Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 194
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 212(12): 1958-1970, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38700420

RESUMEN

Fibroblasts acquire a proinflammatory phenotype in inflammatory bowel disease, but the factors driving this process and how fibroblasts contribute to mucosal immune responses are incompletely understood. TNF superfamily member 12 (TNFSF12, or TNF-like weak inducer of apoptosis [TWEAK]) has gained interest as a mediator of chronic inflammation. In this study, we explore its role as a driver of inflammatory responses in fibroblasts and its contribution to fibroblast-monocyte interaction using human primary colonic fibroblasts, THP-1 and primary monocytes. Recombinant human TWEAK induced the expression of cytokines, chemokines, and immune receptors in primary colonic fibroblasts. The TWEAK upregulated transcriptome shared 29% homology with a previously published transcriptional profile of inflammatory fibroblasts from ulcerative colitis. TWEAK elevated surface expression of activated fibroblast markers and adhesion molecules (podoplanin [PDPN], ICAM-1, and VCAM-1) and secretion of IL-6, CCL2, and CXCL10. In coculture, fibroblasts induced monocyte adhesion and secretion of CXCL1 and IL-8, and they promoted a CD14high/ICAM-1high phenotype in THP-1 cells, which was enhanced when fibroblasts were prestimulated with TWEAK. Primary monocytes in coculture with TWEAK-treated fibroblasts had altered surface expression of CD16 and triggering receptor expressed on myeloid cells-1 (TREM-1) as well as increased CXCL1 and CXCL10 secretion. Conversely, inhibition of the noncanonical NF-κB pathway on colonic fibroblasts with a NF-κB-inducing kinase small molecule inhibitor impaired their ability to induce a CD14high phenotype on monocytes. Our results indicate that TWEAK promotes an inflammatory fibroblast-monocyte crosstalk that may be amenable for therapeutic intervention.


Asunto(s)
Diferenciación Celular , Colon , Citocina TWEAK , Fibroblastos , Monocitos , Humanos , Citocina TWEAK/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Fibroblastos/metabolismo , Fibroblastos/inmunología , Colon/inmunología , Colon/patología , Colon/metabolismo , Diferenciación Celular/inmunología , Comunicación Celular/inmunología , Inflamación/inmunología , Células THP-1 , Técnicas de Cocultivo , Citocinas/metabolismo , Adhesión Celular
2.
Rev Endocr Metab Disord ; 24(5): 921-936, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37402955

RESUMEN

Obesity is a heterogenous disease accompanied by a broad spectrum of cardiometabolic risk profiles. Traditional paradigms for dietary weight management do not address biological heterogeneity between individuals and have catastrophically failed to combat the global pandemic of obesity-related diseases. Nutritional strategies that extend beyond basic weight management to instead target patient-specific pathophysiology are warranted. In this narrative review, we provide an overview of the tissue-level pathophysiological processes that drive patient heterogeneity to shape distinct cardiometabolic phenotypes in obesity. Specifically, we discuss how divergent physiology and postprandial phenotypes can reveal key metabolic defects within adipose, liver, or skeletal muscle, as well as the integrative involvement of the gut microbiome and the innate immune system. Finally, we highlight potential precision nutritional approaches to target these pathways and discuss recent translational evidence concerning the efficacy of such tailored dietary interventions for different obesity phenotypes, to optimise cardiometabolic benefits.


Asunto(s)
Enfermedades Cardiovasculares , Obesidad , Humanos , Obesidad/metabolismo , Estado Nutricional , Hígado/metabolismo , Fenotipo , Enfermedades Cardiovasculares/metabolismo
3.
Eur J Nutr ; 62(5): 2319-2332, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37099211

RESUMEN

PURPOSE: Reporting guidelines facilitate quality and completeness in research reporting. The CONsolidated Standards Of Reporting Trials (CONSORT) statement is widely applied to dietary and nutrition trials but has no extension specific to nutrition. Evidence suggests poor reporting in nutrition research. The Federation of European Nutrition Societies led an initiative to make recommendations for a nutrition extension to the CONSORT statement towards a more robust reporting of the evidence base. METHODS: An international working group was formed of nutrition researchers from 14 institutions in 12 different countries and on five continents. Using meetings over a period of one year, we interrogated the CONSORT statement specifically for its application to report nutrition trials. RESULTS: We provide a total of 28 new nutrition-specific recommendations or emphasised recommendations for the reporting of the introduction (three), methods (twelve), results (five) and discussion (eight). We also added two additional recommendations that were not allocated under the standard CONSORT headings. CONCLUSION: We identify a need to provide guidance in addition to CONSORT to improve the quality and consistency of the reporting and propose key considerations for further development of formal guidelines for the reporting of nutrition trials. Readers are encouraged to engage in this process, provide comments and conduct specific studies to inform further work on the development of reporting guidelines for nutrition trials.


Asunto(s)
Ensayos Clínicos Controlados Aleatorios como Asunto , Proyectos de Investigación , Estado Nutricional , Guías como Asunto
4.
Aging Clin Exp Res ; 35(8): 1651-1660, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37326940

RESUMEN

BACKGROUND: Data on the prevalence of sarcopenia among older adults in Ireland are lacking. AIMS: To assess the prevalence and determinants of sarcopenia among community-dwelling older adults in Ireland. METHODS: This cross-sectional analysis involved n = 308 community-dwelling adults aged ≥ 65 y living in Ireland. Participants were recruited via recreational clubs and primary healthcare services. Sarcopenia was defined according to the 2019 European Working Group on Sarcopenia in Older People (EWGSOP2) criteria. Skeletal muscle mass was estimated using bioelectrical impedance analysis, strength was measured via handgrip dynamometry, and physical performance was assessed using the Short Physical Performance Battery. Detailed information was collected on demographics, health, and lifestyle. Dietary macronutrient intake was measured via a single 24 h recall. Binary logistic regression was used to examine potential demographic, health, lifestyle, and dietary determinants of sarcopenia (where both probable and confirmed sarcopenia were combined). RESULTS: The prevalence of EWGSOP2-defined probable sarcopenia was 20.8% and confirmed sarcopenia was 8.1% (5.8% had severe sarcopenia). Polypharmacy (OR 2.60, 95% confidence interval [CI] 1.3, 5.23), height (OR 0.95, 95% CI 0.91, 0.98), and Instrumental Activities Of Daily Living (IADL) score (OR 0.71, 95% CI 0.59, 0.86) were independently associated with sarcopenia (probable and confirmed combined). There were no independent associations between energy-adjusted macronutrient intakes, as determined by 24 h recall, and sarcopenia. CONCLUSION: Sarcopenia prevalence within this sample of community-dwelling older adults in Ireland is broadly similar to other European cohorts. Polypharmacy, lower height, and lower IADL score were independently associated with EWGSOP2-defined sarcopenia.


Asunto(s)
Sarcopenia , Humanos , Anciano , Sarcopenia/epidemiología , Vida Independiente , Fuerza de la Mano , Actividades Cotidianas , Irlanda/epidemiología , Prevalencia , Estudios Transversales
5.
Molecules ; 28(19)2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37836842

RESUMEN

Milk-derived peptides are known to confer anti-inflammatory effects. We hypothesised that milk-derived cell-penetrating peptides might modulate inflammation in useful ways. Using computational techniques, we identified and synthesised peptides from the milk protein Alpha-S1-casein that were predicted to be cell-penetrating using a machine learning predictor. We modified the interpretation of the prediction results to consider the effects of histidine. Peptides were then selected for testing to determine their cell penetrability and anti-inflammatory effects using HeLa cells and J774.2 mouse macrophage cell lines. The selected peptides all showed cell penetrating behaviour, as judged using confocal microscopy of fluorescently labelled peptides. None of the peptides had an effect on either the NF-κB transcription factor or TNFα and IL-1ß secretion. Thus, the identified milk-derived sequences have the ability to be internalised into the cell without affecting cell homeostatic mechanisms such as NF-κB activation. These peptides are worthy of further investigation for other potential bioactivities or as a naturally derived carrier to promote the cellular internalisation of other active peptides.


Asunto(s)
Péptidos de Penetración Celular , FN-kappa B , Humanos , Ratones , Animales , FN-kappa B/metabolismo , Péptidos de Penetración Celular/farmacología , Células HeLa , Leche/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Antiinflamatorios/farmacología
6.
J Sleep Res ; 30(3): e13202, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32996666

RESUMEN

Intermittent hypoxia (IH) plays a key role in the pathogenesis of insulin resistance (IR) in obstructive sleep apnoea (OSA). IH induces a pro-inflammatory phenotype of the adipose tissue with M1 macrophage polarisation, subsequently impeding adipocyte insulin signalling, and these changes are in striking similarity to those seen in obesity. However, the detailed molecular mechanisms of IH-induced macrophage polarisation are unknown and identification of same should lead to the identification of novel therapeutic targets. In the present study, we tested the hypothesis that IH acts through similar mechanisms as obesity, activating Toll-like-receptor (TLR)4/nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) signalling pathways leading to the upregulation and secretion of the key cytokines interleukin (IL)-1ß and IL-6. Bone-marrow derived macrophages (BMDMs) from lean and obese C57BL/6 male mice were exposed to a state-of-the-art in vitro model of IH. Independent of obesity, IH led to a pro-inflammatory M1 phenotype characterised by increased inducible nitric oxide synthase and IL-6 mRNA expression, robust increase in NF-κB DNA-binding activity and IL-6 secretion. Furthermore, IH significantly increased pro-IL-1ß mRNA and protein expression and mature IL-1ß secretion compared to control treatment. Providing mechanistic insight, pre-treatment with the TLR4 specific inhibitor, TAK-242, prevented IH-induced M1 polarisation and upregulation of IL-1ß mRNA and pro-IL-1ß protein expression. Moreover, IH-induced increase in IL-1ß secretion was prevented in BMDMs isolated from NLRP3 knockout mice. Thus, targeting TLR4/NF-κB and NLRP3 signalling pathways may provide novel therapeutic options for metabolic complications in OSA.


Asunto(s)
Activación de Macrófagos/fisiología , Apnea Obstructiva del Sueño/terapia , Animales , Masculino , Ratones
7.
Br J Nutr ; 125(6): 628-632, 2021 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-32892755

RESUMEN

As COVID-19 continues to spread worldwide, severe disease and mortality have been observed in obese patients. We discuss how obesity and obesity-associated factors such as 'meta-flammation', dietary fat intake and paradoxical suppression of the innate immune response within the pulmonary compartment may be crucial determinants in the host response to a novel viral pathogen. Modulation of immune cell bioenergetics and metabolic potential plays a central role in the innate immune response to infection, and as we strive to combat this new global health threat, immunometabolism of the innate immune system warrants attention.


Asunto(s)
COVID-19/inmunología , Sistema Inmunológico/virología , Obesidad/inmunología , Obesidad/virología , SARS-CoV-2/inmunología , COVID-19/mortalidad , Grasas de la Dieta/inmunología , Ingestión de Alimentos/inmunología , Metabolismo Energético/inmunología , Humanos , Inmunidad Innata/inmunología , Inflamación , Obesidad/mortalidad , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología
8.
J Immunol ; 202(5): 1521-1530, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30700584

RESUMEN

Hypoxia is a common and prominent feature of the microenvironment at sites of bacteria-associated inflammation in inflammatory bowel disease. The prolyl-hydroxylases (PHD1/2/3) and the asparaginyl-hydroxylase factor-inhibiting HIF are oxygen-sensing enzymes that regulate adaptive responses to hypoxia through controlling the activity of HIF and NF-κB-dependent transcriptional pathways. Previous studies have demonstrated that the pan-hydroxylase inhibitor dimethyloxalylglycine (DMOG) is effective in the alleviation of inflammation in preclinical models of inflammatory bowel disease, at least in part, through suppression of IL-1ß-induced NF-κB activity. TLR-dependent signaling in immune cells, such as monocytes, which is important in bacteria-driven inflammation, shares a signaling pathway with IL-1ß. In studies into the effect of pharmacologic hydroxylase inhibition on TLR-induced inflammation in monocytes, we found that DMOG selectively triggers cell death in cultured THP-1 cells and primary human monocytes at concentrations well tolerated in other cell types. DMOG-induced apoptosis was independent of increased caspase-3/7 activity but was accompanied by reduced expression of the inhibitor of apoptosis protein 1 (cIAP1). Based on these data, we hypothesize that pharmacologic inhibition of the HIF-hydroxylases selectively targets monocytes for cell death and that this may contribute to the anti-inflammatory activity of HIF-hydroxylase inhibitors.


Asunto(s)
Aminoácidos Dicarboxílicos/farmacología , Inflamación/tratamiento farmacológico , Oxigenasas de Función Mixta/antagonistas & inhibidores , Monocitos/efectos de los fármacos , Inhibidores de Prolil-Hidroxilasa/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Células Cultivadas , Células HEK293 , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Oxigenasas de Función Mixta/inmunología , Oxigenasas de Función Mixta/metabolismo , Monocitos/inmunología , Monocitos/metabolismo
9.
J Allergy Clin Immunol ; 146(4): 706-720, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32841652

RESUMEN

Metabolic inflammation (metaflammation) is characteristic of obesity-related metabolic disorders, associated with increased risk of development of type 2 diabetes, nonalcoholic fatty liver disease (NAFLD), or cardiovascular disease. Metaflammation refers to a chronic, low-grade systemic inflammation as opposed to the classical transient and acute inflammatory responses of the innate immune system. Metaflammation is driven by a range of adverse dietary factors, including saturated fatty acids and some sugars, suggesting that certain dietary triggers may be particularly relevant beyond simple excessive dietary intake presenting as obesity. Importantly, obese patients with diabetes have a higher risk of infection and display gut microbiota profiles characteristic of dysfunctional immunity. Targeting metaflammation has also emerged as a strategy to attenuate metabolic disease. In this review we explore how different nutrition interventions may reconfigure disrupted metabolic inflammation in type 2 diabetes and nonalcoholic fatty liver disease by reestablishing a conventional proinflammatory program in innate immune cells and/or correcting dysbiosis to dampen systemic inflammation. We begin by reviewing concepts of metabolic inflammation relating to IL-1ß inflammation and how it is induced by dietary and/or metabolic stressors. We then explore whether and how dietary interventions may attenuate processes pertaining to metaflammation, either directly or indirectly via the microbiome. Hence, we hope to bring new perspectives to alleviate the metaflammation typifying metabolic disease.


Asunto(s)
Susceptibilidad a Enfermedades , Metabolismo Energético , Inflamación/etiología , Inflamación/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Enfermedades Cardiovasculares/etiología , Diabetes Mellitus Tipo 2/etiología , Dieta , Humanos , Inflamación/diagnóstico , Enfermedad del Hígado Graso no Alcohólico/etiología , Estado Nutricional , Receptores Toll-Like/metabolismo
10.
Br J Nutr ; 124(3): 247-255, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32122411

RESUMEN

Preliminary evidence has suggested that high-fat diets (HFD) enriched with SFA, but not MUFA, promote hyperinsulinaemia and pancreatic hypertrophy with insulin resistance. The objective of this study was to determine whether the substitution of dietary MUFA within a HFD could attenuate the progression of pancreatic islet dysfunction seen with prolonged SFA-HFD. For 32 weeks, C57BL/6J mice were fed either: (1) low-fat diet, (2) SFA-HFD or (3) SFA-HFD for 16 weeks, then switched to MUFA-HFD for 16 weeks (SFA-to-MUFA-HFD). Fasting insulin was assessed throughout the study; islets were isolated following the intervention. Substituting SFA with MUFA-HFD prevented the progression of hyperinsulinaemia observed in SFA-HFD mice (P < 0·001). Glucose-stimulated insulin secretion from isolated islets was reduced by SFA-HFD, yet not fully affected by SFA-to-MUFA-HFD. Markers of ß-cell identity (Ins2, Nkx6.1, Ngn3, Rfx6, Pdx1 and Pax6) were reduced, and islet inflammation was increased (IL-1ß, 3·0-fold, P = 0·007; CD68, 2·9-fold, P = 0·001; Il-6, 1·1-fold, P = 0·437) in SFA-HFD - effects not seen with SFA-to-MUFA-HFD. Switching to MUFA-HFD can partly attenuate the progression of SFA-HFD-induced hyperinsulinaemia, pancreatic inflammation and impairments in ß-cell function. While further work is required from a mechanistic perspective, dietary fat may mediate its effect in an IL-1ß-AMP-activated protein kinase α1-dependent fashion. Future work should assess the potential translation of the modulation of metabolic inflammation in man.


Asunto(s)
Dieta Alta en Grasa/métodos , Grasas de la Dieta/farmacología , Ácidos Grasos Monoinsaturados/farmacología , Ácidos Grasos/farmacología , Hiperinsulinismo/dietoterapia , Animales , Modelos Animales de Enfermedad , Resistencia a la Insulina/fisiología , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas/efectos de los fármacos
11.
Public Health Nutr ; 23(13): 2373-2383, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31829294

RESUMEN

OBJECTIVE: To apply a dietary modelling approach to investigate the impact of substituting beef intakes with three types of alternative fatty acid (FA) composition of beef on population dietary fat intakes. DESIGN: Cross-sectional, national food consumption survey - the National Adult Nutrition Survey (NANS). The fat content of the beef-containing food codes (n 52) and recipes (n 99) were updated with FA composition data from beef from animals receiving one of three ruminant dietary interventions: grass-fed (GRASS), grass finished on grass silage and concentrates (GSC) or concentrate-fed (CONC). Mean daily fat intakes, adherence to dietary guidelines and the impact of altering beef FA composition on dietary fat sources were characterised. SETTING: Ireland. PARTICIPANTS: Beef consumers (n 1044) aged 18-90 years. RESULTS: Grass-based feeding practices improved dietary intakes of a number of individual FA, wherein myristic acid (C14 : 0) and palmitic acid (C16 : 0) were decreased, with an increase in conjugated linoleic acid (C18 : 2c9,t11) and trans-vaccenic acid (C18 : 1t11; P < 0·05). Improved adherence with dietary recommendations for total fat (98·5 %), SFA (57·4 %) and PUFA (98·8 %) was observed in the grass-fed beef scenario (P < 0·001). Trans-fat intakes were increased significantly in the grass-fed beef scenario (P < 0·001). CONCLUSIONS: To the best of our knowledge, the present study is the first to characterise the impact of grass-fed beef consumption at population level. The study suggests that habitual consumption of grass-fed beef may have potential as a public health strategy to improve dietary fat quality.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Ácidos Grasos/análisis , Carne Roja/análisis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Alimentación Animal , Animales , Bovinos , Estudios Transversales , Dieta , Humanos , Irlanda , Persona de Mediana Edad , Adulto Joven
12.
Scand J Clin Lab Invest ; 80(4): 282-290, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32134347

RESUMEN

Impaired insulin secretion and action are important for development of type 2 diabetes (T2D) and metabolic syndrome (MetS). Despite recognized heterogeneity of these glucometabolic disorders, few data are available of biological variation in insulin secretion and action among individuals with T2D and MetS. The aim of this study was to explore the inter-individual variations using gold standard methods in a cross-sectional study of two independent cohorts of phenotypically well-characterized subjects. Cohort I included 486 subjects with MetS, and cohort II 62 subjects with established T2D. First phase insulin secretion was defined as the incremental area under the curve 0-8 min (iAUC0-8 min) during an intravenous glucose tolerance test (IVGTT). Insulin sensitivity was measured as the insulin sensitivity index (SI) modelled from IVGTT in cohort I, and in II as total glucose disposal (TGD) estimated from a euglycaemic-hyperinsulinaemic clamp. Variation is given as total range and, fold-variation between 5%- and 95%-percentile. The iAUC0-8 min ranged from -60 to 3397 mUL-1min-1 among subjects with MetS and from -263 to 1194 mUL-1min-1 in subjects with T2D, representing a more than 10-fold variation. Insulin sensitivity ranged from SI 0.19 to 15.29 (mU/L)-1min-1 among subjects with MetS and TGD 12.9-101.6 µmolkgFFM-1min-1 in subjects with T2D, representing a 6.8 and 5.5-fold variation, respectively. The other components of MetS; BMI, waist-hip ratio, HDL-cholesterol, triglycerides and blood pressure (BP), showed a 1.4-4.7-fold variation. In conclusion, our data demonstrated extensive inter-individual variations in insulin secretion and sensitivity. These variations may be essential to take into account when planning clinical research and treatment in subjects with T2D and MetS.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Secreción de Insulina , Insulina/sangre , Síndrome Metabólico/sangre , Adulto , Anciano , Área Bajo la Curva , Variación Biológica Individual , Presión Sanguínea/fisiología , Índice de Masa Corporal , HDL-Colesterol/sangre , Estudios Transversales , Diabetes Mellitus Tipo 2/patología , Femenino , Técnica de Clampeo de la Glucosa/métodos , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Masculino , Síndrome Metabólico/patología , Persona de Mediana Edad , Triglicéridos/sangre , Relación Cintura-Cadera
13.
Annu Rev Nutr ; 37: 77-102, 2017 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-28826373

RESUMEN

Worldwide obesity rates have reached epidemic proportions and significantly contribute to the growing prevalence of metabolic diseases. Chronic low-grade inflammation, a hallmark of obesity, involves immune cell infiltration into expanding adipose tissue. In turn, obesity-associated inflammation can lead to complications in other metabolic tissues (e.g., liver, skeletal muscle, pancreas) through lipotoxicity and inflammatory signaling networks. Importantly, although numerous signaling pathways are known to integrate metabolic and inflammatory processes, the nucleotide-binding and oligomerization domain-like receptor, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome is now noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome can be influenced by various metabolites, including fatty acids. Specifically, although saturated fatty acids may promote NLRP3 inflammasome activation, monounsaturated fatty acids and polyunsaturated fatty acids have recently been shown to impede NLRP3 activity. Therefore, the NLRP3 inflammasome and associated metabolic inflammation have key roles in the relationships among fatty acids, metabolites, and metabolic disease. This review focuses on the ability of fatty acids to influence inflammation and the NLRP3 inflammasome across numerous metabolic tissues in the body. In addition, we explore some perspectives for the future, wherein recent work in the immunology field clearly demonstrates that metabolic reprogramming defines immune cell functionality. Although there is a paucity of information about how diet and fatty acids modulate this process, it is possible that this will open up a new avenue of research relating to nutrient-sensitive metabolic inflammation.


Asunto(s)
Ácidos Grasos , Inflamasomas/inmunología , Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Animales , Humanos , Inflamación/patología , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología
14.
Int J Mol Sci ; 19(10)2018 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-30304866

RESUMEN

Nutritional status provides metabolic substrates to activate AMP-Activated Protein Kinase (AMPK), the energy sensor that regulates metabolism. Recent evidence has demonstrated that AMPK has wider functions with respect to regulating immune cell metabolism and function. One such example is the regulatory role that AMPK has on NLRP3-inlflammasome and IL-1ß biology. This in turn can result in subsequent negative downstream effects on glucose, lipid and insulin metabolism. Nutrient stress in the form of obesity can impact AMPK and whole-body metabolism, leading to complications such as type 2 diabetes and cancer risk. There is a lack of data regarding the nature and extent that nutrient status has on AMPK and metabolic-inflammation. However, emerging work elucidates to a direct role of individual nutrients on AMPK and metabolic-inflammation, as a possible means of modulating AMPK activity. The posit being to use such nutritional agents to re-configure metabolic-inflammation towards more oxidative phosphorylation and promote the resolution of inflammation. The complex paradigm will be discussed within the context of if/how dietary components, nutrients including fatty acids and non-nutrient food components, such as resveratrol, berberine, curcumin and the flavonoid genistein, modulate AMPK dependent processes relating to inflammation and metabolism.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético , Inflamación/etiología , Inflamación/metabolismo , Fenómenos Fisiológicos de la Nutrición , Animales , Biomarcadores , Activación Enzimática , Humanos , Resistencia a la Insulina , Neoplasias/etiología , Neoplasias/metabolismo , Estado Nutricional , Transducción de Señal
15.
J Proteome Res ; 16(3): 1280-1287, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28145113

RESUMEN

Epidemiology and clinical studies provide clear evidence of the complex links between diet and health. To understand these links, reliable dietary assessment methods are pivotal. Biomarkers have emerged as more objective measures of intake compared with traditional dietary assessment methods. However, there are only a limited number of putative biomarkers of intake successfully identified and validated. The use of biomarkers that reflect food intake to examine diet related diseases represents the next step in biomarker research. Therefore, the aim of this study was to (1) identify and confirm biomarkers associated with dietary fat intake and (2) examine the relationship between those biomarkers with health parameters. Heatmap analysis identified a panel of 22 lipid biomarkers associated with total dietary fat intake in the Metabolic Challenge (MECHE) Study. Confirmation of four of these biomarkers demonstrated responsiveness to different levels of fat intake in a separate intervention study (NutriTech study). Linear regression identified a significant relationship between the panel of dietary fat biomarkers and HOMA-IR, with three lipid biomarkers (C16, PCaaC36:2, and PCae36:4) demonstrating significant associations. Identifying such links allows us to explore the relationship between diet and health to determine whether these biomarkers can be modulated through diet to improve health outcomes.


Asunto(s)
Dieta , Salud , Lípidos/sangre , Adolescente , Adulto , Biomarcadores/sangre , Estudios de Cohortes , Grasas de la Dieta/sangre , Ingestión de Alimentos , Femenino , Humanos , Irlanda , Masculino , Persona de Mediana Edad , Adulto Joven
16.
J Proteome Res ; 16(11): 4122-4133, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-28950061

RESUMEN

Validated protein biomarkers are needed for assessing health trajectories, predicting and subclassifying disease, and optimizing diagnostic and therapeutic clinical decision-making. The sensitivity, specificity, accuracy, and precision of single or combinations of protein biomarkers may be altered by differences in physiological states limiting the ability to translate research results to clinically useful diagnostic tests. Aptamer based affinity assays were used to test whether low abundant serum proteins differed based on age, sex, and fat mass in a healthy population of 94 males and 102 females from the MECHE cohort. The findings were replicated in 217 healthy male and 377 healthy female participants in the DiOGenes consortium. Of the 1129 proteins in the panel, 141, 51, and 112 proteins (adjusted p < 0.1) were identified in the MECHE cohort and significantly replicated in DiOGenes for sexual dimorphism, age, and fat mass, respectively. Pathway analysis classified a subset of proteins from the 3 phenotypes to the complement and coagulation cascades pathways and to immune and coagulation processes. These results demonstrated that specific proteins were statistically associated with dichotomous (male vs female) and continuous phenotypes (age, fat mass), which may influence the identification and use of biomarkers of clinical utility for health diagnosis and therapeutic strategies.


Asunto(s)
Fenotipo , Proteómica/métodos , Tejido Adiposo , Factores de Edad , Femenino , Humanos , Masculino , Caracteres Sexuales
17.
Circulation ; 133(19): 1838-50, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27081117

RESUMEN

BACKGROUND: Acute inflammation impairs reverse cholesterol transport (RCT) and reduces high-density lipoprotein (HDL) function in vivo. This study hypothesized that obesity-induced inflammation impedes RCT and alters HDL composition, and investigated if dietary replacement of saturated (SFA) for monounsaturated (MUFA) fatty acids modulates RCT. METHODS AND RESULTS: Macrophage-to-feces RCT, HDL efflux capacity, and HDL proteomic profiling was determined in C57BL/6j mice following 24 weeks on SFA- or MUFA-enriched high-fat diets (HFDs) or low-fat diet. The impact of dietary SFA consumption and insulin resistance on HDL efflux function was also assessed in humans. Both HFDs increased plasma (3)H-cholesterol counts during RCT in vivo and ATP-binding cassette, subfamily A, member 1-independent efflux to plasma ex vivo, effects that were attributable to elevated HDL cholesterol. By contrast, ATP-binding cassette, subfamily A, member 1-dependent efflux was reduced after both HFDs, an effect that was also observed with insulin resistance and high SFA consumption in humans. SFA-HFD impaired liver-to-feces RCT, increased hepatic inflammation, and reduced ABC subfamily G member 5/8 and ABC subfamily B member 11 transporter expression in comparison with low-fat diet, whereas liver-to-feces RCT was preserved after MUFA-HFD. HDL particles were enriched with acute-phase proteins (serum amyloid A, haptoglobin, and hemopexin) and depleted of paraoxonase-1 after SFA-HFD in comparison with MUFA-HFD. CONCLUSIONS: Ex vivo efflux assays validated increased macrophage-to-plasma RCT in vivo after both HFDs but failed to capture differential modulation of hepatic cholesterol trafficking. By contrast, proteomics revealed the association of hepatic-derived inflammatory proteins on HDL after SFA-HFD in comparison with MUFA-HFD, which reflected differential hepatic cholesterol trafficking between groups. Acute-phase protein levels on HDL may serve as novel biomarkers of impaired liver-to-feces RCT in vivo.


Asunto(s)
Colesterol/metabolismo , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos Monoinsaturados/administración & dosificación , Ácidos Grasos/administración & dosificación , Lipoproteínas HDL/genética , Proteómica/métodos , Adolescente , Adulto , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Ácidos Grasos/efectos adversos , Ácidos Grasos Monoinsaturados/efectos adversos , Femenino , Humanos , Lipoproteínas HDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Obesidad/etiología , Obesidad/metabolismo , Adulto Joven
18.
Eur Respir J ; 49(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28424360

RESUMEN

Obstructive sleep apnoea (OSA) is increasingly associated with insulin resistance. The underlying pathophysiology remains unclear but intermittent hypoxia (IH)-mediated inflammation and subsequent dysfunction of the adipose tissue has been hypothesised to play a key role.We tested this hypothesis employing a comprehensive translational approach using a murine IH model of lean and diet-induced obese mice, an innovative IH system for cell cultures and a tightly controlled patient cohort.IH led to the development of insulin resistance in mice, corrected for the degree of obesity, and reduced insulin-mediated glucose uptake in 3T3-L1 adipocytes, associated with inhibition of the insulin-signalling pathway and downregulation of insulin-receptor substrate-1 mRNA. Providing mechanistic insight, IH induced a pro-inflammatory phenotype of visceral adipose tissue in mice with pro-inflammatory M1 macrophage polarisation correlating with the severity of insulin resistance. Complimentary in vitro analysis demonstrated that IH led to M1 polarisation of THP1-derived macrophages. In subjects without comorbidities (n=186), OSA was independently associated with insulin resistance. Furthermore, we found an independent correlation of OSA severity with the M1 macrophage inflammatory marker sCD163.This study provides evidence that IH induces a pro-inflammatory phenotype of the adipose tissue, which may be a crucial link between OSA and the development of insulin resistance.


Asunto(s)
Hipoxia/metabolismo , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Obesidad/complicaciones , Apnea Obstructiva del Sueño/metabolismo , Células 3T3-L1 , Adulto , Animales , Humanos , Inflamación/metabolismo , Insulina/metabolismo , Grasa Intraabdominal/metabolismo , Modelos Lineales , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Distribución Aleatoria , Apnea Obstructiva del Sueño/fisiopatología
19.
Biochem Soc Trans ; 45(4): 979-985, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28710289

RESUMEN

Metabolic inflammation is a very topical area of research, wherein aberrations in metabolic and inflammatory pathways probably contribute to atherosclerosis, insulin resistance (IR) and type 2 diabetes. Metabolic insults arising from obesity promote inflammation, which in turn impedes insulin signalling and reverse cholesterol transport (RCT). Key cells in the process are metabolically activated macrophages, which up-regulate both pro- and anti-inflammatory pathways in response to lipid spillover from adipocytes. Peroxisome proliferator-activated receptors and AMP-activated protein kinase (AMPK) are regulators of cellular homeostasis that influence both inflammatory and metabolic pathways. Dietary fats, such as saturated fatty acids (SFAs), can differentially modulate metabolic inflammation. Palmitic acid, in particular, is a well-characterized nutrient that promotes metabolic inflammation via the NLRP3 (the nod-like receptor containing a pyrin domain) inflammasome, which is partly attributable to AMPK inhibition. Conversely, some unsaturated fatty acids are less potent agonists of metabolic inflammation. For example, monounsaturated fatty acid does not reduce AMPK as potently as SFA and n-3 polyunsaturated fatty acids actively resolve inflammation via resolvins and protectins. Nevertheless, the full extent to which nutritional state modulates metabolic inflammation requires greater clarification.


Asunto(s)
Aterosclerosis/etiología , Diabetes Mellitus Tipo 2/etiología , Dieta/efectos adversos , Resistencia a la Insulina , Modelos Inmunológicos , Obesidad/etiología , Adipocitos/inmunología , Adipocitos/metabolismo , Adipocitos/patología , Animales , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología
20.
Br J Nutr ; 118(3): 222-228, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28831958

RESUMEN

Evidence suggests that processed red meat consumption is a risk factor for CVD and type 2 diabetes (T2D). This analysis investigates the association between dietary patterns, their processed red meat contributions, and association with blood biomarkers of CVD and T2D, in 786 Irish adults (18-90 years) using cross-sectional data from a 2011 national food consumption survey. All meat-containing foods consumed were assigned to four food groups (n 502) on the basis of whether they contained red or white meat and whether they were processed or unprocessed. The remaining foods (n 2050) were assigned to twenty-nine food groups. Two-step and k-means cluster analyses were applied to derive dietary patterns. Nutrient intakes, plasma fatty acids and biomarkers of CVD and T2D were assessed. A total of four dietary patterns were derived. In comparison with the pattern with lower contributions from processed red meat, the dietary pattern with greater processed red meat intakes presented a poorer Alternate Healthy Eating Index (21·2 (sd 7·7)), a greater proportion of smokers (29 %) and lower plasma EPA (1·34 (sd 0·72) %) and DHA (2·21 (sd 0·84) %) levels (P<0·001). There were no differences in classical biomarkers of CVD and T2D, including serum cholesterol and insulin, across dietary patterns. This suggests that the consideration of processed red meat consumption as a risk factor for CVD and T2D may need to be re-assessed.


Asunto(s)
Enfermedades Cardiovasculares/epidemiología , Diabetes Mellitus Tipo 2/epidemiología , Dieta , Manipulación de Alimentos , Síndrome Metabólico/epidemiología , Carne Roja/efectos adversos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Glucemia/metabolismo , Enfermedades Cardiovasculares/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Estudios de Cohortes , Estudios Transversales , Diabetes Mellitus Tipo 2/sangre , Ácidos Docosahexaenoicos/sangre , Ácido Eicosapentaenoico/sangre , Ácidos Grasos/sangre , Femenino , Humanos , Insulina/sangre , Masculino , Síndrome Metabólico/sangre , Persona de Mediana Edad , Evaluación Nutricional , Encuestas Nutricionales , Factores de Riesgo , Factores Socioeconómicos , Triglicéridos/sangre , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA