Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 733: 150446, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39067249

RESUMEN

Alzheimer's disease (AD), caused by amyloid beta (Aß) plaques and Tau tangles, is a neurodegenerative disease characterized by progressive memory impairment and cognitive dysfunction. High-fat diet (HFD), which induces type 2 diabetes, exacerbates Aß plaque deposition in the brain. To investigate the function of HFD in Tau-mediated AD, we fed an HFD to the Drosophila Tau model and found that HFD aggravates Tau-induced neurological phenotypes. Since microRNAs (miRNAs) are biomarkers for diabetes and AD, we evaluated the expression levels of common miRNAs of HFD and AD in HFD-fed Tau model fly brains. Among the common miRNAs, the expression levels of Let-7 and miR-34 were increased. We found that the inhibition of these miRNAs alleviates Tau-mediated AD phenotypes. Our research provides valuable insights into how HFD accelerates tau toxicity. Additionally, our work highlights the therapeutic potential of targeting Let-7 and miR-34 to develop innovative treatment approaches for AD.

2.
J Neurogenet ; 37(1-2): 3-9, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36165786

RESUMEN

Neurogenetic research using the Drosophila model has immensely expanded around the world. Likewise, scientists in South Korea have leveraged the advantages of Drosophila genetic tools to understand various neurobiological processes. In this special issue, we will overview the history of Drosophila neurogenetic research in South Korea that led to significant discoveries and notably implications. We will describe how Drosophila system was first introduced to elevate neural developmental studies in 1990s. Establishing Drosophila-related resources has been a key venture, which led to the generation of over 100,000 mutant lines and the launch of the K-Gut initiative with Korea Drosophila Research Center (KDRC). These resources have supported the pioneer studies in modeling human disease and understanding genes and neural circuits that regulate animal behavior and physiology.


Asunto(s)
Drosophila , Neurociencias , Animales , Humanos , Conducta Animal/fisiología , Drosophila/genética , Neurogénesis , República de Corea
3.
Biochem Biophys Res Commun ; 568: 95-102, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34217014

RESUMEN

Sarcopenia is a syndrome characterized by progressive loss of muscle mass and function during aging. Although mitochondrial dysfunction and related metabolic defects precede age-related changes in muscle, their contributions to muscle aging are still not well known. In this study, we used a Drosophila model to investigate the role of lipophorin receptors (LpRs), a Drosophila homologue of the mammalian very low-density lipoprotein receptor (VLDLR), in mitochondrial dynamics and muscle aging. Muscle-specific knockdown of LpR1 or LpR2 resulted in mitochondrial dysfunction and reduced proteostasis, which contributed to muscle aging. Activation of AMP-activated protein kinase (AMPK) ameliorated muscle dysfunction induced by LpR1 knockdown. These results suggest that LpR1/VLDLR is a novel key target that modulates age-dependent lipid remodeling and muscle homeostasis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Mitocondrias/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Técnicas de Silenciamiento del Gen , Longevidad , Masculino , Mitocondrias/genética , Recambio Mitocondrial , Receptores Citoplasmáticos y Nucleares/genética
4.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34445171

RESUMEN

Tauopathy refers to a group of progressive neurodegenerative diseases, including frontotemporal lobar degeneration and Alzheimer's disease, which correlate with the malfunction of microtubule-associated protein Tau (MAPT) due to abnormal hyperphosphorylation, leading to the formation of intracellular aggregates in the brain. Despite extensive efforts to understand tauopathy and develop an efficient therapy, our knowledge is still far from complete. To find a solution for this group of devastating diseases, several animal models that mimic diverse disease phenotypes of tauopathy have been developed. Rodents are the dominating tauopathy models because of their similarity to humans and established disease lines, as well as experimental approaches. However, powerful genetic animal models using Drosophila, zebrafish, and C. elegans have also been developed for modeling tauopathy and have contributed to understanding the pathophysiology of tauopathy. The success of these models stems from the short lifespans, versatile genetic tools, real-time in-vivo imaging, low maintenance costs, and the capability for high-throughput screening. In this review, we summarize the main findings on mechanisms of tauopathy and discuss the current tauopathy models of these non-rodent genetic animals, highlighting their key advantages and limitations in tauopathy research.


Asunto(s)
Modelos Animales de Enfermedad , Tauopatías/genética , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Drosophila/genética , Drosophila/fisiología , Humanos , Tauopatías/fisiopatología , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas tau/genética
5.
PLoS Genet ; 8(8): e1002857, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22876196

RESUMEN

Feeding behavior is one of the most essential activities in animals, which is tightly regulated by neuroendocrine factors. Drosophila melanogaster short neuropeptide F (sNPF) and the mammalian functional homolog neuropeptide Y (NPY) regulate food intake. Understanding the molecular mechanism of sNPF and NPY signaling is critical to elucidate feeding regulation. Here, we found that minibrain (mnb) and the mammalian ortholog Dyrk1a, target genes of sNPF and NPY signaling, [corrected] regulate food intake in Drosophila melanogaster and mice. In Drosophila melanogaster neuronal cells and mouse hypothalamic cells, sNPF and NPY modulated the mnb and Dyrk1a expression through the PKA-CREB pathway. Increased Dyrk1a activated Sirt1 to regulate the deacetylation of FOXO, which potentiated FOXO-induced sNPF/NPY expression and in turn promoted food intake. Conversely, AKT-mediated insulin signaling suppressed FOXO-mediated sNPF/NPY expression, which resulted in decreasing food intake. Furthermore, human Dyrk1a transgenic mice exhibited decreased FOXO acetylation and increased NPY expression in the hypothalamus, and [corrected] increased food intake. Our findings demonstrate that Mnb/Dyrk1a regulates food intake through the evolutionary conserved Sir2-FOXO-sNPF/NPY pathway in Drosophila melanogaster and mammals.


Asunto(s)
Regulación del Apetito/genética , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Regulación de la Expresión Génica , Transducción de Señal/genética , Acetilación , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Hipotálamo/fisiología , Mamíferos/fisiología , Ratones , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Quinasas DyrK
6.
Angew Chem Int Ed Engl ; 54(39): 11472-6, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26178411

RESUMEN

The abnormal assembly of ß-amyloid (Aß) peptides into neurotoxic, ß-sheet-rich amyloid aggregates is a major pathological hallmark of Alzheimer's disease (AD). Light-induced photosensitizing molecules can regulate Aß amyloidogenesis. Multiple photochemical analyses using circular dichroism, atomic force microscopy, dot blot, and native gel electrophoresis verified that photoactivated meso-tetra(4-sulfonatophenyl)porphyrin (TPPS with M = 2H(+), Zn(2+), Cu(2+), Mn(2+)) successfully inhibits Aß aggregation in vitro. Furthermore, Aß toxicity was relieved in the photoexcited-TPPS-treated Drosophila AD model. TPPS suppresses neural cell death, synaptic toxicity, and behavioral defects in the Drosophila AD model under blue light illumination. Behavioral phenotypes, including larval locomotion defect and short lifespan caused by Aß overexpression, were also rescued by blue light-excited TPPS.


Asunto(s)
Péptidos beta-Amiloides/química , Porfirinas/química , Sinapsis/efectos de los fármacos , Péptidos beta-Amiloides/toxicidad , Animales , Drosophila , Procesos Fotoquímicos
7.
Nucleic Acids Res ; 40(1): 75-87, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21911363

RESUMEN

The tumor suppressor p53 responds to a wide variety of cellular stress signals. Among potential regulatory pathways, post-translational modifications such as acetylation by CBP/p300 and PCAF have been suggested for modulation of p53 activity. However, exactly how p53 acetylation is modulated remains poorly understood. Here, we found that SET/TAF-Iß inhibited p300- and PCAF-mediated p53 acetylation in an INHAT (inhibitor of histone acetyltransferase) domain-dependent manner. SET/TAF-Iß interacted with p53 and repressed transcription of p53 target genes. Consequently, SET/TAF-Iß blocked both p53-mediated cell cycle arrest and apoptosis in response to cellular stress. Using different apoptosis analyses, including FACS, TUNEL and BrdU incorporation assays, we also found that SET/TAF-Iß induced cellular proliferation via inhibition of p53 acetylation. Furthermore, we observed that apoptotic Drosophila eye phenotype induced by either dp53 overexpression or UV irradiation was rescued by expression of dSet. Inhibition of dp53 acetylation by dSet was observed in both cases. Our findings provide new insights into the regulation of stress-induced p53 activation by HAT-inhibiting histone chaperone SET/TAF-Iß.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Apoptosis , Línea Celular , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/efectos de la radiación , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/metabolismo , Estrés Fisiológico , Activación Transcripcional , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Rayos Ultravioleta
8.
Z Naturforsch C J Biosci ; 69(1-2): 68-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24772825

RESUMEN

Our previous data demonstrated that CoCl2-induced hypoxia controls endoplasmic reticulum (ER) stress-associated and other intracellular factors. One of them, the transcription factor Pokemon, was differentially regulated by low-dose radiation (LDR). There are limited data regarding how this transcription factor is involved in expression of the unfolded protein response (UPR) under hypoxic conditions. The purpose of this study was to obtain clues on how Pokemon is involved in the UPR. Pokemon was selected as a differentially expressed gene under hypoxic conditions; however, its regulation was clearly repressed by LDR. It was also demonstrated that both expression of ER chaperones and ER stress sensors were affected by hypoxic conditions, and the same results were obtained when cells in which Pokemon was up- or down-regulated were used. The current state of UPR and LDR research associated with the Pokemon pathway offers an important opportunity to understand the oncogenesis, senescence, and differentiation of cells, as well as to facilitate introduction of new therapeutic radiopharmaceuticals.


Asunto(s)
Proteínas Represoras/metabolismo , Respuesta de Proteína Desplegada , Animales , Secuencia de Bases , Hipoxia de la Célula , Cartilla de ADN , Relación Dosis-Respuesta en la Radiación , Células PC12 , Ratas , Proteínas Represoras/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Exp Mol Med ; 54(4): 426-432, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35388147

RESUMEN

Cancer cachexia syndrome is a major cause of morbidity and mortality in cancer patients in the advanced stage. It is a devastating disorder characterized by nutritional impairment, weakness, and wasting, and it affects treatment success and quality of life. Two major symptoms of cancer cachexia are anorexia and weight loss. Weight loss in cachexia is not reversed through increased food intake, suggesting that anorexia and weight loss in cancer patients are regulated by independent molecular mechanisms. Although the wasting phenotype mostly occurs in skeletal muscle and adipose tissue, other organs, such as the brain, liver, pancreas, heart, and gut, are also involved in cachexia. Thus, cachexia is a multiorgan syndrome. Although the molecular basis of cancer cachexia-induced weight loss is known, the mechanism underlying anorexia is poorly understood. Here, we highlight our recent discovery of a new anorexia mechanism by which a tumor-derived humoral factor induces cancer anorexia by regulating feeding-related neuropeptide hormones in the brain. Furthermore, we elucidated the process through which anorexia precedes tissue wasting in cachexia. This review article aims to provide an overview of the key molecular mechanisms of anorexia and tissue wasting caused by cancer cachexia.


Asunto(s)
Caquexia , Neoplasias , Tejido Adiposo , Anorexia/etiología , Anorexia/terapia , Caquexia/complicaciones , Caquexia/genética , Humanos , Neoplasias/complicaciones , Neoplasias/terapia , Calidad de Vida
11.
Mol Cells ; 45(9): 640-648, 2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-35993164

RESUMEN

CD133, also known as prominin-1, was first identified as a biomarker of mammalian cancer and neural stem cells. Previous studies have shown that the prominin-like (promL) gene, an orthologue of mammalian CD133 in Drosophila, plays a role in glucose and lipid metabolism, body growth, and longevity. Because locomotion is required for food sourcing and ultimately the regulation of metabolism, we examined the function of promL in Drosophila locomotion. Both promL mutants and pan-neuronal promL inhibition flies displayed reduced spontaneous locomotor activity. As dopamine is known to modulate locomotion, we also examined the effects of promL inhibition on the dopamine concentration and mRNA expression levels of tyrosine hydroxylase (TH) and DOPA decarboxylase (Ddc), the enzymes responsible for dopamine biosynthesis, in the heads of flies. Compared with those in control flies, the levels of dopamine and the mRNAs encoding TH and Ddc were lower in promL mutant and pan-neuronal promL inhibition flies. In addition, an immunostaining analysis revealed that, compared with control flies, promL mutant and pan-neuronal promL inhibition flies had lower levels of the TH protein in protocerebral anterior medial (PAM) neurons, a subset of dopaminergic neurons. Inhibition of promL in these PAM neurons reduced the locomotor activity of the flies. Overall, these findings indicate that promL expressed in PAM dopaminergic neurons regulates locomotion by controlling dopamine synthesis in Drosophila.


Asunto(s)
Proteínas de Drosophila , Drosophila , Antígeno AC133/metabolismo , Animales , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Locomoción/genética , Mamíferos/metabolismo
12.
Microbiome ; 10(1): 3, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991725

RESUMEN

BACKGROUND: Host tp53 mutations are frequently found during the early stages of colitis-associated colorectal cancer (CAC), but whether such mutations induce gut microbiota dysbiosis and chronic intestinal inflammation that contributes to the development of CAC, remains unknown. RESULTS: We found that zebrafish tp53 mutant larvae exhibited elevated intestinal inflammation, by monitoring the NFκB activity in the mid-distal intestines of zebrafish larvae using an NFκB:EGFP transgenic reporter line in vivo as well as neutrophil infiltration into the intestine. This inflammation was due to dysbiotic gut microbiota with reduced diversity, revealed using both 16S rRNA amplicon sequencing and a germfree larva model. In this dysbiosis, Aeromonas spp. were aberrantly enriched as major pathobionts and exhibited the capacity for aggressive colonization in tp53 mutants. Importantly, the ex-germfree experiments supported the causality of the host tp53 mutation for inducing the inflammation. Transcriptome and high-performance liquid chromatography analyses of the host gastrointestinal tracts identified dysregulated sialic acid (SA) metabolism concomitant with increased host Neu5Gc levels as the key determinant of aberrant inflammation, which was reversed by the sialidase inhibitors oseltamivir and Philippin A. CONCLUSIONS: These results demonstrate a crucial role for host tp53 in maintaining symbiosis and immune homeostasis via SA metabolism. Disturbed SA metabolism via a tp53 mutation may be exploited by specific elements of the gut microbiome, eliciting both dysbiosis and inflammation. Manipulating sialometabolism may therefore provide an efficacious therapeutic strategy for tp53 mutation-induced dysbiosis, inflammation, and ultimately, related cancers. Video Abstract.


Asunto(s)
Disbiosis , Ácido N-Acetilneuramínico , Animales , Disbiosis/inducido químicamente , Inflamación , Mutación , Ácido N-Acetilneuramínico/efectos adversos , ARN Ribosómico 16S/genética , Pez Cebra
13.
J Cell Mol Med ; 15(1): 152-65, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19874423

RESUMEN

Neuropeptide Y (NPY) and NPY receptors are widely expressed in various organs and cell types and have been shown to have pleiotropic functions. However, their presence or role in human embryonic stem cells (hESCs) remains unknown. We now show that undifferentiated hESCs primarily express NPY and its Y1 and Y5 receptors. Inhibition of NPY signalling using either the selective NPY Y1 or Y5 receptor antagonist reduces the maintenance of self-renewal and proliferation of undifferentiated hESCs. We also provide compelling evidence that exogenous NPY supports the long-term growth of undifferentiated hESCs in the absence of feeder cell factors using only knockout serum replacement media. Further, NPY facilitates the use of chemically defined medium made up of N2/B27 supplement and basic fibroblast growth factor (bFGF) for hESC feeder-free culture. Our results indicate that both Y1 and Y5 receptors appear to be involved in the NPY-mediated activation of AKT/protein kinase B and extracellular signal-regulated kinase 1/2 (ERK1/2) in hESCs. Notably, only Y1 receptor, but not Y5 receptor, is responsible for the NPY-induced activation of cAMP-response element binding (CREB) in hESCs. These results provide the first evidence that NPY and its Y1 and Y5 receptors have potential role in maintaining hESC self-renewal and pluripotency. We demonstrate the underlying importance of NPY signalling and its usefulness in the development of a defined and xeno-free culture condition for the large-scale propagation of undifferentiated hESCs.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/metabolismo , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo
14.
BMC Mol Biol ; 12: 25, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21605464

RESUMEN

BACKGROUND: The unfolded protein response (UPR) is an evolutionary conserved adaptive reaction for increasing cell survival under endoplasmic reticulum (ER) stress conditions. X-box-binding protein-1 (Xbp1) is a key transcription factor of UPR that activates genes involved in protein folding, secretion, and degradation to restore ER function. The UPR induced by ER stress was extensively studied in diseases linked to protein misfolding and aggregations. However, in the porcine system, genes in the UPR pathway were not investigated. In this study, we isolated and characterized the porcine Xbp1 (pXbp1) gene in ER stress using porcine embryonic fibroblast (PEF) cells and porcine organs. ER stress was induced by the treatment of tunicamycin and cell viability was investigated by the MTT assay. For cloning and analyzing the expression pattern of pXbp1, RT-PCR analysis and Western blot were used. Knock-down of pXbp1 was performed by the siRNA-mediated gene silencing. RESULTS: We found that the pXbp1 mRNA was the subject of the IRE1α-mediated unconventional splicing by ER stress. Knock-down of pXbp1 enhanced ER stress-mediated cell death in PEF cells. In adult organs, pXbp1 mRNA and protein were expressed and the spliced forms were detected. CONCLUSIONS: It was first found that the UPR mechanisms and the function of pXbp1 in the porcine system. These results indicate that pXbp1 plays an important role during the ER stress response like other animal systems and open a new opportunity for examining the UPR pathway in the porcine model system.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/metabolismo , Porcinos/metabolismo , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada , Secuencia de Aminoácidos , Animales , Muerte Celular , Células Cultivadas , Clonación Molecular , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Fibroblastos/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Datos de Secuencia Molecular , Empalme del ARN , ARN Mensajero/genética , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/análisis , Factores de Transcripción/genética , Tunicamicina/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
15.
Emerg Med J ; 28(1): 40-3, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21131393

RESUMEN

OBJECTIVES: A bag-valve mask (BVM) device is used as one of the first-line pieces of equipment in emergency situations. However, cardiopulmonary support providers do not recognise the exact tidal volume during procedures, and squeezing methods of BVM may not deliver the same tidal volume each time. To supply a regular and sustained tidal volume, adequate finger points were marked on the surface of a BVM. METHODS: In this study, a total of 83 volunteers participated and practised conventional BVM and volume-marked bag-valve mask (VBVM) procedures. The VBVM is simply a conventional BVM with an imaginary axis grid, drawn to guide the placement of the fingers. The VBVM method provides a constant volume of approximately 500-600 ml; the bag is squeezed until the thumb and the middle finger touch slightly. The results were then statistically analysed. RESULTS: The tidal volume delivered by the studied VBVM method is more accurate than the conventional BVM method (421.87±95.19 ml vs 534.21±24.22 ml, p<0.001). There was no statistical correlation except age between the results and the participants' training level or physical characteristics in the study. CONCLUSIONS: As the conventional BVM method cannot deliver a regular and sustained tidal volume, the authors invented the VBVM method. This method delivered a volume of 500-600 ml with more stability each time, which can improve the outcome of emergency patients.


Asunto(s)
Reanimación Cardiopulmonar/instrumentación , Servicio de Urgencia en Hospital , Respiración Artificial/instrumentación , Volumen de Ventilación Pulmonar , Centros Médicos Académicos , Reanimación Cardiopulmonar/métodos , Estudios de Cohortes , Medicina de Emergencia/métodos , Diseño de Equipo , Seguridad de Equipos , Femenino , Humanos , Máscaras , Respiración Artificial/métodos , Insuficiencia Respiratoria/terapia , Sensibilidad y Especificidad , Resultado del Tratamiento
16.
Int J Mol Sci ; 12(7): 4456-64, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21845089

RESUMEN

We demonstrated that up-regulation of gene expression of endoplasmic reticulum (ER) chaperones (BiP, calnexin, calreticulin, ERp29) and ER membrane kinases (IRE1, PERK, ATF6) was induced by radiation in neuronal PC12 cells. However, addition of silkworm, Bombyx mori, hemolymph to irradiated cells resulted in an obvious decrease in expression of these genes, compared with a single radiation treatment. In contrast, one of the ER chaperones, "ischemia-responsive protein 94 kDa" (irp94), was up-regulated by radiation. However, addition of silkworm hemolymph resulted in no change in the expression of irp94, with an expression pattern that differed from that of ER chaperones. Based on these results, we propose that silkworm hemolymph contains factors that regulate a decrease in the expression of ER chaperones under radiation-irradiation conditions, with the exception of irp94, which is not down-regulated. We suggest that this difference in the molecular character of irp94 may provide a clue to the biological functions associated with ER stress pathways, particularly the effects of radiation.


Asunto(s)
Bombyx/metabolismo , Regulación hacia Abajo/efectos de la radiación , Retículo Endoplásmico/metabolismo , Rayos gamma , Proteínas del Helminto/metabolismo , Chaperonas Moleculares/metabolismo , Animales , Estrés del Retículo Endoplásmico , Células PC12 , Ratas , Regulación hacia Arriba
17.
Int J Mol Sci ; 12(11): 7652-61, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174623

RESUMEN

We demonstrated that upregulation of both gene expression of endoplasmic reticulum (ER) stress chaperones (BiP, calnexin, calreticulin, and PDI) and ER stress sensors (ATF6, IRE1 and PERK) was induced by lidocaine, a local anesthetic, in PC12 cells. In addition to gene regulation, lidocaine also induced typical ER stress phenomena such as ART6 proteolytic cleavage, eIF2 alpha phosphorylation, and XBP1 mRNA splicing. In in vivo experiments, while lidocaine downregulated gene expression of antiapoptotic factors (Bcl-2 and Bcl-xl), pro-apoptotic factor (Bak and Bax) gene expression was upregulated. Furthermore, lidocaine induced apoptosis, as measured histochemically, and upregulated PARP1, a DNA damage repair enzyme. These results are the first to show that lidocaine induces apoptosis through ER stress in vitro and in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Lidocaína/farmacología , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Animales , Calnexina/genética , Calnexina/metabolismo , Calreticulina/genética , Calreticulina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Células PC12 , Fosforilación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Empalme del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Proteína 1 de Unión a la X-Box , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
18.
Nat Commun ; 12(1): 3291, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34078905

RESUMEN

The formation of hyperphosphorylated intracellular Tau tangles in the brain is a hallmark of Alzheimer's disease (AD). Tau hyperphosphorylation destabilizes microtubules, promoting neurodegeneration in AD patients. To identify suppressors of tau-mediated AD, we perform a screen using a microRNA (miR) library in Drosophila and identify the miR-9 family as suppressors of human tau overexpression phenotypes. CG11070, a miR-9a target gene, and its mammalian orthologue UBE4B, an E3/E4 ubiquitin ligase, alleviate eye neurodegeneration, synaptic bouton defects, and crawling phenotypes in Drosophila human tau overexpression models. Total and phosphorylated Tau levels also decrease upon CG11070 or UBE4B overexpression. In mammalian neuroblastoma cells, overexpression of UBE4B and STUB1, which encodes the E3 ligase CHIP, increases the ubiquitination and degradation of Tau. In the Tau-BiFC mouse model, UBE4B and STUB1 overexpression also increase oligomeric Tau degradation. Inhibitor assays of the autophagy and proteasome systems reveal that the autophagy-lysosome system is the major pathway for Tau degradation in this context. These results demonstrate that UBE4B, a miR-9 target gene, promotes autophagy-mediated Tau degradation together with STUB1, and is thus an innovative therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas de Drosophila/genética , MicroARNs/genética , Ubiquitina-Proteína Ligasas/genética , Proteínas tau/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Autofagia/genética , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ojo/metabolismo , Ojo/patología , Humanos , Lisosomas/metabolismo , Ratones , MicroARNs/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas tau/metabolismo
19.
Nat Cell Biol ; 23(2): 172-183, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33558728

RESUMEN

In patients with advanced-stage cancer, cancer-associated anorexia affects treatment success and patient survival. However, the underlying mechanism is poorly understood. Here, we show that Dilp8, a Drosophila homologue of mammalian insulin-like 3 peptide (INSL3), is secreted from tumour tissues and induces anorexia through the Lgr3 receptor in the brain. Activated Dilp8-Lgr3 signalling upregulated anorexigenic nucleobinding 1 (NUCB1) and downregulated orexigenic short neuropeptide F (sNPF) and NPF expression in the brain. In the cancer condition, the protein expression of Lgr3 and NUCB1 was significantly upregulated in neurons expressing sNPF and NPF. INSL3 levels were increased in tumour-implanted mice and INSL3-treated mouse hypothalamic cells showed Nucb2 upregulation and Npy downregulation. Food consumption was significantly reduced in intracerebrospinal INSL3-injected mice. In patients with pancreatic cancer, higher serum INSL3 levels increased anorexia. These results indicate that tumour-derived Dilp8/INSL3 induces cancer anorexia by regulating feeding hormones through the Lgr3/Lgr8 receptor in Drosophila and mammals.


Asunto(s)
Anorexia/metabolismo , Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Anorexia/etiología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Neoplasias del Ojo/patología , Conducta Alimentaria , Humanos , Hipotálamo/metabolismo , Insulina/sangre , Insulina/química , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones Endogámicos C57BL , Neoplasias/complicaciones , Neuronas/metabolismo , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/complicaciones , Proteínas/química , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
20.
J Biol Chem ; 284(43): 29454-61, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19720829

RESUMEN

Activation of c-Jun N-terminal kinase (JNK) signaling in neurons increases stress resistance and extends life span, in part through FOXO-mediated transcription in Drosophila. However, the JNK/FOXO target genes are unknown. Here, we identified Jafrac1, a Drosophila homolog of human Peroxiredoxin II (hPrxII), as a downstream effecter of JNK/FOXO signaling in neurons that enhances stress resistance and extends life span. We found that Jafrac1 was expressed in the adult brain and induced by paraquat, a reactive oxygen species-generating chemical. RNA interference-mediated neuronal knockdown of Jafrac1 enhanced, while neuronal overexpression of Jafrac1 and hPrxII suppressed, paraquat-induced lethality in flies. Neuronal expression of Jafrac1 also significantly reduced ROS levels, restored mitochondrial function, and attenuated JNK activation caused by paraquat. Activation of JNK/FOXO signaling in neurons increased the Jafrac1 expression level under both normal and oxidative stressed conditions. Moreover, neuronal knockdown of Jafrac1 shortened, while overexpression of Jafrac1 and hPrxII extended, the life span in flies. These results support the hypothesis that JNK/FOXO signaling extends life span via amelioration of oxidative damage and mitochondrial dysfunction in neurons.


Asunto(s)
Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Longevidad/fisiología , Neuronas/enzimología , Peroxidasas/biosíntesis , Transducción de Señal/fisiología , Animales , Encéfalo/enzimología , Proteínas de Drosophila/genética , Drosophila melanogaster , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Herbicidas/farmacología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Longevidad/efectos de los fármacos , Mitocondrias/enzimología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Paraquat/farmacología , Peroxidasas/genética , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA