Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Epidemiol ; 37(10): 1087-1105, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36245062

RESUMEN

The Food Chain Plus (FoCus) cohort was launched in 2011 for population-based research related to metabolic inflammation. To characterize this novel pathology in a comprehensive manner, data collection included multiple omics layers such as phenomics, microbiomics, metabolomics, genomics, and metagenomics as well as nutrition profiling, taste perception phenotyping and social network analysis. The cohort was set-up to represent a Northern German population of the Kiel region. Two-step recruitment included the randomised enrolment of participants via residents' registration offices and via the Obesity Outpatient Centre of the University Medical Center Schleswig-Holstein (UKSH). Hence, both a population- and metabolic inflammation- based cohort was created. In total, 1795 individuals were analysed at baseline. Baseline data collection took place between 2011 and 2014, including 63% females and 37% males with an age range of 18-83 years. The median age of all participants was 52.0 years [IQR: 42.5; 63.0 years] and the median baseline BMI in the study population was 27.7 kg/m2 [IQR: 23.7; 35.9 kg/m2]. In the baseline cohort, 14.1% of participants had type 2 diabetes mellitus, which was more prevalent in the subjects of the metabolic inflammation group (MIG; 31.8%). Follow-up for the assessment of disease progression, as well as the onset of new diseases with changes in subject's phenotype, diet or lifestyle factors is planned every 5 years. The first follow-up period was finished in 2020 and included 820 subjects.


Asunto(s)
Diabetes Mellitus Tipo 2 , Femenino , Humanos , Masculino , Estudios de Cohortes , Diabetes Mellitus Tipo 2/epidemiología , Cadena Alimentaria , Inflamación , Obesidad/epidemiología , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años
2.
PLoS Genet ; 13(4): e1006717, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28394895

RESUMEN

Environmental conditions experienced during animal development are thought to have sustained impact on maturation and adult lifespan. Here we show that in the model organism C. elegans developmental rate and adult lifespan depend on larval population density, and that this effect is mediated by excreted small molecules. By using the time point of first egg laying as a marker for full maturity, we found that wildtype hermaphrodites raised under high density conditions developed significantly faster than animals raised in isolation. Population density-dependent acceleration of development (Pdda) was dramatically enhanced in fatty acid ß-oxidation mutants that are defective in the biosynthesis of ascarosides, small-molecule signals that induce developmental diapause. In contrast, Pdda is abolished by synthetic ascarosides and steroidal ligands of the nuclear hormone receptor DAF-12. We show that neither ascarosides nor any known steroid hormones are required for Pdda and that another chemical signal mediates this phenotype, in part via the nuclear hormone receptor NHR-8. Our results demonstrate that C. elegans development is regulated by a push-pull mechanism, based on two antagonistic chemical signals: chemosensation of ascarosides slows down development, whereas population-density dependent accumulation of a different chemical signal accelerates development. We further show that the effects of high larval population density persist through adulthood, as C. elegans larvae raised at high densities exhibit significantly reduced adult lifespan and respond differently to exogenous chemical signals compared to larvae raised at low densities, independent of density during adulthood. Our results demonstrate how inter-organismal signaling during development regulates reproductive maturation and longevity.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Longevidad/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/biosíntesis , Ácidos Grasos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Organismos Hermafroditas/genética , Organismos Hermafroditas/crecimiento & desarrollo , Larva/genética , Larva/crecimiento & desarrollo , Neuropéptidos/metabolismo , Densidad de Población , Receptores Citoplasmáticos y Nucleares/biosíntesis , Transducción de Señal
3.
Hum Mol Genet ; 25(13): 2881-2891, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27149984

RESUMEN

Coenzyme Q10 (CoQ10) is a lipophilic redox molecule that is present in membranes of almost all cells in human tissues. CoQ10 is, amongst other functions, essential for the respiratory transport chain and is a modulator of inflammatory processes and gene expression. Rare monogenetic CoQ10 deficiencies show noticeable symptoms in tissues (e.g. kidney) and cell types (e.g. neurons) with a high energy demand. To identify common genetic variants influencing serum CoQ10 levels, we performed a fixed effects meta-analysis in two independent cross-sectional Northern German cohorts comprising 1300 individuals in total. We identified two genome-wide significant susceptibility loci. The best associated single nucleotide polymorphism (SNP) was rs9952641 (P value = 1.31 × 10 -8, ß = 0.063, CI0.95 [0.041, 0.085]) within the COLEC12 gene on chromosome 18. The SNP rs933585 within the NRXN-1 gene on chromosome 2 also showed genome wide significance (P value = 3.64 × 10 -8, ß = -0.034, CI0.95 [-0.046, -0.022]). Both genes have been previously linked to neuronal diseases like Alzheimer's disease, autism and schizophrenia. Among our 'top-10' associated variants, four additional loci with known neuronal connections showed suggestive associations with CoQ10 levels. In summary, this study demonstrates that serum CoQ10 levels are associated with common genetic loci that are linked to neuronal diseases.


Asunto(s)
Degeneración Nerviosa/genética , Ubiquinona/análogos & derivados , Adulto , Anciano , Ataxia/genética , Ataxia/metabolismo , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular Neuronal/genética , Colectinas/genética , Estudios Transversales , Femenino , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Debilidad Muscular/genética , Debilidad Muscular/metabolismo , Degeneración Nerviosa/etiología , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa , Neuronas , Polimorfismo de Nucleótido Simple/genética , Receptores Depuradores/genética , Ubiquinona/sangre , Ubiquinona/deficiencia , Ubiquinona/genética , Ubiquinona/metabolismo
4.
Eur J Immunol ; 45(9): 2602-14, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26140335

RESUMEN

Natural killer (NK) cells are a subset of cytotoxic lymphocytes that recognize and kill tumor- and virus-infected cells without prior stimulation. Killing of target cells is a multistep process including adhesion to target cells, formation of an immunological synapse, and polarization and release of cytolytic granules. The role of distinct potassium channels in this orchestrated process is still poorly understood. The current study reveals that in addition to the voltage-gated KV 1.3 and the calcium-activated KCa 3.1 channels, human NK cells also express the two-pore domain K2 P channel TASK2 (TWIK-related acid-sensitive potassium channel). Expression of Task2 varies among NK-cell subsets and depends on their differentiation and activation state. Despite its different expression in TASK2(high) CD56(bright) CD16(-) and TASK2(low) CD56(dim) CD16(+) NK cells, TASK2 is involved in cytokine-induced proliferation and cytolytic function of both subsets. TASK2 is crucial for leukocyte functional antigen (LFA-1) mediated adhesion of both resting and cytokine-activated NK cells to target cells, an early step in killing of target cells. With regard to the following mechanism, TASK2 plays a role in release of cytotoxic granules by resting, but not IL-15-induced NK cells. Taken together, our data exhibit two-pore potassium channels as important players in NK-cell activation and effector function.


Asunto(s)
Citotoxicidad Inmunológica , Sinapsis Inmunológicas/metabolismo , Células Asesinas Naturales/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Canales de Potasio de Dominio Poro en Tándem/inmunología , Antígeno CD56/genética , Antígeno CD56/inmunología , Adhesión Celular/efectos de los fármacos , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Expresión Génica , Células HEK293 , Humanos , Interleucina-15/farmacología , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Cultivo Primario de Células , Receptores de IgG/genética , Receptores de IgG/inmunología , Transducción de Señal , Análisis de la Célula Individual
5.
Toxicol Appl Pharmacol ; 305: 22-39, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27260674

RESUMEN

Mitragyna speciosa Korth is known for its euphoric properties and is frequently used for recreational purposes. Several poisoning and fatal cases involving mitragynine have been reported but the underlying causes remain unclear. Human ether-a-go-go-related gene (hERG) encodes the cardiac IKr current which is a determinant of the duration of ventricular action potentials and QT interval. On the other hand, IK1, a Kir current mediated by Kir2.1 channel and IKACh, a receptor-activated Kir current mediated by GIRK channel are also known to be important in maintaining the cardiac function. This study investigated the effects of mitragynine on the current, mRNA and protein expression of hERG channel in hERG-transfected HEK293 cells and Xenopus oocytes. The effects on Kir2.1 and GIRK channels currents were also determined in the oocytes. The hERG tail currents following depolarization pulses were inhibited by mitragynine with an IC50 value of 1.62µM and 1.15µM in the transfected cell line and Xenopus oocytes, respectively. The S6 point mutations of Y652A and F656A attenuated the inhibitor effects of mitragynine, indicating that mitragynine interacts with these high affinity drug-binding sites in the hERG channel pore cavity which was consistent with the molecular docking simulation. Interestingly, mitragynine does not affect the hERG expression at the transcriptional level but inhibits the protein expression. Mitragynine is also found to inhibit IKACh current with an IC50 value of 3.32µM but has no significant effects on IK1. Blocking of both hERG and GIRK channels may cause additive cardiotoxicity risks.


Asunto(s)
Canal de Potasio ERG1/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Alcaloides de Triptamina Secologanina/farmacología , Animales , Alcaloides Diterpénicos , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Células HEK293 , Corazón/fisiología , Humanos , Potenciales de la Membrana/efectos de los fármacos , Oocitos/metabolismo , ARN Mensajero/metabolismo , Xenopus
6.
J Clin Biochem Nutr ; 58(3): 240-5, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27257350

RESUMEN

Coenzyme Q10 (CoQ10) is synthesized in almost all human tissues and presumably involved in age-related alterations and diseases. Here, we examined the impact of aging and sex on the serum CoQ10 status in 860 European adults ranging in age from 18 to 82 years. We identified an inverse U-shaped relationship between CoQ10 concentration and age. Women showed lower cholesterol-adjusted CoQ10 levels than men, irrespective of age. As observed in both sexes, the decrease in CoQ10 concentration in older subjects was accompanied by a shift in the redox status in favour of the oxidized form. A strong positive correlation was found for total CoQ10 and cholesterol concentrations (Spearman's, p≤1E-74). We found strong negative correlations between total (Spearman's, p≤1E-07) and between cholesterol-adjusted CoQ10 concentration (Spearman's, p≤1E-14) and the proportion of the oxidized form of CoQ10. These correlations were not dependent on age and sex and were attenuated by supplementation with 150 mg/day reduced CoQ10 for 14 days. Overall, our results are useful to define risk groups with critical CoQ10 status in humans. In particular, older subjects were characterized by impaired CoQ10 status due to their lowered serum CoQ10 concentration and concomitant decrease of CoQ10 redox capacity.

7.
Pediatr Res ; 78(4): 365-70, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26107394

RESUMEN

BACKGROUND: Coenzyme Q10 (CoQ10) is synthesized in most human tissues, with high concentration in the skeletal muscle. CoQ10 functions in the mitochondrial respiratory chain and serves as a potent liphophilic antioxidant in membranes. CoQ10 deficiency impairs mitochondrial ATP synthesis and increases oxidative stress. It has been suggested that plasma CoQ10 status is not a robust proxy for the diagnosis of CoQ10 deficiency. METHODS: We determined the concentration and redox-status of CoQ10 in plasma and muscle tissue from 140 healthy children (0.8-15.3 y) by high-performance liquid chromatography (HPLC) with electrochemical detection. RESULTS: There was no correlation between CoQ10 concentration or redox status between plasma and muscle tissue. Lipid-related CoQ10 plasma concentrations showed a negative correlation with age (Spearman's, P ≤ 0.02), but there was no significant age-related correlation for muscle concentration. In muscle tissue, we found a distinct shift in the redox status in favor of the oxidized proportion with increasing age (Spearman's, P ≤ 0.00001). Reference values for muscle CoQ10 concentration (40.5 ± 12.2 pmol/mg wet tissue) and CoQ10 redox status (46.8 ± 6.8% oxidized within total) were established for healthy children. CONCLUSION: The age-related redox shift in muscle tissue suggests changes in antioxidative defense during childhood. The reference values established here provide a necessary prerequisite for diagnosing early CoQ10 deficiency.


Asunto(s)
Músculos Abdominales/enzimología , Desarrollo del Adolescente , Envejecimiento/metabolismo , Desarrollo Infantil , Ubiquinona/análogos & derivados , Músculos Abdominales/crecimiento & desarrollo , Adolescente , Factores de Edad , Envejecimiento/sangre , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Técnicas Electroquímicas , Femenino , Voluntarios Sanos , Humanos , Lactante , Masculino , Oxidación-Reducción , Valores de Referencia , Ubiquinona/análisis , Ubiquinona/sangre , Regulación hacia Arriba
8.
J Clin Biochem Nutr ; 57(3): 171-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26566301

RESUMEN

Coenzyme Q (CoQ) is necessary for mitochondrial energy production and modulates the expression of genes that are important for inflammatory processes, growth and detoxification reactions. A cellular surveillance-activated detoxification and defenses (cSADDs) pathway has been recently identified in C. elegans. The down-regulation of the components of the cSADDs pathway initiates an aversion behavior of the nematode. Here we hypothesized that CoQ regulates genes of the cSADDs pathway. To verify this we generated CoQ-deficient worms ("CoQ-free") and performed whole-genome expression profiling. We found about 30% (120 genes) of the cSADDs pathway genes were differentially regulated under CoQ-deficient condition. Remarkably, 83% of these genes were down-regulated. The majority of the CoQ-sensitive cSADDs pathway genes encode for proteins involved in larval development (enrichment score (ES) = 38.0, p = 5.0E(-37)), aminoacyl-tRNA biosynthesis, proteasome function (ES 8.2, p = 5.9E(-31)) and mitochondria function (ES 3.4, p = 1.7E(-5)). 67% (80 genes) of these genes are categorized as lethal. Thus it is shown for the first time that CoQ regulates a substantial number of essential genes that function in the evolutionary conserved cellular surveillance-activated detoxification and defenses pathway in C. elegans.

9.
J Cell Sci ; 125(Pt 10): 2486-99, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22357950

RESUMEN

Serotonin receptors 5-HT(1A) and 5-HT(7) are highly coexpressed in brain regions implicated in depression. However, their functional interaction has not been established. In the present study we show that 5-HT(1A) and 5-HT(7) receptors form heterodimers both in vitro and in vivo. Foerster resonance energy transfer-based assays revealed that, in addition to heterodimers, homodimers composed either of 5-HT(1A) or 5-HT(7) receptors together with monomers coexist in cells. The highest affinity for complex formation was obtained for the 5-HT(7)-5-HT(7) homodimers, followed by the 5-HT(7)-5-HT(1A) heterodimers and 5-HT(1A)-5-HT(1A) homodimers. Functionally, heterodimerization decreases 5-HT(1A)-receptor-mediated activation of G(i) protein without affecting 5-HT(7)-receptor-mediated signalling. Moreover, heterodimerization markedly decreases the ability of the 5-HT(1A) receptor to activate G-protein-gated inwardly rectifying potassium channels in a heterologous system. The inhibitory effect on such channels was also preserved in hippocampal neurons, demonstrating a physiological relevance of heteromerization in vivo. In addition, heterodimerization is crucially involved in initiation of the serotonin-mediated 5-HT(1A) receptor internalization and also enhances the ability of the 5-HT(1A) receptor to activate the mitogen-activated protein kinases. Finally, we found that production of 5-HT(7) receptors in the hippocampus continuously decreases during postnatal development, indicating that the relative concentration of 5-HT(1A)-5-HT(7) heterodimers and, consequently, their functional importance undergoes pronounced developmental changes.


Asunto(s)
Receptor de Serotonina 5-HT1A/metabolismo , Receptores de Serotonina/metabolismo , Transducción de Señal , Animales , Encéfalo/metabolismo , Línea Celular Tumoral , Dimerización , Ratones , Neuronas/metabolismo , Unión Proteica , Transporte de Proteínas , Receptor de Serotonina 5-HT1A/química , Receptor de Serotonina 5-HT1A/genética , Receptores de Serotonina/química , Receptores de Serotonina/genética
10.
Biochem Biophys Res Commun ; 452(4): 920-7, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25234594

RESUMEN

Coenzyme Q (CoQ, ubiquinone) is an essential component of the respiratory chain, a cofactor of pyrimidine biosynthesis and acts as an antioxidant in extra mitochondrial membranes. More recently CoQ has been identified as a modulator of apoptosis, inflammation and gene expression. CoQ deficient Caenorhabditis elegans clk-1 mutants show several phenotypes including a delayed postembryonic growth. Using wild type and two clk-1 mutants, here we established an experimental set-up to study the consequences of endogenous CoQ deficiency or exogenous CoQ supply on gene expression and growth. We found that a deficiency of endogenous CoQ synthesis down-regulates a cluster of genes that are important for growth (i.e., RNA polymerase II, eukaryotic initiation factor) and up-regulates oxidation reactions (i.e., cytochrome P450, superoxide dismutase) and protein interactions (i.e., F-Box proteins). Exogenous CoQ supply partially restores the expression of these genes as well as the growth retardation of CoQ deficient clk-1 mutants. On the other hand exogenous CoQ supply does not alter the expression of a further sub-set of genes. These genes are involved in metabolism (i.e., succinate dehydrogenase complex), cell signalling or synthesis of lectins. Thus, our work provides a comprehensive overview of genes which can be modulated in their expression by endogenous or exogenous CoQ. As growth retardation in CoQ deficiency is linked to the gene expression profile we suggest that CoQ promotes growth via gene expression.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Ubiquinona/análogos & derivados , Animales , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Mutación , Ubiquinona/metabolismo , Ubiquinona/farmacología
11.
Cytokine ; 68(1): 65-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24721042

RESUMEN

lincRNAs recently have been discovered as evolutionary conserved transcripts of non-coding DNA sequences and have been implicated in the regulation of cellular differentiation. In humans, molecular studies have suggested a functional role for lincRNAs in cancer development. The aim of the present study was to examine whether these novel molecules are specifically regulated by different cytokines in cells of the innate immune system in humans in vivo and whether lincRNAs thereby might be involved in the pathophysiology of rheumatoid arthritis (RA). Therefore, CD14(+) monocytes were isolated from RA patients before and after anti-IL-6R (tocilizumab) or anti-TNF-α (adalimumab) therapy and lincRNA transcription was analysed by a microarray based experiment. As expected, we found lincRNAs to be present in CD14(+) monocytes of RA patients. However, of the total number of 7.419 lincRNAs examined in this study only a very small number was significantly regulated by either IL-6 or TNF-α (85 lincRNAs, corresponding to 1.1%). The numbers of lincRNAs regulated was higher due to TNF-α compared to IL-6. Interestingly, none of the identified lincRNAs was influenced by both, IL-6 and TNF-α, suggesting the regulation of lincRNA transcription to be highly specific for distinct cytokines. Taken together, our results suggest (1) that lincRNAs are novel intracellular molecular effectors of specific cytokines in cells of the innate immune system in humans in vivo and (2) that lincRNAs might be involved in the molecular pathophysiology of RA.


Asunto(s)
Artritis Reumatoide/genética , Regulación de la Expresión Génica , Inmunidad Innata , Interleucina-6/fisiología , ARN Largo no Codificante/genética , ARN Mensajero/genética , Factor de Necrosis Tumoral alfa/fisiología , Artritis Reumatoide/inmunología , Humanos , Persona de Mediana Edad
12.
J Exp Biol ; 217(Pt 14): 2480-8, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24803455

RESUMEN

Locomotion is crucial for the survival of living organisms, as it allows foraging, flight and mating behaviour. In response to environmental cues, many organisms switch between alternative forms of locomotion, referred to as gaits. The nematode Caenorhabditis elegans exhibits two gaits: swimming in liquids and crawling on dense gels. The kinematics and patterns of muscle activity differ between the two gaits, with swimming being less efficient than crawling. We found that C. elegans when grown on dietary restriction (DR) plates and then tested immediately for swimming activity exhibit an accelerated frequency of body-bending swimming compared with ad libitum-fed worms, resulting in an increased swimming speed. This response is independent of the presence or absence of food bacteria in the assay liquid. In contrast, the crawling speed of DR worms on assay agar plates is decreased and influenced by food availability. Because DR also attenuates the disturbed swimming activity of worms that are deficient in the presynaptic dopamine transporter DAT-1, our data link DR-induced alterations of the swimming gait to synaptic processes. This strongly suggests a biochemical rather than a biomechanical response to DR provoked by changes in the worm's body structure. We conclude that the increase in locomotor activity in response to DR is specific to the swimming gait and might represent a survival strategy, allowing food-deprived nematodes to exit unfavourable environments.


Asunto(s)
Adaptación Fisiológica , Fenómenos Fisiológicos Nutricionales de los Animales , Conducta Animal/fisiología , Caenorhabditis elegans/fisiología , Marcha/fisiología , Locomoción/fisiología , Natación/fisiología , Animales , Agua
13.
Physiol Genomics ; 45(13): 539-51, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23715261

RESUMEN

The metabolic and health-promoting effects of dietary restriction (DR) have been extensively studied in several species. The response to DR with respect to sex is essentially unknown. To address this question, we used the model organism Caenorhabditis elegans to analyze body composition and gene expression in males and hermaphrodites in response to DR. Unexpectedly, DR increased the fat-to-fat-free mass ratio and enlarged lipid droplets in both sexes to a similar extent. These effects were linked to a downregulation of the lipase-like 5 (lipl-5) gene in both sexes at two developmental stages. By contrast, the reductions in body size, protein content, and total RNA content in response to DR were more pronounced in hermaphrodites than in males. Functional enrichment analysis of gene expression data showed a DR-induced downregulation of several embryogenesis-associated genes concomitant with an ongoing expression of sperm-associated genes in hermaphrodites. In conclusion, DR increases fat stores in both sexes of C. elegans in the form of large and possibly lipolysis-resistant lipid droplets and markedly alters the reproductive program in hermaphrodites but not in males.


Asunto(s)
Adiposidad/genética , Composición Corporal/genética , Caenorhabditis elegans/genética , Restricción Calórica , Caracteres Sexuales , Transcriptoma , Animales , Tamaño Corporal/genética , Peso Corporal/genética , Caenorhabditis elegans/anatomía & histología , Análisis por Conglomerados , Colágeno/genética , Colágeno/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Glucosa/metabolismo , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lipasa/genética , Lipasa/metabolismo , Metabolismo de los Lípidos/genética , Masculino , Anotación de Secuencia Molecular , ARN/metabolismo , Saposinas/genética , Saposinas/metabolismo , Espermatozoides/metabolismo , Factores de Tiempo , Trehalosa/metabolismo , Regulación hacia Arriba/genética
14.
Anal Biochem ; 437(1): 88-94, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23467099

RESUMEN

Swine tissues were used as surrogates for human tissues with coenzyme Q10 (CoQ10) as the primary endogenous quinoid to establish a reliable method for the analysis of total CoQ10 concentration and redox status using the reduced and oxidized forms of CoQ9 as internal standards. Specimens of frozen swine tissues were disrupted by bead milling using 2-propanol as the homogenization medium supplemented with the internal standards. After hexane extraction, CoQ10 was analyzed via high-performance liquid chromatography with electrochemical detection. The method is linear (12-60 mg fresh muscle tissue/sample), sensitive (~200 pmol CoQ10/sample), and reproducible (coefficients of variation of 6.0 and 3.2% for total CoQ10 and 2.4 and 3.2% for the redox status of within-day and day-to-day precision, respectively), with analytic recoveries for ubiquinone-10, ubihydroquinone-10, and total Q10 of 91, 104, and 94%, respectively. The concentration and redox status were stable for at least 3 months at -84°C. The total CoQ10 concentrations (pmol/mg fresh tissue) in swine tissues were as follows: lung (17.4±1.42), skeletal muscle (26.7±2.57), brain (40.7±4.02), liver (62.1±31.0), kidney (111.7±37.08), and heart muscle (149.1±36.78). Significant tissue-specific variations were also found for the redox status (% oxidation of total): swine liver (~28), lung (~36), kidney (~37), heart muscle (~57), skeletal muscle (~61), and brain (~67).


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Electroquímica/métodos , Porcinos , Ubiquinona/análogos & derivados , Animales , Electroquímica/normas , Humanos , Oxidación-Reducción , Estándares de Referencia , Ubiquinona/aislamiento & purificación , Ubiquinona/metabolismo
15.
Biochim Biophys Acta ; 1808(8): 2036-44, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21575593

RESUMEN

A variety of ion channels are supposed to orchestrate the homoeostatic volume regulation in T lymphocytes. However, the relative contribution of different potassium channels to the osmotic volume regulation and in particular to the regulatory volume decrease (RVD) in T cells is far from clear. This study explores a putative role of the newly identified K(2P) channels (TASK1, TASK2, TASK3 and TRESK) along with the voltage-gated potassium channel K(V)1.3 and the calcium-activated potassium channel K(Ca)3.1 in the RVD of murine T lymphocytes, using genetic and pharmacological approaches. K(2P) channel knockouts exerted profound effects on the osmotic properties of murine T lymphocytes, as revealed by reduced water and RVD-related solute permeabilities. Moreover, both genetic and pharmacological data proved a key role of K(V)1.3 and TASK2 channels in the RVD of murine T cells exposed to hypotonic saline. Our experiments demonstrate a leading role of potassium channels in the osmoregulation of T lymphocytes under different conditions. In summary, the present study sheds new light on the complex and partially redundant network of potassium channels involved in the basic physiological process of the cellular volume homeostasis and extends the repertoire of potassium channels by the family of K(2P) channels.


Asunto(s)
Tamaño de la Célula , Canal de Potasio Kv1.3/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Linfocitos T/metabolismo , Equilibrio Hidroelectrolítico , Animales , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Soluciones Hipotónicas , Canal de Potasio Kv1.3/antagonistas & inhibidores , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio de Dominio Poro en Tándem/genética , Solución Salina Hipertónica , Linfocitos T/efectos de los fármacos , Factores de Tiempo , Equilibrio Hidroelectrolítico/efectos de los fármacos
16.
IUBMB Life ; 64(7): 644-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22639405

RESUMEN

Epidemiological data from human populations and few reports in rodents suggested that the paternal diet affects offspring adiposity and its related diseases. We tested whether this nongenetic and intergenerational inheritance depends on paternal treatment dose. Using the model organism Caenorhabditis elegans, males undergoing several dietary restriction regimes were crossed with ad libitum fed females. We found an inverted U-shaped relationship between the extent of paternal dietary restriction and the level of fat content of progeny. The relationship was evident in both sexes. Body proportions were not affected in offspring. Overall, our findings extent the concept of developmental and adaptive plasticity to include the extent of paternal food consumption in the origin of phenotypic alterations.


Asunto(s)
Adiposidad/fisiología , Alimentación Animal , Animales , Peso Corporal , Caenorhabditis elegans , Epigénesis Genética , Femenino , Privación de Alimentos , Masculino , Exposición Paterna , Fenotipo , Factores de Tiempo
17.
Mutat Res ; 733(1-2): 61-8, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21964355

RESUMEN

Coenzyme Q(10) (CoQ(10)) is an essential component for electron transport in the mitochondrial respiratory chain and serves as cofactor in several biological processes. The reduced form of CoQ(10) (ubiquinol, Q(10)H(2)) is an effective antioxidant in biological membranes. During the last years, particular interest has been grown on molecular effects of CoQ(10) supplementation on mechanisms related to DNA damage prevention. This review describes recent advances in our understanding about the impact of CoQ(10) on genomic stability in cells, animals and humans. With regard to several in vitro and in vivo studies, CoQ(10) provides protective effects on several markers of oxidative DNA damage and genomic stability. In comparison to the number of studies reporting preventive effects of CoQ(10) on oxidative stress biomarkers, CoQ(10) intervention studies in humans with a direct focus on markers of DNA damage are limited. Thus, more well-designed studies in healthy and disease populations with long-term follow up results are needed to substantiate the reported beneficial effects of CoQ(10) on prevention of DNA damage.


Asunto(s)
Daño del ADN , Ubiquinona/análogos & derivados , Animales , Células Cultivadas , Suplementos Dietéticos , Inestabilidad Genómica , Humanos , Peroxidación de Lípido , Micronutrientes/genética , Estrés Oxidativo , Ubiquinona/metabolismo , Ubiquinona/fisiología
18.
Cell Res ; 32(1): 72-88, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34702947

RESUMEN

It remains largely unclear how thymocytes translate relative differences in T cell receptor (TCR) signal strength into distinct developmental programs that drive the cell fate decisions towards conventional (Tconv) or regulatory T cells (Treg). Following TCR activation, intracellular calcium (Ca2+) is the most important second messenger, for which the potassium channel K2P18.1 is a relevant regulator. Here, we identify K2P18.1 as a central translator of the TCR signal into the thymus-derived Treg (tTreg) selection process. TCR signal was coupled to NF-κB-mediated K2P18.1 upregulation in tTreg progenitors. K2P18.1 provided the driving force for sustained Ca2+ influx that facilitated NF-κB- and NFAT-dependent expression of FoxP3, the master transcription factor for Treg development and function. Loss of K2P18.1 ion-current function induced a mild lymphoproliferative phenotype in mice, with reduced Treg numbers that led to aggravated experimental autoimmune encephalomyelitis, while a gain-of-function mutation in K2P18.1 resulted in increased Treg numbers in mice. Our findings in human thymus, recent thymic emigrants and multiple sclerosis patients with a dominant-negative missense K2P18.1 variant that is associated with poor clinical outcomes indicate that K2P18.1 also plays a role in human Treg development. Pharmacological modulation of K2P18.1 specifically modulated Treg numbers in vitro and in vivo. Finally, we identified nitroxoline as a K2P18.1 activator that led to rapid and reversible Treg increase in patients with urinary tract infections. Conclusively, our findings reveal how K2P18.1 translates TCR signals into thymic T cell fate decisions and Treg development, and provide a basis for the therapeutic utilization of Treg in several human disorders.


Asunto(s)
Canales de Potasio , Receptores de Antígenos de Linfocitos T , Linfocitos T Reguladores , Animales , Diferenciación Celular , Factores de Transcripción Forkhead , Humanos , Ratones , FN-kappa B , Timocitos , Timo
19.
J Lipid Res ; 52(6): 1281-1293, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21421847

RESUMEN

The proportions of body fat and fat-free mass are determining factors of adiposity-associated diseases. Work in Caenorhabditis elegans has revealed evolutionarily conserved pathways of fat metabolism. Nevertheless, analysis of body composition and fat distribution in the nematodes has only been partially unraveled because of methodological difficulties. We characterized metabolic C. elegans mutants by using novel and feasible BODIPY 493/503-based fat staining and flow cytometry approaches. Fixative as well as vital BODIPY staining procedures visualize major fat stores, preserve native lipid droplet morphology, and allow quantification of fat content per body volume of individual worms. Colocalization studies using coherent anti-Stokes Raman scattering microscopy, Raman microspectroscopy, and imaging of lysosome-related organelles as well as biochemical measurement confirm our approaches. We found that the fat-to-volume ratio of dietary restriction, TGF-ß, and germline mutants are specific for each strain. In contrast, the proportion of fat-free mass is constant between the mutants, although their volumes differ by a factor of 3. Our approaches enable sensitive, accurate, and high-throughput assessment of adiposity in large C. elegans populations at a single-worm level.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad , Caenorhabditis elegans/metabolismo , Ensayos Analíticos de Alto Rendimiento , Obesidad/metabolismo , Coloración y Etiquetado/métodos , Tejido Adiposo/química , Animales , Compuestos Azo/análisis , Compuestos de Boro/análisis , Caenorhabditis elegans/genética , Modelos Animales de Enfermedad , Fijadores/análisis , Fijadores/metabolismo , Citometría de Flujo , Fluorescencia , Mutación de Línea Germinal , Metabolismo de los Lípidos , Microscopía , Especificidad de la Especie , Espectrometría Raman , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/biosíntesis
20.
J Cell Mol Med ; 15(4): 909-27, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20345851

RESUMEN

Despite intensive efforts on annotation of eukaryotic transcriptoms, little is known about the regulation of low-abundance transcripts. To address this question, we analysed the regulation of novel low-abundance transcript variants of human acyl-CoA binding protein (ACBP), an important multifunctional housekeeping protein, which we have identified by screening of human expressed sequence tags in combination with ab initio gene prediction. By using RT-, real-time RT- and rapid amplification of cDNA ends-PCR in five human tissues, we find these transcripts, which are generated by a consequent use of alternative promoters and alternate first or first two exons, to be authentic ones. They show a tissue-specific distribution and intrinsic responsiveness to glucose and insulin. Promoter analyses of the corresponding transcripts revealed a differential regulation mediated by sterol regulatory element-binding protein-2, hepatocyte nuclear factor-4α and nuclear factor κB (NF-κB), central transcription factors of fat and glucose metabolism and inflammation. Subcellular localization studies of deduced isoforms in liver HepG2 cells showed that they are distributed in different compartments. By demonstrating that ACBP is a target of NF-κB, our findings link fatty acid metabolism with inflammation. Furthermore, our findings show that low-abundance transcripts are regulated in a similar mode than their high-abundance counterparts.


Asunto(s)
Empalme Alternativo/genética , Inhibidor de la Unión a Diazepam/metabolismo , Empalme Alternativo/efectos de los fármacos , Secuencia de Aminoácidos , Bacterias/metabolismo , Secuencia de Bases , Citocinas/farmacología , ADN Complementario/genética , Inhibidor de la Unión a Diazepam/química , Etiquetas de Secuencia Expresada , Perfilación de la Expresión Génica , Sitios Genéticos , Glucosa/farmacología , Factor Nuclear 4 del Hepatocito , Humanos , Insulina/farmacología , Datos de Secuencia Molecular , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Análisis de Secuencia de ADN , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Sitio de Iniciación de la Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA