Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33593901

RESUMEN

Reduced activity of insulin/insulin-like growth factor signaling (IIS) increases healthy lifespan among diverse animal species. Downstream of IIS, multiple evolutionarily conserved transcription factors (TFs) are required; however, distinct TFs are likely responsible for these effects in different tissues. Here we have asked which TFs can extend healthy lifespan within distinct cell types of the adult nervous system in Drosophila Starting from published single-cell transcriptomic data, we report that forkhead (FKH) is endogenously expressed in neurons, whereas forkhead-box-O (FOXO) is expressed in glial cells. Accordingly, we find that neuronal FKH and glial FOXO exert independent prolongevity effects. We have further explored the role of neuronal FKH in a model of Alzheimer's disease-associated neuronal dysfunction, where we find that increased neuronal FKH preserves behavioral function and reduces ubiquitinated protein aggregation. Finally, using transcriptomic profiling, we identify Atg17, a member of the Atg1 autophagy initiation family, as one FKH-dependent target whose neuronal overexpression is sufficient to extend healthy lifespan. Taken together, our results underscore the importance of cell type-specific mapping of TF activity to preserve healthy function with age.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Longevidad , Neuroglía/metabolismo , Neuronas/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Masculino , Neuroglía/citología , Neuronas/citología , Transcriptoma
2.
Development ; 144(13): 2445-2455, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28533206

RESUMEN

Growth factors of the TGFß superfamily play key roles in regulating neuronal and muscle function. Myostatin (or GDF8) and GDF11 are potent negative regulators of skeletal muscle mass. However, expression of myostatin and its cognate receptors in other tissues, including brain and peripheral nerves, suggests a potential wider biological role. Here, we show that Myoglianin (MYO), the Drosophila homolog of myostatin and GDF11, regulates not only body weight and muscle size, but also inhibits neuromuscular synapse strength and composition in a Smad2-dependent manner. Both myostatin and GDF11 affected synapse formation in isolated rat cortical neuron cultures, suggesting an effect on synaptogenesis beyond neuromuscular junctions. We also show that MYO acts in vivo to inhibit synaptic transmission between neurons in the escape response neural circuit of adult flies. Thus, these anti-myogenic proteins act as important inhibitors of synapse function and neuronal growth.


Asunto(s)
Forma de la Célula , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Miostatina/metabolismo , Neuronas/citología , Neuronas/metabolismo , Sinapsis/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Peso Corporal , Regulación hacia Abajo/genética , Drosophila melanogaster/citología , Silenciador del Gen , Glucógeno Sintasa Quinasa 3/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Larva/metabolismo , Células Musculares/metabolismo , Neuroglía/metabolismo , Unión Neuromuscular/metabolismo , Ratas , Transducción de Señal , Transmisión Sináptica
3.
PLoS Biol ; 15(9): e2001655, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28902870

RESUMEN

Lowered insulin/insulin-like growth factor (IGF) signaling (IIS) can extend healthy lifespan in worms, flies, and mice, but it can also have adverse effects (the "insulin paradox"). Chronic, moderately lowered IIS rescues age-related decline in neurotransmission through the Drosophila giant fiber system (GFS), a simple escape response neuronal circuit, by increasing targeting of the gap junctional protein innexin shaking-B to gap junctions (GJs). Endosomal recycling of GJs was also stimulated in cultured human cells when IIS was reduced. Furthermore, increasing the activity of the recycling small guanosine triphosphatases (GTPases) Rab4 or Rab11 was sufficient to maintain GJs upon elevated IIS in cultured human cells and in flies, and to rescue age-related loss of GJs and of GFS function. Lowered IIS thus elevates endosomal recycling of GJs in neurons and other cell types, pointing to a cellular mechanism for therapeutic intervention into aging-related neuronal disorders.


Asunto(s)
Envejecimiento/fisiología , Drosophila/fisiología , Insulina/metabolismo , Somatomedinas/metabolismo , Transmisión Sináptica , Animales , Conexinas/metabolismo , Reacción de Fuga/fisiología , Femenino , Uniones Comunicantes/fisiología , Masculino , Proteínas de Unión al GTP rab/metabolismo
4.
PLoS Genet ; 13(3): e1006593, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28253260

RESUMEN

Nrf2, a transcriptional activator of cell protection genes, is an attractive therapeutic target for the prevention of neurodegenerative diseases, including Alzheimer's disease (AD). Current Nrf2 activators, however, may exert toxicity and pathway over-activation can induce detrimental effects. An understanding of the mechanisms mediating Nrf2 inhibition in neurodegenerative conditions may therefore direct the design of drugs targeted for the prevention of these diseases with minimal side-effects. Our study provides the first in vivo evidence that specific inhibition of Keap1, a negative regulator of Nrf2, can prevent neuronal toxicity in response to the AD-initiating Aß42 peptide, in correlation with Nrf2 activation. Comparatively, lithium, an inhibitor of the Nrf2 suppressor GSK-3, prevented Aß42 toxicity by mechanisms independent of Nrf2. A new direct inhibitor of the Keap1-Nrf2 binding domain also prevented synaptotoxicity mediated by naturally-derived Aß oligomers in mouse cortical neurons. Overall, our findings highlight Keap1 specifically as an efficient target for the re-activation of Nrf2 in AD, and support the further investigation of direct Keap1 inhibitors for the prevention of neurodegeneration in vivo.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Perfilación de la Expresión Génica/métodos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Factor 2 Relacionado con NF-E2/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Animales Modificados Genéticamente , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Cloruro de Litio/farmacología , Longevidad/efectos de los fármacos , Longevidad/genética , Ratones , Microscopía Confocal , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiadiazoles/farmacología , Triazoles/farmacología
5.
Brain Commun ; 3(2): fcab053, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33977265

RESUMEN

Accumulation of amyloid beta peptides is thought to initiate the pathogenesis of Alzheimer's disease. However, the precise mechanisms mediating their neurotoxicity are unclear. Our microarray analyses show that, in Drosophila models of amyloid beta 42 toxicity, genes involved in the unfolded protein response and metabolic processes are upregulated in brain. Comparison with the brain transcriptome of early-stage Alzheimer's patients revealed a common transcriptional signature, but with generally opposing directions of gene expression changes between flies and humans. Among these differentially regulated genes, lactate dehydrogenase (Ldh) was up-regulated by the greatest degree in amyloid beta 42 flies and the human orthologues (LDHA and LDHB) were down-regulated in patients. Functional analyses revealed that either over-expression or inhibition of Ldh by RNA interference (RNAi) slightly exacerbated climbing defects in both healthy and amyloid beta 42-induced Drosophila. This suggests that metabolic responses to lactate dehydrogenase must be finely-tuned, and that its observed upregulation following amyloid beta 42 production could potentially represent a compensatory protection to maintain pathway homeostasis in this model, with further manipulation leading to detrimental effects. The increased Ldh expression in amyloid beta 42 flies was regulated partially by unfolded protein response signalling, as ATF4 RNAi diminished the transcriptional response and enhanced amyloid beta 42-induced climbing phenotypes. Further functional studies are required to determine whether Ldh upregulation provides compensatory neuroprotection against amyloid beta 42-induced loss of activating transcription factor 4 activity and endoplasmatic reticulum stress. Our study thus reveals dysregulation of lactate dehydrogenase signalling in Drosophila models and patients with Alzheimer's disease, which may lead to a detrimental loss of metabolic homeostasis. Importantly, we observed that down-regulation of ATF4-dependent endoplasmic reticulum-stress signalling in this context appears to prevent Ldh compensation and to exacerbate amyloid beta 42-dependent neuronal toxicity. Our findings, therefore, suggest caution in the use of therapeutic strategies focussed on down-regulation of this pathway for the treatment of Alzheimer's disease, since its natural response to the toxic peptide may induce beneficial neuroprotective effects.

6.
Neurobiol Aging ; 66: 149-157, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29579685

RESUMEN

The insulin family of growth factors plays an important role in development and function of the nervous system. Reduced insulin and insulin-growth-factor signaling (IIS), however, can improve symptoms of neurodegenerative diseases in laboratory model organisms and protect against age-associated decline in neuronal function. Recently, we showed that chronic, moderately lowered IIS rescues age-related decline in neurotransmission through the Drosophila giant fiber escape response circuit. Here, we expand our initial findings by demonstrating that reduced functional output in the giant fiber system of aging flies can be prevented by increasing proteasomal activity within the circuit. Manipulations of IIS in neurons can also affect longevity, underscoring the relevance of the nervous system for aging.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/fisiología , Insulina/metabolismo , Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Neuronas/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Transducción de Señal/fisiología , Somatomedinas/metabolismo , Somatomedinas/fisiología , Transmisión Sináptica/fisiología , Animales , Células Cultivadas , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , GTP Fosfohidrolasas/metabolismo , Longevidad , Proteínas de Unión al GTP rab/metabolismo
7.
Fly (Austin) ; 11(4): 284-289, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28837401

RESUMEN

Myoglianin, the Drosophila homolog of the secreted vertebrate proteins Myostatin and GDF-11, is an important regulator of neuronal modeling, and synapse function and morphology. While Myoglianin suppression during development elicits positive effects on the neuromuscular system, genetic manipulations of myoglianin expression levels have a varied effect on the outcome of performance tests in aging flies. Specifically, Myoglianin preserves jumping ability, has no effect on negative geotaxis, and negatively regulates flight performance in aging flies. In addition, Myoglianin exhibits a tissue-specific effect on longevity, with myoglianin upregulation in glial cells increasing the median lifespan. These findings indicate complex role for this TGF-ß-like protein in governing neuromuscular signaling and consequent behavioral outputs and lifespan in adult flies.


Asunto(s)
Envejecimiento/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Drosophila/metabolismo , Drosophila/fisiología , Regulación de la Expresión Génica , Músculos/fisiología , Miostatina/metabolismo , Transducción de Señal , Alas de Animales/fisiología
8.
Cell Rep ; 21(3): 641-653, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045833

RESUMEN

Reduced activity of nutrient-sensing signaling networks can extend organismal lifespan, yet the underlying biology remains unclear. We show that the anti-aging effects of rapamycin and reduced intestinal insulin/insulin growth factor (IGF) signaling (IIS) require the Drosophila FoxA transcription factor homolog Fork Head (FKH). Intestinal FKH induction extends lifespan, highlighting a role for the gut. FKH binds to and is phosphorylated by AKT and Target of Rapamycin. Gut-specific FKH upregulation improves gut barrier function in aged flies. Additionally, it increases the expression of nutrient transporters, as does lowered IIS. Evolutionary conservation of this effect of lowered IIS is suggested by the upregulation of related nutrient transporters in insulin receptor substrate 1 knockout mouse intestine. Our study highlights a critical role played by FKH in the gut in mediating anti-aging effects of reduced IIS. Malnutrition caused by poor intestinal absorption is a major problem in the elderly, and a better understanding of the mechanisms involved will have important therapeutic implications for human aging.


Asunto(s)
Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiología , Alimentos , Factores de Transcripción Forkhead/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Longevidad , Proteínas Nucleares/metabolismo , Animales , Restricción Calórica , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Femenino , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Absorción Intestinal/efectos de los fármacos , Intestinos/citología , Longevidad/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/farmacología , Somatomedinas/metabolismo , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA