Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuroreport ; 32(7): 562-568, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33850086

RESUMO

Mutations in the factor-induced-gene 4 (FIG 4) gene are associated with multiple disorders, including Charcot-Marie-Tooth disease (CMT), epilepsy with polymicrogyria, Yunis-Varón syndrome and amyotrophic lateral sclerosis. The wide spectrum of disorders associated with FIG 4 may be related to the dysregulated epigenetics. Using Gene Expression Omnibus, we found that HDAC1 binds to the FIG 4 gene locus in the genome of human CD4+ T cells. Rpd3 is a well-known Drosophila homolog of human HDAC1. We previously established Drosophila models targeting Drosophila FIG 4 (dFIG 4) that exhibited defective locomotive ability, abnormal synapse morphology at neuromuscular junctions, enlarged vacuoles in the fat body and aberrant compound eye morphology. Genetic crossing experiments followed by physiological and immunocytochemical analyses revealed that Rpd3 mutations suppressed these defects induced by dFIG 4 knockdown. This demonstrated Rpd3 to be an important epigenetic regulator of dFIG 4, suggesting that the inhibition of HDAC1 represses the pathogenesis of FIG 4-associated disorders, including CMT. Defects in epigenetic regulators, such as HDAC1, may also explain the diverse symptoms of FIG 4-associated disorders.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas de Drosophila/genética , Epigênese Genética , Histona Desacetilase 1/genética , Atividade Motora/genética , Junção Neuromuscular/genética , Animais , Doença de Charcot-Marie-Tooth/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/metabolismo , Junção Neuromuscular/metabolismo
2.
Exp Cell Res ; 386(1): 111711, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31704059

RESUMO

The eye imaginal disc-specific knockdown of dFIG4, a Drosophila homolog of FIG4 that is one of the Charcot-Marie-Tooth disease (CMT)-causing genes, induces an aberrant adult compound eye morphology, the so-called rough eye phenotype. We previously performed modifier screening on the dFIG4 knockdown-induced rough eye phenotype and identified several genes, including CR18854, encoding a long non-coding RNA (lncRNA) as genetic interactants with dFIG4. In the present study, in more extensive genetic screening, we found that the deletion of a gene locus encoding both Odorant rector 46a (Or46a) and lncRNA CR43467 effectively suppressed the rough eye phenotype induced by the knockdown of dFIG4. Both genes were located on the same locus, but oriented in opposite directions. In order to identify which of these genes is responsible for the suppression of the rough eye phenotype, we established a CR43467-specific knockdown line using the CRISPR-dCas9 system. By using this system, we demonstrated that the CR43467 gene, but not the Or46a gene, genetically interacted with the dFIG4 gene. The knockdown of CR43467 rescued the reductions in the length of synaptic branches and number of boutons at neuromuscular junctions induced by the knockdown of dFIG4. The vacuole enlargement phenotype induced by the fat body-specific dFIG4 knockdown was also effectively suppressed by the knockdown of CR43467. The knockdown of CR43467 also suppressed the rough eye phenotype induced by other peripheral neuropathy-related genes, such as dCOA7, dHADHB, and dPDHB. We herein identified another gene encoding lncRNA, CR43467 as a genetic interactant with the CMT-causing gene.


Assuntos
Genes Supressores , Monoéster Fosfórico Hidrolases/genética , RNA Longo não Codificante/genética , Animais , Olho Composto de Artrópodes/citologia , Olho Composto de Artrópodes/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Junção Neuromuscular/metabolismo , Fenótipo
3.
Exp Cell Res ; 385(2): 111673, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31614134

RESUMO

Mitochondrial dysfunction causes various diseases. Mutations in the SLC25A46 gene have been identified in mitochondrial diseases that are sometimes classified as Charcot-Marie-Tooth disease type 2, optic atrophy, and Leigh syndrome. A homolog of SLC25A46 was identified in Drosophila and designated as dSLC25A46 (CG5755). We previously established mitochondrial disease model targeting of dSLC25A46, which causes locomotive dysfunction and morphological defects at neuromuscular junctions, such as reduced synaptic branch lengths and decreased numbers of boutons. The diverse symptoms of mitochondrial diseases carrying mutations in SLC25A46 may be associated with the dysregulation of some epigenetic regulators. To investigate the involvement of epigenetic regulators in mitochondrial diseases, we examined candidate epigenetic regulators that interact with human SLC25A46 by searching Gene Expression Omnibus (GEO). We discovered that HDAC1 binds to several SLC25A46 genomic regions in human cultured CD4 (+) cells, and attempted to prove this in an in vivo Drosophila model. By demonstrating that Rpd3, Drosophila HDAC1, regulates the histone H4K8 acetylation state in dSLC25A46 genomic regions, we confirmed that Rpd3 is a novel epigenetic regulator modifying the phenotypes observed with the mitochondrial disease model targeting of dSLC25A46. The functional reduction of Rpd3 rescued the deficient locomotive ability and aberrant morphology of motoneurons at presynaptic terminals induced by the dSLC25A46 knockdown. The present results suggest that the inhibition of HDAC1 suppresses the pathogenic processes that lead to the degeneration of motoneurons in mitochondrial diseases.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Epigênese Genética , Histona Desacetilase 1/metabolismo , Locomoção , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Neurônios Motores/metabolismo , Proteínas de Transporte de Fosfato/genética , Animais , Células Cultivadas , Drosophila melanogaster , Código das Histonas , Histona Desacetilase 1/genética , Humanos , Neurônios Motores/patologia , Neurônios Motores/fisiologia
4.
Neuroreport ; 30(15): 1039-1047, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31503204

RESUMO

AlkB family proteins are enzymes that repair alkylated DNA and RNA by oxidative demethylation. Nine homologs have been identified and characterized in mammals. ALKBH1 is conserved among metazoans including Drosophila. Although the ALKBH1 mouse homolog, Alkbh1 functions in neurogenesis, it currently remains unclear whether ALKBH1 plays a role in neuronal disorders induced by ultraviolet-induced DNA damage. We herein demonstrated that the Drosophila ALKBH1 homolog, AlkB contributed to recovery from neuronal disorders induced by ultraviolet damage. The knockdown of AlkB resulted in not only learning defects but also altered crawling behavior in Drosophila larvae after ultraviolet irradiation. A molecular analysis revealed that AlkB contributed to the repair of ultraviolet-induced DNA damage in the central nervous system of larvae. Therefore, we propose that ALKBH1 plays a role in the repair of ultraviolet-induced DNA damage in central nervous system. Ultraviolet-induced DNA damage is involved in the pathogenesis of xeroderma pigmentosum, and has recently been implicated in Parkinson's disease. The present results will contribute to our understanding of neuronal diseases induced by ultraviolet-induced DNA damage.


Assuntos
Enzimas AlkB/fisiologia , Neurônios/patologia , Neurônios/efeitos da radiação , Raios Ultravioleta , Enzimas AlkB/genética , Animais , Sistema Nervoso Central/patologia , Sistema Nervoso Central/efeitos da radiação , Dano ao DNA/efeitos da radiação , Drosophila , Anormalidades do Olho/genética , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Larva , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/psicologia , Locomoção/efeitos da radiação
5.
Brain Res ; 1708: 207-219, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30578769

RESUMO

piRNAs, small non-coding RNAs, were considered to be restricted to germline cells. Although they have recently been detected in somatic cells including neurons, it remains unclear how piRNA biogenesis is involved in neuronal diseases. We herein examined the possible roles of Aubergine (Aub), a Piwi-family protein (PIWI) responsible for piRNA biogenesis, in the neuronal disorders, using the Cabeza (Caz) knockdown Drosophila. Caz is a Drosophila homologue of FUS, which is one of the genes causing amyotrophic lateral sclerosis (ALS). Aub overexpression enhanced the mobility defects accompanied by anatomical defects in motoneurons at neuromuscular junctions induced by the neuron-specific knockdown of Caz. In order to elucidate the underlying mechanisms, we examined pre-piRNA and mature-size piRNA levels under these conditions. qRT-PCR and RNA-seq analyses revealed that the Caz knockdown increased pre-piRNA levels, but reduced mature-size piRNA levels in the central nervous system (CNS), suggesting a role in the pre-piRNAs production. Aub overexpression did not increase mature-size piRNA levels. These results suggest that the accumulated pre-piRNAs are abnormal abortive pre-piRNAs that cannot be further processed by slicers, including Aub. We also demonstrated a relationship between Caz and pre-piRNAs in the CNS by RNA immunoprecipitation. Aub overexpression induced the abnormal cytoplasmic localization of Caz. Based on these results, we propose a model in which Caz knockdown-induced abnormal pre-piRNAs associate with Caz, then translocate and accumulate in the cytoplasm, a process that may be mediated by Aub. The novel roles for Caz and Aub demonstrated herein using the Caz-knockdown fly will contribute to a deeper understanding of the pathogenesis of ALS.


Assuntos
Proteínas de Drosophila/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , RNA Interferente Pequeno/biossíntese , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Drosophila melanogaster/metabolismo , Masculino , Neurônios Motores/metabolismo , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Processamento Pós-Transcricional do RNA , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/metabolismo , Fator de Transcrição TFIID/metabolismo
6.
Exp Neurol ; 310: 1-13, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30165075

RESUMO

Neuron-specific knockdown of the dFIG4 gene, a Drosophila homologue of human FIG4 and one of the causative genes for Charcot-Marie-Tooth disease (CMT), reduces the locomotive abilities of adult flies, as well as causing defects at neuromuscular junctions, such as reduced synaptic branch length in presynaptic terminals of the motor neurons in third instar larvae. Eye imaginal disc-specific knockdown of dFIG4 induces abnormal morphology of the adult compound eye, the rough eye phenotype. In this study, we carried out modifier screening of the dFIG4 knockdown-induced rough eye phenotype using a set of chromosomal deficiency lines on the second chromosome. By genetic screening, we detected 9 and 15 chromosomal regions whose deletions either suppressed or enhanced the rough eye phenotype induced by the dFIG4 knockdown. By further genetic screening with mutants of individual genes in one of these chromosomal regions, we identified the gene CR18854 that suppressed the rough eye phenotype and the loss-of-cone cell phenotype. The CR18854 gene encodes a long non-coding RNA (lncRNA) consisting of 2566 bases. Mutation and knockdown of CR18854 patially suppressed the enlarged lysosome phenotype induced by Fat body-specific knockdown of dFIG4. Further characterization of CR18854, and a few other lncRNAs in relation to dFIG4 in neuron, using neuron-specific dFIG4 knockdown flies indicated a genetic link between the dFIG4 gene and lncRNAs including CR18854 and hsrω. We also obtained data indicating genetic interaction between CR18854 and Cabeza, a Drosophila homologue of human FUS, which is one of the causing genes for amyotrophic lateral sclerosis (ALS). These results suggest that lncRNAs such as CR18854 and hsrω are involved in a common pathway in CMT and ALS pathogenesis.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Epistasia Genética/genética , Flavoproteínas/genética , Testes Genéticos , Mutação/genética , Monoéster Fosfórico Hidrolases/genética , RNA Longo não Codificante/genética , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Olho/ultraestrutura , Flavoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Lisossomos/genética , Lisossomos/ultraestrutura , Microscopia Eletrônica de Varredura , Movimento/fisiologia , Junção Neuromuscular/genética , Junção Neuromuscular/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Monoéster Fosfórico Hidrolases/metabolismo , Pupa/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retina/citologia
7.
Neuroreport ; 29(10): 856-862, 2018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29742619

RESUMO

Charcot-Marie-Tooth disease (CMT) is the most common hereditary neuropathy, and more than 80 CMT-causing genes have been identified to date. CMT4J is caused by a loss-of-function mutation in the Factor-Induced-Gene 4 (FIG4) gene, the product of which plays important roles in endosome-lysosome homeostasis. We hypothesized that Mammalian sterile 20-like kinase (MST) 1 and 2, tumor-suppressor genes, are candidate modifiers of CMT4J. We therefore examined the interaction between dFIG4 and Hippo (hpo), Drosophila counterparts of FIG4 and MSTs, respectively, using the Drosophila CMT4J model with the knockdown of dFIG4. The loss-of-function allele of hpo improved the rough eye morphology, locomotive dysfunction accompanied by structural defects in the presynaptic terminals of motoneurons, and the enlargement of lysosomes caused by the knockdown of dFIG4. Therefore, we identified hpo as a modifier of phenotypes induced by the knockdown of dFIG4. These results in Drosophila may provide an insight into the pathogenesis of CMT4J and contribute toward the development of disease-modifying therapy for CMT. We also identified the regulation of endosome-lysosome homeostasis as a novel probable function of Hippo/MST.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas de Drosophila/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lisossomos/genética , Mutação/genética , Proteínas Serina-Treonina Quinases/genética , Alelos , Animais , Drosophila melanogaster , Técnicas de Silenciamento de Genes/métodos , Neurônios Motores/metabolismo , Fenótipo
8.
Brain Res ; 1689: 30-44, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29604258

RESUMO

Mutations in SLC25A46 gene have been identified in mitochondrial diseases that are sometimes classified as Charcot-Marie-Tooth disease type 2, optic atrophy and Leigh syndrome. Human SLC25A46 functions as a transporter across the outer mitochondrial membrane. However, it is still unknown how the neurodegeneration occurring in these diseases relates to the loss of SLC25A46 function. Drosophila has CG5755 (dSLC25A46) as a single human SLC25A46 homolog. Here we established pan-neuron specific dSLC25A46 knockdown flies, and examined their phenotypes. Neuron specific knockdown of dSLC25A46 resulted in an impaired motility in both larvae and adults. Defects at neuromuscular junctions (NMJs), such as reduced synaptic branch length, decreased number and size of bouton, reduced density and size of active zone were also observed with the dSLC25A46 knockdown flies. Mitochondrial hyperfusion in synapse at NMJ, accumulation of reactive oxygen species and reduction of ATP were also observed in the dSLC25A46 knockdown flies. These results indicate that depletion of SLC25A46 induces mitochondrial defects accompanied with aberrant morphology of motoneuron and reduction of active zone that results in defect in locomotive ability. In addition, it is known that SLC25A46 mutations in human cause optic atrophy and knockdown of dSLC25A46 induces aberrant morphology of optic stalk of photoreceptor neurons in third instar larvae. Morphology and development of optic stalk of photoreceptor neurons in Drosophila are precisely regulated via cell proliferation and migration. Immunocytochemical analyses of subcellular localization of dSLC25A46 revealed that dSLC25A46 localizes not only in mitochondria, but also in plasma membrane. These observations suggest that in addition to the role in mitochondrial function, plasma membrane-localized dSLC25A46 plays a role in cell proliferation and/or migration to control optic stalk formation. The dSLC25A46 knockdown fly thus recapitulates most of the phenotypes in mitochondrial disease patients, providing a useful tool to study these diseases.


Assuntos
Modelos Animais de Doenças , Drosophila , Doenças Mitocondriais , Trifosfato de Adenosina/metabolismo , Animais , Animais Geneticamente Modificados , Membrana Celular/metabolismo , Membrana Celular/patologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Olho Composto de Artrópodes/crescimento & desenvolvimento , Olho Composto de Artrópodes/metabolismo , Olho Composto de Artrópodes/patologia , Drosophila/genética , Técnicas de Silenciamento de Genes , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Proteínas Mitocondriais/genética , Atividade Motora/fisiologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Proteínas de Transporte de Fosfato/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Glândulas Salivares/crescimento & desenvolvimento , Glândulas Salivares/metabolismo , Glândulas Salivares/patologia , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA