Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 223: 116171, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38552854

RESUMEN

Upper-body adiposity is adversely associated with metabolic health whereas the opposite is observed for the lower-body. The neck is a unique upper-body fat depot in adult humans, housing thermogenic brown adipose tissue (BAT), which is increasingly recognised to influence whole-body metabolic health. Loss of BAT, concurrent with replacement by white adipose tissue (WAT), may contribute to metabolic disease, and specific accumulation of neck fat is seen in certain conditions accompanied by adverse metabolic consequences. Yet, few studies have investigated the relationships between neck fat mass (NFM) and cardiometabolic risk, and the influence of sex and metabolic status. Typically, neck circumference (NC) is used as a proxy for neck fat, without considering other determinants of NC, including variability in neck lean mass. In this study we develop and validate novel methods to quantify NFM using dual x-ray absorptiometry (DEXA) imaging, and subsequently investigate the associations of NFM with metabolic biomarkers across approximately 7000 subjects from the Oxford BioBank. NFM correlated with systemic insulin resistance (Homeostatic Model Assessment for Insulin Resistance; HOMA-IR), low-grade inflammation (plasma high-sensitivity C-Reactive Protein; hsCRP), and metabolic markers of adipose tissue function (plasma triglycerides and non-esterified fatty acids; NEFA). NFM was higher in men than women, higher in type 2 diabetes mellitus compared with non-diabetes, after adjustment for total body fat, and also associated with overall cardiovascular disease risk (calculated QRISK3 score). This study describes the development of methods for accurate determination of NFM at scale and suggests a specific relationship between NFM and adverse metabolic health.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Adulto , Masculino , Humanos , Femenino , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Factores de Riesgo , Tejido Adiposo , Obesidad/metabolismo , Tejido Adiposo Pardo/diagnóstico por imagen , Tejido Adiposo Pardo/metabolismo
2.
Adipocyte ; 12(1): 2179339, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36763512

RESUMEN

While there is no standardized protocol for the differentiation of human adipocytes in culture, common themes exist in the use of supra-physiological glucose and hormone concentrations, and an absence of exogenous fatty acids. These factors can have detrimental effects on some aspects of adipogenesis and adipocyte function. Here, we present methods for modifying the adipogenic differentiation protocol to overcome impaired glucose uptake and insulin signalling in human adipose-derived stem cell lines derived from the stromal vascular fraction of abdominal and gluteal subcutaneous adipose tissue. By reducing the length of exposure to adipogenic hormones, in combination with a physiological glucose concentration (5 mM), and the provision of exogenous fatty acids (reflecting typical dietary fatty acids), we were able to restore early insulin signalling events and glucose uptake, which were impaired by extended use of hormones and a high glucose concentration, respectively. Furthermore, the addition of exogenous fatty acids greatly increased the storage of triglycerides and removed the artificial demand to synthesize all fatty acids by de novo lipogenesis. Thus, modifying the adipogenic cocktail can enhance functional aspects of human adipocytes in vitro and is an important variable to consider prior to in vitro investigations into adipocyte biology.


Asunto(s)
Adipogénesis , Insulina , Humanos , Adipogénesis/fisiología , Insulina/metabolismo , Diferenciación Celular , Técnicas de Cultivo de Célula , Ácidos Grasos , Glucosa , Tejido Adiposo/metabolismo
3.
Cell Rep ; 40(4): 111136, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35905723

RESUMEN

Mechanisms governing regional human adipose tissue (AT) development remain undefined. Here, we show that the long non-coding RNA HOTAIR (HOX transcript antisense RNA) is exclusively expressed in gluteofemoral AT, where it is essential for adipocyte development. We find that HOTAIR interacts with polycomb repressive complex 2 (PRC2) and we identify core HOTAIR-PRC2 target genes involved in adipocyte lineage determination. Repression of target genes coincides with PRC2 promoter occupancy and H3K27 trimethylation. HOTAIR is also involved in modifying the gluteal adipocyte transcriptome through alternative splicing. Gluteal-specific expression of HOTAIR is maintained by defined regions of open chromatin across the HOTAIR promoter. HOTAIR expression levels can be modified by hormonal (estrogen, glucocorticoids) and genetic variation (rs1443512 is a HOTAIR eQTL associated with reduced gynoid fat mass). These data identify HOTAIR as a dynamic regulator of the gluteal adipocyte transcriptome and epigenome with functional importance for human regional AT development.


Asunto(s)
Complejo Represivo Polycomb 2 , ARN Largo no Codificante/genética , Cromatina , Estrógenos , Humanos , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante/metabolismo , Transcriptoma/genética
4.
Eur J Endocrinol ; 186(3): 367-377, 2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35038311

RESUMEN

OBJECTIVE: Metformin is a first-line pharmacotherapy in the treatment of type 2 diabetes, a condition closely associated with non-alcoholic fatty liver disease (NAFLD). Although metformin promotes weight loss and improves insulin sensitivity, its effect on intrahepatic triglyceride (IHTG) remains unclear. We investigated the effect of metformin on IHTG, hepatic de novo lipogenesis (DNL), and fatty acid (FA) oxidation in vivo in humans. DESIGN AND METHODS: Metabolic investigations, using stable-isotope tracers, were performed in ten insulin-resistant, overweight/obese human participants with NAFLD who were treatment naïve before and after 12 weeks of metformin treatment. The effect of metformin on markers of s.c. adipose tissue FA metabolism and function, along with the plasma metabolome, was investigated. RESULTS: Twelve weeks of treatment with metformin resulted in a significant reduction in body weight and improved insulin sensitivity, but IHTG content and FA oxidation remained unchanged. Metformin treatment was associated with a significant decrease in VLDL-triglyceride (TG) concentrations and a significant increase in the relative contribution of DNL-derived FAs to VLDL-TG. There were subtle and relatively few changes in s.c. adipose tissue FA metabolism and the plasma metabolome with metformin treatment. CONCLUSIONS: We demonstrate the mechanisms of action of metformin whereby it improves insulin sensitivity and promotes weight loss, without improvement in IHTG; these observations are partly explained through increased hepatic DNL and a lack of change in FA oxidation.


Asunto(s)
Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina/fisiología , Lipogénesis/fisiología , Hígado/metabolismo , Metformina/uso terapéutico , Triglicéridos/metabolismo , Adulto , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Estudios de Cohortes , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Hipoglucemiantes/farmacología , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Metformina/farmacología , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Sobrepeso/tratamiento farmacológico , Sobrepeso/metabolismo
5.
J Vis Exp ; (170)2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33938882

RESUMEN

Extracellular vesicles (EVs) are lipid enclosed envelopes that carry biologically active material such as proteins, RNA, metabolites and lipids. EVs can modulate the cellular status of other cells locally in tissue microenvironments or through liberation into peripheral blood. Adipocyte-derived EVs are elevated in the peripheral blood and show alterations in their cargo (RNA and protein) during metabolic disturbances, including obesity and diabetes. Adipocyte-derived EVs can regulate the cellular status of neighboring vascular cells, such as endothelial cells and adipose tissue resident macrophages to promote adipose tissue inflammation. Investigating alterations in adipocyte-derived EVs in vivo is complex because EVs derived from peripheral blood are highly heterogenous and contain EVs from other sources, namely platelets, endothelial cells, erythrocytes and muscle. Therefore, the culture of human adipocytes provides a model system for the study of adipocyte derived EVs. Here, we provide a detailed protocol for the extraction of total small EVs from cell culture media of human gluteal and abdominal adipocytes using filtration and ultracentrifugation. We further demonstrate the use of Nanoparticle Tracking Analysis (NTA) for quantification of EV size and concentration and show the presence of EV-protein tumor susceptibility gene 101 (TSG101) in the gluteal and abdominal adipocyte derived-EVs. Isolated EVs from this protocol can be used for downstream analysis, including transmission electron microscopy, proteomics, metabolomics, small RNA-sequencing, microarrays and can be utilized in functional in vitro/in vivo studies.


Asunto(s)
Tejido Adiposo/citología , Vesículas Extracelulares , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Vesículas Extracelulares/metabolismo , Filtración , Humanos , Factores de Transcripción/metabolismo , Ultracentrifugación
6.
Physiol Rep ; 8(13): e14482, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32643289

RESUMEN

BACKGROUND AND AIMS: Nonalcoholic fatty liver disease (NAFLD) begins with steatosis, where a mixed macrovesicular pattern of large and small lipid droplets (LDs) develops. Since in vitro models recapitulating this are limited, the aims of this study were to develop mixed macrovesicular steatosis in immortalized hepatocytes and investigate effects on intracellular metabolism by altering nutritional substrates. METHODS: Huh7 cells were cultured in 11 mM glucose and 2% human serum (HS) for 7 days before additional sugars and fatty acids (FAs), either with 200 µM FAs (low fat low sugar; LFLS), 5.5 mM fructose + 200 µM FAs (low fat high sugar; LFHS), or 5.5 mM fructose + 800 µM FAs (high fat high sugar; HFHS), were added for 7 days. FA metabolism, lipid droplet characteristics, and transcriptomic signatures were investigated. RESULTS: Between the LFLS and LFHS conditions, there were few notable differences. In the HFHS condition, intracellular triacylglycerol (TAG) was increased and the LD pattern and distribution was similar to that found in primary steatotic hepatocytes. HFHS-treated cells had lower levels of de novo-derived FAs and secreted larger, TAG-rich lipoprotein particles. RNA sequencing and gene set enrichment analysis showed changes in several pathways including those involved in metabolism and cell cycle. CONCLUSIONS: Repeated doses of HFHS treatment resulted in a cellular model of NAFLD with a mixed macrovesicular LD pattern and metabolic dysfunction. Since these nutrients have been implicated in the development of NAFLD in humans, the model provides a good physiological basis for studying NAFLD development or regression in vitro.


Asunto(s)
Ácidos Grasos/metabolismo , Glucosa/metabolismo , Hepatocitos/metabolismo , Gotas Lipídicas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Línea Celular Tumoral , Células Cultivadas , Hepatocitos/patología , Humanos , Gotas Lipídicas/patología , Enfermedad del Hígado Graso no Alcohólico/genética , Transcriptoma
7.
Nat Commun ; 11(1): 2797, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32493999

RESUMEN

Fat distribution is an independent cardiometabolic risk factor. However, its molecular and cellular underpinnings remain obscure. Here we demonstrate that two independent GWAS signals at RSPO3, which are associated with increased body mass index-adjusted waist-to-hip ratio, act to specifically increase RSPO3 expression in subcutaneous adipocytes. These variants are also associated with reduced lower-body fat, enlarged gluteal adipocytes and insulin resistance. Based on human cellular studies RSPO3 may limit gluteofemoral adipose tissue (AT) expansion by suppressing adipogenesis and increasing gluteal adipocyte susceptibility to apoptosis. RSPO3 may also promote upper-body fat distribution by stimulating abdominal adipose progenitor (AP) proliferation. The distinct biological responses elicited by RSPO3 in abdominal versus gluteal APs in vitro are associated with differential changes in WNT signalling. Zebrafish carrying a nonsense rspo3 mutation display altered fat distribution. Our study identifies RSPO3 as an important determinant of peripheral AT storage capacity.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Distribución de la Grasa Corporal , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trombospondinas/metabolismo , Proteínas de Pez Cebra/metabolismo , Adipocitos/efectos de los fármacos , Tejido Adiposo/metabolismo , Adiposidad/genética , Adulto , Alelos , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Doxiciclina/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Persona de Mediana Edad , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Caracteres Sexuales , Células Madre/metabolismo , Trombospondinas/genética , Relación Cintura-Cadera , Vía de Señalización Wnt/efectos de los fármacos , Pez Cebra/genética , Proteínas de Pez Cebra/genética
8.
Br J Cancer ; 122(2): 258-265, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819193

RESUMEN

BACKGROUND: Epidemiological studies suggest that metformin may reduce the incidence of cancer in patients with diabetes and multiple late phase clinical trials assessing the potential of repurposing this drug are underway. Transcriptomic profiling of tumour samples is an excellent tool to understand drug bioactivity, identify candidate biomarkers and assess for mechanisms of resistance to therapy. METHODS: Thirty-six patients with untreated primary breast cancer were recruited to a window study and transcriptomic profiling of tumour samples carried out before and after metformin treatment. RESULTS: Multiple genes that regulate fatty acid oxidation were upregulated at the transcriptomic level and there was a differential change in expression between two previously identified cohorts of patients with distinct metabolic responses. Increase in expression of a mitochondrial fatty oxidation gene composite signature correlated with change in a proliferation gene signature. In vitro assays showed that, in contrast to previous studies in models of normal cells, metformin reduces fatty acid oxidation with a subsequent accumulation of intracellular triglyceride, independent of AMPK activation. CONCLUSIONS: We propose that metformin at clinical doses targets fatty acid oxidation in cancer cells with implications for patient selection and drug combinations. CLINICAL TRIAL REGISTRATION: NCT01266486.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Ácidos Grasos/metabolismo , Metformina/farmacología , Proteínas Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Transcriptoma/efectos de los fármacos
9.
Int J Obes (Lond) ; 43(12): 2593, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31641215

RESUMEN

We erroneously published the original Article with an incorrect Copyright line. This has been updated in the XML, PDF and HTML versions of this Article.

10.
Int J Obes (Lond) ; 43(12): 2458-2468, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31324879

RESUMEN

BACKGROUND: Bone morphogenetic proteins (BMPs) regulate adipogenesis but it is not clear whether they influence regional adipose tissue (AT) development in humans. OBJECTIVE: To characterise BMP2 expression, BMP2-SMAD1/5/8 signalling, and BMP2's potential effect on proliferation and adipogenesis in human subcutaneous abdominal and gluteal AT and its constituent preadipocytes. METHODS: BMP2 expression was measured in whole AT and immortalised preadipocytes via qPCR and Western blot; secreted/circulating BMP2 was measured by ELISA. The effect of BMP2 on preadipocyte proliferation was evaluated using a fluorescent assay. BMP2's effect on adipogenesis in immortalised preadipocytes was determined via qPCR of adipogenic markers and cellular triacylglycerol (TAG) accumulation. BMP2-SMAD1/5/8 signalling was assessed in immortalised preadipocytes via Western blot and qPCR of ID1 expression. RESULTS: BMP2 was expressed and released by abdominal and gluteal AT and preadipocytes. Exogenous BMP2 dose dependently promoted adipogenesis in abdominal preadipocytes only; 50 ng/ml BMP2 increased PPARG2 expression (10-fold compared to vehicle, p < 0.001) and TAG accumulation (3-fold compared to vehicle; p < 0.001). BMP2 stimulated SMAD1/5/8 phosphorylation and ID1 expression in abdominal and gluteal preadipocytes but this was blocked by 500 nM K02288, a type 1 BMP receptor inhibitor (p < 0.001). Co-administration of 500 nM K02288 also inhibited the pro-adipogenic effect of 50 ng/ml BMP2 in abdominal cells; >90% inhibition of TAG accumulation (p < 0.001) and ~50% inhibition of PPARG2 expression (p < 0.001). The endogenous iron regulator erythroferrone reduced BMP2-SMAD1/5/8 signalling by ~30% specifically in subcutaneous abdominal preadipocytes (p < 0.01), suggesting it plays a role in restricting the expansion of the body's largest AT depot during energy deficiency. Additionally, a waist-hip ratio-increasing common polymorphism near BMP2 is an eQTL associated with ~15% lower BMP2 expression in abdominal and gluteal AT (p < 0.05) as well as altered adipocyte size in male abdominal AT (p < 0.05). CONCLUSIONS: These data implicate BMP2-SMAD1/5/8 signalling in depot-specific preadipocyte development and abdominal AT expansion in humans.


Asunto(s)
Adipogénesis/fisiología , Proteína Morfogenética Ósea 2/metabolismo , Transducción de Señal/fisiología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Adulto , Anciano , Índice de Masa Corporal , Proliferación Celular/fisiología , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas Smad Reguladas por Receptores/metabolismo
11.
EBioMedicine ; 44: 467-475, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31151930

RESUMEN

BACKGROUND: Abdominal fat mass is associated with metabolic risk whilst gluteal femoral fat is paradoxically protective. MicroRNAs are known to be necessary for adipose tissue formation and function but their role in regulating human fat distribution remains largely unexplored. METHODS: An initial microarray screen of abdominal subcutaneous and gluteal adipose tissue, with validatory qPCR, identified microRNA-196a as being strongly differentially expressed between gluteal and abdominal subcutaneous adipose tissue. FINDINGS: We found that rs11614913, a SNP within pre-miR-196a-2 at the HOXC locus, is an eQTL for miR-196a expression in abdominal subcutaneous adipose tissue (ASAT). Observations in large cohorts showed that rs11614913 increased waist-to-hip ratio, which was driven specifically by an expansion in ASAT. In further experiments, rs11614913 was associated with adipocyte size. Functional studies and transcriptomic profiling of miR-196a knock-down pre-adipocytes revealed a role for miR-196a in regulating pre-adipocyte proliferation and extracellular matrix pathways. INTERPRETATION: These data identify a role for miR-196a in regulating human body fat distribution. FUND: This work was supported by the Medical Research Council and Novo Nordisk UK Research Foundation (G1001959) and Swedish Research Council. We acknowledge the OBB-NIHR Oxford Biomedical Research Centre and the British Heart Foundation (BHF) (RG/17/1/32663). Work performed at the MRC Epidemiology Unit was funded by the United Kingdom's Medical Research Council through grants MC_UU_12015/1, MC_PC_13046, MC_PC_13048 and MR/L00002/1.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Adipocitos/metabolismo , Adulto , Alelos , Línea Celular , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Interferencia de ARN , Transducción de Señal , Transcriptoma
12.
PLoS One ; 14(5): e0217644, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31145760

RESUMEN

Waist-to-hip ratio (WHR) is a prominent cardiometabolic risk factor that increases cardio-metabolic disease risk independently of BMI and for which multiple genetic loci have been identified. However, WHR is a relatively crude proxy for fat distribution and it does not capture all variation in fat distribution. We here present a study of the role of coding genetic variants on fat mass in 6 distinct regions of the body, based on dual-energy X-ray absorptiometry imaging on more than 17k participants. We find that the missense variant CCDC92S70C, previously associated with WHR, is associated specifically increased leg fat mass and reduced visceral but not subcutaneous central fat. The minor allele-carrying transcript of CCDC92 is constitutively more highly expressed in adipose tissue samples. In addition, we identify two coding variants in SPATA20 and UQCC1 that are associated with arm fat mass. SPATA20K422R is a low-frequency variant with a large effect on arm fat only, and UQCC1R51Q is a common variant reaching significance for arm but showing similar trends in other subcutaneous fat depots. Our findings support the notion that different fat compartments are regulated by distinct genetic factors.


Asunto(s)
Tejido Adiposo/diagnóstico por imagen , Obesidad/diagnóstico por imagen , Grasa Subcutánea/diagnóstico por imagen , Relación Cintura-Cadera/métodos , Absorciometría de Fotón , Tejido Adiposo/fisiopatología , Adulto , Composición Corporal/fisiología , Distribución de la Grasa Corporal , Índice de Masa Corporal , Enfermedades Cardiovasculares/diagnóstico por imagen , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Humanos , Masculino , Enfermedades Metabólicas/diagnóstico por imagen , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/fisiopatología , Persona de Mediana Edad , Obesidad/fisiopatología , Factores de Riesgo , Grasa Subcutánea/fisiopatología
13.
J Physiol ; 597(14): 3527-3537, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30883738

RESUMEN

Excessive consumption of free sugars (which typically includes a composite of glucose and fructose) is associated with an increased risk of developing chronic metabolic diseases including obesity, non-alcoholic fatty liver disease (NAFLD), type 2 diabetes and cardiovascular disease. Determining the utilisation, storage and fate of dietary sugars in metabolically relevant tissues is fundamental to understanding their contribution to metabolic disease risk. To date, the study of fructose metabolism has primarily focused on the liver, where it has been implicated in impaired insulin sensitivity, increased fat accumulation and dyslipidaemia. Yet we still have only a limited understanding of the mechanisms by which consumption of fructose, as part of a mixed meal, may alter hepatic fatty acid synthesis and partitioning. Moreover, surprisingly little is known about the metabolism of fructose within other organs, specifically subcutaneous adipose tissue, which is the largest metabolically active organ in the human body and is consistently exposed to nutrient fluxes. This review summarises what is known about fructose metabolism in the liver and adipose tissue and examines evidence for tissue-specific and sex-specific responses to fructose.


Asunto(s)
Fructosa/metabolismo , Enfermedades Metabólicas/metabolismo , Tejido Adiposo/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos/metabolismo , Humanos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo
14.
Methods Mol Biol ; 1862: 83-96, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30315461

RESUMEN

Stable isotopes are powerful tools for tracing the metabolic fate of molecules in the human body. In this chapter, we focus on the use of deuterium (2H), a stable isotope of hydrogen, in the study of human lipid metabolism within the liver in vivo in humans and in vitro using hepatocyte cellular models. The measurement of de novo lipogenesis (DNL) will be focussed on, as the synthesis of fatty acids, specifically palmitate, has been gathering momentum as being implicated in cellular dysfunction, which may be involved in the development of non-alcoholic fatty liver disease (NAFLD). Therefore, this chapter focusses specifically on the use of 2H2O (heavy water) to measure hepatic DNL.


Asunto(s)
Deuterio/administración & dosificación , Lipoproteínas/aislamiento & purificación , Metabolómica/métodos , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Medios de Cultivo/química , Deuterio/química , Hepatocitos/metabolismo , Humanos , Lipogénesis , Lipoproteínas/química , Lipoproteínas/metabolismo , Hígado/metabolismo , Hígado/patología , Espectrometría de Masas/instrumentación , Espectrometría de Masas/métodos , Metabolómica/instrumentación , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Enfermedad del Hígado Graso no Alcohólico/patología , Extracción en Fase Sólida/instrumentación , Extracción en Fase Sólida/métodos , Ultracentrifugación/instrumentación , Ultracentrifugación/métodos
15.
Mol Metab ; 16: 172-179, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30100245

RESUMEN

OBJECTIVE: The composition of the extracellular matrix (ECM) impacts adipocyte function and might determine adipose tissue (AT) function and distribution. Cartilage oligomeric matrix protein (COMP), a matricellular protein usually studied in bone and cartilage, is highly differentially expressed between subcutaneous abdominal and gluteal AT. This study aimed to explore COMP's role in human subcutaneous abdominal and gluteal AT and preadipocyte biology. METHODS: COMP mRNA levels were measured in whole AT and immortalised preadipocytes via quantitative (q)-PCR. Tissue and cellular COMP protein were measured via Western blot and immunohistochemistry; plasma COMP was measured by ELISA. The effect of COMP on adipogenesis in immortalised preadipocytes was evaluated by qPCR of adipogenic markers and cellular triacylglycerol (TAG) accumulation. RESULTS: qPCR analysis of paired subcutaneous abdominal and gluteal AT biopsies (n = 190) across a range of BMI (20.7-45.5 kg/m2) indicated ∼3-fold higher COMP expression in gluteal AT (P = 1.7 × 10-31); protein levels mirrored this. Immunohistochemistry indicated COMP was abundant in gluteal AT ECM and co-localised with collagen-1. AT COMP mRNA levels and circulating COMP protein levels were positively associated with BMI/adiposity but unrelated to AT distribution. COMP expression changed dynamically during adipogenesis (time × depot, P = 0.01). Supplementation of adipogenic medium with exogenous COMP protein (500 ng/ml) increased PPARG2 expression ∼1.5-fold (P = 0.0003) and TAG accumulation ∼1.25-fold in abdominal and gluteal preadipocytes (P = 0.02). CONCLUSIONS: We confirmed that COMP is an ECM protein which is differentially expressed between subcutaneous abdominal and gluteal AT. Despite its depot-specific expression pattern, however, AT COMP mRNA levels and plasma COMP concentration correlated positively with overall obesity but not body fat distribution. Exogenous COMP enhanced adipogenesis. These data identify COMP as a novel regulator of AT and highlight the importance of the ECM to AT biology.


Asunto(s)
Adipogénesis/fisiología , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Grasa Subcutánea/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adiposidad , Adulto , Biopsia con Aguja , Distribución de la Grasa Corporal , Índice de Masa Corporal , Nalgas , Proteína de la Matriz Oligomérica del Cartílago/genética , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Obesidad/metabolismo , Grasa Subcutánea/fisiología , Tejido Subcutáneo/metabolismo
16.
Adipocyte ; 6(1): 40-55, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-28452592

RESUMEN

Upper-body adiposity is associated with increased metabolic disease risk, while lower-body adiposity is paradoxically protective. Efforts to understand the underlying mechanisms require appropriate and reproducible in vitro culture models. We have therefore generated immortalised (im) human preadipocyte (PAD) cell lines derived from paired subcutaneous abdominal and gluteal adipose tissue. These cell lines, denoted imAPAD and imGPAD display enhanced proliferation and robust adipogenic capacities. Differentiated imAPAD and imGPAD adipocytes synthesize triglycerides de novo and respond lipolytically to catecholamine-stimulation. Importantly the cells retain their depot-of-origin 'memory' as reflected by inherent differences in fatty acid metabolism and expression of depot-specific developmental genes. These features make these cell lines an invaluable tool for the in vitro investigation of depot-specific human adipocyte biology.


Asunto(s)
Adipocitos/citología , Adipogénesis/fisiología , Grasa Abdominal/metabolismo , Grasa Abdominal/fisiología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adiposidad/fisiología , Distribución de la Grasa Corporal , Nalgas/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Humanos , Modelos Biológicos , Obesidad/metabolismo , Grasa Subcutánea/citología
17.
Biochim Biophys Acta ; 1851(5): 686-96, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25668679

RESUMEN

In this review we discuss the role of developmental transcription factors in adipose tissue biology with a focus on how these developmental genes may contribute to regional variation in adipose tissue distribution and function. Regional, depot-specific, differences in lipid handling and signalling (lipolysis, lipid storage and adipokine/lipokine signalling) are important determinants of metabolic health. At a cellular level, preadipocytes removed from their original depot and cultured in vitro retain depot-specific functional properties, implying that these are intrinsic to the cells and not a function of their environment in situ. High throughput screening has identified a number of developmental transcription factors involved in embryological development, including members of the Homeobox and T-Box gene families, that are strongly differentially expressed between regional white adipose tissue depots and also between brown and white adipose tissue. However, the significance of depot-specific developmental signatures remains unclear. Developmental transcription factors determine body patterning during embryogenesis. The divergent developmental origins of regional adipose tissue depots may explain their differing functional characteristics. There is evidence from human genetics that developmental genes determine adipose tissue distribution: in GWAS studies a number of developmental genes have been identified as being correlated with anthropometric measures of adiposity and fat distribution. Additionally, compelling functional studies have recently implicated developmental genes in both white adipogenesis and the so-called 'browning' of white adipose tissue. Understanding the genetic and developmental pathways in adipose tissue may help uncover novel ways to intervene with the function of adipose tissue in order to promote health.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Factores de Transcripción/metabolismo , Adipoquinas/metabolismo , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Blanco/embriología , Tejido Adiposo Blanco/crecimiento & desarrollo , Adiposidad , Animales , Metabolismo Energético , Regulación del Desarrollo de la Expresión Génica , Humanos , Lipogénesis , Lipólisis , Morfogénesis , Transducción de Señal , Factores de Transcripción/genética
18.
Nat Rev Endocrinol ; 11(2): 90-100, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25365922

RESUMEN

The distribution of adipose tissue in the body has wide-ranging and reproducible associations with health and disease. Accumulation of adipose tissue in the upper body (abdominal obesity) is associated with the development of cardiovascular disease, insulin resistance, type 2 diabetes mellitus and even all-cause mortality. Conversely, accumulation of fat in the lower body (gluteofemoral obesity) shows opposite associations with cardiovascular disease and type 2 diabetes mellitus when adjusted for overall fat mass. The abdominal depots are characterized by rapid uptake of predominantly diet-derived fat and a high lipid turnover that is easily stimulated by adrenergic receptor activation. The lower-body fat stores have a reduced lipid turnover with a capacity to accommodate fat undergoing redistribution. Lower-body adipose tissue also seems to retain the capacity to recruit additional adipocytes as a result of weight gain and demonstrates fewer signs of inflammatory insult. New data suggest that the profound functional differences between the upper-body and lower-body tissues are controlled by site-specific sets of developmental genes, such as HOXA6, HOXA5, HOXA3, IRX2 and TBX5 in subcutaneous abdominal adipose tissue and HOTAIR, SHOX2 and HOXC11 in gluteofemoral adipose tissue, which are under epigenetic control. This Review discusses the developmental and functional differences between upper-body and lower-body fat depots and provides mechanistic insight into the disease-protective effects of lower-body fat.


Asunto(s)
Tejido Adiposo/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología
19.
Cell Rep ; 9(1): 349-365, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25263561

RESUMEN

An in vivo model of antiangiogenic therapy allowed us to identify genes upregulated by bevacizumab treatment, including Fatty Acid Binding Protein 3 (FABP3) and FABP7, both of which are involved in fatty acid uptake. In vitro, both were induced by hypoxia in a hypoxia-inducible factor-1α (HIF-1α)-dependent manner. There was a significant lipid droplet (LD) accumulation in hypoxia that was time and O2 concentration dependent. Knockdown of endogenous expression of FABP3, FABP7, or Adipophilin (an essential LD structural component) significantly impaired LD formation under hypoxia. We showed that LD accumulation is due to FABP3/7-dependent fatty acid uptake while de novo fatty acid synthesis is repressed in hypoxia. We also showed that ATP production occurs via ß-oxidation or glycogen degradation in a cell-type-dependent manner in hypoxia-reoxygenation. Finally, inhibition of lipid storage reduced protection against reactive oxygen species toxicity, decreased the survival of cells subjected to hypoxia-reoxygenation in vitro, and strongly impaired tumorigenesis in vivo.


Asunto(s)
Ácidos Grasos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metabolismo de los Lípidos , Oxígeno/metabolismo , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Glioblastoma , Humanos , Oxidación-Reducción
20.
Diabetes ; 63(11): 3785-97, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24947352

RESUMEN

Upper- and lower-body fat depots exhibit opposing associations with obesity-related metabolic disease. We defined the relationship between DEXA-quantified fat depots and diabetes/cardiovascular risk factors in a healthy population-based cohort (n = 3,399). Gynoid fat mass correlated negatively with insulin resistance after total fat mass adjustment, whereas the opposite was seen for abdominal fat. Paired transcriptomic analysis of gluteal subcutaneous adipose tissue (GSAT) and abdominal subcutaneous adipose tissue (ASAT) was performed across the BMI spectrum (n = 49; 21.4-45.5 kg/m(2)). In both depots, energy-generating metabolic genes were negatively associated and inflammatory genes were positively associated with obesity. However, associations were significantly weaker in GSAT. At the systemic level, arteriovenous release of the proinflammatory cytokine interleukin-6 (n = 34) was lower from GSAT than ASAT. Isolated preadipocytes retained a depot-specific transcriptional "memory" of embryonic developmental genes and exhibited differential promoter DNA methylation of selected genes (HOTAIR, TBX5) between GSAT and ASAT. Short hairpin RNA-mediated silencing identified TBX5 as a regulator of preadipocyte proliferation and adipogenic differentiation in ASAT. In conclusion, intrinsic differences in the expression of developmental genes in regional adipocytes provide a mechanistic basis for diversity in adipose tissue (AT) function. The less inflammatory nature of lower-body AT offers insight into the opposing metabolic disease risk associations between upper- and lower-body obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Enfermedades Cardiovasculares/metabolismo , Obesidad/metabolismo , Grasa Abdominal/metabolismo , Adulto , Metilación de ADN , Femenino , Humanos , Grasa Intraabdominal/metabolismo , Masculino , Persona de Mediana Edad , Factores de Riesgo , Grasa Subcutánea Abdominal/metabolismo , Proteínas de Dominio T Box/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...