the third carried T182M and G511R missense mutations, overall revealing that all six patients harbour a missense mutation in the subdomain of TLDc between residues 500 and 511. We solved the crystal structure of the conserved Drosophila TLDc domain. This allowed us to predict destabilizing effects of the G501R and G511R mutations and, to a lesser degree, of R360H and potentially A500V. Next, we characterized the functional consequences of a strong and a weak TLDc mutation (TBC1D24G501R and TBC1D24R360H) using Drosophila, where TBC1D24/Skywalker regulates synaptic vesicle trafficking. In a Drosophila model neuronally expressing human TBC1D24, we demonstrated that the TBC1D24G501R TLDc mutation causes activity-induced locomotion and synaptic vesicle trafficking defects, while TBC1D24R360H is benign. The neuronal phenotypes of the TBC1D24G501R mutation are consistent with exacerbated oxidative stress sensitivity, which is rescued by treating TBC1D24G501R mutant animals with antioxidants N-acetylcysteine amide or α-tocopherol as indicated by restored synaptic vesicle trafficking levels and sustained behavioural activity. Our data thus show that mutations in the TLDc domain of TBC1D24 cause Rolandic-type focal motor epilepsy and exercise-induced dystonia. The humanized TBC1D24G501R fly model exhibits sustained activity and vesicle transport defects. We propose that the TBC1D24/Sky TLDc domain is a reactive oxygen species sensor mediating synaptic vesicle trafficking rates that, when dysfunctional, causes a movement disorder in patients and flies. The TLDc and TBC domain mutations' response to antioxidant treatment we observed in the animal model suggests a potential for combining antioxidant-based therapeutic approaches to TBC1D24-associated disorders with previously described lipid-altering strategies for TBC domain mutations.


Asunto(s)
Acetilcisteína/análogos & derivados , Antioxidantes/uso terapéutico , Modelos Animales de Enfermedad , Drosophila melanogaster/fisiología , Distonía/tratamiento farmacológico , Epilepsia Rolándica/genética , Proteínas Activadoras de GTPasa/genética , Esfuerzo Físico , alfa-Tocoferol/uso terapéutico , Acetilcisteína/uso terapéutico , Adolescente , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Transporte Biológico/efectos de los fármacos , Dominio Catalítico/genética , Niño , Preescolar , Cristalografía por Rayos X , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Distonía/etiología , Epilepsia Rolándica/tratamiento farmacológico , Femenino , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/fisiología , Humanos , Lactante , Locomoción/genética , Locomoción/fisiología , Masculino , Modelos Moleculares , Mutación Missense , Neuronas/fisiología , Estrés Oxidativo , Linaje , Conformación Proteica , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Eliminación de Secuencia , Homología de Secuencia de Aminoácido , Vesículas Sinápticas/metabolismo , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/genética
14.
J Neurosci ; 36(6): 1914-29, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26865615

RESUMEN

Mitochondria play an important role in the regulation of neurotransmission, and mitochondrial impairment is a key event in neurodegeneration. Cells rely on mitochondrial carrier proteins of the SLC25 family to shuttle ions, cofactors, and metabolites necessary for enzymatic reactions. Mutations in these carriers often result in rare but severe pathologies in the brain, and some of the genes, including SLC25A39 and SLC25A40, reside in susceptibility loci of severe forms of epilepsy. However, the role of most of these carriers has not been investigated in neurons in vivo. We identified shawn, the Drosophila homolog of SLC25A39 and SLC25A40, in a genetic screen to identify genes involved in neuronal function. Shawn localizes to mitochondria, and missense mutations result in an accumulation of reactive oxygen species, mitochondrial dysfunction, and neurodegeneration. Shawn regulates metal homeostasis, and we found in shawn mutants increased levels of manganese, calcium, and mitochondrial free iron. Mitochondrial mutants often cannot maintain synaptic transmission under demanding conditions, but shawn mutants do, and they also do not display endocytic defects. In contrast, shawn mutants harbor a significant increase in neurotransmitter release. Our work provides the first functional annotation of these essential mitochondrial carriers in the nervous system, and the results suggest that metal imbalances and mitochondrial dysfunction may contribute to defects in synaptic transmission and neuronal survival. SIGNIFICANCE STATEMENT: We describe for the first time the role of the mitochondrial carrier Shawn/SLC25A39/SLC25A40 in the nervous system. In humans, these genes reside in susceptibility loci for epilepsy, and, in flies, we observe neuronal defects related to mitochondrial dysfunction and metal homeostasis defects. Interestingly, shawn mutants also harbor increased neurotransmitter release and neurodegeneration. Our data suggest a connection between maintaining a correct metal balance and mitochondrial function to regulate neuronal survival and neurotransmitter release.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Animales , Animales Modificados Genéticamente , Supervivencia Celular/genética , Humanos , Larva/metabolismo , Potencial de la Membrana Mitocondrial/genética , Potencial de la Membrana Mitocondrial/fisiología , Metales/metabolismo , Mitocondrias/metabolismo , Mutación Missense/genética , Neuronas/fisiología , Neurotransmisores/metabolismo , Técnicas de Placa-Clamp , Sinapsis/fisiología , Transmisión Sináptica/genética
15.
Hum Mol Genet ; 24(23): 6736-55, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26376863

RESUMEN

ATP6AP2, an essential accessory component of the vacuolar H+ ATPase (V-ATPase), has been associated with intellectual disability (ID) and Parkinsonism. ATP6AP2 has been implicated in several signalling pathways; however, little is known regarding its role in the nervous system. To decipher its function in behaviour and cognition, we generated and characterized conditional knockdowns of ATP6AP2 in the nervous system of Drosophila and mouse models. In Drosophila, ATP6AP2 knockdown induced defective phototaxis and vacuolated photoreceptor neurons and pigment cells when depleted in eyes and altered short- and long-term memory when depleted in the mushroom body. In mouse, conditional Atp6ap2 deletion in glutamatergic neurons (Atp6ap2(Camk2aCre/0) mice) caused increased spontaneous locomotor activity and altered fear memory. Both Drosophila ATP6AP2 knockdown and Atp6ap2(Camk2aCre/0) mice presented with presynaptic transmission defects, and with an abnormal number and morphology of synapses. In addition, Atp6ap2(Camk2aCre/0) mice showed autophagy defects that led to axonal and neuronal degeneration in the cortex and hippocampus. Surprisingly, axon myelination was affected in our mutant mice, and axonal transport alterations were observed in Drosophila. In accordance with the identified phenotypes across species, genome-wide transcriptome profiling of Atp6ap2(Camk2aCre/0) mouse hippocampi revealed dysregulation of genes involved in myelination, action potential, membrane-bound vesicles and motor behaviour. In summary, ATP6AP2 disruption in mouse and fly leads to cognitive impairment and neurodegeneration, mimicking aspects of the neuropathology associated with ATP6AP2 mutations in humans. Our results identify ATP6AP2 as an essential gene for the nervous system.


Asunto(s)
Trastornos del Conocimiento/etiología , Proteínas de Drosophila/genética , Proteínas de la Membrana/genética , Degeneración Nerviosa/etiología , ATPasas de Translocación de Protón/genética , Receptores de Superficie Celular/genética , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Drosophila , Femenino , Técnicas de Silenciamiento del Gen , Discapacidad Intelectual/genética , Masculino , Ratones , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/fisiología , Neuronas/ultraestructura , Trastornos Parkinsonianos/genética , Sinapsis/metabolismo , Sinapsis/fisiología , Sinapsis/ultraestructura
16.
J Cell Sci ; 128(3): 541­52, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25501810

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with Parkinson's disease, but the precise physiological function of the protein remains ill-defined. Recently, our group proposed a model in which LRRK2 kinase activity is part of an EndoA phosphorylation cycle that facilitates efficient vesicle formation at synapses in the Drosophila melanogaster neuromuscular junctions.Flies harbor only one Lrrk gene, which might encompass the functions of both mammalian LRRK1 and LRRK2. We therefore studied the role of LRRK2 in mammalian synaptic function and provide evidence that knockout or pharmacological inhibition of LRRK2 results in defects in synaptic vesicle endocytosis, altered synaptic morphology and impairments in neurotransmission. In addition, our data indicate that mammalian endophilin A1 (EndoA1,also known as SH3GL2) is phosphorylated by LRRK2 in vitro at T73 and S75, two residues in the BAR domain. Hence, our results indicate that LRRK2 kinase activity has an important role in the regulation of clathrin-mediated endocytosis of synaptic vesicles and subsequent neurotransmission at the synapse.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endocitosis/genética , Proteínas Serina-Treonina Quinasas/genética , Transmisión Sináptica/genética , Vesículas Sinápticas/genética , Animales , Células Cultivadas , Clatrina/metabolismo , Drosophila melanogaster , Dinamina I/antagonistas & inhibidores , Endocitosis/efectos de los fármacos , Hipocampo/citología , Hidrazonas/farmacología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Long-Evans , Sacarosa/farmacología , Transmisión Sináptica/efectos de los fármacos
17.
Metab Eng ; 43(Pt B): 187-197, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27847310

RESUMEN

Mutations in succinate dehydrogenase (SDH) are associated with tumor development and neurodegenerative diseases. Only in tumors, loss of SDH activity is accompanied with the loss of complex I activity. Yet, it remains unknown whether the metabolic phenotype of SDH mutant tumors is driven by loss of complex I function, and whether this contributes to the peculiarity of tumor development versus neurodegeneration. We addressed this question by decoupling loss of SDH and complex I activity in cancer cells and neurons. We found that sole loss of SDH activity was not sufficient to recapitulate the metabolic phenotype of SDH mutant tumors, because it failed to decrease mitochondrial respiration and to activate reductive glutamine metabolism. These metabolic phenotypes were only induced upon the additional loss of complex I activity. Thus, we show that complex I function defines the metabolic differences between SDH mutation associated tumors and neurodegenerative diseases, which could open novel therapeutic options against both diseases.


Asunto(s)
Complejo I de Transporte de Electrón , Mutación , Proteínas de Neoplasias , Neoplasias , Succinato Deshidrogenasa , Línea Celular Tumoral , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Neuronas/enzimología , Neuronas/patología , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo
18.
Hum Mol Genet ; 23(19): 5227-42, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24852371

RESUMEN

Loss-of-function mutations in PARK2, the gene encoding the E3 ubiquitin ligase Parkin, are the most frequent cause of recessive Parkinson's disease (PD). Parkin translocates from the cytosol to depolarized mitochondria, ubiquitinates outer mitochondrial membrane proteins and induces selective autophagy of the damaged mitochondria (mitophagy). Here, we show that ubiquitin-specific protease 15 (USP15), a deubiquitinating enzyme (DUB) widely expressed in brain and other organs, opposes Parkin-mediated mitophagy, while a panel of other DUBs and a catalytically inactive version of USP15 do not. Moreover, knockdown of USP15 rescues the mitophagy defect of PD patient fibroblasts with PARK2 mutations and decreased Parkin levels. USP15 does not affect the ubiquitination status of Parkin or Parkin translocation to mitochondria, but counteracts Parkin-mediated mitochondrial ubiquitination. Knockdown of the DUB CG8334, the closest homolog of USP15 in Drosophila, largely rescues the mitochondrial and behavioral defects of parkin RNAi flies. These data identify USP15 as an antagonist of Parkin and suggest that USP15 inhibition could be a therapeutic strategy for PD cases caused by reduced Parkin levels.


Asunto(s)
Mitocondrias/metabolismo , Mitofagia , Ubiquitina-Proteína Ligasas/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación , Animales , Línea Celular , Drosophila , Activación Enzimática , Epistasis Genética , Femenino , Fibroblastos/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Mitocondrias/genética , Mitofagia/genética , Modelos Biológicos , Mutación , Especificidad de Órganos/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Unión Proteica , Ubiquitina-Proteína Ligasas/genética , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación/genética
19.
PLoS Genet ; 9(4): e1003478, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23637640

RESUMEN

The PTEN-induced kinase 1 (PINK1) is a mitochondrial kinase, and pink1 mutations cause early onset Parkinson's disease (PD) in humans. Loss of pink1 in Drosophila leads to defects in mitochondrial function, and genetic data suggest that another PD-related gene product, Parkin, acts with pink1 to regulate the clearance of dysfunctional mitochondria (mitophagy). Consequently, pink1 mutants show an accumulation of morphologically abnormal mitochondria, but it is unclear if other factors are involved in pink1 function in vivo and contribute to the mitochondrial morphological defects seen in specific cell types in pink1 mutants. To explore the molecular mechanisms of pink1 function, we performed a genetic modifier screen in Drosophila and identified aconitase (acon) as a dominant suppressor of pink1. Acon localizes to mitochondria and harbors a labile iron-sulfur [4Fe-4S] cluster that can scavenge superoxide to release hydrogen peroxide and iron that combine to produce hydroxyl radicals. Using Acon enzymatic mutants, and expression of mitoferritin that scavenges free iron, we show that [4Fe-4S] cluster inactivation, as a result of increased superoxide in pink1 mutants, results in oxidative stress and mitochondrial swelling. We show that [4Fe-4S] inactivation acts downstream of pink1 in a pathway that affects mitochondrial morphology, but acts independently of parkin. Thus our data indicate that superoxide-dependent [4Fe-4S] inactivation defines a potential pathogenic cascade that acts independent of mitophagy and links iron toxicity to mitochondrial failure in a PD-relevant model.


Asunto(s)
Aconitato Hidratasa , Drosophila , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Humanos , Hierro/metabolismo , Mitocondrias/genética , Enfermedad de Parkinson/genética
20.
PLoS Genet ; 9(10): e1003911, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204314

RESUMEN

Intellectual Disability (ID) disorders, defined by an IQ below 70, are genetically and phenotypically highly heterogeneous. Identification of common molecular pathways underlying these disorders is crucial for understanding the molecular basis of cognition and for the development of therapeutic intervention strategies. To systematically establish their functional connectivity, we used transgenic RNAi to target 270 ID gene orthologs in the Drosophila eye. Assessment of neuronal function in behavioral and electrophysiological assays and multiparametric morphological analysis identified phenotypes associated with knockdown of 180 ID gene orthologs. Most of these genotype-phenotype associations were novel. For example, we uncovered 16 genes that are required for basal neurotransmission and have not previously been implicated in this process in any system or organism. ID gene orthologs with morphological eye phenotypes, in contrast to genes without phenotypes, are relatively highly expressed in the human nervous system and are enriched for neuronal functions, suggesting that eye phenotyping can distinguish different classes of ID genes. Indeed, grouping genes by Drosophila phenotype uncovered 26 connected functional modules. Novel links between ID genes successfully predicted that MYCN, PIGV and UPF3B regulate synapse development. Drosophila phenotype groups show, in addition to ID, significant phenotypic similarity also in humans, indicating that functional modules are conserved. The combined data indicate that ID disorders, despite their extreme genetic diversity, are caused by disruption of a limited number of highly connected functional modules.


Asunto(s)
Ojo/metabolismo , Discapacidad Intelectual/genética , Redes y Vías Metabólicas/genética , Sinapsis/genética , Animales , Animales Modificados Genéticamente , Drosophila/genética , Ojo/crecimiento & desarrollo , Técnicas de Silenciamiento del Gen , Variación Genética , Humanos , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Neuronas/metabolismo , Fenotipo , Interferencia de ARN , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda