Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biol Res ; 57(1): 26, 2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38735981

RESUMEN

BACKGROUND: Vitamin C (ascorbate) is a water-soluble antioxidant and an important cofactor for various biosynthetic and regulatory enzymes. Mice can synthesize vitamin C thanks to the key enzyme gulonolactone oxidase (Gulo) unlike humans. In the current investigation, we used Gulo-/- mice, which cannot synthesize their own ascorbate to determine the impact of this vitamin on both the transcriptomics and proteomics profiles in the whole liver. The study included Gulo-/- mouse groups treated with either sub-optimal or optimal ascorbate concentrations in drinking water. Liver tissues of females and males were collected at the age of four months and divided for transcriptomics and proteomics analysis. Immunoblotting, quantitative RT-PCR, and polysome profiling experiments were also conducted to complement our combined omics studies. RESULTS: Principal component analyses revealed distinctive differences in the mRNA and protein profiles as a function of sex between all the mouse cohorts. Despite such sexual dimorphism, Spearman analyses of transcriptomics data from females and males revealed correlations of hepatic ascorbate levels with transcripts encoding a wide array of biological processes involved in glucose and lipid metabolisms as well as in the acute-phase immune response. Moreover, integration of the proteomics data showed that ascorbate modulates the abundance of various enzymes involved in lipid, xenobiotic, organic acid, acetyl-CoA, and steroid metabolism mainly at the transcriptional level, especially in females. However, several proteins of the mitochondrial complex III significantly correlated with ascorbate concentrations in both males and females unlike their corresponding transcripts. Finally, poly(ribo)some profiling did not reveal significant enrichment difference for these mitochondrial complex III mRNAs between Gulo-/- mice treated with sub-optimal and optimal ascorbate levels. CONCLUSIONS: Thus, the abundance of several subunits of the mitochondrial complex III are regulated by ascorbate at the post-transcriptional levels. Our extensive omics analyses provide a novel resource of altered gene expression patterns at the transcriptional and post-transcriptional levels under ascorbate deficiency.


Asunto(s)
Ácido Ascórbico , Hígado , Proteómica , Animales , Ácido Ascórbico/metabolismo , Hígado/metabolismo , Hígado/efectos de los fármacos , Femenino , Masculino , Ratones , L-Gulonolactona Oxidasa/genética , L-Gulonolactona Oxidasa/metabolismo , Perfilación de la Expresión Génica , Transcriptoma , Análisis de Componente Principal , Antioxidantes/metabolismo
2.
J Proteome Res ; 20(11): 5036-5053, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34643398

RESUMEN

A suboptimal blood vitamin C (ascorbate) level increases the risk of several chronic diseases. However, the detection of hypovitaminosis C is not a simple task, as ascorbate is unstable in blood samples. In this study, we examined the serum proteome of mice lacking the gulonolactone oxidase (Gulo) required for the ascorbate biosynthesis. Gulo-/- mice were supplemented with different concentrations of ascorbate in drinking water, and serum was collected to identify proteins correlating with serum ascorbate levels using an unbiased label-free liquid chromatography-tandem mass spectrometry global quantitative proteomic approach. Parallel reaction monitoring was performed to validate the correlations. We uncovered that the serum proteome profiles differ significantly between male and female mice. Also, unlike Gulo-/- males, a four-week ascorbate treatment did not entirely re-establish the serum proteome profile of ascorbate-deficient Gulo-/- females to the optimal profile exhibited by Gulo-/- females that never experienced an ascorbate deficiency. Finally, the serum proteins involved in retinoid metabolism, cholesterol, and lipid transport were similarly affected by ascorbate levels in males and females. In contrast, the proteins regulating serum peptidases and the protein of the acute phase response were different between males and females. These proteins are potential biomarkers correlating with blood ascorbate levels and require further study in standard clinical settings. The complete proteomics data set generated in this study has been deposited to the public repository ProteomeXchange with the data set identifier: PXD027019.


Asunto(s)
Ácido Ascórbico , Proteoma , Animales , Suplementos Dietéticos , Femenino , L-Gulonolactona Oxidasa/metabolismo , Masculino , Ratones , Proteoma/genética , Proteómica
3.
Nucleic Acids Res ; 47(14): 7532-7547, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31219578

RESUMEN

Fanconi Anemia (FA) clinical phenotypes are heterogenous and rely on a mutation in one of the 22 FANC genes (FANCA-W) involved in a common interstrand DNA crosslink-repair pathway. A critical step in the activation of FA pathway is the monoubiquitination of FANCD2 and its binding partner FANCI. To better address the clinical phenotype associated with FANCI and the epistatic relationship with FANCD2, we created the first conditional inactivation model for FANCI in mouse. Fanci -/- mice displayed typical FA features such as delayed development in utero, microphtalmia, cellular sensitivity to mitomycin C, occasional limb abnormalities and hematological deficiencies. Interestingly, the deletion of Fanci leads to a strong meiotic phenotype and severe hypogonadism. FANCI was localized in spermatocytes and spermatids and in the nucleus of oocytes. Both FANCI and FANCD2 proteins co-localized with RPA along meiotic chromosomes, albeit at different levels. Consistent with a role in meiotic recombination, FANCI interacted with RAD51 and stimulated D-loop formation, unlike FANCD2. The double knockout Fanci-/- Fancd2-/- also showed epistatic relationship for hematological defects while being not epistatic with respect to generating viable mice in crosses of double heterozygotes. Collectively, this study highlights common and distinct functions of FANCI and FANCD2 during mouse development, meiotic recombination and hematopoiesis.


Asunto(s)
Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Espermatocitos/metabolismo
4.
FASEB J ; 32(7): 3623-3640, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29452565

RESUMEN

Werner syndrome (WS) is a premature aging disorder caused by mutations in a RecQ-family DNA helicase (WRN). Mice lacking part of the helicase domain of the WRN ortholog exhibit several phenotypic features of WS. In this study, we generated a Wrn mutant line that, like humans, relies entirely on dietary sources of vitamin C (ascorbate) to survive, by crossing them to mice that lack the gulonolactone oxidase enzyme required for ascorbate synthesis. In the presence of 0.01% ascorbate (w/v) in drinking water, double-mutant mice exhibited a severe reduction in lifespan, small size, sterility, osteopenia, and metabolic profiles different from wild-type (WT) mice. Although increasing the dose of ascorbate to 0.4% improved dramatically the phenotypes of double-mutant mice, the metabolic and cytokine profiles were different from age-matched WT mice. Finally, double-mutant mice treated with 0.01% ascorbate revealed a permanent activation of all the 3 branches of the ER stress response pathways due to a severe chronic oxidative stress in the ER compartment. In addition, markers associated with the ubiquitin-proteasome-dependent ER-associated degradation pathway were increased. Augmenting the dose of ascorbate reversed the activation of this pathway to WT levels rendering this pathway a potential therapeutic target in WS.-Aumailley, L., Dubois, M. J., Brennan, T. A., Garand, C., Paquet, E. R., Pignolo, R. J., Marette, A., Lebel, M. Serum vitamin C levels modulate the lifespan and endoplasmic reticulum stress response pathways in mice synthesizing a nonfunctional mutant WRN protein.


Asunto(s)
Ácido Ascórbico/sangre , Estrés del Retículo Endoplásmico , Longevidad , Helicasa del Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Animales , Ácido Ascórbico/uso terapéutico , Femenino , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Endogámicos C57BL , Síndrome de Werner/tratamiento farmacológico , Síndrome de Werner/genética
5.
Brain Behav Immun ; 73: 450-469, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29908963

RESUMEN

Werner syndrome (WS) is a premature aging disorder caused by mutations in a RecQ-family DNA helicase, WRN. Mice lacking part of the helicase domain of the WRN orthologue exhibit many phenotypic features of WS, including metabolic abnormalities and a shorter lifespan. Yet, little is known about the impact of WRN mutations on the central nervous system in both humans and mouse models of WS. In the current study, we have performed a longitudinal behavioral assessment on mice bearing a Wrn helicase deletion. Behavioral tests demonstrated a loss of motor activity and coordination, reduction in perception, increase in repetitive behavior, and deficits in both spatial and social novelty memories in Wrn mutant mice compared to age-matched wild type mice. These neurological deficits were associated with biochemical and histological changes in the brain of aged Wrn mutant mice. Microglia, resident immune cells that regulate neuronal plasticity and function in the brain, were hyper-ramified in multiple regions involved with the behavioral deficits of Wrn mutant mice. Furthermore, western analyses indicated that Wrn mutant mice exhibited an increase of oxidative stress markers in the prefrontal cortex. Supporting these findings, electron microscopy studies revealed increased cellular aging and oxidative stress features, among microglia and neurons respectively, in the prefrontal cortex of aged Wrn mutant mice. In addition, multiplex immunoassay of serum identified significant changes in the expression levels of several pro- and anti-inflammatory cytokines. Taken together, these findings indicate that microglial dysfunction and neuronal oxidative stress, associated with peripheral immune system alterations, might be important driving forces leading to abnormal neurological symptoms in WS thus suggesting potential therapeutic targets for interventions.


Asunto(s)
Helicasa del Síndrome de Werner/fisiología , Síndrome de Werner/genética , Animales , Senescencia Celular/fisiología , Daño del ADN/fisiología , Modelos Animales de Enfermedad , Femenino , Estudios Longitudinales , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Actividad Motora/genética , Actividad Motora/fisiología , Proteínas Mutantes , Neuronas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Síndrome de Werner/inmunología , Síndrome de Werner/fisiopatología , Helicasa del Síndrome de Werner/genética
6.
BMC Genomics ; 15: 940, 2014 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-25346348

RESUMEN

BACKGROUND: Werner Syndrome (WS) is a rare disorder characterized by the premature onset of a number of age-related diseases. The gene responsible for WS encodes a DNA helicase/exonuclease protein believed to affect different aspects of transcription, replication, and DNA repair. Caenorhabditis elegans (C. elegans) with a nonfunctional wrn-1 DNA helicase ortholog also exhibits a shorter life span, which can be rescued by vitamin C. In this study, we analyzed the impact of a mutation in the wrn-1 gene and the dietary supplementation of vitamin C on the global mRNA expression of the whole C. elegans by the RNA-seq technology. RESULTS: Vitamin C increased the mean life span of the wrn-1(gk99) mutant and the N2 wild type strains at 25°C. However, the alteration of gene expression by vitamin C is different between wrn-1(gk99) and wild type strains. We observed alteration in the expression of 1522 genes in wrn-1(gk99) worms compared to wild type animals. Such genes significantly affected the metabolism of lipid, cellular ketone, organic acid, and carboxylic acids. Vitamin C, in return, altered the expression of genes in wrn-1(gk99) worms involved in locomotion and anatomical structure development. Proteolysis was the only biological process significantly affected by vitamin C in wild type worms. CONCLUSIONS: Expression profiling of wrn-1(gk99) worms revealed a very different response to the addition of vitamin C compared to wild type worms. Finally, vitamin C extended the life span of wrn-1(gk99) animals by altering biological processes involved mainly in locomotion and anatomical structure development.


Asunto(s)
Ácido Ascórbico/farmacología , Caenorhabditis elegans/efectos de los fármacos , Longevidad/efectos de los fármacos , Mutación , Homología de Secuencia de Ácido Nucleico , Transcriptoma/efectos de los fármacos , Síndrome de Werner/genética , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/fisiología , Longevidad/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN
7.
Aging (Albany NY) ; 16(10): 8417-8445, 2024 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-38795389

RESUMEN

Werner syndrome (WS) is a progeroid disorder caused by mutations in a protein containing both a DNA exonuclease and DNA helicase domains. Previous studies indicated that males lacking the helicase domain of the Wrn protein orthologue exhibited hepatic transcriptomic and metabolic alterations. In this study, we used a label-free liquid chromatography-tandem mass spectrometry approach to uncover proteins abundance associated with specific biological processes that differed depending on the age (four or ten months) and/or the genotype (wild type or Wrn mutant) in the serum and liver of mice. Principal component analysis of the proteomic data from both serum and hepatic tissue revealed a sexual dimorphism regardless of the age and the genotype of the mice. Moreover, although all Wrn mutant mice exhibited fatty liver by the age of ten months, a significant age and genotype dependent enrichment of proteins involved in lipid and fatty acid metabolic processes were uncovered only in males. Also, a genotype dependent increase in serum oxidant detoxification processes was observed in the serum of Wrn mutant males. Despite these sexual differences, several aspects of the immune system were affected in both females and males. Finally, an increase of specific immunoglobulin molecules was common in the liver and serum of both older Wrn mutant females and males. Such results suggest that specific immunoglobulin variants maybe associated with fatty liver progression in WS.


Asunto(s)
Envejecimiento , Modelos Animales de Enfermedad , Hígado , Proteómica , Caracteres Sexuales , Helicasa del Síndrome de Werner , Síndrome de Werner , Animales , Masculino , Femenino , Síndrome de Werner/genética , Hígado/metabolismo , Ratones , Envejecimiento/inmunología , Helicasa del Síndrome de Werner/genética , Helicasa del Síndrome de Werner/metabolismo
8.
J Nutr Biochem ; 125: 109538, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38030046

RESUMEN

Vitamin C (ascorbic acid) is an important water-soluble antioxidant associated with decreased oxidative stress in type 2 diabetes (T2D) patients. A previous targeted plasma proteomic study has indicated that ascorbic acid is associated with markers of the immune system in healthy subjects. However, the association between the levels of ascorbic acid and blood biomarkers in subjects at risk of developing T2D is still unknown. Serum ascorbic acid was measured by ultra-performance liquid chromatography and serum proteins were quantified by untargeted liquid-chromatography mass spectrometry in 25 hyperinsulinemia subjects that were randomly assigned a high dairy intake diet or an adequate dairy intake diet for 6 weeks, then crossed-over after a 6-week washout period. Spearman correlation followed by gene ontology analyses were performed to identify biological pathways associated with ascorbic acid. Finally, machine learning analysis was performed to obtain a specific serum protein signature that could predict ascorbic acid levels. After adjustments for waist circumference, LDL, HDL, fasting insulin, fasting blood glucose, age, gender, and dairy intake; serum ascorbic acid correlated positively with different aspects of the immune system. Machine learning analysis indicated that a signature composed of 21 features that included 17 proteins (mainly from the immune system), age, sex, waist circumference, and LDL could predict serum ascorbic acid levels in hyperinsulinemia subjects. In conclusion, the result reveals a correlation as well as modulation between serum ascorbic acid levels and proteins that play vital roles in regulating different aspects of the immune response in individuals at risk of T2D. The development of a predictive signature for ascorbic acid will further help the assessment of ascorbic acid status in clinical settings.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperinsulinismo , Humanos , Ácido Ascórbico , Proteínas Sanguíneas , Lipoproteínas LDL , Proteómica , Circunferencia de la Cintura , Masculino , Femenino
9.
BMC Cancer ; 13: 303, 2013 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-23800275

RESUMEN

BACKGROUND: The X-linked ribosomal protein S4 (RPS4X), which is involved in cellular translation and proliferation, has previously been identified as a partner of the overexpressed multifunctional protein YB-1 in several breast cancer cells. Depletion of RPS4X results in consistent resistance to cisplatin in such cell lines. METHODS: As platinum-based chemotherapy is a standard first line therapy used to treat patients with ovarian cancer, we evaluated the prognostic value of RPS4X and YB-1 at the protein level in specimen from 192 high-grade serous epithelial ovarian cancer patients. RESULTS: Immunohistochemistry studies indicated that high expression of RPS4X was associated with a lower risk of death and later disease progression (HR = 0.713, P = 0.001 and HR = 0.761, P = 0.001, respectively) as compared to low expression of RPS4X. In contrast, YB-1 was not significantly associated with either recurrence or survival time in this cohort. Finally, the depletion of RPS4X with different siRNAs in two different ovarian cancer cell lines reduced their proliferative growth rate but more importantly increased their resistance to cisplatin. CONCLUSION: Altogether, these results suggest that the levels of RPS4X could be a good indicator for resistance to platinum-based therapy and a prognostic marker for ovarian cancer. Our study also showed that RPS4X is an independent prognostic factor in patients with serous epithelial ovarian cancer.


Asunto(s)
Cistadenocarcinoma Seroso/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Ribosómicas/biosíntesis , Biomarcadores de Tumor/análisis , Western Blotting , Carcinoma Epitelial de Ovario , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/mortalidad , Resistencia a Antineoplásicos/fisiología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Persona de Mediana Edad , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/mortalidad , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , Proteínas Ribosómicas/análisis , Análisis de Matrices Tisulares , Transfección , Proteína 1 de Unión a la Caja Y/análisis , Proteína 1 de Unión a la Caja Y/biosíntesis
10.
Exp Cell Res ; 318(14): 1620-32, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22659133

RESUMEN

Werner syndrome (WS) is a rare autosomal disease characterized by the premature onset of several age-associated pathologies. The protein defective in WS patients (WRN) is a helicase/exonuclease involved in DNA repair, replication, transcription and telomere maintenance. Hypoxia-inducible factor-1 (HIF-1) is a decisive element for the transcriptional regulation of genes essential for adaptation to low oxygen conditions. HIF-1 is also implicated in the molecular mechanisms of ageing. Here, we show that the cellular depletion of WRN protein (by siRNA targeting) leads to increased HIF-1 complex stabilization and activation. HIF-1 activation in the absence of WRN involves the generation of mitochondrial reactive oxygen species (mtROS) since SkQ1, a mitochondrial-targeted antioxidant, and stigmatellin, an inhibitor of mitochondrial complex III, blocked increased HIF-1 levels. Ascorbate, an essential co-factor involved in HIF-1 stability, was decreased in WRN-depleted cells. Interestingly, expression levels of GLUT1, a known dehydroascorbic acid transporter, were also decreased in WRN-depleted cells. Ascorbate supplementation of WRN-depleted cells led to a dose-dependent inhibition of HIF-1 activation. These results indicate that WRN protein regulates HIF-1 activation by affecting mitochondrial ROS production and intracellular ascorbate levels. This work provides a novel mechanistic link between HIF-1 activity and different age-associated pathologies.


Asunto(s)
Exodesoxirribonucleasas/metabolismo , Proteínas de Neoplasias/metabolismo , RecQ Helicasas/metabolismo , Síndrome de Werner/genética , Células Cultivadas , Exodesoxirribonucleasas/genética , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mitocondrias/metabolismo , Proteínas Mitocondriales , Especies Reactivas de Oxígeno/metabolismo , RecQ Helicasas/genética , Helicasa del Síndrome de Werner
11.
Nutrients ; 15(22)2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-38004101

RESUMEN

Bile acids regulate glucose homeostasis and lipid metabolism. Further, the levels of bile acids can be influenced by the intake of dairy products. Although the serum proteome can provide information on the biological pathways associated with different metabolites, it is unknown whether the intake of dairy modifies such associations between bile acids and the proteome. The objectives of this study were to examine plasma bile acid profiles, find the correlations between bile acids and lipid as well as glycemic markers, and to uncover the correlation between bile acids and proteins after high dairy (HD) and adequate dairy (AD) intake among 25 overweight individuals with hyperinsulinemia. In this randomized crossover-trial study, hyperinsulinemia adults were randomized to both HD (≥4 servings/day) and AD (≤2 servings/day) for 6 weeks. Measurements and analyses were performed on before- as well as after- AD and HD conditions. The results indicated that plasma 7α-hydroxy-4-cholesten-3-one (7AC4) increased after HD in comparison with before HD intake (p = 0.03). After adjusting for BMI, age, and sex, 7AC4 positively correlated with triglyceride levels in the pre-AD (r = 0.44; p = 0.03) and post-HD (r = 0.42; p = 0.04). Further, 7AC4 correlated positively with proteins associated with high-density lipoprotein particle remodeling pathway and reverse cholesterol transport only after HD consumption. Thus, the consumption of higher dairy intake modifies the association between 7AC4-a biomarker for bile acid synthesis-and serum proteins involved in cholesterol clearance. Overall, higher dairy consumption may have a positive effect on cholesterol metabolism in subjects at risk of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperinsulinismo , Adulto , Humanos , Ácidos y Sales Biliares , Proteoma , Colesterol , Productos Lácteos , Proteínas Sanguíneas
12.
Gut Microbes ; 15(2): 2271597, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37876025

RESUMEN

Shigella spp. are the causative agents of bacterial dysentery and shigellosis, mainly in children living in developing countries. The study of Shigella entire life cycle in vivo and the evaluation of vaccine candidates' protective efficacy have been hampered by the lack of a suitable animal model of infection. None of the studies evaluated so far (rabbit, guinea pig, mouse) allowed the recapitulation of full shigellosis symptoms upon Shigella oral challenge. Historical reports have suggested that dysentery and scurvy are both metabolic diseases associated with ascorbate deficiency. Mammals, which are susceptible to Shigella infection (humans, non-human primates and guinea pigs) are among the few species unable to synthesize ascorbate. We optimized a low-ascorbate diet to induce moderate ascorbate deficiency, but not scurvy, in guinea pigs to investigate whether poor vitamin C status increases the progression of shigellosis. Moderate ascorbate deficiency increased shigellosis symptom severity during an extended period of time (up to 48 h) in all strains tested (Shigella sonnei, Shigella flexneri 5a, and 2a). At late time points, an important influx of neutrophils was observed both within the disrupted colonic mucosa and in the luminal compartment, although Shigella was able to disseminate deep into the organ to reach the sub-mucosal layer and the bloodstream. Moreover, we found that ascorbate deficiency also increased Shigella penetration into the colon epithelium layer in a Gulo-/- mouse infection model. The use of these new rodent models of shigellosis opens new doors for the study of both Shigella infection strategies and immune responses to Shigella infection.


Asunto(s)
Disentería Bacilar , Microbioma Gastrointestinal , Shigella , Cobayas , Humanos , Animales , Conejos , Ratones , Disentería Bacilar/microbiología , Modelos Animales de Enfermedad , Shigella flexneri , Ácido Ascórbico , Mamíferos
13.
Redox Biol ; 57: 102491, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36179436

RESUMEN

Ascorbate is a crucial antioxidant and essential cofactor of biosynthetic and regulatory enzymes. Unlike humans, mice can synthesize ascorbate thanks to the key enzyme gulonolactone oxidase (Gulo). In the present study, we used the Gulo-/- mouse model, which cannot synthesize their own ascorbate to determine the impact of this vitamin on the liver proteome of specific subcellular organelles. We performed label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) global quantitative proteomic profiling to identify and quantify proteins in microsomal enriched liver extracts (MEE) from Gulo-/- mice treated with 0-0.4% (w/v) ascorbate in drinking water until the age of four months. Using a principal component analysis on normalized and imputed data of the label-free protein quantifications, a sex-based difference in MEE proteome profiles was observed for all the different ascorbate treated mice. Suboptimal hepatic ascorbate concentrations affected the levels of more proteins and hence biochemical processes in females than in males. Nevertheless, Pearson correlation analyses revealed that the MS intensities of various proteins involved in complement activation inversely correlated with liver ascorbate concentrations in both Gulo-/- males and females. Moreover, the correlation analyses also indicated that several proteins in the mitochondrial complex III of the electron transport chain positively correlated with liver ascorbate concentrations in both Gulo-/- females and males. Consequently, the mitochondrial complex III activity in Gulo-/- female and male mice treated with suboptimal hepatic concentrations of ascorbate was significantly lower than Gulo-/- mice treated with optimal ascorbate concentration. Finally, the whole liver of ascorbate-deficient Gulo-/- mice exhibited lower ATP levels and increased reactive oxygen species. These findings provide new information on how ascorbate deficiency potentially induces mitochondrial dysfunction in the liver of mice.

14.
J Proteome Res ; 10(3): 1216-27, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21210717

RESUMEN

Werner syndrome (WS) is characterized by the premature onset of several age-associated pathologies. The protein defective in WS patients (WRN) is a helicase/exonuclease involved in DNA repair, replication, telomere maintenance, and transcription. Here, we present the results of a large-scale proteome analysis to determine protein partners of WRN. We expressed fluorescent tagged-WRN (eYFP-WRN) in human 293 embryonic kidney cells and detected interacting proteins by co-immunoprecipitation from cell extract. We identified by mass spectrometry 220 nuclear proteins that complexed with WRN. This number was reduced to 40 when broad-spectrum nucleases were added to the lysate. We consider these 40 proteins as directly interacting with WRN. Some of these proteins have previously been shown to interact with WRN, whereas most are new partners. Among the top 15 hits, we find the new interactors TMPO, HNRNPU, RPS3, RALY, RPS9 DDX21, and HNRNPM. These proteins are likely important components in understanding the function of WRN in preventing premature aging and deserve further investigation. We have confirmed endogenous WRN interaction with endogenous RPS3, a ribosomal protein with endonuclease activities involved in oxidative DNA damage recognition. Our results suggest that the use of nucleases during cell lysis severely restricts interacting protein partners and thus enhances specificity.


Asunto(s)
Desoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/metabolismo , Proteoma/análisis , RecQ Helicasas/química , RecQ Helicasas/metabolismo , Animales , Cromatografía Liquida/métodos , Exodesoxirribonucleasas/genética , Células HEK293 , Células HeLa , Humanos , Unión Proteica , RecQ Helicasas/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reproducibilidad de los Resultados , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Espectrometría de Masas en Tándem/métodos , Síndrome de Werner/patología , Síndrome de Werner/fisiopatología , Helicasa del Síndrome de Werner
15.
Mol Cancer ; 10: 145, 2011 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-22118625

RESUMEN

BACKGROUND: YB-1 is a multifunctional protein that affects transcription, splicing, and translation. Overexpression of YB-1 in breast cancers causes cisplatin resistance. Recent data have shown that YB-1 is also overexpress in colorectal cancer. In this study, we tested the hypothesis that YB-1 also confers oxaliplatin resistance in colorectal adenocarcinomas. RESULTS: We show for the first time that transfection of YB-1 cDNA confers oxaliplatin resistance in two colorectal cancer cell lines (SW480 and HT29 cell lines). Furthermore, we identified by mass spectrometry analyses important YB-1 interactors required for such oxaliplatin resistance in these colorectal cancer cell lines. A tagged YB-1 construct was used to identify proteins interacting directly to YB-1 in such cells. We then focused on proteins that are potentially involved in colorectal cancer progression based on the Oncomine microarray database. Genes encoding for these YB-1 interactors were also examined in the public NCBI comparative genomic hybridization database to determine whether these genes are localized to regions of chromosomes rearranged in colorectal cancer tissues. From these analyses, we obtained a list of proteins interacting with YB-1 and potentially involved in oxaliplatin resistance. Oxaliplatin dose response curves of SW480 and HT29 colorectal cancer cell lines transfected with several siRNAs corresponding to each of these YB-1 interactors were obtained to identify proteins significantly affecting oxaliplatin sensitivity upon gene silencing. Only the depletion of either NONO or RALY sensitized both colorectal cancer cell lines to oxaliplatin. Furthermore, depletion of NONO or RALY sensitized otherwise oxaliplatin resistant overexpressing YB-1 SW480 or HT29 cells. CONCLUSION: These results suggest knocking down NONO or RALY significant counteracts oxaliplatin resistance in colorectal cancers overexpressing the YB-1 protein.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Ribonucleoproteína Heterogénea-Nuclear Grupo C/genética , Proteínas Asociadas a Matriz Nuclear/genética , Factores de Transcripción de Octámeros/genética , Compuestos Organoplatinos/farmacología , Proteínas de Unión al ARN/genética , Proteína 1 de Unión a la Caja Y/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Proteínas de Unión al ADN , Resistencia a Antineoplásicos , Ribonucleoproteína Heterogénea-Nuclear Grupo C/metabolismo , Humanos , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/metabolismo , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino , Proteínas de Unión al ARN/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo
16.
Cancer Sci ; 102(7): 1410-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21466612

RESUMEN

The Y-box binding protein 1 (YB-1) is a multifunctional protein that affects transcription, splicing, and translation. Overexpression of YB-1 in breast cancers causes cisplatin resistance. The exact mechanism by which YB-1 confers cisplatin resistance is unknown. The aim of the present study was to identify, using mass spectrometry, proteins that interact with YB-1 that are important for cisplatin resistance in two breast cancer cell lines, namely MCF7 and MDA-MB-231. A tagged YB-1 construct was used to identify proteins interacting directly with YB-1 in breast cancer cells. We then focused on proteins that are potentially involved in breast cancer progression based on the ONCOMINE public microarray database. Genes encoding for these YB-1-interacting proteins were examined in the public NCBI comparative genomic hybridization database to determine whether they are localized to regions of chromosomes that are rearranged in breast cancer tissues. From these analyses, we generated a list of proteins potentially involved in cisplatin resistance. Cisplatin dose-response curves were constructed in MCF7 and MDA-MB-231 transfected with four siRNA corresponding to each of these YB-1 interactors to identify proteins significantly affecting cisplatin sensitivity upon gene silencing. Depletion of only the X-linked ribosomal protein S4 (RPS4X) resulted in consistent resistance to cisplatin in both cell lines with at least three different siRNA sequences against RPS4X. Further analyses indicated that the knock down of RPS4X decreased DNA synthesis, induced cisplatin resistance, and is equivalent to the overexpression of YB-1 in both MCF7 and MDA-MB-231 cells. These results suggest that the RPS4X/YB-1 complex is a significant potential target to counteract cisplatin resistance in breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Cisplatino/farmacología , Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Proteínas Ribosómicas/fisiología , Neoplasias de la Mama/patología , Bromodesoxiuridina/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/análisis , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Femenino , Humanos , Proteínas Nucleares/análisis , ARN Interferente Pequeño/genética , Proteínas Ribosómicas/análisis , Proteína 1 de Unión a la Caja Y
17.
FASEB J ; 24(1): 158-72, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19741171

RESUMEN

Werner syndrome (WS) is a premature aging disorder caused by mutations in a RecQ-like DNA helicase. Mice lacking the helicase domain of the WRN homologue exhibit many phenotypic features of WS, including a prooxidant status and a shorter mean life span compared to wild-type animals. Here, we show that Wrn mutant mice also develop premature liver sinusoidal endothelial defenestration along with inflammation and metabolic syndrome. Vitamin C supplementation rescued the shorter mean life span of Wrn mutant mice and reversed several age-related abnormalities in adipose tissues and liver endothelial defenestration, genomic integrity, and inflammatory status. At the molecular level, phosphorylation of age-related stress markers like Akt kinase-specific substrates and the transcription factor NF-kappaB, as well as protein kinase Cdelta and Hif-1alpha transcription factor levels, which are increased in the liver of Wrn mutants, were normalized by vitamin C. Vitamin C also increased the transcriptional regulator of lipid metabolism PPARalpha. Finally, microarray and gene set enrichment analyses on liver tissues revealed that vitamin C decreased genes normally up-regulated in human WS fibroblasts and cancers, and it increased genes involved in tissue injury response and adipocyte dedifferentiation in obese mice. Vitamin C did not have such effect on wild-type mice. These results indicate that vitamin C supplementation could be beneficial for patients with WS.


Asunto(s)
Envejecimiento/efectos de los fármacos , Ácido Ascórbico/uso terapéutico , Síndrome de Werner/tratamiento farmacológico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Secuencia de Bases , ADN Mitocondrial/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Glutatión/sangre , Glutatión/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Longevidad/efectos de los fármacos , Longevidad/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Electrónica de Rastreo , Estrés Oxidativo , PPAR alfa/genética , RecQ Helicasas/genética , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Síndrome de Werner/patología , Helicasa del Síndrome de Werner
18.
Antioxid Redox Signal ; 34(11): 856-874, 2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33202145

RESUMEN

Significance: Werner syndrome (WS) is a rare autosomal recessive malady typified by a pro-oxidant/proinflammatory status, genetic instability, and by the early onset of numerous age-associated illnesses. The protein malfunctioning in WS individuals (WRN) is a helicase/exonuclease implicated in transcription, DNA replication/repair, and telomere maintenance. Recent Advances: In the last two decades, a series of important biological systems were created to comprehend at the molecular level the effect of a defective WRN protein. Such biological tools include mouse and worm (Caenorhabditis elegans) with a mutation in the Wrn helicase ortholog as well as human WS-induced pluripotent stem cells that can ultimately be differentiated into most cell lineages. Such WS models have identified anomalies related to the hallmarks of aging. Most importantly, vitamin C counteracts these age-related cellular phenotypes in these systems. Critical Issues: Vitamin C is the only antioxidant agent capable of reversing the cellular aging-related phenotypes in those biological systems. Since vitamin C is a cofactor for many hydroxylases and mono- or dioxygenase, it adds another level of complexity in deciphering the exact molecular pathways affected by this vitamin. Moreover, it is still unclear whether a short- or long-term vitamin C supplementation in human WS patients who already display aging-related phenotypes will have a beneficial impact. Future Directions: The discovery of new molecular markers specific to the modified biological pathways in WS that can be used for novel imaging techniques or as blood markers will be necessary to assess the favorable effect of vitamin C supplementation in WS. Antioxid. Redox Signal. 34, 856-874.


Asunto(s)
Ácido Ascórbico/uso terapéutico , Helicasa del Síndrome de Werner/genética , Síndrome de Werner/dietoterapia , Animales , Ácido Ascórbico/genética , Ácido Ascórbico/metabolismo , Caenorhabditis elegans/genética , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Suplementos Dietéticos , Modelos Animales de Enfermedad , Humanos , Ratones , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Síndrome de Werner/patología
19.
BMC Genomics ; 11: 127, 2010 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-20175907

RESUMEN

BACKGROUND: Werner Syndrome (WS) is a rare disorder characterized by the premature onset of a number of age-related diseases. The gene responsible for WS encodes a DNA helicase/exonuclease protein believed to affect different aspects of transcription, replication, and/or DNA repair. In addition to genomic instability, human WS cells exhibit oxidative stress. In this report, we have examined the impact of exogenous hydrogen peroxide on the expression profile of mouse embryonic fibroblasts lacking part of the helicase domain of the WRN homologue (here referred to as Wrn Delta hel/Delta hel). RESULTS: Wrn Delta hel/Delta hel mutant mouse embryonic fibroblasts exhibit increased oxidative stress. This was reflected by increased intracellular reactive oxygen species (ROS), increased oxidative damage in genomic DNA, changes in ATP/ADP ratios, and a disruption of the inner mitochondrial transmembrane potential when compared to wild type mouse embryonic fibroblasts. Expression profile analyses of hydrogen peroxide-treated wild type cells have indicated significant decreases in the expression of genes involved in mitosis, glycolysis, fatty acid metabolism, nucleic acid metabolism, and cell cycle control, as well as protein modification and stability. Such decreases in these biological processes were not observed in hydrogen peroxide-treated Wrn Delta hel/Delta hel cells. Importantly, untreated Wrn Delta hel/Delta hel cells already exhibited down regulation of several biological processes decreased in wild type cells that had been treated with hydrogen peroxide. CONCLUSION: Expression profiling of Wrn Delta hel/Delta hel mutant cells revealed a very different response to exogenous addition of hydrogen peroxide in culture compared to wild type cells. This is due in part to the fact that Wrn Delta hel/Delta hel mutant cells already exhibited a modest chronic intracellular oxidative stress.


Asunto(s)
Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo , RecQ Helicasas/genética , Animales , Biología Computacional , Daño del ADN , Embrión de Mamíferos , Fibroblastos/efectos de los fármacos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Especies Reactivas de Oxígeno/análisis , Eliminación de Secuencia , Síndrome de Werner/genética , Helicasa del Síndrome de Werner
20.
DNA Repair (Amst) ; 8(6): 704-19, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19272840

RESUMEN

Maintenance of the mitochondrial genome (mtDNA) is essential for proper cellular function. The accumulation of damage and mutations in the mtDNA leads to diseases, cancer, and aging. Mammalian mitochondria have proficient base excision repair, but the existence of other DNA repair pathways is still unclear. Deficiencies in DNA mismatch repair (MMR), which corrects base mismatches and small loops, are associated with DNA microsatellite instability, accumulation of mutations, and cancer. MMR proteins have been identified in yeast and coral mitochondria; however, MMR proteins and function have not yet been detected in human mitochondria. Here we show that human mitochondria have a robust mismatch-repair activity, which is distinct from nuclear MMR. Key nuclear MMR factors were not detected in mitochondria, and similar mismatch-binding activity was observed in mitochondrial extracts from cells lacking MSH2, suggesting distinctive pathways for nuclear and mitochondrial MMR. We identified the repair factor YB-1 as a key candidate for a mitochondrial mismatch-binding protein. This protein localizes to mitochondria in human cells, and contributes significantly to the mismatch-binding and mismatch-repair activity detected in HeLa mitochondrial extracts, which are significantly decreased when the intracellular levels of YB-1 are diminished. Moreover, YB-1 depletion in cells increases mitochondrial DNA mutagenesis. Our results show that human mitochondria contain a functional MMR repair pathway in which YB-1 participates, likely in the mismatch-binding and recognition steps.


Asunto(s)
Reparación de la Incompatibilidad de ADN , ADN Mitocondrial/genética , Proteínas de Unión al ADN/metabolismo , Mitocondrias/metabolismo , Proteínas Nucleares/metabolismo , Núcleo Celular/metabolismo , Resistencia al Cloranfenicol , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Células HeLa , Humanos , Proteínas Nucleares/genética , Consumo de Oxígeno , Fracciones Subcelulares , Proteína 1 de Unión a la Caja Y
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA