Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2794: 305-311, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38630239

RESUMEN

Brain defects often lead to motor dysfunctions in humans. Drosophila melanogaster has been one of the most useful organisms in the study of neuronal biology due to its similarities with humans and has contributed to a more detailed understanding of the effects of genetic dysfunctions in the brain on behavior. We herein present modified protocols for the crawling assay with larvae and the climbing assay with adult flies that are simple to perform as well as a series of commands for ImageJ to automatically analyze data for the crawling assay.


Asunto(s)
Artrópodos , Drosophila , Adulto , Humanos , Animales , Larva , Drosophila melanogaster , Bioensayo
2.
Biochem Biophys Res Commun ; 578: 35-41, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34536827

RESUMEN

Neuron-restrictive silencing factor (NRSF), also known as RE-1 silencing transcription factor (REST), has pivotal functions in many neuron-specific genes. Previous studies revealed that neuron-specific alternative splicing (AS) of REST produces divergent forms of REST variants and provides regulatory complexity in the nervous system. However, the biological significance of these variants in the regulation of neuronal activities remains to be clarified. Here, we revealed that Charlatan (Chn), a Drosophila REST-like molecule, is also regulated by neuron-specific AS. Neuron-specific AS produced six divergent variants of Chn proteins, one of which preferentially localized to axons. A small sequence of this variant was especially important for the axonal localization. Our data suggest that some variants have roles beyond the transcriptional regulation of neuronal activities.


Asunto(s)
Axones/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Empalme Alternativo , Animales , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas Represoras/genética , Factores de Transcripción/metabolismo
3.
Adv Exp Med Biol ; 1076: 25-40, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29951813

RESUMEN

Alzheimer's disease (AD) is the most epidemic neuronal dysfunctions among elderly people. It is accompanied by neuronal disorders along with learning and memory defects, as well as massive neurodegeneration phenotype. The presence of intracellular neurofibrillary tangles (NFTs) and extracellular amyloid plaques, called senile plaques (SPs), and brain atrophy are typically observed in the brains of AD patients. It has been over 20 years since the discovery that small peptide, called beta-amyloid (Aß), has pivotal role for the disease formation. Since then, a variety of drugs have been developed to cure AD; however, there is currently no effective drug for the disorder. This therapeutic void reflects lacks of ideal model system, which can evaluate the progression of AD in a short period. Recently, large numbers of AD model system have been established using Drosophila melanogaster by overproducing Aß molecules in the brain. These systems successfully reflect some of the symptoms along with AD. In this review, we would like to point out "pros and cons" of Drosophila AD models.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Drosophila melanogaster , Animales , Humanos
4.
Sci Rep ; 8(1): 8345, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29844375

RESUMEN

The assembly and deposition of amyloid ß protein (Aß) is a fundamental event during the early stages of Alzheimer's disease (AD) and cerebral amyloid angiopathy. A growing body of evidence indicates that gangliosides form a pathological platform for the generation of ganglioside-bound Aß, which facilitates the assembly of soluble Aßs; however, the molecular mechanisms underlying the binding of Aß to gangliosides in the brain remain unclear due to the lack of an in vivo system that may address this issue. In insects, including the fruit fly Drosophila melanogaster, gangliosides are not intrinsically present at a detectable level. We herein demonstrate that ganglioside expression is inducible in Drosophila via the expression of transgenes of ganglioside synthesis enzymes and the feeding of exogenous sialic acid, and also that the induction of ganglioside synthesis significantly accelerates Aß assembly in vivo. Our results support the hypothesis that gangliosides are responsible for Aß assembly in vivo and also provide an opportunity to develop a valuable model for basic research as well as a therapeutic strategy for AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Gangliósidos/metabolismo , Acetiltransferasas/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Gangliósido G(M1)/metabolismo , Galactosiltransferasas/metabolismo , Humanos , Ácido N-Acetilneuramínico/farmacología , Unión Proteica/fisiología , Transgenes/genética
5.
Hum Mol Genet ; 26(23): 4642-4656, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973191

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder among the elderly. During the progression of AD, massive neuronal degeneration occurs in the late stage of the disease; however, the molecular mechanisms responsible for this neuronal loss remain unknown. AßpE3-42 (an N-terminal-truncated amyloid-ß peptide that begins with pyroglutamate at the third position) is produced during late-stage AD. It also aggregates more rapidly in vitro and exhibits greater toxicity in neurons than full-length Aß1-42. In the present study, we established a Drosophila melanogaster model that expresses Aß3-42E3Q, which effectively produces AßpE3-42, and investigated the function of AßpE3-42 using the photoreceptor neurons of Drosophila. AßpE3-42 induced caspase-dependent apoptosis and caused progressive degeneration in photoreceptor neurons. Mutations in ER stress response genes or the administration of an inhibitor of the ER stress response markedly suppressed the degeneration phenotype, suggesting that the ER stress response plays an important role in neurodegeneration caused by AßpE3-42. We also confirmed that human Tau-dependent apoptotic induction was strongly enhanced by AßpE3-42. Thus, AßpE3-42 expression system in the fly may be a promising new tool for studying late-onset neurodegeneration in AD.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Estrés del Retículo Endoplásmico/fisiología , Enfermedades Neurodegenerativas/metabolismo , Fragmentos de Péptidos/biosíntesis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Apoptosis/fisiología , Encéfalo/metabolismo , Caspasas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Ácido Pirrolidona Carboxílico/metabolismo
6.
Acta Neuropathol Commun ; 4(1): 109, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27717375

RESUMEN

Several species of ß-amyloid peptides (Aß) exist as a result of differential cleavage from amyloid precursor protein (APP) to yield various C-terminal Aß peptides. Several N-terminal modified Aß peptides have also been identified in Alzheimer's disease (AD) brains, the most common of which is pyroglutamate-modified Aß (AßpE3-42). AßpE3-42 peptide has an increased propensity to aggregate, appears to accumulate in the brain before the appearance of clinical symptoms of AD, and precedes Aß1-42 deposition. Moreover, in vitro studies have shown that AßpE3-42 can act as a seed for full length Aß1-42. In this study, we characterized the Drosophila model of AßpE3-42 toxicity by expressing the peptide in specific sets of neurons using the GAL4-UAS system, and measuring different phenotypic outcomes. We found that AßpE3-42 peptide had an increased propensity to aggregate. Expression of AßpE3-42 in the neurons of adult flies led to behavioural dysfunction and shortened lifespan. Expression of AßpE3-42 constitutively in the eyes led to disorganised ommatidia, and activation of the c-Jun N-terminal kinase (JNK) signaling pathway. The eye disruption was almost completely rescued by co-expressing a candidate Aß degrading enzyme, neprilysin2. Furthermore, we found that neprilysin2 was capable of degrading AßpE3-42. Also, we tested the seeding hypothesis for AßpE3-42 in vivo, and measured its effect on Aß1-42 levels. We found that Aß1-42 levels were significantly increased when Aß1-42 and AßpE3-42 peptides were co-expressed. Furthermore, we found that AßpE3-42 enhanced Aß1-42 toxicity in vivo. Our findings implicate AßpE3-42 as an important source of toxicity in AD, and suggest that its specific degradation could be therapeutic.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/toxicidad , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ojo/metabolismo , Ojo/patología , Femenino , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Actividad Motora/fisiología , Neprilisina/genética , Neprilisina/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/toxicidad , Estabilidad Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Am J Neurodegener Dis ; 5(1): 62-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27073743

RESUMEN

Transducin ß-like 1 (TBL1), a transcriptional co-repressor complex, is a causative factor for late-onset hearing impairments. Transcriptional co-repressor complexes play pivotal roles in gene expression by making a complex with divergent transcription factors. However, it remained to be clarified how co-repressor complex regulates cellular survival. We herein demonstrated that ebi, a Drosophila homologue of TBL1, suppressed photoreceptor cell degeneration in the presence of excessive innate immune signaling. We also showed that the balance between NF-κB and AP-1 is a key component of cellular survival under stress conditions. Given that Ebi plays an important role in innate immune responses by regulating NF-κB activity and inhibition of apoptosis induced by associating with AP-1, it may be involved in the regulation of photoreceptor cell survival by modulating cross-talk between NF-κB and AP-1.

8.
Aging (Albany NY) ; 8(3): 427-39, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26959388

RESUMEN

Increasing evidence indicates that defects in the sensory system are highly correlated with age-related neurodegenerative diseases, including Alzheimer's disease (AD). This raises the possibility that sensory cells possess some commonalities with neurons and may provide a tool for studying AD. The sensory system, especially the auditory system, has the advantage that depression in function over time can easily be measured with electrophysiological methods. To establish a new mouse AD model that takes advantage of this benefit, we produced transgenic mice expressing amyloid-ß (Aß), a causative element for AD, in their auditory hair cells. Electrophysiological assessment indicated that these mice had hearing impairment, specifically in high-frequency sound perception (>32 kHz), at 4 months after birth. Furthermore, loss of hair cells in the basal region of the cochlea, which is known to be associated with age-related hearing loss, appeared to be involved in this hearing defect. Interestingly, overexpression of human microtubule-associated protein tau, another factor in AD development, synergistically enhanced the Aß-induced hearing defects. These results suggest that our new system reflects some, if not all, aspects of AD progression and, therefore, could complement the traditional AD mouse model to monitor Aß-induced neuronal dysfunction quantitatively over time.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/metabolismo , Modelos Animales de Enfermedad , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva de Alta Frecuencia/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Femenino , Células Ciliadas Auditivas/patología , Pérdida Auditiva de Alta Frecuencia/metabolismo , Pérdida Auditiva de Alta Frecuencia/patología , Masculino , Ratones Transgénicos , Proteínas tau/metabolismo
9.
PLoS One ; 10(11): e0141457, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26524764

RESUMEN

The innate immune response and stress-induced apoptosis are well-established signaling pathways related to cellular defense. NF-κB and AP-1 are redox-sensitive transcription factors that play important roles in those pathways. Here we show that Ebi, a Drosophila homolog of the mammalian co-repressor molecule transducin ß-like 1 (TBL1), variously regulates the expression of specific genes that are targets of redox-sensitive transcription factors. In response to different stimuli, Ebi activated gene expression to support the acute immune response in fat bodies, whereas Ebi repressed genes that are involved in apoptosis in photoreceptor cells. Thus, Ebi seems to act as a regulatory switch for genes that are activated or repressed in response to different external stimuli. Our results offer clear in vivo evidence that the Ebi-containing co-repressor complex acts in a distinct manner to regulate transcription that is required for modulating the output of various processes during Drosophila development.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/inmunología , Cuerpo Adiposo/inmunología , Proteínas de Unión al GTP/metabolismo , Animales , Apoptosis , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Inmunidad Innata , Oxidación-Reducción , Células Fotorreceptoras de Invertebrados/citología , Regiones Promotoras Genéticas , Transducción de Señal , Transcripción Genética
10.
Sci Rep ; 5: 10821, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26027742

RESUMEN

Methylene blue (MB) inhibits the aggregation of tau, a main constituent of neurofibrillary tangles. However, MB's mode of action in vivo is not fully understood. MB treatment reduced the amount of sarkosyl-insoluble tau in Drosophila that express human wild-type tau. MB concurrently ameliorated the climbing deficits of transgenic tau flies to a limited extent and diminished the climbing activity of wild-type flies. MB also decreased the survival rate of wild-type flies. Based on its photosensitive efficacies, we surmised that singlet oxygen generated through MB under light might contribute to both the beneficial and toxic effects of MB in vivo. We identified rose bengal (RB) that suppressed tau accumulation and ameliorated the behavioral deficits to a lesser extent than MB. Unlike MB, RB did not reduce the survival rate of flies. Our findings indicate that singlet oxygen generators with little toxicity may be suitable drug candidates for treating tauopathies.


Asunto(s)
Oxígeno Singlete/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Animales Modificados Genéticamente , Autofagia , Conducta Animal , Modelos Animales de Enfermedad , Drosophila , Femenino , Humanos , Masculino , Azul de Metileno/farmacología , Estrés Oxidativo , Solubilidad , Tauopatías/tratamiento farmacológico , Proteínas tau/química
11.
Development ; 141(13): 2621-32, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24961799

RESUMEN

Intestinal stem cells (ISCs) in the adult Drosophila midgut can respond to tissue damage and support repair. We used genetic manipulation to increase the number of ISC-like cells in the adult midgut and performed gene expression profiling to identify potential ISC regulators. A detailed analysis of one of these potential regulators, the zinc-finger protein Charlatan, was carried out. MARCM clonal analysis and RNAi in precursor cells showed that loss of Chn function caused severe ISC division defects, including loss of EdU incorporation, phosphorylated histone 3 staining and expression of the mitotic protein Cdc2. Loss of Charlatan also led to a much reduced histone acetylation staining in precursor cells. Both the histone acetylation and ISC division defects could be rescued by the simultaneous decrease of the Histone Deacetylase 2. The overexpression of Charlatan blocked differentiation reversibly, but loss of Charlatan did not lead to automatic differentiation. The results together suggest that Charlatan does not simply act as an anti-differentiation factor but instead functions to maintain a chromatin structure that is compatible with stem cell properties, including proliferation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Drosophila/fisiología , Drosophila/genética , Intestinos/citología , Células Madre/fisiología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular/genética , Drosophila/fisiología , Perfilación de la Expresión Génica , Análisis por Micromatrices , Microscopía Fluorescente , Interferencia de ARN
12.
Dev Growth Differ ; 56(5): 358-67, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24738783

RESUMEN

Neuronal network consists of many types of neuron and glial cells. This diversity is guaranteed by the constant cell proliferation of neuronal stem cells following stop cell cycle re-entry, which leads to differentiation during development. Neuronal differentiation occurs mainly at the specific cell cycle phase, the G1 phase. Therefore, cell cycle exit at the G1 phase is quite an important issue in understanding the process of neuronal cell development. Recent studies have revealed that aberrant S phase re-entry from the G1 phase often links cellular survival. In this review we discuss the different types of G1 arrest on the process of neuronal development in Drosophila. We also describe the issue that aberrant S phase entry often causes apoptosis, and the same mechanism might contribute to sensory organ defects, such as deafness.


Asunto(s)
Drosophila/citología , Drosophila/metabolismo , Animales , Apoptosis , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular , Drosophila/embriología , Proteínas de Drosophila/metabolismo , Fase G1 , Proteínas de Unión al GTP/metabolismo , Humanos , Modelos Animales , Neurogénesis , Fase S
13.
Am J Neurodegener Dis ; 3(3): 134-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25628964

RESUMEN

Aging is a major risk factor for Alzheimer's disease (AD). Aggregation of amyloid beta (Aß) in cerebral cortex and hippocampus is a hallmark of AD. Many factors have been identified as causative elements for onset and progression of AD; for instance, tau seems to mediate the neuronal toxicity of Aß, and downregulation of macroautophagy (autophagy) is thought to be a causative element of AD pathology. Expression of autophagy-related genes is reduced with age, which leads to increases in oxidative stress and aberrant protein accumulation. In this study, we found that expression of the autophagy-related genes atg1, atg8a, and atg18 in Drosophila melanogaster was regulated with aging as well as their own activities. In addition, the level of atg18 was maintained by dfoxo (foxo) and dsir2 (sir2) activities in concert with aging. These results indicate that some autophagy-related gene expression is regulated by foxo/sir2-mediated aging processes. We further found that reduced autophagy activity correlated with late-onset neuronal dysfunction caused by neuronal induction of Aß. These data support the idea that age-related dysfunction of autophagy is a causative element in onset and progression of AD.

14.
Genes Cells ; 18(11): 934-45, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23919485

RESUMEN

The IMD pathway is one of the major regulators of the innate immune response in Drosophila. Although extensive analysis of the IMD pathway has been carried out, precise mechanisms for how each target gene of the pathway is down-regulated remain to be clarified. Here, we carried out genetic screening and found that fat facets (faf), which encodes a deubiquitinating enzyme, inhibited the expression of the target genes of the IMD pathway. Overexpression of faf suppressed the infection-induced expression of Diptericin and increased susceptibility to bacterial infection in flies, whereas faf loss-of-function mutants decreased susceptibility. Time course analysis revealed that specific subsets of the target genes of the IMD pathway were affected by faf. Biochemical analysis showed that Faf made a complex with Imd, and both Faf and Imd were polyubiquitinated when they were co-overexpressed. Given that faf-dependent Imd polyubiquitination did not seem to cause protein degradation of Imd, Faf might inhibit the IMD pathway by modulating the state of Imd ubiquitination and/or stability.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/inmunología , Endopeptidasas/metabolismo , Adenosina Monofosfato/genética , Adenosina Monofosfato/metabolismo , Animales , Bacillus subtilis , Drosophila/metabolismo , Drosophila/microbiología , Endopeptidasas/genética , Enterobacter cloacae , Inmunidad Innata , Mutación , Transducción de Señal , Ubiquitinación
15.
Genes Cells ; 18(11): 909-20, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23919509

RESUMEN

As multicellular organisms develop, many cells permanently stop dividing and undergo terminal differentiation. The G1 phase of the cell cycle is thought to be the critical decision point for differentiation. Many growth factors, such as epidermal growth factor, are involved in regulating the G1 to S phase transition, and aberrant activation of growth factor signaling is one of the critical causes of tumor formation. Therefore, each cell must have proper mechanisms to suppress inappropriate/excessive activation of growth factor signaling, but the underlying molecular mechanisms remain undefined. Here, we found that ebi, a Drosophila homologue of genes encoding transducin-ß-like 1 and transducin-ß-like 1-related protein, mitigated excess growth stimulation by taking advantage of its distinct epigenetic functions. Ebi acted as a corepressor of transcription by forming a complex with retinoblastoma family protein (RBF), a Drosophila homologue of retinoblastoma, and regulating the expression of specific target genes of the Rbf/E2F pathway. Furthermore, ebi also sustained expression of certain genes, including Rbf, encoding factors that inhibit progression out of G1. Our genetic studies suggest that the antagonistic function of ebi against the Polycomb group silencing complex plays a role in the G1/S phase transition.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Epigénesis Genética , Proteínas de Unión al GTP/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Ojo Compuesto de los Artrópodos/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores ErbB/genética , Proteínas del Ojo/metabolismo , Fase G1 , Proteínas de Unión al GTP/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas del Grupo Polycomb/antagonistas & inhibidores , Proteína de Retinoblastoma/metabolismo , Fase S , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
16.
PLoS One ; 7(5): e37028, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666340

RESUMEN

Sensory organs are constantly exposed to physical and chemical stresses that collectively threaten the survival of sensory neurons. Failure to protect stressed neurons leads to age-related loss of neurons and sensory dysfunction in organs in which the supply of new sensory neurons is limited, such as the human auditory system. Transducin ß-like protein 1 (TBL1) is a candidate gene for ocular albinism with late-onset sensorineural deafness, a form of X-linked age-related hearing loss. TBL1 encodes an evolutionarily conserved F-box-like and WD40 repeats-containing subunit of the nuclear receptor co-repressor/silencing mediator for retinoid and thyroid hormone receptor and other transcriptional co-repressor complexes. Here we report that a Drosophila homologue of TBL1, Ebi, is required for maintenance of photoreceptor neurons. Loss of ebi function caused late-onset neuronal apoptosis in the retina and increased sensitivity to oxidative stress. Ebi formed a complex with activator protein 1 (AP-1) and was required for repression of Drosophila pro-apoptotic and anti-apoptotic genes expression. These results suggest that Ebi/AP-1 suppresses basal transcription levels of apoptotic genes and thereby protects sensory neurons from degeneration.


Asunto(s)
Apoptosis/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Proteínas de Unión al GTP/metabolismo , Silenciador del Gen , Células Fotorreceptoras/citología , Factor de Transcripción AP-1/metabolismo , Animales , Sitios de Unión , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Supervivencia Celular/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Masculino , Neuropéptidos/genética , Células Fotorreceptoras/metabolismo , Regiones Promotoras Genéticas/genética , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Eliminación de Secuencia , Factores de Tiempo
17.
Aging (Albany NY) ; 3(11): 1098-109, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22156377

RESUMEN

The O-type forkhead domain transcription factor (FOXO) is involved in many biological processes such as aging, the oxidative stress response, and growth regulation. FOXO activity is tightly controlled within cells. In particular, growth factor signaling pathways and the oxidative stress response can both stimulate nuclear translocation of this transcription factor. Here, we show that tetrahydrocurcumin (THC), a curcumin metabolite, regulates the oxidative stress response and aging via FOXO. In NIH3T3 cells, THC induced nuclear accumulation of FOXO4, a member of the FOXO family of transcription factors, by inhibiting phosphorylation of protein kinase B (PKB)/Akt. In Drosophila melanogaster, THC attenuated the oxidative stress response, an effect that was blocked in a foxo mutant background. THC also extended the life span of Drosophila under normal conditions, and loss of either foxo or Sir2 activity eliminated this effect. Based on these results, THC may regulate the aging process via an evolutionarily conserved signaling pathway that includes both foxo and Sir2.


Asunto(s)
Curcumina/análogos & derivados , Factores de Transcripción Forkhead/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Western Blotting , Curcumina/farmacología , Drosophila melanogaster , Inmunohistoquímica , Esperanza de Vida , Ratones , Células 3T3 NIH , Estrés Oxidativo/fisiología
18.
Genes Cells ; 16(8): 896-909, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21762412

RESUMEN

Sensory bristle formation in Drosophila is a well-characterized system for studying sensory organ development at the molecular level. The master proneural genes of the achaete-scute (ac-sc) complex, which encode basic-helix-loop-helix (bHLH) transcription factors, are necessary and sufficient for sensory bristle formation. charlatan (chn) was originally identified as a transcriptional activator of ac-sc gene expression through interaction with its enhancer, an activity that promotes sensory bristle development. In contrast, Chn was also identified as a functional homologue of mammalian neuron-restrictive silencing factor or RE1 silencing transcription factor (NRSF/REST), an important transcriptional repressor during vertebrate neurogenesis and stem cell development that acts through epigenetic gene silencing. Here, we report that Chn acts as a repressor of extramacrochaetae (emc) and hairy, molecules that inhibit ac-sc expression. This double-negative mechanism, together with direct activation via the achaete enhancer, increases expression of achaete and ensures robust development of sensory neurons. A mutation in the C-terminal repressor motif of Chn, which causes Chn to lose its repression activity, converted Chn to an activator of emc and hairy, suggesting that Chn is a dual functional regulator of transcription. Because chn-like sequences are found among arthropods, regulation of neuronal development by Chn-like molecules may be widely conserved.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Represoras/metabolismo , Células Receptoras Sensoriales/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Evolución Molecular , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Represoras/genética , Alineación de Secuencia , Factores de Transcripción/genética , Transcripción Genética , Activación Transcripcional
19.
Development ; 135(7): 1355-64, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18305002

RESUMEN

The downregulation of E-cadherin by Src promotes epithelial to mesenchymal transition and tumorigenesis. However, a simple loss of cell adhesion is not sufficient to explain the diverse developmental roles of Src and metastatic behavior of viral Src-transformed cells. Here, we studied the functions of endogenous and activated forms of Drosophila Src in the context of tracheal epithelial development, during which extensive remodeling of adherens junctions takes place. We show that Src42A is selectively activated in the adherens junctions of epithelia undergoing morphogenesis. Src42A and Src64B are required for tracheal development and to increase the rate of adherens junction turnover. The activation of Src42A caused opposing effects: it reduced the E-cadherin protein level but stimulated transcription of the E-cadherin gene through the activation of Armadillo and TCF. This TCF-dependent pathway was essential for the maintenance of E-cadherin expression and for tissue integrity under conditions of high Src activity. Our data suggest that the two opposing outcomes of Src activation on E-cadherin facilitate the efficient exchange of adherens junctions, demonstrating the key role of Src in the maintenance of epithelial integrity.


Asunto(s)
Uniones Adherentes/metabolismo , Células Epiteliales/citología , Tráquea/citología , Familia-src Quinasas/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Drosophila/embriología , Proteínas de Drosophila/metabolismo , Activación Enzimática , Morfogénesis , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Tráquea/metabolismo
20.
Genetics ; 176(2): 927-36, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17435247

RESUMEN

Allelic mutants exhibiting growth defects in Drosophila were isolated. Molecular cloning identified the responsible gene as a budding yeast Tim50 ortholog, and thus it was named tiny tim 50 (ttm50). The weak allele (ttm50(Gp99)) produced small flies due to reduced cell size and number, and growth terminated at the larval stage in the strong alleles (ttm50(IE1) and ttm50(IE2)). Twin-spot analysis showed fewer cells in ttm50(Gp99) clones, whereas ttm50(IE1) clones did not proliferate, suggesting that the gene has an essential cellular function. Tim50 is known to maintain mitochondrial membrane potential (MMP) while facilitating inner-membrane protein transport. We found that tagged Ttm50 also localized to mitochondria and that mitochondrial morphology and MMP were affected in mutants, indicating that mitochondrial dysfunction causes the developmental phenotype. Conversely, ttm50 overexpression increased MMP and apoptosis. Co-expression of p35 suppressed this apoptosis, resulting in cell overproliferation. Interestingly, ttm50 transcription was tissue specific, corresponding to elevated MMP in the larval midgut, which was decreased in the mutant. The correlation of ttm50 expression levels with differences in MMP match its proposed role in mitochondrial permeability barrier maintenance. Thus a mitochondrial protein translocase component can play active roles in regulating metabolic levels, possibly for modulation of physiological function or growth in development.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Proteínas Mitocondriales/genética , Consumo de Oxígeno/fisiología , Animales , División Celular , ADN/genética , ADN/aislamiento & purificación , Cartilla de ADN , ADN Complementario/genética , Drosophila/citología , Drosophila/fisiología , Femenino , Mutagénesis Insercional , Mutagénesis Sitio-Dirigida , Óvulo/fisiología , Cromosoma X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA