Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Nutrients ; 13(11)2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34836252

RESUMEN

Postprandial lipemia (PPL) is an important risk factor for cardiovascular disease. Inter-individual variation in the dietary response to a meal is known to be influenced by genetic factors, yet genes that dictate variation in postprandial lipids are not completely characterized. Genetic studies of the plasma lipidome can help to better understand postprandial metabolism by isolating lipid molecular species which are more closely related to the genome. We measured the plasma lipidome at fasting and 6 h after a standardized high-fat meal in 668 participants from the Genetics of Lipid-Lowering Drugs and Diet Network study (GOLDN) using ultra-performance liquid chromatography coupled to (quadrupole) time-of-flight mass spectrometry. A total of 413 unique lipids were identified. Heritable and responsive lipid species were examined for association with single-nucleotide polymorphisms (SNPs) genotyped on the Affymetrix 6.0 array. The most statistically significant SNP findings were replicated in the Amish Heredity and Phenotype Intervention (HAPI) Heart Study. We further followed up findings from GOLDN with a regional analysis of cytosine-phosphate-guanine (CpGs) sites measured on the Illumina HumanMethylation450 array. A total of 132 lipids were both responsive to the meal challenge and heritable in the GOLDN study. After correction for multiple testing of 132 lipids (α = 5 × 10-8/132 = 4 × 10-10), no SNP was statistically significantly associated with any lipid response. Four SNPs in the region of a known lipid locus (fatty acid desaturase 1 and 2/FADS1 and FADS2) on chromosome 11 had p < 8.0 × 10-7 for arachidonic acid FA(20:4). Those SNPs replicated in HAPI Heart with p < 3.3 × 10-3. CpGs around the FADS1/2 region were associated with arachidonic acid and the relationship of one SNP was partially mediated by a CpG (p = 0.005). Both SNPs and CpGs from the fatty acid desaturase region on chromosome 11 contribute jointly and independently to the diet response to a high-fat meal.


Asunto(s)
Genómica , Hipolipemiantes/farmacología , Lipidómica , Periodo Posprandial/efectos de los fármacos , Periodo Posprandial/genética , Adulto , Anciano , delta-5 Desaturasa de Ácido Graso/genética , Ácido Graso Desaturasas/genética , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Lípidos , Masculino , Comidas , Persona de Mediana Edad , Fenotipo , Plasma , Polimorfismo de Nucleótido Simple
2.
JCI Insight ; 6(19)2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34622807

RESUMEN

The signaling mechanisms by which dietary fat and cholesterol signals regulate central pathways of glucose homeostasis are not completely understood. By using a hepatocyte-specific PKCß-deficient (PKCßHep-/-) mouse model, we demonstrated the role of hepatic PKCß in slowing disposal of glucose overload by suppressing glycogenesis and increasing hepatic glucose output. PKCßHep-/- mice exhibited lower plasma glucose under the fed condition, modestly improved systemic glucose tolerance and mildly suppressed gluconeogenesis, increased hepatic glycogen accumulation and synthesis due to elevated glucokinase expression and activated glycogen synthase (GS), and suppressed glucose-6-phosphatase expression compared with controls. These events were independent of hepatic AKT/GSK-3α/ß signaling and were accompanied by increased HNF-4α transactivation, reduced FoxO1 protein abundance, and elevated expression of GS targeting protein phosphatase 1 regulatory subunit 3C in the PKCßHep-/- liver compared with controls. The above data strongly imply that hepatic PKCß deficiency causes hypoglycemia postprandially by promoting glucose phosphorylation via upregulating glucokinase and subsequently redirecting more glucose-6-phosphate to glycogen via activating GS. In summary, hepatic PKCß has a unique and essential ability to induce a coordinated response that negatively affects glycogenesis at multiple levels under physiological postprandial conditions, thereby integrating nutritional fat intake with dysregulation of glucose homeostasis.


Asunto(s)
Glucemia/metabolismo , Grasas de la Dieta , Glucógeno/biosíntesis , Hígado/metabolismo , Proteína Quinasa C beta/genética , Animales , Colesterol en la Dieta , Proteína Forkhead Box O1/metabolismo , Glucoquinasa/metabolismo , Gluconeogénesis/genética , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/metabolismo , Glucógeno Sintasa/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Ratones , Ratones Noqueados , Periodo Posprandial/genética , Proteína Quinasa C beta/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal
3.
Diabetes ; 70(12): 2932-2946, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34610981

RESUMEN

Humans spend the greater part of the day in a postprandial state. However, the genetic basis of postprandial blood measures is relatively uncharted territory. We examined the genetics of variation in concentrations of postprandial metabolites (t = 150 min) in response to a liquid mixed meal through genome-wide association studies (GWAS) performed in the Netherlands Epidemiology of Obesity (NEO) study (n = 5,705). The metabolite response GWAS identified an association between glucose change and rs10830963:G in the melatonin receptor 1B (ß [SE] -0.23 [0.03], P = 2.15 × 10-19). In addition, the ANKRD55 locus led by rs458741:C showed strong associations with extremely large VLDL (XXLVLDL) particle response (XXLVLDL total cholesterol: ß [SE] 0.17 [0.03], P = 5.76 × 10-10; XXLVLDL cholesterol ester: ß [SE] 0.17 [0.03], P = 9.74 × 10-10), which also revealed strong associations with body composition and diabetes in the UK Biobank (P < 5 × 10-8). Furthermore, the associations between XXLVLDL response and insulinogenic index, HOMA-ß, Matsuda insulin sensitivity index, and HbA1c in the NEO study implied the role of chylomicron synthesis in diabetes (with false discovery rate-corrected q <0.05). To conclude, genetic studies of metabolomics change after a liquid meal illuminate novel pathways for glucose and lipid metabolism. Further studies are warranted to corroborate biological pathways of the ANKRD55 locus underlying diabetes.


Asunto(s)
Metabolismo de los Hidratos de Carbono/genética , Glucosa/metabolismo , Metabolismo de los Lípidos/genética , Comidas/fisiología , Metaboloma/genética , Anciano , Femenino , Estudio de Asociación del Genoma Completo , Técnicas de Genotipaje , Humanos , Resistencia a la Insulina/genética , Masculino , Metabolómica/métodos , Persona de Mediana Edad , Países Bajos , Periodo Posprandial/genética , Soluciones
4.
Neurosci Lett ; 764: 136230, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34500001

RESUMEN

S-Adenosylmethionine (SAM) is the major endogenous methyl donor for methyltransferase reactions, while 5-Azacytidine (AZA) is a synthetic drug inhibiting DNA methyltransferase activity. Both molecules can thus influence DNA methylation patterns in an organism and thereby affect gene expression and ultimately behavior in the long-term. Whether or not effects on behavior are exerted on a shorter time scale is unclear. The goal of this study was to explore the direct effects of SAM and AZA on appetite regulation, using broiler chicken and Japanese quail as the animal models. Fed or 180 min-fasted broilers (at day 4 post-hatch) or 360 min-fasted quail (at day 7 post-hatch) were intracerebroventricularly injected with SAM or AZA and food intake was measured for 360 min. For broilers, there was no effect of AZA, at any dose, on food intake in either fed or fasted chicks at any time point. In contrast, 1 and 10 µg doses of SAM reduced food intake in fed chicks at 60 min post-injection. In fasted chicks, although there were no differences for the first 30 min post-injection, SAM suppressed food intake during the second 30-min period. For quail, however, AZA (25 µg dose) decreased food intake at 60 and 150-360 min post-injection in fasted birds. A reduction in food intake was also observed at 120- and 360-min post-injection in fed quail in response to 5 and 25 µg doses of AZA, respectively. SAM had no effect when quail were fasted, whereas 1 µg dose of SAM suppressed food consumption in fed quail during the third 30-min period. Thus, when administered directly into the central nervous system, SAM may act as a transient appetite suppressant in both broilers and quail, whereas the direct inhibitory effect of AZA on food consumption depends on species and nutritional states.


Asunto(s)
Apetito/efectos de los fármacos , Azacitidina/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , S-Adenosilmetionina/administración & dosificación , Animales , Apetito/genética , Pollos/fisiología , Coturnix/fisiología , Metilación de ADN/efectos de los fármacos , Ingestión de Alimentos/genética , Ayuno , Femenino , Inyecciones Intraventriculares , Masculino , Modelos Animales , Periodo Posprandial/efectos de los fármacos , Periodo Posprandial/genética , Especificidad de la Especie
5.
J Nutr ; 151(4): 848-856, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33693945

RESUMEN

BACKGROUND: Previous acute studies suggest the Glu298Asp polymorphism (rs1799983) may influence vascular reactivity in response to long-chain n-3 PUFA intake. However, the effects of this genotype on postprandial vascular function after meals rich in SFAs, n-6 PUFAs, and MUFAs are unclear. OBJECTIVES: This study determined the impact of the Glu298Asp polymorphism on changes in vascular function and cardiometabolic risk biomarkers in response to sequential meals of varying fat composition. METHODS: In a randomized, double-blind, crossover, acute study, 32 postmenopausal women (mean ± SD age: 58 ± 5 y; BMI: 25.9 ± 4.1 kg/m2) consumed mixed meals (breakfast: 0 min, 50 g fat; lunch: 330 min, 30 g fat) containing SFAs, n-6 PUFAs, or MUFAs on 3 occasions. Blood samples for cardiometabolic disease risk markers and real-time measures of vascular reactivity [including flow-mediated dilatation (FMD; primary outcome)] were collected/performed before and regularly for 480 min after breakfast. Participants were retrospectively genotyped for the Glu298Asp (rs1799983) polymorphism. Data were analyzed using linear mixed models. RESULTS: For the postprandial %FMD response, a test fat × genotype interaction was observed for the AUC (P = 0.019) but not incremental AUC (IAUC), with the AUC being ∼24% greater after MUFA- than after SFA- and n-6 PUFA-rich meals in the Glu298 homozygotes (P ≤ 0.026). Test fat × genotype interactions were also evident for postprandial insulin (P ≤ 0.005), with the MUFA-rich meals demonstrating significantly higher AUC (12.8%/14.9%), IAUC (14.6%/20.0%), and maximum concentration (20.0%/34.5%) than the SFA- and n-6 PUFA-rich meals, respectively, in Asp298 carriers (P < 0.05). Genotype did not influence other study outcome measures in response to the test fats. CONCLUSIONS: Our findings suggest the Glu298Asp polymorphism may represent a potential determinant of the inter-individual variability in postprandial responsiveness of %FMD and insulin to acute meal fat composition in postmenopausal women. Further studies are required to confirm these observations.This trial was registered at clinicaltrials.gov as NCT02144454.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Insulina/sangre , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/fisiología , Polimorfismo de Nucleótido Simple , Vasodilatación/genética , Vasodilatación/fisiología , Biomarcadores/sangre , Factores de Riesgo Cardiometabólico , Estudios Cruzados , Grasas de la Dieta/análisis , Método Doble Ciego , Ácidos Grasos/administración & dosificación , Ácidos Grasos Monoinsaturados/administración & dosificación , Ácidos Grasos Omega-6/administración & dosificación , Femenino , Humanos , Persona de Mediana Edad , Posmenopausia/sangre , Posmenopausia/genética , Posmenopausia/fisiología , Periodo Posprandial/genética , Periodo Posprandial/fisiología
6.
Nat Med ; 27(2): 321-332, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33432175

RESUMEN

The gut microbiome is shaped by diet and influences host metabolism; however, these links are complex and can be unique to each individual. We performed deep metagenomic sequencing of 1,203 gut microbiomes from 1,098 individuals enrolled in the Personalised Responses to Dietary Composition Trial (PREDICT 1) study, whose detailed long-term diet information, as well as hundreds of fasting and same-meal postprandial cardiometabolic blood marker measurements were available. We found many significant associations between microbes and specific nutrients, foods, food groups and general dietary indices, which were driven especially by the presence and diversity of healthy and plant-based foods. Microbial biomarkers of obesity were reproducible across external publicly available cohorts and in agreement with circulating blood metabolites that are indicators of cardiovascular disease risk. While some microbes, such as Prevotella copri and Blastocystis spp., were indicators of favorable postprandial glucose metabolism, overall microbiome composition was predictive for a large panel of cardiometabolic blood markers including fasting and postprandial glycemic, lipemic and inflammatory indices. The panel of intestinal species associated with healthy dietary habits overlapped with those associated with favorable cardiometabolic and postprandial markers, indicating that our large-scale resource can potentially stratify the gut microbiome into generalizable health levels in individuals without clinically manifest disease.


Asunto(s)
Microbioma Gastrointestinal/genética , Metagenoma/genética , Microbiota/genética , Obesidad/microbiología , Adulto , Biomarcadores/metabolismo , Blastocystis/genética , Glucemia/metabolismo , Niño , Dieta/efectos adversos , Ayuno/metabolismo , Conducta Alimentaria , Femenino , Microbiología de Alimentos , Glucosa/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo , Periodo Posprandial/genética , Prevotella/genética , Prevotella/aislamiento & purificación
7.
Mol Nutr Food Res ; 64(21): e2000319, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32844586

RESUMEN

SCOPE: It is aimed to investigate how intake of high-fat meals composed of different dairy products with a similar fat content affects postprandial peripheral blood mononuclear cell (PBMC) expression of inflammation-related genes, as well as circulating inflammatory markers and metabolites. METHODS AND RESULTS: Healthy subjects (n = 47) consume four different high-fat meals composed of either butter, cheese, whipped cream, or sour cream in a randomized controlled cross-over study. Fasting and postprandial PBMC gene expression, plasma metabolites, and circulating inflammatory markers are measured. Using a linear mixed model, it is found that expression of genes related to lymphocyte activation, cytokine signaling, chemokine signaling, and cell adhesion is differentially altered between the four meals. In general, intake of the fermented products cheese and sour cream reduces, while intake of the non-fermented products butter and whipped cream increases, expression of these genes. Plasma amino acid concentrations increase after intake of cheese compared to the other meals, and the amino acid changes correlate with several of the differentially altered genes. CONCLUSION: Intake of fermented dairy products, especially cheese, induces a less inflammatory postprandial PBMC gene expression response than non-fermented dairy products. These findings may partly explain inconsistent findings in studies on health effects of dairy products.


Asunto(s)
Productos Lácteos Cultivados , Expresión Génica/fisiología , Inflamación/sangre , Leucocitos Mononucleares/fisiología , Periodo Posprandial/genética , Adulto , Biomarcadores/sangre , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Inflamación/dietoterapia , Metabolismo de los Lípidos , Masculino , Persona de Mediana Edad , Periodo Posprandial/fisiología , Adulto Joven
8.
Int J Mol Sci ; 21(9)2020 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-32397573

RESUMEN

Gut-derived satiety hormones provide negative feedback to suppress food intake and maintain metabolic function in peripheral tissues. Despite the wealth of knowledge of the systemic effects of these hormones, very little is known concerning the mechanisms by which nutrients, such as dietary fats, can promote the expression of genes involved in L-cell hormone production. We have tested the role of various dietary fats and found that after hydrolysis into free fatty acids (FFA's), there is a differential response in the extent to which they induce PYY gene and protein production. The effect of FFA's also seems to relate to triglyceride (TG) re-esterification rate, with MUFA re-esterifying faster with lower PYY production. We have also found that there are differences in potency of FFA's based on their desaturation patterns in vitro. The potency effect of FFA's is influenced by the rate of TG re-esterification, such that the longer FFA's are in contact with L-cells, the more PYY they produce. We found that chronic consumption of high-fat diets enables the small intestine to re-esterify FFA's into TG faster and earlier which resulted in a blunted postprandial PYY response. Lastly, we found that FFA's induce X-box-binding protein-1 activation (Xbp1s) in L-cells and that adenoviral delivery of Xbp1s was sufficient to induce PYY gene expression. Taken together, the present work indicates that dietary fat can induce satiety, in part, prior to re-esterification. Chronic high-fat diet consumption increases the rate of re-esterification which diminishes satiety and may lead to increased food intake. Targeting intestinal TG synthesis may prove beneficial in restoring obesity-associated reductions in postprandial satiety.


Asunto(s)
Ácidos Grasos Monoinsaturados/farmacología , Ácidos Grasos/farmacología , Péptido YY/metabolismo , Periodo Posprandial/efectos de los fármacos , Empalme del ARN/genética , Triglicéridos/biosíntesis , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Línea Celular Tumoral , Dieta Alta en Grasa , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Ácidos Grasos no Esterificados/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células L , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Péptido YY/genética , Periodo Posprandial/genética , Empalme del ARN/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Respuesta de Saciedad/fisiología , Triglicéridos/metabolismo , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/farmacología
9.
Peptides ; 128: 170309, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32259550

RESUMEN

CRF system, structural conservation, has an association with feeding regulation in mammals. However, mammals and fish have different physiological mechanisms, the potential role of CRF system for feeding regulation in teleost fish are most unknown. To better explore possible feeding mechanisms of CRF system in Acipenser dabryanus, the gene expression patterns of CRF system have been investigated after different energy status. CRF and two receptors have been studied in Acipenser dabryanus in previous study, thus, four components of CRF system (UI, UCN2, UCN3 and CRF-BP) have been studied in this study. Results showed post-prandial increased UCNs mRNA expressions, and 10 days fasting decreased UCNs mRNA expressions, and the mRNA abundance of CRF-BP has no significant differences. Above, this study confirmed the CRF system has potential role for feeding regulation in Acipenser dabryanus.


Asunto(s)
Hormona Liberadora de Corticotropina/genética , Conducta Alimentaria/fisiología , Proteínas de Peces/genética , Peces/fisiología , ARN Mensajero/metabolismo , Receptores de Hormona Liberadora de Corticotropina/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular/métodos , Hormona Liberadora de Corticotropina/metabolismo , ADN Complementario/genética , Proteínas de Peces/metabolismo , Peces/genética , Peces/metabolismo , Filogenia , Periodo Posprandial/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Homología de Secuencia
10.
PLoS One ; 15(2): e0229495, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32101585

RESUMEN

PURPOSE: "Quantile-dependent expressivity" describes an effect of the genotype that depends upon the level of the phenotype (e.g., whether a subject's triglycerides are high or low relative to its population distribution). Prior analyses suggest that the effect of a genetic risk score (GRS) on fasting plasma triglyceride levels increases with the percentile of the triglyceride distribution. Postprandial lipemia is well suited for testing quantile-dependent expressivity because it exposes each individual's genotype to substantial increases in their plasma triglyceride concentrations. Ninety-seven published papers were identified that plotted mean triglyceride response vs. time and genotype, which were converted into quantitative data. Separately, for each published graph, standard least-squares regression analysis was used to compare the genotype differences at time t (dependent variable) to average triglyceride concentrations at time t (independent variable) to assess whether the genetic effect size increased in association with higher triglyceride concentrations and whether the phenomenon could explain purported genetic interactions with sex, diet, disease, BMI, and drugs. RESULTS: Consistent with the phenomenon, genetic effect sizes increased (P≤0.05) with increasing triglyceride concentrations for polymorphisms associated with ABCA1, ANGPTL4, APOA1, APOA2, APOA4, APOA5, APOB, APOC3, APOE, CETP, FABP2, FATP6, GALNT2, GCKR, HL, IL1b, LEPR, LOX-1, LPL, MC4R, MTTP, NPY, SORT1, SULF2, TNFA, TCF7L2, and TM6SF2. The effect size for these polymorphisms showed a progressively increasing dose-response, with intermediate effect sizes at intermediate triglyceride concentrations. Quantile-dependent expressivity provided an alternative interpretation to their interactions with sex, drugs, disease, diet, and age, which have been traditionally ascribed to gene-environment interactions and genetic predictors of drug efficacy (i.e., personalized medicine). CONCLUSION: Quantile-dependent expressivity applies to the majority of genetic variants affecting postprandial triglycerides, which may arise because the impaired functionalities of these variants increase at higher triglyceride concentrations. Purported gene-drug interactions may be the manifestations of quantile-dependent expressivity, rather than genetic predictors of drug efficacy.


Asunto(s)
Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Periodo Posprandial/fisiología , Adulto , Apolipoproteína C-III/genética , Apolipoproteínas A/genética , HDL-Colesterol/sangre , Femenino , Genotipo , Humanos , Hiperlipidemias/fisiopatología , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Periodo Posprandial/genética , Análisis de Regresión , Triglicéridos/análisis , Triglicéridos/sangre
11.
J Clin Lipidol ; 14(1): 66-76.e11, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31859127

RESUMEN

BACKGROUND: Humans spend most of the time in the postprandial state, yet most knowledge about high-density lipoproteins (HDL) derives from the fasted state. HDL protein and lipid cargo mediate HDL's antiatherogenic effects, but whether these HDL constituents change in the postprandial state and are affected by dietary macronutrients remains unknown. OBJECTIVES: This study aimed to assess changes in HDL protein and lipid composition after the consumption of a high-carbohydrate or high saturated fat (HSF) meal. METHODS: We isolated HDL from plasma collected during a randomized, cross-over study of metabolically healthy subjects. Subjects consumed isocaloric meals consisting predominantly of either carbohydrate or fat. At baseline and at 3 and 6 hours postprandial, we quantified HDL protein and lipid composition by liquid chromatography-mass spectrometry. RESULTS: A total of 15 subjects were included (60% female, aged 34 ± 15 years, body mass index: 24.1 ± 2.7 kg/m2). Consumption of the HSF meal led to HDL enrichment in total lipid (P = .006), triglyceride (P = .02), and phospholipid (P = .008) content and a corresponding depletion in protein content. After the HSF meal, 16 of the 25 measured phosphatidylcholine species significantly increased in abundance (P values range from .027 to <.001), along with several sphingolipids including ceramides (P < .004), lactosylceramide (P = .023), and sphingomyelin-14 (P = .013). Enrichment in apolipoprotein A-I (P = .001) was the only significant change in HDL protein composition after the HSF meal. The high-carbohydrate meal conferred only minimal changes in HDL composition. CONCLUSION: Meal macronutrient content acutely affects HDL composition in the postprandial state, with the HSF meal resulting in enrichment of HDL phospholipid content with possible consequences for HDL function.


Asunto(s)
Carbohidratos/administración & dosificación , Grasas de la Dieta/administración & dosificación , Ácidos Grasos/sangre , Lipoproteínas HDL/sangre , Obesidad/sangre , Adulto , Glucemia/genética , Índice de Masa Corporal , Carbohidratos/efectos adversos , LDL-Colesterol/sangre , Grasas de la Dieta/efectos adversos , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Ayuno , Femenino , Humanos , Lipidómica/métodos , Masculino , Comidas , Obesidad/dietoterapia , Obesidad/genética , Obesidad/patología , Periodo Posprandial/genética , Triglicéridos/sangre
12.
Nutrients ; 11(9)2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31480637

RESUMEN

Apolipoprotein (APO) E (ε) genotype is considered to play an important role in lipid responses to dietary fat manipulation but the impact on novel cardiometabolic risk markers is unclear. To address this knowledge gap, we investigated the relationship between the APOE genotype and cardiometabolic risk markers in response to acute and chronic dietary fat intakes. Associations with fasting (baseline) outcome measures (n = 218) were determined using data from the chronic DIVAS (n = 191/195 adults at moderate cardiovascular disease risk) and acute DIVAS-2 (n = 27/32 postmenopausal women) studies examining the effects of diets/meals varying in saturated, polyunsaturated and monounsaturated (MUFA) fatty acid composition. Participants were retrospectively genotyped for APOE (rs429358, rs7412). For baseline cardiometabolic outcomes, E4 carriers had higher fasting total and low-density lipoprotein-cholesterol (LDL-C), total cholesterol: high-density lipoprotein-cholesterol (HDL-C) and LDL-C: HDL-C ratios, but lower C-reactive protein (CRP) than E3/E3 and E2 carriers (p ≤ 0.003). Digital volume pulse stiffness index was higher in E2 carriers than the E3/E3 group (p = 0.011). Following chronic dietary fat intake, the significant diet × genotype interaction was found for fasting triacylglycerol (p = 0.010), with indication of a differential responsiveness to MUFA intake between the E3/E3 and E4 carriers (p = 0.006). Test fat × genotype interactions were observed for the incremental area under the curve for the postprandial apolipoprotein B (apoB; p = 0.022) and digital volume pulse reflection index (DVP-RI; p = 0.030) responses after the MUFA-rich meals, with a reduction in E4 carriers and increase in the E3/E3 group for the apoB response, but an increase in E4 carriers and decrease in the E3/E3 group for the DVP-RI response. In conclusion, baseline associations between the APOE genotype and fasting lipids and CRP confirm previous findings, although a novel interaction with digital volume pulse arterial stiffness was observed in the fasted state and differential postprandial apoB and DVP-RI responses after the MUFA-rich meals. The reported differential impact of the APOE genotype on cardiometabolic markers in the acute and chronic state requires confirmation.


Asunto(s)
Apolipoproteínas E/genética , Grasas de la Dieta/farmacología , Ayuno/sangre , Comidas/fisiología , Periodo Posprandial/genética , Anciano , Apolipoproteínas E/sangre , Proteína C-Reactiva/análisis , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Femenino , Genotipo , Humanos , Lípidos/sangre , Masculino , Persona de Mediana Edad , Posmenopausia/sangre , Estudios Retrospectivos
13.
J Nutr Biochem ; 72: 108209, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31473510

RESUMEN

White blood cells are among the first responders to dietary components and their metabolites absorbed from the gut. The objective of this study was to determine the whole blood transcriptome response to high-fat challenge meals. A total of 45 fasting and postprandial (3-h and 6-h) whole blood transcriptomes from 5 subjects in a crossover intervention trial of a high-fat meal supplemented with placebo, blueberry powder or docosahexaenoic acid (DHA) were analyzed using RNA sequencing. Select target genes were validated by quantitative reverse-transcription polymerase chain reaction in 180 samples from 20 subjects. The largest contributor to variance was the subject (13,856 genes differentially expressed), followed by the subject on a specific day (2276 genes), followed by the subject's postprandial response (651 genes). After determining the nonsignificance of individual dietary treatments (blueberry, DHA, placebo), treatments were used as replicates to examine postprandial responses to a high-fat meal. The universal postprandial response (95 genes) was associated with lipid utilization, fatty acid beta-oxidation and circadian rhythms. Subject-specific postprandial responses were enriched for genes involved in the innate immune response, particularly those of pattern recognition receptors and their downstream signaling components. Genes involved in innate immune responses are differentially expressed in a subject-specific and time-dependent manner in response to the high-fat meals. These genes can serve as biomarkers to assess individual responsiveness to a high-fat diet in inducing postprandial inflammation. Furthermore, the dynamic temporal change in gene expression in postprandial blood suggests that monitoring these genes at multiple time points is necessary to reveal responders to dietary intervention.


Asunto(s)
Sangre/inmunología , Grasas de la Dieta/administración & dosificación , Inmunidad Innata/genética , Periodo Posprandial/genética , Transcriptoma , Adulto , Arándanos Azules (Planta)/química , Dieta Alta en Grasa/efectos adversos , Ácidos Docosahexaenoicos/farmacología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Placebos , Adulto Joven
14.
Physiol Genomics ; 51(9): 411-431, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31282806

RESUMEN

Carnivorous rainbow trout exhibit prolonged postprandial hyperglycemia when fed a diet exceeding 20% carbohydrate content. This poor capacity to utilize carbohydrates has led to rainbow trout being classified as "glucose-intolerant" (GI). The metabolic phenotype has spurred research to identify the underlying cellular and molecular mechanisms of glucose intolerance, largely because carbohydrate-rich diets provide economic and ecological advantages over traditionally used fish meal, considered unsustainable for rainbow trout aquaculture operations. Evidence points to a contribution of hepatic intermediary carbohydrate and lipid metabolism, as well as upstream insulin signaling. Recently, microRNAs (miRNAs), small noncoding RNAs acting as negative posttranscriptional regulators affecting target mRNA stability and translation, have emerged as critical regulators of hepatic control of glucose-homeostasis in mammals, revealing that dysregulated hepatic miRNAs might play a role in organismal hyperglycemia in metabolic disease. To determine whether hepatic regulatory miRNA networks may contribute to GI in rainbow trout, we induced prolonged postprandial hyperglycemia in rainbow trout by using a carbohydrate-rich diet and profiled genome-wide hepatic miRNAs in hyperglycemic rainbow trout compared with fasted trout and trout fed a diet devoid of carbohydrates. Using small RNA next-generation sequencing and real-time RT-PCR validation, we identified differentially regulated hepatic miRNAs between these groups and used an in silico approach to predict bona fide mRNA targets and enriched pathways. Diet-induced hyperglycemia resulted in differential regulation of hepatic miRNAs compared with fasted fish. Some of the identified miRNAs, such as miRNA-27b-3p and miRNA-200a-3p, are known to be responsive to hyperglycemia in the liver of hyperglycemic glucose-tolerant fish and mammals, suggesting an evolutionary conserved regulation. Using Gene Ontology term-based enrichment analysis, we identify intermediate carbohydrate and lipid metabolism and insulin signaling as potential targets of posttranscriptional regulation by hyperglycemia-regulated miRNAs and provide correlative expression analysis of specific predicted miRNA-target pairs. This study identifies hepatic miRNAs in rainbow trout that exhibit differential postprandial expression in response to diets with different carbohydrate content and predicts posttranscriptionally regulated target mRNAs enriched for pathways involved in glucoregulation. Together, these results provide a framework for testable hypotheses of functional involvement of specific hepatic miRNAs in GI in rainbow trout.


Asunto(s)
Dieta de Carga de Carbohidratos/efectos adversos , Hiperglucemia/etiología , Hígado/metabolismo , MicroARNs/genética , Oncorhynchus mykiss/genética , Transcriptoma , Animales , Regulación de la Expresión Génica , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Insulina/metabolismo , Periodo Posprandial/genética , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
15.
J Nutr ; 149(6): 915-922, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31049566

RESUMEN

BACKGROUND: The transcription factor 7-like 2 (TCF7L2) gene confers one of the strongest genetic predispositions to type 2 diabetes, but diabetes development can be modified by diet. OBJECTIVE: The aim of our study was to evaluate postprandial metabolic alterations in healthy men with a high genetic risk of diabetes, after two meals with varying macronutrient content. METHODS: The study was conducted in 21 homozygous nondiabetic men carrying the high-risk (HR, n = 8, age: 31.2 ± 6.3 y, body mass index (BMI, kg/m2) 28.5 ± 8.1) or low-risk (LR, n = 13, age: 35.2 ± 10.3 y, BMI: 28.1 ± 6.4) genotypes at the rs7901695 locus. During two meal challenge test visits subjects received standardized isocaloric (450 kcal) liquid meals: high-carbohydrate (HC, carbohydrates: 89% of energy) and normo-carbohydrate (NC, carbohydrates: 45% of energy). Fasting (0 min) and postprandial (30, 60, 120, 180 min) plasma samples were analyzed for metabolite profiles through untargeted metabolomics. Metabolic fingerprinting was performed on an ultra-high-performance liquid chromatography (UHPLC) system connected to an iFunnel quadrupole-time-of-flight (Q-TOF) mass spectrometer. RESULTS: In HR-genotype men, after the intake of an HC-meal, we noted a significantly lower area under the curves (AUCs) of postprandial plasma concentrations of most of the phospholipids (-37% to -53%, variable importance in the projection (VIP) = 1.2-1.5), lysophospholipids (-29% to -86%, VIP = 1.1-2.6), sphingolipids (-32% to -47%, VIP = 1.1-1.3), as well as arachidonic (-36%, VIP = 1.4) and oleic (-63%, VIP = 1.3) acids, their metabolites: keto- and hydoxy-fatty acids (-38% to -78%, VIP = 1.3-2.5), leukotrienes (-65% to -83%, VIP = 1.4-2.2), uric acid (-59%, VIP = 1.5), and pyroglutamic acid (-65%, VIP = 1.8). The AUCs of postprandial sphingosine concentrations were higher (125-832%, VIP = 1.9-3.2) after the NC-meal, AUCs of acylcarnitines were lower (-21% to -61%, VIP = 1.1-2.4), and AUCs of fatty acid amides were higher (51-508%, VIP = 1.7-3.1) after the intake of both meals. CONCLUSIONS: In nondiabetic men carrying the TCF7L2 HR genotype, subtle but detectable modifications in intermediate lipid metabolism are induced by an HC-meal. This trial was registered at www.clinicaltrials.gov as NCT03792685.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Carbohidratos de la Dieta/administración & dosificación , Metabolismo de los Lípidos/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Adulto , Estudios Cruzados , Predisposición Genética a la Enfermedad , Genotipo , Homocigoto , Humanos , Lípidos/sangre , Masculino , Metabolómica , Polimorfismo de Nucleótido Simple , Periodo Posprandial/genética , Periodo Posprandial/fisiología , Factores de Riesgo , Método Simple Ciego
16.
PLoS Biol ; 16(12): e2006249, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30532187

RESUMEN

Hepatic circadian gene transcription is tightly coupled to feeding behavior, which has a profound impact on metabolic disorders associated with diet-induced obesity. Here, we describe a genomics approach to uncover mechanisms controlling hepatic postprandial gene expression. Combined transcriptomic and cistromic analysis identified hundreds of circadian-regulated genes and enhancers controlled by feeding. Postprandial suppression of enhancer activity was associated with reduced glucocorticoid receptor (GR) and Forkhead box O1 (FOXO1) occupancy of chromatin correlating with reduced serum corticosterone levels and increased serum insulin levels. Despite substantial co-occupancy of feeding-regulated enhancers by GR and FOXO1, selective disruption of corticosteroid and/or insulin signaling resulted in dysregulation of specific postprandial regulated gene programs. In combination, these signaling pathways operate a major part of the genes suppressed by feeding. Importantly, the feeding response was disrupted in diet-induced obese animals, which was associated with dysregulation of several corticosteroid- and insulin-regulated genes, providing mechanistic insights to dysregulated circadian gene transcription associated with obesity.


Asunto(s)
Insulina/metabolismo , Periodo Posprandial/genética , Receptores de Glucocorticoides/metabolismo , Animales , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Hepatocitos/metabolismo , Insulina/genética , Resistencia a la Insulina , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Receptores de Glucocorticoides/genética , Transducción de Señal/efectos de los fármacos
17.
PLoS Genet ; 14(11): e1007767, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30457986

RESUMEN

Behavior and physiology are orchestrated by neuropeptides acting as central neuromodulators and circulating hormones. An outstanding question is how these neuropeptides function to coordinate complex and competing behaviors. In Drosophila, the neuropeptide leucokinin (LK) modulates diverse functions, but mechanisms underlying these complex interactions remain poorly understood. As a first step towards understanding these mechanisms, we delineated LK circuitry that governs various aspects of post-feeding physiology and behavior. We found that impaired LK signaling in Lk and Lk receptor (Lkr) mutants affects diverse but coordinated processes, including regulation of stress, water homeostasis, feeding, locomotor activity, and metabolic rate. Next, we sought to define the populations of LK neurons that contribute to the different aspects of this physiology. We find that the calcium activity in abdominal ganglia LK neurons (ABLKs), but not in the two sets of brain neurons, increases specifically following water consumption, suggesting that ABLKs regulate water homeostasis and its associated physiology. To identify targets of LK peptide, we mapped the distribution of Lkr expression, mined a brain single-cell transcriptome dataset for genes coexpressed with Lkr, and identified synaptic partners of LK neurons. Lkr expression in the brain insulin-producing cells (IPCs), gut, renal tubules and chemosensory cells, correlates well with regulatory roles detected in the Lk and Lkr mutants. Furthermore, these mutants and flies with targeted knockdown of Lkr in IPCs displayed altered expression of insulin-like peptides (DILPs) and transcripts in IPCs and increased starvation resistance. Thus, some effects of LK signaling appear to occur via DILP action. Collectively, our data suggest that the three sets of LK neurons have different targets, but modulate the establishment of post-prandial homeostasis by regulating distinct physiological processes and behaviors such as diuresis, metabolism, organismal activity and insulin signaling. These findings provide a platform for investigating feeding-related neuroendocrine regulation of vital behavior and physiology.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Neuropéptidos/genética , Neuropéptidos/fisiología , Animales , Animales Modificados Genéticamente , Conducta Animal/fisiología , Diuresis/genética , Diuresis/fisiología , Proteínas de Drosophila/deficiencia , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Insulina/fisiología , Masculino , Actividad Motora/genética , Actividad Motora/fisiología , Mutación , Neuronas/clasificación , Neuronas/fisiología , Neuropéptidos/deficiencia , Periodo Posprandial/genética , Periodo Posprandial/fisiología , Receptores de Neuropéptido/deficiencia , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/fisiología , Transducción de Señal
18.
PLoS One ; 13(11): e0206727, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30395627

RESUMEN

MicroRNAs (miRNAs) are a class of small non-coding RNAs which are known to posttranscriptionally regulate the expression of most genes in both animals and plants. Meanwhile, studies have shown that numbers of miRNAs are present in body fluids including the plasma. Despite the mode of action of these circulating miRNAs still remains unknown, they have been found to be promising biomarkers for disease diagnosis, prognosis and response to treatment. In order to evaluate the potential of miRNAs as non-invasive biomarkers in aquaculture, a time-course experiment was implemented to investigate the postprandial regulation of miRNAs levels in liver and plasma as well as the hepatic expression of genes involved in cholesterol metabolism. We showed that miR-1, miR-33a, miR-122, miR-128 and miR-223 were expressed in the liver of rainbow trout and present at detectable level in the plasma. We also demonstrated that hepatic expression of miR-1, miR-122 and miR-128 were regulated by feed intake and reached their highest levels 12 hours after the meal. Interestingly, we observed that circulating levels of miR-128 and miR-223 are subjected to postprandial regulations similar to that observed in their hepatic counterparts. Statistical correlations were observed between liver and plasma for miR-128 and miR-223 and between hepatic and circulating miR-122, miR-128 and miR-223 and expression of genes related to cholesterol synthesis and efflux or glucose phosphorylation. These results demonstrated that circulating miR-122, miR-128 and miR-223 are potential biomarkers of cholesterol metabolism in rainbow trout.


Asunto(s)
Colesterol/genética , Colesterol/metabolismo , MicroARNs/sangre , MicroARNs/genética , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/metabolismo , Animales , Glucemia/metabolismo , Colesterol/sangre , Perfilación de la Expresión Génica , Metabolismo de los Lípidos/genética , Lipogénesis/genética , Hígado/metabolismo , MicroARNs/metabolismo , Oncorhynchus mykiss/sangre , Periodo Posprandial/genética , Triglicéridos/sangre
19.
J Nutr Biochem ; 57: 45-55, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29674246

RESUMEN

The postprandial hypertriglyceridemia is an important and largely silent disturbance involved in the genesis of numerous pathological conditions. Exaggerated and prolonged states of postprandial hypertriglyceridemia are frequently related to the ingestion of meals enriched in saturated fatty acids (SFAs). MicroRNAs are noncoding RNAs that function as gene regulators and play significant roles in both health and disease. However, differential miRNA expression between fasting and postprandial states has never been elucidated. Here, we studied the impact of a high-saturated-fat meal, mainly rich in palmitic acid, on the miRNA signature in peripheral blood mononuclear cells (PBMCs) of nine male healthy individuals in the postprandial period by using a two-step analysis: miRNA array and validation through quantitative real-time polymerase chain reaction. Compared with miRNA expression signature in PBMCs at fasting, 36 miRNAs were down-regulated and 43 miRNAs were up-regulated in PBMCs at postprandial hypertriglyceridemic peak. Six chromosomes (3, 7, 8, 12, 14 and 19) had nearly half (48.1%) of dysregulated miRNA-gene-containing regions. Down-regulated miR-300 and miR-369-3p and up-regulated miR-495-3p, miR-129-5p and miR-7-2-3p had the highest number of target genes. The differentially expressed miRNAs and their predicted target genes involved pathways in cancer, MAPK signaling pathway, endocytosis and axon guidance. Only down-regulated miRNAs notably targeted PI3K-Akt signaling pathways, whereas only up-regulated miRNAs targeted focal adhesion, Wnt signaling pathway, transcriptional misregulation in cancer and ubiquitin-mediated proteolysis. This is the first study of miRNA expression analysis of human PBMCs during postprandial hypertriglyceridemia and offers insight into new potential mechanisms by which dietary SFAs influence health or disease.


Asunto(s)
Ácidos Grasos/farmacología , MicroARNs , Periodo Posprandial/genética , Adolescente , Simulación por Computador , Ácidos Grasos/farmacocinética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertrigliceridemia/genética , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Ácido Palmítico/farmacología , Adulto Joven
20.
PLoS One ; 13(2): e0192947, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29489876

RESUMEN

The metabolic health benefits of fermented milks have already been investigated using clinical biomarkers but the development of transcriptomic analytics in blood offers an alternative approach that may help to sensitively characterise such effects. We aimed to assess the effects of probiotic yoghurt intake, compared to non-fermented, acidified milk intake, on clinical biomarkers and gene expression in peripheral blood. To this end, a randomised, crossover study was conducted in fourteen healthy, young men to test the two dairy products. For a subset of seven subjects, RNA sequencing was used to measure gene expression in blood collected during postprandial tests and after two weeks daily intake. We found that the postprandial response in insulin was different for probiotic yoghurt as compared to that of acidified milk. Moreover changes in several clinical biomarkers were associated with changes in the expression of genes representing six metabolic genesets. Assessment of the postprandial effects of each dairy product on gene expression by geneset enrichment analysis revealed significant, similar modulation of inflammatory and glycolytic genes after both probiotic yoghurt and acidified milk intake, although distinct kinetic characteristics of the modulation differentiated the dairy products. The aryl hydrocarbon receptor was a major contributor to the down-regulation of the inflammatory genesets and was also positively associated with changes in circulating insulin at 2h after yoghurt intake (p = 0.05). Daily intake of the dairy products showed little effect on the fasting blood transcriptome. Probiotic yoghurt and acidified milk appear to affect similar gene pathways during the postprandial phase but differences in the timing and the extent of this modulation may lead to different physiological consequences. The functional relevance of these differences in gene expression is supported by their associations with circulating biomarkers.


Asunto(s)
Leche , Probióticos , Transcriptoma/genética , Yogur , Adulto , Animales , Apetito , Biomarcadores/sangre , Estudios Cruzados , Productos Lácteos Cultivados , Método Doble Ciego , Perfilación de la Expresión Génica , Marcadores Genéticos , Humanos , Masculino , Periodo Posprandial/genética , ARN/sangre , ARN/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...