Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Hum Mol Genet ; 32(10): 1673-1682, 2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-36611007

RESUMEN

The abnormal expansion of GGGGCC hexanucleotide repeats within the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The accumulation of GGGGCC repeat-containing RNAs as RNA foci, and the deposition of dipeptide repeat proteins (DPR) produced from these repeat RNAs by unconventional translation are major pathological hallmarks of C9orf72-linked ALS/FTD (C9-ALS/FTD), and are both thought to play a crucial role in the pathogenesis of these diseases. Because GGGGCC repeat RNA is likely to be the most upstream therapeutic target in the pathogenic cascade of C9-ALS/FTD, lowering the cellular level of GGGGCC repeat RNA is expected to mitigate repeat RNA toxicity, and will therefore be a disease-modifying therapeutic strategy for the treatment of C9-ALS/FTD. In this study, we demonstrated using a Drosophila model of C9-ALS/FTD that elevated expression of a subset of human RNA-binding proteins that bind to GGGGCC repeat RNA, including hnRNPA3, IGF2BP1, hnRNPA2B1, hnRNPR and SF3B3, reduces the level of GGGGCC repeat RNA, resulting in the suppression of neurodegeneration. We further showed that hnRNPA3-mediated reduction of GGGGCC repeat RNA suppresses disease pathology, such as RNA foci and DPR accumulation. These results demonstrate that hnRNPA3 and other RNA-binding proteins negatively regulate the level of GGGGCC repeat RNA, and mitigate repeat RNA toxicity in vivo, indicating the therapeutic potential of the repeat RNA-lowering approach mediated by endogenous RNA-binding proteins for the treatment of C9-ALS/FTD.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedad de Pick , Animales , Humanos , Demencia Frontotemporal/patología , Esclerosis Amiotrófica Lateral/metabolismo , ARN/genética , ARN/metabolismo , Proteína C9orf72/genética , Drosophila/genética , Drosophila/metabolismo , Enfermedad de Pick/genética , Proteínas/genética , Dipéptidos/genética , Expansión de las Repeticiones de ADN/genética
2.
Hum Mol Genet ; 27(5): 823-836, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29309590

RESUMEN

Mutations in DNAJC13 gene have been linked to familial form of Parkinson's disease (PD) with Lewy pathology. DNAJC13 is an endosome-related protein and believed to regulate endosomal membrane trafficking. However, the mechanistic link between DNAJC13 mutation and α-synuclein (αSYN) pathology toward neurodegeneration remains poorly understood. In this study, we showed that PD-linked N855S-mutant DNAJC13 caused αSYN accumulation in the endosomal compartment, presumably due to defective cargo trafficking from the early endosome to the late and/or recycling endosome. In vivo experiments using human αSYN transgenic flies showed that mutant DNAJC13 not only increased the amount of insoluble αSYN in fly head but also induced dopaminergic neurodegeneration, rough eye phenotype and age-dependent locomotor impairment. Together, these findings suggest that DNAJC13 mutation perturbs multi-directional endosomal trafficking, resulting in the aberrant endosomal retention of αSYN, which might predispose to the neurodegenerative process that leads to PD.


Asunto(s)
Endosomas/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Chaperonas Moleculares/genética , Mutación , Enfermedad de Parkinson/genética , Actinas/metabolismo , Animales , Animales Modificados Genéticamente , Transporte Biológico , Células COS , Chlorocebus aethiops , Neuronas Dopaminérgicas/patología , Drosophila/genética , Endosomas/genética , Ojo/patología , Humanos , Proteínas de Filamentos Intermediarios/genética , Locomoción/genética , Chaperonas Moleculares/metabolismo , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/genética , Enfermedad de Parkinson/fisiopatología
3.
Brain ; 142(9): 2845-2859, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31312839

RESUMEN

Mutations in lysosomal genes increase the risk of neurodegenerative diseases, as is the case for Parkinson's disease. Here, we found that pathogenic and protective mutations in arylsulfatase A (ARSA), a gene responsible for metachromatic leukodystrophy, a lysosomal storage disorder, are linked to Parkinson's disease. Plasma ARSA protein levels were changed in Parkinson's disease patients. ARSA deficiency caused increases in α-synuclein aggregation and secretion, and increases in α-synuclein propagation in cells and nematodes. Despite being a lysosomal protein, ARSA directly interacts with α-synuclein in the cytosol. The interaction was more extensive with protective ARSA variant and less with pathogenic ARSA variant than wild-type. ARSA inhibited the in vitro fibrillation of α-synuclein in a dose-dependent manner. Ectopic expression of ARSA reversed the α-synuclein phenotypes in both cell and fly models of synucleinopathy, the effects correlating with the extent of the physical interaction between these molecules. Collectively, these results suggest that ARSA is a genetic modifier of Parkinson's disease pathogenesis, acting as a molecular chaperone for α-synuclein.


Asunto(s)
Cerebrósido Sulfatasa/fisiología , Chaperonas Moleculares/metabolismo , Mutación Missense , Enfermedad de Parkinson/metabolismo , Mutación Puntual , alfa-Sinucleína/metabolismo , Adulto , Anciano , Animales , Animales Modificados Genéticamente , Encéfalo/enzimología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células Cultivadas , Cerebrósido Sulfatasa/sangre , Cerebrósido Sulfatasa/genética , Demencia/sangre , Demencia/etiología , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Técnicas de Inactivación de Genes , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/psicología , Linaje , Agregación Patológica de Proteínas/genética , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/metabolismo
4.
Exp Cell Res ; 371(2): 311-321, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30092221

RESUMEN

Mutations in the Fused in Sarcoma (FUS) gene have been identified in familial ALS in human. Drosophila contains a single ortholog of human FUS called Cabeza (Caz). We previously established Drosophila models of ALS targeted to Caz, which developed the locomotive dysfunction and caused anatomical defects in presynaptic terminals of motoneurons. Accumulating evidence suggests that ALS and cancer share defects in many cellular processes. The Hippo pathway was originally discovered in Drosophila and plays a role as a tumor suppressor in mammals. We aimed to determine whether Hippo pathway genes modify the ALS phenotype using Caz knockdown flies. We found a genetic link between Caz and Hippo (hpo), the Drosophila ortholog of human Mammalian sterile 20-like kinase (MST) 1 and 2. Loss-of-function mutations of hpo rescued Caz knockdown-induced eye- and neuron-specific defects. The decreased Caz levels in nuclei induced by Caz knockdown were also rescued by loss of function mutations of hpo. Moreover, hpo mRNA level was dramatically increased in Caz knockdown larvae, indicating that Caz negatively regulated hpo. Our results demonstrate that hpo, Drosophila MST, is a novel modifier of Drosophila FUS. Therapeutic targets that inhibit the function of MST could modify the pathogenic processes of ALS.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Larva/genética , Neurogénesis/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al ARN/genética , Factor de Transcripción TFIID/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Ojo/metabolismo , Ojo/ultraestructura , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Larva/citología , Larva/crecimiento & desarrollo , Larva/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Degeneración Nerviosa , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factor de Transcripción TFIID/deficiencia
5.
Adv Exp Med Biol ; 1076: 63-78, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29951815

RESUMEN

Repeat expansion disorders are a group of inherited neuromuscular diseases, which are caused by expansion mutations of repeat sequences in the disease-causing genes. Repeat expansion disorders include a class of diseases caused by repeat expansions in the coding region of the genes, producing mutant proteins with amino acid repeats, mostly the polyglutamine (polyQ) diseases, and another class of diseases caused by repeat expansions in the noncoding regions, producing aberrant RNA with expanded repeats, which are called noncoding repeat expansion diseases. A variety of Drosophila disease models have been established for both types of diseases, and they have made significant contributions toward elucidating the molecular mechanisms of and developing therapies for these neuromuscular diseases.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila melanogaster , Enfermedades Neuromusculares , Expansión de Repetición de Trinucleótido , Animales , Humanos , Enfermedades Neuromusculares/genética , Expansión de Repetición de Trinucleótido/genética
6.
J Biol Chem ; 290(3): 1442-53, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25480790

RESUMEN

Oligomer formation and accumulation of pathogenic proteins are key events in the pathomechanisms of many neurodegenerative diseases, such as Alzheimer disease, ALS, and the polyglutamine (polyQ) diseases. The autophagy-lysosome degradation system may have therapeutic potential against these diseases because it can degrade even large oligomers. Although p62/sequestosome 1 plays a physiological role in selective autophagy of ubiquitinated proteins, whether p62 recognizes and degrades pathogenic proteins in neurodegenerative diseases has remained unclear. In this study, to elucidate the role of p62 in such pathogenic conditions in vivo, we used Drosophila models of neurodegenerative diseases. We found that p62 predominantly co-localizes with cytoplasmic polyQ protein aggregates in the MJDtr-Q78 polyQ disease model flies. Loss of p62 function resulted in significant exacerbation of eye degeneration in these flies. Immunohistochemical analyses revealed enhanced accumulation of cytoplasmic aggregates by p62 knockdown in the MJDtr-Q78 flies, similarly to knockdown of autophagy-related genes (Atgs). Knockdown of both p62 and Atgs did not show any additive effects in the MJDtr-Q78 flies, implying that p62 function is mediated by autophagy. Biochemical analyses showed that loss of p62 function delays the degradation of the MJDtr-Q78 protein, especially its oligomeric species. We also found that loss of p62 function exacerbates eye degeneration in another polyQ disease fly model as well as in ALS model flies. We therefore conclude that p62 plays a protective role against polyQ-induced neurodegeneration, by the autophagic degradation of polyQ protein oligomers in vivo, indicating its therapeutic potential for the polyQ diseases and possibly for other neurodegenerative diseases.


Asunto(s)
Autofagia , Proteínas de Drosophila/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Péptidos/química , Células Fotorreceptoras de Invertebrados/metabolismo , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/metabolismo , Animales , Citoplasma/metabolismo , Drosophila , Inmunohistoquímica , Microscopía Electrónica de Rastreo , Fosforilación , Células Fotorreceptoras de Invertebrados/ultraestructura , Desnaturalización Proteica , Pliegue de Proteína , Transgenes , Proteínas Ubiquitinadas/química
7.
Hum Mol Genet ; 23(13): 3467-80, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24497576

RESUMEN

In humans, mutations in the fused in sarcoma (FUS) gene have been identified in sporadic and familial forms of amyotrophic lateral sclerosis (ALS). Cabeza (Caz) is the Drosophila ortholog of human FUS. Previously, we established Drosophila models of ALS harboring Caz-knockdown. These flies develop locomotive deficits and anatomical defects in motoneurons (MNs) at neuromuscular junctions; these phenotypes indicate that loss of physiological FUS functions in the nucleus can cause MN degeneration similar to that seen in FUS-related ALS. Here, we aimed to explore molecules that affect these ALS-like phenotypes of our Drosophila models with eye-specific and neuron-specific Caz-knockdown. We examined several previously reported ALS-related genes and found genetic links between Caz and ter94, the Drosophila ortholog of human Valosin-containing protein (VCP). Genetic crossing the strongest loss-of-function allele of ter94 with Caz-knockdown strongly enhanced the rough-eye phenotype and the MN-degeneration phenotype caused by Caz-knockdown. Conversely, the overexpression of wild-type ter94 in the background of Caz-knockdown remarkably suppressed those phenotypes. Our data demonstrated that expression levels of Drosophila VCP ortholog dramatically modified the phenotypes caused by Caz-knockdown in either direction, exacerbation or remission. Our results indicate that therapeutic agents that up-regulate the function of human VCP could modify the pathogenic processes that lead to the degeneration of MNs in ALS.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Neuronas Motoras/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Factor de Transcripción TFIID/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Ciclo Celular/genética , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Ojo Compuesto de los Artrópodos/metabolismo , Ojo Compuesto de los Artrópodos/patología , Drosophila , Proteínas de Drosophila/genética , Mutación , Proteína FUS de Unión a ARN/genética , Proteínas de Unión al ARN/genética , Factor de Transcripción TFIID/genética , Proteína que Contiene Valosina
8.
Acta Neuropathol Commun ; 12(1): 20, 2024 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-38311779

RESUMEN

The abnormal aggregation of TDP-43 into cytoplasmic inclusions in affected neurons is a major pathological hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although TDP-43 is aberrantly accumulated in the neurons of most patients with sporadic ALS/FTD and other TDP-43 proteinopathies, how TDP-43 forms cytoplasmic aggregates remains unknown. In this study, we show that a deficiency in DCTN1, a subunit of the microtubule-associated motor protein complex dynactin, perturbs the dynamics of stress granules and drives the formation of TDP-43 cytoplasmic aggregation in cultured cells, leading to the exacerbation of TDP-43 pathology and neurodegeneration in vivo. We demonstrated using a Drosophila model of ALS/FTD that genetic knockdown of DCTN1 accelerates the formation of ubiquitin-positive cytoplasmic inclusions of TDP-43. Knockdown of components of other microtubule-associated motor protein complexes, including dynein and kinesin, also increased the formation of TDP-43 inclusions, indicating that intracellular transport along microtubules plays a key role in TDP-43 pathology. Notably, DCTN1 knockdown delayed the disassembly of stress granules in stressed cells, leading to an increase in the formation of pathological cytoplasmic inclusions of TDP-43. Our results indicate that a deficiency in DCTN1, as well as disruption of intracellular transport along microtubules, is a modifier that drives the formation of TDP-43 pathology through the dysregulation of stress granule dynamics.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteínas de Unión al ADN , Proteínas de Drosophila , Complejo Dinactina , Demencia Frontotemporal , Animales , Humanos , Esclerosis Amiotrófica Lateral/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila/metabolismo , Complejo Dinactina/genética , Demencia Frontotemporal/patología , Gránulos de Estrés , Proteínas de Drosophila/genética
9.
Sci Rep ; 13(1): 22826, 2023 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-38129650

RESUMEN

Nucleotide repeat expansion of GGGGCC (G4C2) in the non-coding region of C9orf72 is the most common genetic cause underlying amyotrophic lateral sclerosis and frontotemporal dementia. Transcripts harboring this repeat expansion undergo the translation of dipeptide repeats via a non-canonical process known as repeat-associated non-AUG (RAN) translation. In order to ascertain the essential components required for RAN translation, we successfully recapitulated G4C2-RAN translation using an in vitro reconstituted translation system comprising human factors, namely the human PURE system. Our findings conclusively demonstrate that the presence of fundamental translation factors is sufficient to mediate the elongation from the G4C2 repeat. Furthermore, the initiation mechanism proceeded in a 5' cap-dependent manner, independent of eIF2A or eIF2D. In contrast to cell lysate-mediated RAN translation, where longer G4C2 repeats enhanced translation, we discovered that the expansion of the G4C2 repeats inhibited translation elongation using the human PURE system. These results suggest that the repeat RNA itself functions as a repressor of RAN translation. Taken together, our utilization of a reconstituted RAN translation system employing minimal factors represents a distinctive and potent approach for elucidating the intricacies underlying RAN translation mechanism.


Asunto(s)
Proteína C9orf72 , Biosíntesis de Proteínas , Extensión de la Cadena Peptídica de Translación , Factores de Elongación de Péptidos/metabolismo , Humanos , Proteína C9orf72/genética , Sistema de Lectura Ribosómico , Iniciación de la Cadena Peptídica Traduccional , Técnicas In Vitro , Células HeLa , Esclerosis Amiotrófica Lateral/genética , Demencia Frontotemporal/genética
10.
Elife ; 122023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37461319

RESUMEN

Abnormal expansions of GGGGCC repeat sequence in the noncoding region of the C9orf72 gene is the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). The expanded repeat sequence is translated into dipeptide repeat proteins (DPRs) by noncanonical repeat-associated non-AUG (RAN) translation. Since DPRs play central roles in the pathogenesis of C9-ALS/FTD, we here investigate the regulatory mechanisms of RAN translation, focusing on the effects of RNA-binding proteins (RBPs) targeting GGGGCC repeat RNAs. Using C9-ALS/FTD model flies, we demonstrated that the ALS/FTD-linked RBP FUS suppresses RAN translation and neurodegeneration in an RNA-binding activity-dependent manner. Moreover, we found that FUS directly binds to and modulates the G-quadruplex structure of GGGGCC repeat RNA as an RNA chaperone, resulting in the suppression of RAN translation in vitro. These results reveal a previously unrecognized regulatory mechanism of RAN translation by G-quadruplex-targeting RBPs, providing therapeutic insights for C9-ALS/FTD and other repeat expansion diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Humanos , Esclerosis Amiotrófica Lateral/patología , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Demencia Frontotemporal/patología , ARN/metabolismo , Proteína FUS de Unión a ARN/genética , Proteínas de Unión al ARN/genética , Drosophila/genética
11.
Nat Commun ; 12(1): 236, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431896

RESUMEN

Synthetic small molecules modulating RNA structure and function have therapeutic potential for RNA diseases. Here we report our discovery that naphthyridine carbamate dimer (NCD) targets disease-causing r(UGGAA)n repeat RNAs in spinocerebellar ataxia type 31 (SCA31). Structural analysis of the NCD-UGGAA/UGGAA complex by nuclear magnetic resonance (NMR) spectroscopy clarifies the mode of binding that recognizes four guanines in the UGGAA/UGGAA pentad by hydrogen bonding with four naphthyridine moieties of two NCD molecules. Biological studies show that NCD disrupts naturally occurring RNA foci built on r(UGGAA)n repeat RNA known as nuclear stress bodies (nSBs) by interfering with RNA-protein interactions resulting in the suppression of nSB-mediated splicing events. Feeding NCD to larvae of the Drosophila model of SCA31 alleviates the disease phenotype induced by toxic r(UGGAA)n repeat RNA. These studies demonstrate that small molecules targeting toxic repeat RNAs are a promising chemical tool for studies on repeat expansion diseases.


Asunto(s)
Drosophila/genética , ARN/genética , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Células HeLa , Humanos , Intrones/genética , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación de Ácido Nucleico , Fenotipo , Fosforilación , Proteínas de Unión al ARN/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Temperatura
12.
PLoS One ; 14(6): e0218261, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31242217

RESUMEN

Parkinson's disease (PD) is one of the most common neurodegenerative diseases, which is characterized by progressive motor dysfunction as well as non-motor symptoms. Pathological and genetic studies have demonstrated that α-synuclein (αSyn) plays key roles in the pathogenesis of PD. Although several missense mutations in the αSyn gene have been identified as causes of familial PD, the mechanisms underlying the variance in the clinical phenotypes of familial PD caused by different mutations remain elusive. Here, we established novel Drosophila models expressing either wild-type (WT) αSyn or one of five αSyn mutants (A30P, E46K, H50Q, G51D, and A53T) using site-specific transgenesis, which express transgenes at equivalent levels. Expression of either WT or mutant αSyn in the compound eyes by the GMR-GAL4 driver caused mild rough eye phenotypes with no obvious difference among the mutants. Upon pan-neuronal expression by the nSyb-GAL4 driver, these αSyn-expressing flies showed a progressive decline in locomotor function. Notably, we found that E46K, H50Q, G51D, and A53T αSyn-expressing flies showed earlier onset of locomotor dysfunction than WT αSyn-expressing flies, suggesting their enhanced toxic effects. Whereas mRNA levels of WT and mutant αSyn were almost equivalent, we found that protein expression levels of E46K αSyn were higher than those of WT αSyn. In vivo chase experiments using the drug-inducible GMR-GeneSwitch driver demonstrated that degradation of E46K αSyn protein was significantly slower than WT αSyn protein, indicating that the E46K αSyn mutant gains resistance to degradation in vivo. We therefore conclude that our novel site-specific transgenic fly models expressing either WT or mutant αSyn are useful to explore the mechanisms by which different αSyn mutants gain toxic functions in vivo.


Asunto(s)
Mutación Missense , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila , Humanos , Enfermedad de Parkinson/patología , Proteolisis , alfa-Sinucleína/genética
13.
Am J Neurodegener Dis ; 7(1): 11-31, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29531866

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease characterized by the motor neuron degeneration that eventually leads to complete paralysis and death within 2-5 years after disease onset. One of the major pathological hallmark of ALS is abnormal accumulation of inclusions containing TAR DNA-binding protein-43 (TDP-43). TDP-43 is normally found in the nucleus, but in ALS, it localizes in the cytoplasm as inclusions as well as in the nucleus. Loss of nuclear TDP-43 functions likely contributes to neurodegeneration. TBPH is the Drosophila ortholog of human TDP-43. In the present study, we confirmed that Drosophila models harboring TBPH knockdown develop locomotive deficits and degeneration of motoneurons (MNs) due to loss of its nuclear functions, recapitulating the human ALS phenotypes. We previously suggested that ter94, the Drosophila ortholog of human Valosin-containing protein (VCP), is a modulator of degeneration in MNs induced by knockdown of Caz, the Drosophila ortholog of human FUS. In this study, to determine the effects of VCP on TDP-43-assosiated ALS pathogenic processes, we examined genetic interactions between TBPH and ter94. Overexpression of ter94 suppressed the compound eye degeneration caused by TBPH knockdown and suppressed the morbid phenotypes caused by neuron-specific TBPH knockdown, such as locomotive dysfunction and degeneration of MN terminals. Further immunocytochemical analyses revealed that the suppression is caused by restoring the cytoplasmically mislocalized TBPH back to the nucleus. In consistent with these observations, a loss-of-function mutation of ter94 enhanced the compound eye degeneration caused by TBPH knockdown, and partially enhanced the locomotive dysfunction caused by TBPH knockdown. Our data demonstrated that expression levels of ter94 influenced the phenotypes caused by TBPH knockdown, and indicate that reagents that up-regulate the function of human VCP could modify MN degeneration in ALS caused by TDP-43 mislocalization.

14.
Sci Rep ; 8(1): 11291, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30050143

RESUMEN

Fused in sarcoma (FUS) was identified as a component of typical inclusions in frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). In FTLD, both nuclear and cytoplasmic inclusions with wild-type FUS exist, while cytoplasmic inclusions with a mutant-form of FUS occur in many ALS cases. These observations imply that FUS plays a role across these two diseases. In this study, we examined the effect of several proteins including molecular chaperons on the aberrant eye morphology phenotype induced by overexpression of wild-type human FUS (hFUS) in Drosophila eye imaginal discs. By screening, we found that the co-expression of nucleophosmin-human myeloid leukemia factor 1 (NPM-hMLF1) fusion protein could suppress the aberrant eye morphology phenotype induced by hFUS. The driving of hFUS expression at 28 °C down-regulated levels of hFUS and endogenous cabeza, a Drosophila homolog of hFUS. The down-regulation was mediated by proteasome dependent degradation. Co-expression of NPM-hMLF1 suppressed this down-regulation. In addition, co-expression of NPM-hMLF1 partially rescued pharate adult lethal phenotype induced by hFUS in motor neurons. These findings with a Drosophila model that mimics FTLD provide clues for the development of novel FTLD therapies.


Asunto(s)
Animales Modificados Genéticamente , Degeneración Lobar Frontotemporal/patología , Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de la radiación , Drosophila , Ojo/embriología , Anomalías del Ojo/prevención & control , Humanos , Discos Imaginales/embriología , Proteínas Nucleares/genética , Nucleofosmina , Proteínas/genética , Proteína FUS de Unión a ARN/genética , Proteínas Recombinantes de Fusión/genética , Temperatura
15.
Neuron ; 94(1): 108-124.e7, 2017 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-28343865

RESUMEN

Microsatellite expansion disorders are pathologically characterized by RNA foci formation and repeat-associated non-AUG (RAN) translation. However, their underlying pathomechanisms and regulation of RAN translation remain unknown. We report that expression of expanded UGGAA (UGGAAexp) repeats, responsible for spinocerebellar ataxia type 31 (SCA31) in Drosophila, causes neurodegeneration accompanied by accumulation of UGGAAexp RNA foci and translation of repeat-associated pentapeptide repeat (PPR) proteins, consistent with observations in SCA31 patient brains. We revealed that motor-neuron disease (MND)-linked RNA-binding proteins (RBPs), TDP-43, FUS, and hnRNPA2B1, bind to and induce structural alteration of UGGAAexp. These RBPs suppress UGGAAexp-mediated toxicity in Drosophila by functioning as RNA chaperones for proper UGGAAexp folding and regulation of PPR translation. Furthermore, nontoxic short UGGAA repeat RNA suppressed mutated RBP aggregation and toxicity in MND Drosophila models. Thus, functional crosstalk of the RNA/RBP network regulates their own quality and balance, suggesting convergence of pathomechanisms in microsatellite expansion disorders and RBP proteinopathies.


Asunto(s)
Proteínas de Unión al ADN/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Repeticiones de Microsatélite/genética , Enfermedad de la Neurona Motora/genética , Pliegue del ARN/genética , Proteína FUS de Unión a ARN/genética , Ataxias Espinocerebelosas/genética , Anciano , Anciano de 80 o más Años , Animales , Animales Modificados Genéticamente , Expansión de las Repeticiones de ADN , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Humanos , Masculino , Persona de Mediana Edad , Chaperonas Moleculares/genética , Células PC12 , Biosíntesis de Proteínas/genética , Proteínas de Unión al ARN/genética , Ratas
16.
Curr Biol ; 26(17): 2223-32, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27498563

RESUMEN

Wolbachia, endosymbiotic bacteria prevalent in invertebrates, manipulate their hosts in a variety of ways: they induce cytoplasmic incompatibility, male lethality, male-to-female transformation, and parthenogenesis. However, little is known about the molecular basis for host manipulation by these bacteria. In Drosophila melanogaster, Wolbachia infection makes otherwise sterile Sex-lethal (Sxl) mutant females capable of producing mature eggs. Through a functional genomic screen for Wolbachia genes with growth-inhibitory effects when expressed in cultured Drosophila cells, we identified the gene WD1278 encoding a novel protein we call toxic manipulator of oogenesis (TomO), which phenocopies some of the Wolbachia effects in Sxl mutant D. melanogaster females. We demonstrate that TomO enhances the maintenance of germ stem cells (GSCs) by elevating Nanos (Nos) expression via its interaction with nos mRNA, ultimately leading to the restoration of germ cell production in Sxl mutant females that are otherwise without GSCs.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Proteínas de Unión al ARN/genética , Células Madre/metabolismo , Wolbachia/fisiología , Animales , Proteínas Bacterianas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/microbiología , Femenino , Células Germinativas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Wolbachia/genética
17.
Insect Biochem Mol Biol ; 35(8): 911-20, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15944086

RESUMEN

In Drosophila melanogaster, desat1 is involved in the synthesis of fatty acids (FAs), some of which are precursors in the production of unsaturated hydrocarbons (HCs) in position 7 (7-HC) that play an important role in mating behaviour. Three GS lines with P-element insertion in the desat1 promoter showed more or less decrease in 7-HC, depending on the site of insertion. The forced transcription of genomic 5'P-flanking sequence led to opposite effects upon 7-HC, depending on the orientation of the insertions. Homozygous GS12251 flies showed particularly low 7-HC levels and severely affected courtship parameters (courtship latency doubled, number of copulation attempts decreased by half). After transposon excision, the HC phenotype was reversed in most lines, showing that the location of the transposon was responsible for the mutant phenotype. In homozygous GS12251 flies, the amounts of FAs and desat1 transcripts were reduced by half, compared to the amounts in heterozygous or wild-type flies. Relative proportions among FAs were quite similar to those of wild-type, with the exception of a slight decrease in myristoleic, palmitoleic and vaccenic acid. As the reduction of desat1 activity in the mutant resulted in a large decrease in both unsaturated and saturated FAs, it could impair FA and lipid metabolism, as it is known in vertebrates.


Asunto(s)
Cortejo , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Ácido Graso Desaturasas/genética , Ácidos Grasos/metabolismo , Atractivos Sexuales/biosíntesis , Animales , Línea Celular , Proteínas de Drosophila , Femenino , Masculino , Mutación
18.
Genes Genet Syst ; 78(1): 29-36, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12655136

RESUMEN

In Drosophila females, mating is known to cause a reduction in life span, which is referred to as 'the cost of mating'. Since mating enhances oogenesis and oviposition, the cost of mating may be regarded as a trade-off between reproduction and longevity. We examined whether the cost of mating exists in mutant females that are unable to produce eggs. Three different mutant alleles of ovarian tumors (otu) and an allele of dunce (dnc(M11)) of Drosophila melanogaster were used to sterilize females. For all the female sterile mutants tested, mating dramatically decreased the life span of homozygous sterile females. Even more extreme shortening of life spans were observed when the sex peptide gene (Acp70A) was expressed in homozygous otu females, though they were virgin, indicating that the shortening in life span is due to seminal factors. These results indicate that the cost of mating is greater in females defective in oogenesis than that in normally fertile females.


Asunto(s)
Proteínas de Drosophila , Drosophila/genética , Infertilidad Femenina/genética , Mutación , Conducta Sexual Animal/fisiología , Animales , Animales Modificados Genéticamente , Drosophila/fisiología , Femenino , Heterocigoto , Péptidos y Proteínas de Señalización Intercelular , Longevidad/genética , Péptidos/genética , Péptidos/metabolismo , Semen/metabolismo
19.
PLoS One ; 7(1): e30045, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22291901

RESUMEN

Male killing, induced by different bacterial taxa of maternally inherited microorganisms, resulting in highly distorted female-biased sex-ratios, is a common phenomenon among arthropods. Some strains of the endosymbiont bacteria Wolbachia have been shown to induce this phenotype in particular insect hosts. High altitude populations of Drosophila bifasciata infected with Wolbachia show selective male killing during embryonic development. However, since this was first reported, circa 60 years ago, the interaction between Wolbachia and its host has remained unclear. Herein we show that D. bifasciata male embryos display defective chromatin remodeling, improper chromatid segregation and chromosome bridging, as well as abnormal mitotic spindles and gradual loss of their centrosomes. These defects occur at different times in the early development of male embryos leading to death during early nuclear division cycles or large defective areas of the cellular blastoderm, culminating in abnormal embryos that die before eclosion. We propose that Wolbachia affects the development of male embryos by specifically targeting male chromatin remodeling and thus disturbing mitotic spindle assembly and chromosome behavior. These are the first observations that demonstrate fundamental aspects of the cytological mechanism of male killing and represent a solid base for further molecular studies of this phenomenon.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Drosophila/microbiología , Infecciones por Rickettsiaceae/mortalidad , Caracteres Sexuales , Wolbachia/fisiología , Wolbachia/patogenicidad , Animales , Ensamble y Desensamble de Cromatina/genética , Drosophila/embriología , Drosophila/genética , Embrión no Mamífero , Desarrollo Embrionario/fisiología , Femenino , Predisposición Genética a la Enfermedad , Masculino , Meiosis/genética , Meiosis/fisiología , Infecciones por Rickettsiaceae/embriología , Infecciones por Rickettsiaceae/microbiología , Infecciones por Rickettsiaceae/veterinaria , Huso Acromático/genética , Huso Acromático/metabolismo , Huso Acromático/fisiología
20.
PLoS One ; 5(7): e11557, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20644630

RESUMEN

Walker-Warburg syndrome, a progressive muscular dystrophy, is a severe disease with various kinds of symptoms such as muscle weakness and occasional seizures. The genes of protein O-mannosyltransferases 1 and 2 (POMT1 and POMT2), fukutin, and fukutin-related protein are responsible for this syndrome. In our previous study, we cloned Drosophila orthologs of human POMT1 and POMT2 and identified their activity. However, the mechanism of onset of this syndrome is not well understood. Furthermore, little is known about the behavioral properties of the Drosophila POMT1 and POMT2 mutants, which are called rotated abdomen (rt) and twisted (tw), respectively. First, we performed various kinds of behavioral tests and described in detail the muscle structures by using these mutants. The mutant flies exhibited abnormalities in heavy exercises such as climbing or flight but not in light movements such as locomotion. Defective motor function in mutants appeared immediately after eclosion and was exaggerated with aging. Along with motor function, muscle ultrastructure in the tw mutant was altered, as seen in human patients. We demonstrated that expression of RNA interference (RNAi) for the rt gene and the tw mutant was almost completely lethal and semi-lethal, respectively. Flies expressing RNAi had reduced lifespans. These findings clearly demonstrate that Drosophila POMT mutants are models for human muscular dystrophy. We then observed a high density of myoblasts with an enhanced degree of apoptosis in the tw mutant, which completely lost enzymatic activity. In this paper, we propose a novel mechanism for the development of muscular dystrophy: POMT mutation causes high myoblast density and position derangement, which result in apoptosis, muscle disorganization, and muscle cell defects.


Asunto(s)
Apoptosis/fisiología , Distrofias Musculares/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Animales , Apoptosis/genética , Western Blotting , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Inmunohistoquímica , Locomoción/genética , Locomoción/fisiología , Longevidad/genética , Longevidad/fisiología , Manosiltransferasas/genética , Manosiltransferasas/metabolismo , Microscopía Electrónica de Transmisión , Distrofias Musculares/genética , Reacción en Cadena de la Polimerasa , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA