Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Sci Rep ; 14(1): 10453, 2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714725

RESUMEN

Recent research has highlighted the importance of the gut microbiome in regulating aging, and probiotics are interventions that can promote gut health. In this study, we surveyed several novel lactic acid bacteria to examine their beneficial effect on organismal health and lifespan in C. elegans. We found that animals fed some lactic acid bacteria, including L. acidophilus 1244 and L. paracasei subsp. paracasei 2004, grew healthy. Supplementation with the lactic acid bacterial strains L. acidophilus 1244 or L. paracasei subsp. paracasei 2004 significantly improved health, including food consumption, motility, and resistance to oxidative stressor, hydrogen peroxide. Our RNA-seq analysis showed that supplementation with L. paracasei subsp. paracasei 2004 significantly increased the expression of daf-16, a C. elegans FoxO homolog, as well as genes related to the stress response. Furthermore, daf-16 deletion inhibited the longevity effect of L. paracasei subsp. paracasei 2004 supplementation. Our results suggest that L. paracasei subsp. paracasei 2004 improves health and lifespan in a DAF-16-dependent manner.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Factores de Transcripción Forkhead , Longevidad , Probióticos , Animales , Caenorhabditis elegans/fisiología , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Lacticaseibacillus paracasei/fisiología , Lacticaseibacillus paracasei/genética , Estrés Oxidativo , Microbioma Gastrointestinal
2.
Life Sci Alliance ; 6(12)2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37802655

RESUMEN

The protein kinase DYRK1A encoded in human chromosome 21 is the major contributor to the multiple symptoms observed in Down syndrome patients. In addition, DYRK1A malfunction is associated with various other neurodevelopmental disorders such as autism spectrum disorder. Here, we identified FAM53C with no hitherto known biological function as a novel suppressive binding partner of DYRK1A. FAM53C is bound to the catalytic protein kinase domain of DYRK1A, whereas DCAF7/WDR68, the major DYRK1A-binding protein, binds to the N-terminal domain of DYRK1A. The binding of FAM53C inhibited autophosphorylation activity of DYRK1A and its kinase activity to an exogenous substrate, MAPT/Tau. FAM53C did not bind directly to DCAF7/WDR68, whereas DYRK1A tethered FAM53C and DCAF7/WDR68 by binding concurrently to both of them, forming a tri-protein complex. DYRK1A possesses an NLS and accumulates in the nucleus when overexpressed in cells. Co-expression of FAM53C induced cytoplasmic re-localization of DYRK1A, revealing the cytoplasmic anchoring function of FAM53C to DYRK1A. Moreover, the binding of FAM53C to DYRK1A suppressed the DYRK1A-dependent nuclear localization of DCAF7/WDR68. All the results show that FAM53C binds to DYRK1A, suppresses its kinase activity, and anchors it in the cytoplasm. In addition, FAM53C is bound to the DYRK1A-related kinase DYRK1B with an Hsp90/Cdc37-independent manner. The results explain for the first time why endogenous DYRK1A is distributed in the cytoplasm in normal brain tissue. FAM53C-dependent regulation of the kinase activity and intracellular localization of DYRK1A may play a significant role in gene expression regulation caused by normal and aberrant levels of DYRK1A.


Asunto(s)
Encéfalo , Proteínas Portadoras , Proteínas Quinasas , Humanos , Trastorno del Espectro Autista/metabolismo , Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Fosforilación , Factores de Transcripción , Síndrome de Down/metabolismo , Encéfalo/metabolismo , Proteínas Quinasas/metabolismo , Quinasas DyrK
3.
iScience ; 24(7): 102706, 2021 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-34235410

RESUMEN

Previous studies have revealed the importance of inter-tissue communications for lifespan regulation. However, the inter-tissue network responsible for lifespan regulation is not well understood, even in a simple organism Caenorhabditis elegans. To understand the mechanisms underlying systemic lifespan regulation, we focused on lifespan regulation by the insulin/insulin-like growth factor-1 signaling (IIS) pathway; IIS reduction activates the DAF-16/FOXO transcription factor, which results in lifespan extension. Our tissue-specific knockdown and knockout analyses demonstrated that IIS reduction in neurons and the intestine markedly extended lifespan. DAF-16 activation in neurons resulted in DAF-16 activation in the intestine and vice versa. Our dual gene manipulation method revealed that intestinal and neuronal DAF-16 mediate longevity induced by daf-2 knockout in neurons and the intestine, respectively. In addition, the systemic regulation of intestinal DAF-16 required the IIS pathway in intestinal and neurons. Collectively, these results highlight the importance of the neuronal DAF-16-to-intestinal DAF-16 communication for organismal lifespan regulation.

4.
Biochim Biophys Acta Mol Cell Res ; 1868(10): 119081, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34147560

RESUMEN

The DYRK (Dual-specificity tYrosine-phosphorylation Regulated protein Kinase) family consists of five related protein kinases (DYRK1A, DYRK1B, DYRK2, DYRK3, DYRK4). DYRKs show homology to Drosophila Minibrain, and DYRK1A in human chromosome 21 is responsible for various neuronal disorders including human Down syndrome. Here we report identification of cellular proteins that associate with specific members of DYRKs. Cellular proteins with molecular masses of 90, 70, and 50-kDa associated with DYRK1B and DYRK4. These proteins were identified as molecular chaperones Hsp90, Hsp70, and Cdc37, respectively. Microscopic analysis of GFP-DYRKs showed that DYRK1A and DYRK1B were nuclear, while DYRK2, DYRK3, and DYRK4 were mostly cytoplasmic in COS7 cells. Overexpression of DYRK1B induced nuclear re-localization of these chaperones with DYRK1B. Treatment of cells with specific Hsp90 inhibitors, geldanamycin and 17-AAG, abolished the association of Hsp90 and Cdc37 with DYRK1B and DYRK4, but not of Hsp70. Inhibition of Hsp90 chaperone activity affected intracellular dynamics of DYRK1B and DYRK4. DYRK1B and DYRK4 underwent rapid formation of cytoplasmic punctate dots after the geldanamycin treatment, suggesting that the chaperone function of Hsp90 is required for prevention of protein aggregation of the target kinases. Prolonged inhibition of Hsp90 by geldanamycin, 17-AAG, or ganetespib, decreased cellular levels of DYRK1B and DYRK4. Finally, DYRK1B and DYRK4 were ubiquitinated in cells, and ubiquitinated DYRK1B and DYRK4 further increased by Hsp90 inhibition with geldanamycin. Taken together, these results indicate that Hsp90 and Cdc37 discriminate specific members of the DYRK kinase family and play an important role in quality control of these client kinases in cells.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Chaperoninas/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Benzoquinonas/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Lactamas Macrocíclicas/farmacología , Quinasas DyrK
5.
Commun Biol ; 4(1): 207, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594200

RESUMEN

Environmental conditions can cause phenotypic changes, part of which can be inherited by subsequent generations via soma-to-germline communication. However, the signaling molecules or pathways that mediate intertissue communication remain unclear. Here, we show that intertissue small RNA communication systems play a key role in the acquisition and inheritance of hormesis effects - stress-induced stress resistance - in Caenorhabditis elegans. The miRNA-processing enzyme DRSH-1 is involved in both the acquisition and the inheritance of hormesis, whereas worm-specific Argonaute (WAGO) proteins, which function with endo-siRNAs, are involved only in its inheritance. Further analyses demonstrate that the miRNA production system in the neuron and the small RNA transport machinery in the intestine are both essential for its acquisition and that both the transport of small RNAs in the germline and the germline Argonaute HRDE-1 complex are required for its inheritance. Our results thus demonstrate that overlapping and distinct roles of small RNA systems in the acquisition and inheritance of hormesis effects.


Asunto(s)
Proteínas Argonautas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Hormesis/genética , MicroARNs/metabolismo , Transporte de ARN , ARN Interferente Pequeño/metabolismo , Ribonucleasa III/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Argonautas/genética , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Herencia , Histona Metiltransferasas/genética , Histona Metiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Intestinos , MicroARNs/genética , Neuronas/metabolismo , Presión Osmótica , Estrés Oxidativo , Interferencia de ARN , ARN Interferente Pequeño/genética , Ribonucleasa III/genética
6.
Cell Rep ; 30(10): 3207-3217.e4, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32160530

RESUMEN

Changes in epigenetic states affect organismal homeostasis, including stress resistance. However, the mechanisms coordinating epigenetic states and systemic stress resistance remain largely unknown. Here, we identify the intestine-to-germline communication of epigenetic states, which intergenerationally enhances stress resistance in C. elegans. The alterations in epigenetic states by deficiency of the histone H3K4me3 modifier ASH-2 in the intestine or germline increase organismal stress resistance, which is abrogated by knockdown of the H3K4 demethylase RBR-2. Remarkably, the increase in stress resistance induced by ASH-2 deficiency in the intestine is abrogated by RBR-2 knockdown in the germline, suggesting the intestine-to-germline transmission of epigenetic information. This communication from intestine to germline in the parental generation increases stress resistance in the next generation. Moreover, the intertissue communication is mediated partly by transcriptional regulation of F08F1.3. These results reveal that intertissue communication of epigenetic information provides mechanisms for intergenerational regulation of systemic stress resistance.


Asunto(s)
Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Epigénesis Genética , Células Germinativas/metabolismo , Patrón de Herencia/genética , Intestinos/fisiología , Estrés Fisiológico/genética , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Regulación hacia Abajo/genética , Estrés Oxidativo
7.
Genes Cells ; 25(2): 86-99, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31788928

RESUMEN

Neurotrophic signaling regulates neural cell behaviors in development and physiology, although its role in regeneration has not been fully investigated. Here, we examined the role of neurotrophic signaling in Xenopus laevis tadpole tail regeneration. After the tadpole tails were amputated, the expression of neurotrophin ligand family genes, especially ngf and bdnf, was up-regulated as regeneration proceeded. Moreover, notochordal expression of the NGF receptor gene TrkA, but not that of other neurotrophin receptor genes TrkB and TrkC, became prominent in the regeneration bud, a structure arising from the tail stump after tail amputation. The regenerated tail length was significantly shortened by the pan-Trk inhibitor K252a or the TrkA inhibitor GW-441756, but not by the TrkB inhibitor ANA-12, suggesting that TrkA signaling is involved in elongation of regenerating tails. Furthermore, during Xenopus laevis embryonic development, TrkA expression was detected in the dorsal mesoderm at the gastrula stage and in the notochord at the neurula stage, and its knockdown led to gastrulation defects with subsequent shortening of the body axis length. These results suggest that Xenopus laevis TrkA signaling, which can act in the mesoderm/notochord, plays a key role in body axis elongation during embryogenesis as well as tail elongation during tadpole regeneration.


Asunto(s)
Desarrollo Embrionario/genética , Larva/genética , Receptor trkA/genética , Receptor trkA/metabolismo , Regeneración/genética , Transducción de Señal , Cola (estructura animal)/fisiología , Xenopus laevis/anomalías , Xenopus laevis/genética , Animales , Azepinas/farmacología , Benzamidas/farmacología , Carbazoles/farmacología , Regulación del Desarrollo de la Expresión Génica , Alcaloides Indólicos/farmacología , Factor de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Receptor trkA/antagonistas & inhibidores , Receptor trkC/genética , Receptores de Factor de Crecimiento Nervioso/genética , Transducción de Señal/efectos de los fármacos , Cola (estructura animal)/anatomía & histología
8.
J Biol Chem ; 294(1): 195-209, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30409903

RESUMEN

Lineage specification of the three germ layers occurs during early embryogenesis and is critical for normal development. The nucleosome remodeling and deacetylase (NuRD) complex is a repressive chromatin modifier that plays a role in lineage commitment. However, the role of chromodomain helicase DNA-binding protein 4 (CHD4), one of the core subunits of the NuRD complex, in neural lineage commitment is poorly understood. Here, we report that the CHD4/NuRD complex plays a critical role in neural differentiation of mouse embryonic stem cells (ESCs). We found that RNAi-mediated Chd4 knockdown suppresses neural differentiation, as did knockdown of methyl-CpG-binding domain protein Mbd3, another NuRD subunit. Chd4 and Mbd3 knockdowns similarly affected changes in global gene expression during neural differentiation and up-regulated several mesendodermal genes. However, inhibition of mesendodermal genes by knocking out the master regulators of mesendodermal lineages, Brachyury and Eomes, through a CRISPR/Cas9 approach could not restore the impaired neural differentiation caused by the Chd4 knockdown, suggesting that CHD4 controls neural differentiation by not repressing other lineage differentiation processes. Notably, Chd4 knockdown increased the acetylation levels of p53, resulting in increased protein levels of p53. Double knockdown of Chd4 and p53 restored the neural differentiation rate. Furthermore, overexpression of BCL2, a downstream factor of p53, partially rescued the impaired neural differentiation caused by the Chd4 knockdown. Our findings reveal that the CHD4/NuRD complex regulates neural differentiation of ESCs by down-regulating p53.


Asunto(s)
Diferenciación Celular , ADN Helicasas/metabolismo , Regulación hacia Abajo , Neuronas/metabolismo , Nucleosomas/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Animales , Línea Celular , ADN Helicasas/genética , Técnicas de Silenciamiento del Gen , Ratones , Células Madre Embrionarias de Ratones , Neuronas/citología , Nucleosomas/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína p53 Supresora de Tumor/genética
9.
Inflamm Regen ; 38: 22, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555601

RESUMEN

Throughout life, organisms are subjected to a variety of environmental perturbations, including temperature, nutrient conditions, and chemical agents. Exposure to external signals induces diverse changes in the physiological conditions of organisms. Genetically identical individuals exhibit highly phenotypic variations, which suggest that environmental variations among individuals can affect their phenotypes in a cumulative and inhomogeneous manner. The organismal phenotypes mediated by environmental conditions involve development, metabolic pathways, fertility, pathological processes, and even lifespan. It is clear that genetic factors influence the lifespan of organisms. Likewise, it is now increasingly recognized that environmental factors also have a large impact on the regulation of aging. Multiple studies have reported on the contribution of epigenetic signatures to the long-lasting phenotypic effects induced by environmental signals. Nevertheless, the mechanism of how environmental stimuli induce epigenetic changes at specific loci, which ultimately elicit phenotypic variations, is still largely unknown. Intriguingly, in some cases, the altered phenotypes associated with epigenetic changes could be stably passed on to the next generations. In this review, we discuss the environmental regulation of organismal viability, that is, longevity and stress resistance, and the relationship between this regulation and epigenetic factors, focusing on studies in the nematode C. elegans.

10.
Genes Cells ; 23(4): 254, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29637706
11.
J Biol Chem ; 293(22): 8342-8361, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29674317

RESUMEN

Epithelia contribute to physical barriers that protect internal tissues from the external environment and also support organ structure. Accordingly, establishment and maintenance of epithelial architecture are essential for both embryonic development and adult physiology. Here, using gene knockout and knockdown techniques along with gene profiling, we show that extracellular signal-regulated kinase 3 (ERK3), a poorly characterized atypical mitogen-activated protein kinase (MAPK), regulates the epithelial architecture in vertebrates. We found that in Xenopus embryonic epidermal epithelia, ERK3 knockdown impairs adherens and tight-junction protein distribution, as well as tight-junction barrier function, resulting in epidermal breakdown. Moreover, in human epithelial breast cancer cells, inhibition of ERK3 expression induced thickened epithelia with aberrant adherens and tight junctions. Results from microarray analyses suggested that transcription factor AP-2α (TFAP2A), a transcriptional regulator important for epithelial gene expression, is involved in ERK3-dependent changes in gene expression. Of note, TFAP2A knockdown phenocopied ERK3 knockdown in both Xenopus embryos and human cells, and ERK3 was required for full activation of TFAP2A-dependent transcription. Our findings reveal that ERK3 regulates epithelial architecture, possibly together with TFAP2A.


Asunto(s)
Neoplasias de la Mama/patología , Embrión no Mamífero/enzimología , Células Epiteliales/química , Proteína Quinasa 6 Activada por Mitógenos/metabolismo , Factor de Transcripción AP-2/metabolismo , Xenopus laevis/fisiología , Animales , Neoplasias de la Mama/enzimología , Sistemas CRISPR-Cas , Adhesión Celular , Membrana Celular , Células Cultivadas , Embrión no Mamífero/citología , Células Epiteliales/enzimología , Células Epiteliales/patología , Femenino , Células Hep G2 , Humanos , Proteína Quinasa 6 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 6 Activada por Mitógenos/genética , Uniones Estrechas , Factor de Transcripción AP-2/antagonistas & inhibidores , Factor de Transcripción AP-2/genética , Xenopus laevis/embriología
12.
Cell Rep ; 21(13): 3646-3652, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29281814

RESUMEN

Organismal lifespan is highly plastic in response to environmental cues, and dietary restriction (DR) is the most robust way to extend lifespan in various species. Recent studies have shown that sex also is an important factor for lifespan regulation; however, it remains largely unclear how these two factors, food and sex, interact in lifespan regulation. The nematode Caenorhabditis elegans has two sexes, hermaphrodite and male, and only the hermaphrodites are essential for the short-term succession of the species. Here, we report an extreme sexual dimorphism in the responsiveness to DR in C. elegans; the essential hermaphrodites show marked longevity responses to various forms of DR, but the males show few longevity responses and sustain reproductive ability. Our analysis reveals that the sex determination pathway and the steroid hormone receptor DAF-12 regulate the sex-specific DR responsiveness, integrating sex and environmental cues to determine organismal lifespan.


Asunto(s)
Caenorhabditis elegans/fisiología , Restricción Calórica , Caracteres Sexuales , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Femenino , Organismos Hermafroditas/fisiología , Longevidad/fisiología , Masculino , Modelos Genéticos , Transcripción Genética
13.
EMBO Rep ; 2017 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-28794203

RESUMEN

The well-known link between longevity and the Sir2 histone deacetylase family suggests that histone deacetylation, a modification associated with repressed chromatin, is beneficial to longevity. However, the molecular links between histone acetylation and longevity remain unclear. Here, we report an unexpected finding that the MYST family histone acetyltransferase complex (MYS-1/TRR-1 complex) promotes rather than inhibits stress resistance and longevity in Caenorhabditis elegans Our results show that these beneficial effects are largely mediated through transcriptional up-regulation of the FOXO transcription factor DAF-16. MYS-1 and TRR-1 are recruited to the promoter regions of the daf-16 gene, where they play a role in histone acetylation, including H4K16 acetylation. Remarkably, we also find that the human MYST family Tip60/TRRAP complex promotes oxidative stress resistance by up-regulating the expression of FOXO transcription factors in human cells. Tip60 is recruited to the promoter regions of the foxo1 gene, where it increases H4K16 acetylation levels. Our results thus identify the evolutionarily conserved role of the MYST family acetyltransferase as a key epigenetic regulator of DAF-16/FOXO transcription factors.

14.
FEBS Lett ; 591(11): 1584-1600, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28471520

RESUMEN

The process of cell reprogramming has been characterized considerably since the successful generation of induced pluripotent stem cells. However, the importance of cell-cell communications for cellular reprogramming remains largely unknown. Secreted factors, which are expressed and secreted during reprogramming, may influence the reprogramming efficiency. Here, we have identified Sostdc1, Glb1l2, Fetub, Dpp4, Gdf3, Trh, and Tdgf1 as prominently upregulated secreted factors during reprogramming. Our detailed analysis reveals that these seven factors may be categorized into four groups based on their expression patterns in relation to the reprogramming stages. Remarkably, knockdown of Sostdc1, which is the most prominently upregulated factor and which is expressed earlier than the other six factors, results in reduced reprogramming efficiency, suggesting its involvement in the reprogramming process.


Asunto(s)
Reprogramación Celular/genética , Regulación de la Expresión Génica , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Fetuína-B/genética , Fetuína-B/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Factor 3 de Diferenciación de Crecimiento/genética , Factor 3 de Diferenciación de Crecimiento/metabolismo , Immunoblotting , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Análisis por Micromatrices , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Mol Cell Biol ; 37(15)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28483913

RESUMEN

Deregulated activation of RAS/extracellular signal-regulated kinase (ERK) signaling and defects in retinoic acid receptor (RAR) signaling are both implicated in many types of cancers. However, interrelationships between these alterations in regulating cancer cell fates have not been fully elucidated. Here, we show that RAS/ERK and RAR signaling pathways antagonistically interact with each other to regulate colorectal cancer (CRC) cell fates. We show that RAR signaling activation promotes spontaneous differentiation of CRC cells, while ERK activation suppresses it. Our microarray analyses identify genes whose expression levels are upregulated by RAR signaling. Notably, one of these genes, MKP4, encoding a member of dual-specificity phosphatases for mitogen-activated protein (MAP) kinases, mediates ERK inactivation upon RAR activation, thereby promoting the differentiation of CRC cells. Moreover, our results also show that RA induction of RAR target genes is suppressed by the ERK pathway activation. This suppression results from the inhibition of RAR transcriptional activity, which is shown to be mediated through an RIP140/histone deacetylase (HDAC)-mediated mechanism. These results identify antagonistic interactions between RAS/ERK and RAR signaling in the cell fate decision of CRC cells and define their underlying molecular mechanisms.


Asunto(s)
Colon/patología , Neoplasias Colorrectales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Ácido Retinoico/metabolismo , Recto/patología , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Colon/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/metabolismo , Humanos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Proteínas Nucleares/metabolismo , Proteína de Interacción con Receptores Nucleares 1 , Regiones Promotoras Genéticas , Recto/metabolismo
16.
J Biol Chem ; 292(27): 11300-11309, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28507100

RESUMEN

Intermittent fasting (IF) is a dietary restriction regimen that extends the lifespans of Caenorhabditis elegans and mammals by inducing changes in gene expression. However, how IF induces these changes and promotes longevity remains unclear. One proposed mechanism involves gene regulation by microRNAs (miRNAs), small non-coding RNAs (∼22 nucleotides) that repress gene expression and whose expression can be altered by fasting. To test this proposition, we examined the role of the miRNA machinery in fasting-induced transcriptional changes and longevity in C. elegans We revealed that fasting up-regulated the expression of the miRNA-induced silencing complex (miRISC) components, including Argonaute and GW182, and the miRNA-processing enzyme DRSH-1 (the ortholog of the Drosophila Drosha enzyme). Our lifespan measurements demonstrated that IF-induced longevity was suppressed by knock-out or knockdown of miRISC components and was completely inhibited by drsh-1 ablation. Remarkably, drsh-1 ablation inhibited the fasting-induced changes in the expression of the target genes of DAF-16, the insulin/IGF-1 signaling effector in C. elegans Fasting-induced transcriptome alterations were substantially and modestly suppressed in the drsh-1 null mutant and the null mutant of ain-1, a gene encoding GW182, respectively. Moreover, miRNA array analyses revealed that the expression levels of numerous miRNAs changed after 2 days of fasting. These results indicate that components of the miRNA machinery, especially the miRNA-processing enzyme DRSH-1, play an important role in mediating IF-induced longevity via the regulation of fasting-induced changes in gene expression.


Asunto(s)
Proteínas de Caenorhabditis elegans/biosíntesis , Caenorhabditis elegans/metabolismo , Proteínas Portadoras/biosíntesis , Ayuno , Regulación de la Expresión Génica , Longevidad/fisiología , MicroARNs/metabolismo , Ribonucleasa III/biosíntesis , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas Portadoras/genética , MicroARNs/genética , Ribonucleasa III/genética
17.
Nat Cell Biol ; 19(5): 530-541, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28414314

RESUMEN

Recent studies have revealed that newly emerging transformed cells are often apically extruded from epithelial tissues. During this process, normal epithelial cells can recognize and actively eliminate transformed cells, a process called epithelial defence against cancer (EDAC). Here, we show that mitochondrial membrane potential is diminished in RasV12-transformed cells when they are surrounded by normal cells. In addition, glucose uptake is elevated, leading to higher lactate production. The mitochondrial dysfunction is driven by upregulation of pyruvate dehydrogenase kinase 4 (PDK4), which positively regulates elimination of RasV12-transformed cells. Furthermore, EDAC from the surrounding normal cells, involving filamin, drives the Warburg-effect-like metabolic alteration. Moreover, using a cell-competition mouse model, we demonstrate that PDK-mediated metabolic changes promote the elimination of RasV12-transformed cells from intestinal epithelia. These data indicate that non-cell-autonomous metabolic modulation is a crucial regulator for cell competition, shedding light on the unexplored events at the initial stage of carcinogenesis.


Asunto(s)
Comunicación Celular , Transformación Celular Neoplásica/metabolismo , Metabolismo Energético , Células Epiteliales/metabolismo , Animales , Línea Celular Transformada , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Técnicas de Cocultivo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Perros , Femenino , Genes ras , Glucosa/metabolismo , Glucólisis , Ácido Láctico/metabolismo , Células de Riñón Canino Madin Darby , Masculino , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal , Técnicas de Cultivo de Tejidos , Transfección
18.
Genes Cells ; 22(2): 189-202, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28097777

RESUMEN

The Activin/Nodal/TGF-ß signaling pathway plays a major role in maintaining mouse epiblast stem cells (EpiSCs). The EpiSC-maintaining medium, which contains Activin A and bFGF, induces differentiation of mouse embryonic stem cells (ESCs) to EpiSCs. Here, we show that Activin A also has an ability to efficiently propagate ESCs without differentiation to EpiSCs when combined with a MEK inhibitor PD0325901. ESCs cultured in Activin+PD retained high-level expression of naive pluripotency-related transcription factors. Genomewide analysis showed that the gene expression profile of ESCs cultured in Activin+PD resembles that of ESCs cultured in 2i. ESCs cultured in Activin+PD also showed features common to the naive pluripotency of ESCs, including the preferential usage of the Oct4 distal enhancer and the self-renewal response to Wnt pathway activation. Our finding shows a role of Activin/Nodal/TGF-ß signaling in stabilizing self-renewal gene regulatory networks in ESCs.


Asunto(s)
Activinas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Activinas/química , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt
19.
Genes Cells ; 22(2): 210-219, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28105749

RESUMEN

Dietary restriction regimens lead to enhanced stress resistance and extended life span in many species through the regulation of fasting and/or diet-responsive mechanisms. The fasting stimulus is perceived by sensory neurons and causes behavioral and metabolic adaptations. Octopamine (OA), one of the Caenorhabditis elegans neurotransmitters, is involved in behavioral adaptations, and its levels are increased under fasting conditions. However, it remains largely unknown how OA contributes to the fasting responses. In this study, we found that OA administration enhanced organismal resistance to oxidative stress. This enhanced resistance was suppressed by a mutation of the OA receptors, SER-3 and SER-6. Moreover, we found that OA administration promoted the nuclear translocation of DAF-16, the key transcription factor in fasting responses, and that the OA-induced enhancement of stress resistance required DAF-16. Altogether, our results suggest that OA signaling, which is triggered by the absence of food, shifts the organismal state to a more protective one to prepare for environmental stresses.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Factores de Transcripción Forkhead/metabolismo , Octopamina/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Caenorhabditis elegans , Ayuno/fisiología , Longevidad , Mutación , Estrés Oxidativo/fisiología , Transducción de Señal
20.
Nat Commun ; 8: 14031, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28067237

RESUMEN

Hormesis is a biological phenomenon, whereby exposure to low levels of toxic agents or conditions increases organismal viability. It thus represents a beneficial aspect of adaptive responses to harmful environmental stimuli. Here we show that hormesis effects induced in the parental generation can be passed on to the descendants in Caenorhabditis elegans. Animals subjected to various stressors during developmental stages exhibit increased resistance to oxidative stress and proteotoxicity. The increased resistance is transmitted to the subsequent generations grown under unstressed conditions through epigenetic alterations. Our analysis reveal that the insulin/insulin-like growth factor (IGF) signalling effector DAF-16/FOXO and the heat-shock factor HSF-1 in the parental somatic cells mediate the formation of epigenetic memory, which is maintained through the histone H3 lysine 4 trimethylase complex in the germline across generations. The elicitation of memory requires the transcription factor SKN-1/Nrf in somatic tissues. We propose that germ-to-soma communication across generations is an essential framework for the transgenerational inheritance of acquired traits, which provides the offspring with survival advantages to deal with environmental perturbation.


Asunto(s)
Adaptación Fisiológica/genética , Caenorhabditis elegans/efectos de los fármacos , Epigénesis Genética , Hormesis , Patrón de Herencia , Larva/efectos de los fármacos , Animales , Arsenitos/farmacología , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células Germinativas , Histonas/genética , Histonas/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Longevidad/efectos de los fármacos , Concentración Osmolar , Péptidos/farmacología , Carácter Cuantitativo Heredable , Transducción de Señal , Cloruro de Sodio/farmacología , Compuestos de Sodio/farmacología , Inanición , Estrés Fisiológico , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA