Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 35(2): 108972, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852856

RESUMEN

Disruption of sphingolipid homeostasis is known to cause neurological disorders, but the mechanisms by which specific sphingolipid species modulate pathogenesis remain unclear. The last step of de novo sphingolipid synthesis is the conversion of dihydroceramide to ceramide by dihydroceramide desaturase (human DEGS1; Drosophila Ifc). Loss of ifc leads to dihydroceramide accumulation, oxidative stress, and photoreceptor degeneration, whereas human DEGS1 variants are associated with leukodystrophy and neuropathy. In this work, we demonstrate that DEGS1/ifc regulates Rac1 compartmentalization in neuronal cells and that dihydroceramide alters the association of active Rac1 with organelle-mimicking membranes. We further identify the Rac1-NADPH oxidase (NOX) complex as the major cause of reactive oxygen species (ROS) accumulation in ifc-knockout (ifc-KO) photoreceptors and in SH-SY5Y cells with the leukodystrophy-associated DEGS1H132R variant. Suppression of Rac1-NOX activity rescues degeneration of ifc-KO photoreceptors and ameliorates oxidative stress in DEGS1H132R-carrying cells. Therefore, we conclude that DEGS1/ifc deficiency causes dihydroceramide accumulation, resulting in Rac1 mislocalization and NOX-dependent neurodegeneration.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ácido Graso Desaturasas/genética , Proteínas de la Membrana/genética , NADPH Oxidasas/genética , Proteína de Unión al GTP rac1/genética , Animales , Línea Celular Tumoral , Ceramidas/metabolismo , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/metabolismo , Electrorretinografía , Ácido Graso Desaturasas/antagonistas & inhibidores , Ácido Graso Desaturasas/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/deficiencia , NADPH Oxidasas/metabolismo , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Mutación Puntual , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retina/metabolismo , Retina/patología , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
2.
Exp Eye Res ; 201: 108291, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33049273

RESUMEN

Retinitis pigmentosa (RP) is a collection of heterogenous genetic retinal disorders resulting in cumulative retinal deterioration involving progressive loss of photoreceptors and eventually in total blindness. Oxidative stress plays a central role in this photoreceptor loss. Gypenosides (Gyp) are the main functional component isolated from the climbing vine Gynostemma pentaphyllum and have been shown to defend cells against the effects of oxidative stress and inflammation, providing protection in experimentally-induced optic neuritis. The zebrafish model has been used to investigate a range of human diseases. Previously we reported early retinal degeneration in a mutant zebrafish line carrying a point-nonsense mutation in the retinitis pigmentosa GTPase regulator interacting protein 1 (rpgrip1) gene that is mutated in RP patients. The current study investigated the potential protective effects of Gyp against photoreceptor degeneration in the Rpgrip1 deleted zebrafish. Rpgrip1 mutant zebrafish were treated with 5 µg/ml of Gyp in E3 medium from 6 h post fertilization (hpf) till 1 month post fertilization (mpf). Rpgrip1 mutant zebrafish treated with 5 µg/ml of Gyp showed a significant decrease by 68.41% (p = 0.0002) in photoreceptor cell death compared to that of untreated mutant zebrafish. Expression of antioxidant genes catalase, sod1, sod2, gpx1, gclm, nqo-1 and nrf-2 was significantly decreased in rpgrip1 mutant zebrafish eyes by 61.51%, 77.40%, 60.11%, 81.17%, 72.07%, 78.95% and 85.42% (all p < 0.0001), respectively, when compared to that of wildtype zebrafish; superoxide dismutase and catalase activities, and glutathione levels in rpgrip1 mutant zebrafish eyes were significantly decreased by 87.21%, 21.55% and 96.51% (all p < 0.0001), respectively. There were marked increases in the production of reactive oxygen species (ROS) and malondialdehyde (MDA) by 2738.73% and 510.69% (all p < 0.0001), respectively, in rpgrip1 mutant zebrafish eyes; expression of pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α was also significantly increased by 150.11%, 267.79% and 190.72% (all p < 0.0001), respectively, in rpgrip1 mutant zebrafish eyes, compared to that of wildtype zebrafish. Treatment with Gyp significantly counteracted these effects. This study indicates that Gyp has a potential role in the treatment of RP.


Asunto(s)
Estrés Oxidativo , Células Fotorreceptoras de Invertebrados/efectos de los fármacos , Retina/efectos de los fármacos , Retinitis Pigmentosa/tratamiento farmacológico , Animales , Gynostemma , Inmunohistoquímica , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/metabolismo , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Rodopsina/metabolismo , Pez Cebra
3.
PLoS Genet ; 16(6): e1008869, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32569302

RESUMEN

We investigate mutations in trß2, a splice variant of thrb, identifying changes in function, structure, and behavior in larval and adult zebrafish retinas. Two N-terminus CRISPR mutants were identified. The first is a 6BP+1 insertion deletion frameshift resulting in a truncated protein. The second is a 3BP in frame deletion with intact binding domains. ERG recordings of isolated cone signals showed that the 6BP+1 mutants did not respond to red wavelengths of light while the 3BP mutants did respond. 6BP+1 mutants lacked optomotor and optokinetic responses to red/black and green/black contrasts. Both larval and adult 6BP+1 mutants exhibit a loss of red-cone contribution to the ERG and an increase in UV-cone contribution. Transgenic reporters show loss of cone trß2 activation in the 6BP+1 mutant but increase in the density of cones with active blue, green, and UV opsin genes. Antibody reactivity for red-cone LWS1 and LWS2 opsin was absent in the 6BP+1 mutant, as was reactivity for arrestin3a. Our results confirm a critical role for trß2 in long-wavelength cone development.


Asunto(s)
Visión de Colores/genética , Regulación del Desarrollo de la Expresión Génica , Genes erbA/genética , Retina/crecimiento & desarrollo , Receptores beta de Hormona Tiroidea/genética , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Opsinas de los Conos/genética , Opsinas de los Conos/metabolismo , Mutación del Sistema de Lectura , Mutación INDEL , Larva , Modelos Animales , Células Fotorreceptoras de Invertebrados/patología , Retina/citología , Retina/patología , Eliminación de Secuencia , Transactivadores/genética , Transactivadores/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Mol Biol Cell ; 29(13): 1640-1651, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29742016

RESUMEN

Synthesis and maturation of the light sensor, rhodopsin, are critical for the maintenance of light sensitivity and for photoreceptor homeostasis. In Drosophila, the main rhodopsin, Rh1, is synthesized in the endoplasmic reticulum and transported to the rhabdomere through the secretory pathway. In an unbiased genetic screen for factors involved in rhodopsin homeostasis, we identified mutations in vha68-1, which encodes the vacuolar proton-translocating ATPase (V-ATPase) catalytic subunit A isoform 1 of the V1 component. Loss of vha68-1 in photoreceptor cells disrupted post-Golgi anterograde trafficking of Rh1, reduced light sensitivity, increased secretory vesicle pH, and resulted in incomplete Rh1 deglycosylation. In addition, vha68-1 was required for activity-independent photoreceptor cell survival. Importantly, vha68-1 mutants exhibited phenotypes similar to those exhibited by mutations in the V0 component of V-ATPase, vha100-1. These data demonstrate that the V1 and V0 components of V-ATPase play key roles in post-Golgi trafficking of Rh1 and that Drosophila may represent an important animal model system for studying diseases associated with V-ATPase dysfunction.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Rodopsina/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Animales , Proteínas de Drosophila/genética , Glicosilación , Homeostasis , Concentración de Iones de Hidrógeno , Fototransducción , Modelos Biológicos , Mutación/genética , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Células Fotorreceptoras de Invertebrados/ultraestructura , Transporte de Proteínas , Degeneración Retiniana/patología , Vesículas Secretoras/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Vías Visuales/metabolismo
5.
Exp Cell Res ; 366(2): 92-102, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29501567

RESUMEN

Pyruvate dehydrogenase complex deficiency (PDCD) is a common primary cause of defects in mitochondrial function and also can lead to peripheral neuropathy. Pyruvate dehydrogenase E1 component subunit beta (PDHB) is a subunit of pyruvate dehydrogenase E1, which is a well-known component of PDC. In Drosophila melanogaster, the CG11876 (dPDHB) gene is a homolog of human PDHB. In this study, we established a Drosophila model with neuron-specific knockdown of dPDHB to investigate its role in neuropathy pathogenesis. Knockdown of dPDHB in pan-neurons induced locomotor defects in both larval and adult stages, which were consistent with abnormal morphology of the motor neuron terminals at neuromuscular junctions and mitochondrial fragmentation in brains. Moreover, neuron-specific knockdown of dPDHB also shortened the lifespan of adult flies. In addition, flies with knockdown of dPDHB manifested a rough eye phenotype and aberrant photoreceptor axon targeting. These results with the Drosophila model suggest the involvement of PDHB in peripheral neuropathy.


Asunto(s)
Axones/patología , Drosophila melanogaster/fisiología , Locomoción , Longevidad , Neuronas Motoras/patología , Células Fotorreceptoras de Invertebrados/patología , Piruvato Deshidrogenasa (Lipoamida)/antagonistas & inhibidores , Animales , Animales Modificados Genéticamente/fisiología , Axones/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Masculino , Mitocondrias/metabolismo , Mitocondrias/patología , Neuronas Motoras/metabolismo , Fenotipo , Células Fotorreceptoras de Invertebrados/metabolismo , Piruvato Deshidrogenasa (Lipoamida)/genética , Piruvato Deshidrogenasa (Lipoamida)/metabolismo
6.
Hum Mol Genet ; 27(8): 1353-1365, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29425337

RESUMEN

Aggregation of fused in sarcoma (FUS) protein, and mutations in FUS gene, are causative to a range of neurodegenerative disorders including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. To gain insights into the molecular mechanism whereby FUS causes neurodegeneration, we generated transgenic Drosophila melanogaster overexpressing human FUS in the photoreceptor neurons, which exhibited mild retinal degeneration. Expression of familial ALS-mutant FUS aggravated the degeneration, which was associated with an increase in cytoplasmic localization of FUS. A carboxy-terminally truncated R495X mutant FUS also was localized in cytoplasm, whereas the degenerative phenotype was diminished. Double expression of R495X and wild-type FUS dramatically exacerbated degeneration, sequestrating wild-type FUS into cytoplasmic aggregates. Notably, replacement of all tyrosine residues within the low-complexity domain, which abolished self-assembly of FUS, completely eliminated the degenerative phenotypes. Taken together, we propose that self-assembly of FUS through its low-complexity domain contributes to FUS-induced neurodegeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Demencia Frontotemporal/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/genética , Células Fotorreceptoras de Invertebrados/metabolismo , Proteínas Recombinantes de Fusión/genética , Degeneración Retiniana/genética , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Expresión Génica , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/química , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Células Fotorreceptoras de Invertebrados/patología , Dominios Proteicos , Pliegue de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Tirosina/química , Tirosina/metabolismo
7.
BMC Dev Biol ; 18(1): 1, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382313

RESUMEN

BACKGROUND: The compound eyes of insects allow them to catch photons and convert the energy into electric signals. All compound eyes consist of numerous ommatidia, each comprising a fixed number of photoreceptors. Different ommatidial types are characterized by a specific set of photoreceptors differing in spectral sensitivity. In honey bees, males and females possess different ommatidial types forming distinct retinal mosaics. However, data are lacking on retinal ontogeny and the mechanisms by which the eyes are patterned. In this study, we investigated the intrinsic temporal and circadian expression patterns of the opsins that give rise to the ultraviolet, blue and green sensitive photoreceptors, as well as the morphological maturation of the retina during pupal development of honey bees. RESULTS: qPCR and histological labeling revealed that temporal opsin mRNA expression differs between sexes and correlates with rhabdom elongation during photoreceptor development. In the first half of the pupal stage, when the rhabdoms of the photoreceptors are still short, worker and (dorsal) drone retinae exhibit similar expression patterns with relatively high levels of UV (UVop) and only marginal levels of blue (BLop) and green (Lop1) opsin mRNA. In the second half of pupation, when photoreceptors and rhabdoms elongate, opsin expression in workers becomes dominated by Lop1 mRNA. In contrast, the dorsal drone eye shows high expression levels of UVop and BLop mRNA, whereas Lop1 mRNA level decreases. Interestingly, opsin expression levels increase up to 22-fold during early adult life. We also found evidence that opsin expression in adult bees is under the control of the endogenous clock. CONCLUSIONS: Our data indicate that the formation of the sex-specific retinal composition of photoreceptors takes place during the second half of the pupal development, and that opsin mRNA expression levels continue to increase in young bees, which stands in contrast to Drosophila, where the highest expression levels are found during the late pupal stage and remain constant in adults. From an evolutionary perspective, we hypothesize that the delayed retinal maturation during the early adult phase is linked to the delayed transition from indoor to outdoor activities in bees, when vision becomes important.


Asunto(s)
Abejas/crecimiento & desarrollo , Abejas/metabolismo , Opsinas/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Animales , Ritmo Circadiano/genética , Regulación del Desarrollo de la Expresión Génica , Opsinas/genética , Pupa/crecimiento & desarrollo , Pupa/metabolismo , Retina/crecimiento & desarrollo , Retina/metabolismo , Factores de Tiempo
8.
Glia ; 66(4): 874-888, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29285794

RESUMEN

Mitochondrial defects associated with respiratory chain complex I deficiency lead to heterogeneous fatal syndromes. While the role of NDUFS8, an essential subunit of the core assembly of the complex I, is established in mitochondrial diseases, the mechanisms underlying neuropathology are poorly understood. We developed a Drosophila model of NDUFS8 deficiency by knocking down the expression of its fly homologue in neurons or in glial cells. Downregulating ND23 in neurons resulted in shortened lifespan, and decreased locomotion. Although total brain ATP levels were decreased, histological analysis did not reveal any signs of neurodegeneration except for photoreceptors of the retina. Interestingly, ND23 deficiency-associated phenotypes were rescued by overexpressing the glucose transporter hGluT3 demonstrating that boosting glucose metabolism in neurons was sufficient to bypass altered mitochondrial functions and to confer neuroprotection. We then analyzed the consequences of ND23 knockdown in glial cells. In contrast to neuronal knockdown, loss of ND23 in glia did not lead to significant behavioral defects nor to reduced lifespan, but induced brain degeneration, as visualized by numerous vacuoles found all over the nervous tissue. This phenotype was accompanied by the massive accumulation of lipid droplets at the cortex-neuropile boundaries, suggesting an alteration of lipid metabolism in glia. These results demonstrate that complex I deficiency triggers metabolic alterations both in neurons and glial cells which may contribute to the neuropathology.


Asunto(s)
Proteínas de Drosophila/deficiencia , Metabolismo de los Lípidos/fisiología , Enfermedades Mitocondriales/patología , NADH Deshidrogenasa/deficiencia , Enfermedades Neurodegenerativas/patología , Neuroglía/patología , Adenosina Trifosfato/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Femenino , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Homeostasis/fisiología , Humanos , Enfermedades Mitocondriales/metabolismo , Actividad Motora/fisiología , NADH Deshidrogenasa/genética , Enfermedades Neurodegenerativas/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Neuronas/patología , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Interferencia de ARN , ARN Mensajero/metabolismo
9.
Neurobiol Dis ; 108: 238-248, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28888970

RESUMEN

Duplication 15q syndrome (Dup15q) is an autism-associated disorder co-incident with high rates of pediatric epilepsy. Additional copies of the E3 ubiquitin ligase UBE3A are thought to cause Dup15q phenotypes, yet models overexpressing UBE3A in neurons have not recapitulated the epilepsy phenotype. We show that Drosophila endogenously expresses Dube3a (fly UBE3A homolog) in glial cells and neurons, prompting an investigation into the consequences of glial Dube3a overexpression. Here we expand on previous work showing that the Na+/K+ pump ATPα is a direct ubiquitin ligase substrate of Dube3a. A robust seizure-like phenotype was observed in flies overexpressing Dube3a in glial cells, but not neurons. Glial-specific knockdown of ATPα also produced seizure-like behavior, and this phenotype was rescued by simultaneously overexpressing ATPα and Dube3a in glia. Our data provides the basis of a paradigm shift in Dup15q research given that clinical phenotypes have long been assumed to be due to neuronal UBE3A overexpression.


Asunto(s)
Proteínas de Drosophila/metabolismo , Neuroglía/metabolismo , Convulsiones/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Sinapsis/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Modificados Genéticamente , Cromosomas Humanos Par 15/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Drosophila , Proteínas de Drosophila/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Potasio/metabolismo , Convulsiones/patología , ATPasa Intercambiadora de Sodio-Potasio/genética , Sinapsis/patología , Trisomía/patología , Ubiquitina-Proteína Ligasas/genética
10.
Dis Model Mech ; 10(8): 993-1003, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28623239

RESUMEN

The myotonic dystrophies are prototypic toxic RNA gain-of-function diseases. Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are caused by different unstable, noncoding microsatellite repeat expansions - (CTG)DM1 in DMPK and (CCTG)DM2 in CNBP Although transcription of mutant repeats into (CUG)DM1 or (CCUG)DM2 appears to be necessary and sufficient to cause disease, their pathomechanisms remain incompletely understood. To study the mechanisms of (CCUG)DM2 toxicity and develop a convenient model for drug screening, we generated a transgenic DM2 model in the fruit fly Drosophila melanogaster with (CCUG)n repeats of variable length (n=16 and 106). Expression of noncoding (CCUG)106, but not (CCUG)16, in muscle and retinal cells led to the formation of ribonuclear foci and mis-splicing of genes implicated in DM pathology. Mis-splicing could be rescued by co-expression of human MBNL1, but not by CUGBP1 (CELF1) complementation. Flies with (CCUG)106 displayed strong disruption of external eye morphology and of the underlying retina. Furthermore, expression of (CCUG)106 in developing retinae caused a strong apoptotic response. Inhibition of apoptosis rescued the retinal disruption in (CCUG)106 flies. Finally, we tested two chemical compounds that have shown therapeutic potential in DM1 models. Whereas treatment of (CCUG)106 flies with pentamidine had no effect, treatment with a PKR inhibitor blocked both the formation of RNA foci and apoptosis in retinae of (CCUG)106 flies. Our data indicate that expression of expanded (CCUG)DM2 repeats is toxic, causing inappropriate cell death in affected fly eyes. Our Drosophila DM2 model might provide a convenient tool for in vivo drug screening.


Asunto(s)
Apoptosis/efectos de los fármacos , Expansión de las Repeticiones de ADN/genética , Drosophila melanogaster/genética , Distrofia Miotónica/genética , ARN/toxicidad , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efectos de los fármacos , Humanos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Pentamidina/farmacología , Células Fotorreceptoras de Invertebrados/efectos de los fármacos , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Inhibidores de Proteínas Quinasas/farmacología , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Retina/anomalías , Retina/efectos de los fármacos , Retina/patología , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/metabolismo
11.
EMBO Rep ; 18(7): 1150-1165, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28507162

RESUMEN

Dihydroceramide desaturases are evolutionarily conserved enzymes that convert dihydroceramide (dhCer) to ceramide (Cer). While elevated Cer levels cause neurodegenerative diseases, the neuronal activity of its direct precursor, dhCer, remains unclear. We show that knockout of the fly dhCer desaturase gene, infertile crescent (ifc), results in larval lethality with increased dhCer and decreased Cer levels. Light stimulation leads to ROS increase and apoptotic cell death in ifc-KO photoreceptors, resulting in activity-dependent neurodegeneration. Lipid-containing Atg8/LC3-positive puncta accumulate in ifc-KO photoreceptors, suggesting lipophagy activation. Further enhancing lipophagy reduces lipid droplet accumulation and rescues ifc-KO defects, indicating that lipophagy plays a protective role. Reducing dhCer synthesis prevents photoreceptor degeneration and rescues ifc-KO lethality, while supplementing downstream sphingolipids does not. These results pinpoint that dhCer accumulation is responsible for ifc-KO defects. Human dhCer desaturase rescues ifc-KO larval lethality, and rapamycin reverses defects caused by dhCer accumulation in human neuroblastoma cells, suggesting evolutionarily conserved functions. This study demonstrates a novel requirement for dhCer desaturase in neuronal maintenance in vivo and shows that lipophagy activation prevents activity-dependent degeneration caused by dhCer accumulation.


Asunto(s)
Autofagia , Ceramidas/metabolismo , Metabolismo de los Lípidos , Animales , Apoptosis , Línea Celular Tumoral , Ceramidas/análisis , Drosophila , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Ácido Graso Desaturasas/genética , Técnicas de Inactivación de Genes , Humanos , Luz/efectos adversos , Lipólisis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Enfermedades Neurodegenerativas/prevención & control , Células Fotorreceptoras de Invertebrados/patología , Células Fotorreceptoras de Invertebrados/efectos de la radiación , Esfingolípidos/metabolismo
12.
Exp Neurol ; 277: 86-95, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26708557

RESUMEN

Mutations in Factor-Induced-Gene 4 (FIG4) gene have been identified in Charcot-Marie-Tooth disease type 4J (CMT4J), Yunis-Varon syndrome and epilepsy with polymicrogyria. FIG4 protein regulates a cellular abundance of phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), a signaling lipid on the cytosolic surface of membranes of the late endosomal compartment. PI(3,5)P2 is required for retrograde membrane trafficking from lysosomal and late endosomal compartments to the Golgi. However, it is still unknown how the neurodegeneration that occurs in these diseases is related to the loss of FIG4 function. Drosophila has CG17840 (dFIG4) as a human FIG4 homolog. Here we specifically knocked down dFIG4 in various tissues, and investigated their phenotypes. Neuron-specific knockdown of dFIG4 resulted in axonal targeting aberrations of photoreceptor neurons, shortened presynaptic terminals of motor neurons in 3rd instar larvae and reduced climbing ability in adulthood and life span. Fat body-specific knockdown of dFIG4 resulted in enlarged lysosomes in cells that were detected by staining with LysoTracker. In addition, eye imaginal disk-specific knockdown of dFIG4 disrupted differentiation of pupal ommatidial cell types, such as cone cells and pigment cells, suggesting an additional role of dFIG4 during eye development.


Asunto(s)
Axones/patología , Anomalías del Ojo/genética , Trastornos Neurológicos de la Marcha/genética , Trastornos Neurológicos de la Marcha/patología , Longevidad/genética , Neuronas Motoras/patología , Monoéster Fosfórico Hidrolasas/deficiencia , Animales , Animales Modificados Genéticamente , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Flavoproteínas/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Discos Imaginales/patología , Lisosomas/patología , Unión Neuromuscular/genética , Unión Neuromuscular/patología , Monoéster Fosfórico Hidrolasas/genética , Células Fotorreceptoras de Invertebrados/patología , Trastornos Psicomotores/genética , Alineación de Secuencia
13.
Acta Neuropathol ; 130(1): 35-47, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25862636

RESUMEN

The involvement of Amyloid-ß (Aß) in the pathogenesis of Alzheimer's disease (AD) is well established. However, it is becoming clear that the amyloid load in AD brains consists of a heterogeneous mixture of Aß peptides, implying that a thorough understanding of their respective role and toxicity is crucial for the development of efficient treatments. Besides the well-studied Aß40 and Aß42 species, recent data have raised the possibility that Aß43 peptides might be instrumental in AD pathogenesis, because they are frequently observed in both dense and diffuse amyloid plaques from human AD brains and are highly amyloidogenic in vitro. However, whether Aß43 is toxic in vivo is currently unclear. Using Drosophila transgenic models of amyloid pathology, we show that Aß43 peptides are mainly insoluble and highly toxic in vivo, leading to the progressive loss of photoreceptor neurons, altered locomotion and decreased lifespan when expressed in the adult fly nervous system. In addition, we demonstrate that Aß43 species are able to trigger the aggregation of the typically soluble and non-toxic Aß40, leading to synergistic toxic effects on fly lifespan and climbing ability, further suggesting that Aß43 peptides could act as a nucleating factor in AD brains. Altogether, our study demonstrates high pathogenicity of Aß43 species in vivo and supports the idea that Aß43 contributes to the pathological events leading to neurodegeneration in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Péptidos beta-Amiloides/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila , Femenino , Cabeza/patología , Cabeza/fisiología , Actividad Motora/fisiología , Fragmentos de Péptidos/genética , Células Fotorreceptoras de Invertebrados/patología , Células Fotorreceptoras de Invertebrados/fisiología , Agregado de Proteínas , ARN Mensajero/metabolismo , Solubilidad , Análisis de Supervivencia
14.
Channels (Austin) ; 9(1): 14-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25664921

RESUMEN

Cytoplasmic Ca2+ overload is known to trigger autophagy and ER-stress. Furthermore, ER-stress and autophagy are commonly associated with degenerative pathologies, but their role in disease progression is still a matter of debate, in part, owing to limitations of existing animal model systems. The Drosophila eye is a widely used model system for studying neurodegenerative pathologies. Recently, we characterized the Drosophila protein, Calphotin, as a cytosolic immobile Ca2+ buffer, which participates in Ca2+ homeostasis in Drosophila photoreceptor cells. Exposure of calphotin hypomorph flies to continuous illumination, which induces Ca2+ influx into photoreceptor cells, resulted in severe Ca2+-dependent degeneration. Here we show that this degeneration is autophagy and ER-stress related. Our studies thus provide a new model in which genetic manipulations trigger changes in cellular Ca2+ distribution. This model constitutes a framework for further investigations into the link between cytosolic Ca2+, ER-stress and autophagy in human disorders and diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Calcio/farmacología , Drosophila/citología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Modelos Genéticos , Células Fotorreceptoras de Invertebrados/efectos de los fármacos , Células Fotorreceptoras de Invertebrados/patología , Animales , Autofagia/genética , Modelos Animales de Enfermedad , Drosophila/genética , Estrés del Retículo Endoplásmico/genética
15.
Dev Neurobiol ; 74(12): 1210-25, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24909306

RESUMEN

Accumulations of Tau, a microtubule-associated protein (MAP), into neurofibrillary tangles is a hallmark of Alzheimer's disease and other tauopathies. However, the mechanisms leading to this pathology are still unclear: the aggregates themselves could be toxic or the sequestration of Tau into tangles might prevent Tau from fulfilling its normal functions, thereby inducing a loss of function defect. Surprisingly, the consequences of losing normal Tau expression in vivo are still not well understood, in part due to the fact that Tau knockout mice show only subtle phenotypes, presumably due to the fact that mammals express several MAPs with partially overlapping functions. In contrast, flies express fewer MAP, with Tau being the only member of the Tau/MAP2/MAP4 family. Therefore, we used Drosophila to address the physiological consequences caused by the loss of Tau. Reducing the levels of fly Tau (dTau) ubiquitously resulted in developmental lethality, whereas deleting Tau specifically in neurons or the eye caused progressive neurodegeneration. Similarly, chromosomal mutations affecting dTau also caused progressive degeneration in both the eye and brain. Although photoreceptor cells initially developed normally in dTau knockdown animals, they subsequently degenerated during late pupal stages whereas weaker dTau alleles caused an age-dependent defect in rhabdomere structure. Expression of wild type human Tau partially rescued the neurodegenerative phenotype caused by the loss of endogenous dTau, suggesting that the functions of Tau proteins are functionally conserved from flies to humans.


Asunto(s)
Proteínas de Drosophila/deficiencia , Degeneración Nerviosa/fisiopatología , Células Fotorreceptoras de Invertebrados/fisiología , Degeneración Retiniana/fisiopatología , Proteínas tau/deficiencia , Proteínas tau/metabolismo , Empalme Alternativo , Animales , Animales Modificados Genéticamente , Axones/patología , Axones/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Drosophila , Proteínas de Drosophila/genética , Técnicas de Silenciamiento del Gen , Humanos , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Mutación , Degeneración Nerviosa/patología , Células Fotorreceptoras de Invertebrados/patología , Isoformas de Proteínas , Degeneración Retiniana/patología , Proteínas tau/genética
16.
J Neurosci ; 33(44): 17444-57, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24174678

RESUMEN

Intraocular pressure (IOP) elevation is a principal risk factor for glaucoma. Using a microbead injection technique to chronically raise IOP for 15 or 30 d in mice, we identified the early changes in visual response properties of different types of retinal ganglion cells (RGCs) and correlated these changes with neuronal morphology before cell death. Microbead-injected eyes showed reduced optokinetic tracking as well as cell death. In such eyes, multielectrode array recordings revealed that four RGC types show diverse alterations in their light responses upon IOP elevation. OFF-transient RGCs exhibited a more rapid decline in both structural and functional organizations compared with other RGCs. In contrast, although the light-evoked responses of OFF-sustained RGCs were perturbed, the dendritic arbor of this cell type remained intact. ON-transient and ON-sustained RGCs had normal functional receptive field sizes but their spontaneous and light-evoked firing rates were reduced. ON- and OFF-sustained RGCs lost excitatory synapses across an otherwise structurally normal dendritic arbor. Together, our observations indicate that there are changes in spontaneous activity and light-evoked responses in RGCs before detectable dendritic loss. However, when dendrites retract, we found corresponding changes in receptive field center size. Importantly, the effects of IOP elevation are not uniformly manifested in the structure and function of diverse RGC populations, nor are distinct RGC types perturbed within the same time-frame by such a challenge.


Asunto(s)
Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glaucoma/patología , Glaucoma/fisiopatología , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/fisiología , Potenciales de Acción/fisiología , Animales , Muerte Celular/fisiología , Femenino , Presión Intraocular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estimulación Luminosa/métodos , Células Fotorreceptoras de Invertebrados/patología , Células Fotorreceptoras de Invertebrados/fisiología , Distribución Aleatoria
17.
Genetics ; 195(3): 857-70, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24037265

RESUMEN

Trinucleotide CAG repeat disorders are caused by expansion of polyglutamine (polyQ) domains in certain proteins leading to fatal neurodegenerative disorders and are characterized by accumulation of inclusion bodies in the neurons. Clearance of these inclusion bodies holds the key to improve the disease phenotypes, which affects basic cellular processes such as transcription, protein degradation and cell signaling. In the present study, we show that P-glycoprotein (P-gp), originally identified as a causative agent of multidrug-resistant cancer cells, plays an important role in ameliorating the disease phenotype. Using a Drosophila transgenic strain that expresses a stretch of 127 glutamine repeats, we demonstrate that enhancing P-gp levels reduces eye degeneration caused by expression of polyQ, whereas reducing it increases the severity of the disease. Increase in polyQ inclusion bodies represses the expression of mdr genes, suggesting a functional link between P-gp and polyQ. P-gp up-regulation restores the defects in the actin organization and precise array of the neuronal connections caused by inclusion bodies. ß-Catenin homolog, Armadillo, also interacts with P-gp and regulates the accumulation of inclusion bodies. These results thus show that P-gp and polyQ interact with each other, and changing P-gp levels can directly affect neurodegeneration.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Péptidos/genética , Péptidos/metabolismo , Expansión de Repetición de Trinucleótido , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/antagonistas & inhibidores , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Técnicas de Silenciamiento del Gen , Genes de Insecto , Genes MDR , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Modelos Biológicos , Degeneración Nerviosa/patología , Fenotipo , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Verapamilo/farmacología
18.
Curr Biol ; 23(14): 1349-54, 2013 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-23850283

RESUMEN

Inherited retinal degeneration in humans is caused by mutations in a wide spectrum of genes that regulate photoreceptor development and homeostasis. Many of these genes are structurally and functionally conserved in Drosophila, making the fly eye an ideal system in which to study the cellular and molecular basis of blindness. DLin-7, the ortholog of vertebrate MALS/Veli, is a core component of the evolutionarily conserved Crumbs complex. Mutations in any core member of the Crb complex lead to retinal degeneration in Drosophila. Strikingly, mutations in the human ortholog, CRB1, result in retinitis pigmentosa 12 (RP12) and Leber congenital amaurosis, two severe retinal dystrophies. Unlike Crumbs, DLin-7 is expressed not only in photoreceptor cells but also in postsynaptic lamina neurons. Here, we show that DLin-7 is required in postsynaptic neurons, but not in photoreceptors such as Crumbs, to prevent light-dependent retinal degeneration. At the photoreceptor synapse, DLin-7 acts as part of a conserved DLin-7/CASK/DlgS97 complex required to control the number of capitate projections and active zones, important specializations in the photoreceptor synapse that are essential for proper neurotransmission. These results are the first to demonstrate that a postsynaptically acting protein prevents light-dependent photoreceptor degeneration and describe a novel, Crumbs-independent mechanism for photoreceptor degeneration.


Asunto(s)
Moléculas de Adhesión Celular/genética , Proteínas de Drosophila/genética , Drosophila/genética , Luz , Células Fotorreceptoras de Invertebrados/metabolismo , Degeneración Retiniana/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Inmunoprecipitación , Microscopía Electrónica de Transmisión , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/ultraestructura , Células Fotorreceptoras de Invertebrados/patología , Degeneración Retiniana/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
19.
PLoS Genet ; 9(6): e1003559, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23754968

RESUMEN

Rhodopsin has been used as a prototype system to investigate G protein-coupled receptor (GPCR) internalization and endocytic sorting mechanisms. Failure of rhodopsin recycling upon light activation results in various degenerative retinal diseases. Accumulation of internalized rhodopsin in late endosomes and the impairment of its lysosomal degradation are associated with unregulated cell death that occurs in dystrophies. However, the molecular basis of rhodopsin accumulation remains elusive. We found that the novel norpA(P24) suppressor, diehard4, is responsible for the inability of endo-lysosomal rhodopsin trafficking and retinal degeneration in Drosophila models of retinal dystrophies. We found that diehard4 encodes Osiris 21. Loss of its function suppresses retinal degeneration in norpA(P24), rdgC(306), and trp(1), but not in rdgB(2), suggesting a common cause of photoreceptor death. In addition, the loss of Osiris 21 function shifts the membrane balance between late endosomes and lysosomes as evidenced by smaller late endosomes and the proliferation of lysosomal compartments, thus facilitating the degradation of endocytosed rhodopsin. Our results demonstrate the existence of negative regulation in vesicular traffic between endosomes and lysosomes. We anticipate that the identification of additional components and an in-depth description of this specific molecular machinery will aid in therapeutic interventions of various retinal dystrophies and GPCR-related human diseases.


Asunto(s)
Proteínas de Drosophila/genética , Endocitosis/genética , Proteínas de la Membrana/genética , Fosfolipasa C beta/genética , Distrofias Retinianas/genética , Rodopsina/genética , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Endosomas/genética , Endosomas/metabolismo , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Distrofias Retinianas/metabolismo , Distrofias Retinianas/patología , Rodopsina/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
20.
Hum Mol Genet ; 22(22): 4474-84, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23804749

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by progressive and selective loss of motor neurons. The discovery of mutations in the gene encoding an RNA-binding protein, TAR DNA-binding protein of 43 kD (TDP-43), in familial ALS, strongly implicated abnormalities in RNA processing in the pathogenesis of ALS, although the mechanisms whereby TDP-43 leads to neurodegeneration remain elusive. To clarify the mechanism of degeneration caused by TDP-43, we generated transgenic Drosophila melanogaster expressing a series of systematically modified human TDP-43 genes in the retinal photoreceptor neurons. Overexpression of wild-type TDP-43 resulted in vacuolar degeneration of the photoreceptor neurons associated with thinning of the retina, which was significantly exacerbated by mutations of TDP-43 linked to familial ALS or disrupting its nuclear localization signal (NLS). Remarkably, these degenerative phenotypes were completely normalized by addition of a mutation or deletion of the RNA recognition motif that abolishes the RNA binding ability of TDP-43. Altogether, our results suggest that RNA binding is key to the neurodegeneration caused by overexpression of TDP-43, and that abnormalities in RNA processing may be crucial to the pathogenesis of TDP-43 proteinopathy.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila melanogaster/genética , ARN/metabolismo , Proteinopatías TDP-43/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Animales Modificados Genéticamente , Sitios de Unión , Citosol/metabolismo , Proteínas de Unión al ADN/química , Modelos Animales de Enfermedad , Drosophila melanogaster/metabolismo , Humanos , Degeneración Nerviosa , Fenotipo , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Proteinopatías TDP-43/genética , Proteinopatías TDP-43/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...