Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(1-2): 343-58, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439387

RESUMEN

Genome-wide association studies have revealed numerous risk loci associated with diverse diseases. However, identification of disease-causing variants within association loci remains a major challenge. Divergence in gene expression due to cis-regulatory variants in noncoding regions is central to disease susceptibility. We show that integrative computational analysis of phylogenetic conservation with a complexity assessment of co-occurring transcription factor binding sites (TFBS) can identify cis-regulatory variants and elucidate their mechanistic role in disease. Analysis of established type 2 diabetes risk loci revealed a striking clustering of distinct homeobox TFBS. We identified the PRRX1 homeobox factor as a repressor of PPARG2 expression in adipose cells and demonstrate its adverse effect on lipid metabolism and systemic insulin sensitivity, dependent on the rs4684847 risk allele that triggers PRRX1 binding. Thus, cross-species conservation analysis at the level of co-occurring TFBS provides a valuable contribution to the translation of genetic association signals to disease-related molecular mechanisms.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Polimorfismo de Nucleótido Simple , Animales , Línea Celular , Células Cultivadas , Secuencia Conservada , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/metabolismo , Humanos , Resistencia a la Insulina , PPAR gamma/genética , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 43(6): 1054-1065, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37051928

RESUMEN

BACKGROUND: Adipose tissue insulin resistance is linked to altered plasma levels of triglycerides and HDL (high-density lipoprotein)-cholesterol. However, its degree of independence from liver resistance and different metabolic traits (lipolysis, lipogenesis) effected is not clear and was presently investigated. METHODS: In 3290 adult subjects, plasma levels of triglycerides and HDL-cholesterol were cross-sectionally measured and related to interindividual variations in measures of insulin resistance in the liver (homeostasis mode assessment of insulin resistance index) or adipose tissue (Adipo-IR index). In subgroups, insulin-induced antilipolysis and lipogenesis in isolated subcutaneous fat cells (n=578) were determined alongside global adipose tissue gene expression (n=132). RESULTS: Using linear regression, homeostasis mode assessment of insulin resistance and Adipo-IR strongly correlated with the plasma lipids explaining 33% of the variations in triglycerides. Together with other variables (age, sex, body mass index, cardiometabolic disorders, nicotine use, ethnicity, and physical activity) in multiple regression, homeostasis mode assessment of insulin resistance, and Adipo-IR each remained an important regressor for triglycerides and HDL-cholesterol (P<0.0001). In fat cells, half-maximum effective concentration but not maximum effect of insulin on antilipolysis and lipogenesis contributed independently to variations in triglycerides and HDL-cholesterol (P=0.001 or lower). This was linked to expression of the insulin receptor, insulin receptor substrate-1, and AKT serine/threonine kinase 2 in adipose tissue. CONCLUSIONS: Markers of insulin resistance in the liver and adipose tissue each associate strongly, and independently of each other, to elevated triglycerides and decreased HDL levels. At the fat cell, early insulin receptor signaling and sensitivity, but not maximum insulin action contributes to the variations in circulating triglycerides and HDL-cholesterol.


Asunto(s)
Dislipidemias , Resistencia a la Insulina , Adulto , Humanos , Receptor de Insulina , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Triglicéridos , Insulina , HDL-Colesterol , Hígado/metabolismo , Dislipidemias/diagnóstico , Dislipidemias/genética
3.
Int J Obes (Lond) ; 44(2): 377-387, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31164724

RESUMEN

BACKGROUND: Transcriptome analysis of abdominal subcutaneous white adipose tissue (sWAT) has identified important obesity-associated disturbances. However, the relation between sWAT transcriptome and long-term future changes in body weight remains elusive. OBJECTIVE: To investigate sWAT transcriptome signatures before and after long-term weight changes and assess their predictive value for body weight changes. DESIGN: A total of 56 women were followed longitudinally and subdivided into weight-stable (WS, n = 25), weight-gaining (WG, n = 14) and weight-losing (WL, n = 17) groups between baseline and follow-up (13 ± 1 years). The fasting sWAT transcriptome was analyzed by gene microarray at baseline and follow-up. Key genes associated with weight changes were validated using quantitative real-time PCR. RESULTS: In total 285 transcripts exhibited difference (FDR < 30%) in expression fold change over time between WL and WS women. WL women displayed decreased pro-inflammatory (NLRP3) but increased insulin-response gene (FASN and GLUT4) expression over time. In comparison, 461 transcripts displayed difference in expression fold change over time between WG and WS women (P < 0.05). Genes involved in autophagic processes (CDK5, SQSTM1 and FBXL2) were generally upregulated in WG women. At baseline, 307 and 302 transcripts were differentially expressed (FDR < 30%) in WL and WG women, respectively, when independently compared against WS women. Baseline expression of adipogenic and lipogenic genes (PPARG, IRS2 and HACD2) was lower, while pro-fibrotic (COL6A1) was higher, in WL than WS women; whereas protein processing genes were lower expressed in WG than in WS women. CONCLUSION: In adult women, long-term body weight change associates with altered sWAT transcriptome. Expression of genes associated with inflammation, insulin response, adipogenesis and lipogenesis are linked to weight loss. However, other pathways such as autophagy not only associate but also predict future weight gain suggesting that intrinsic factors in sWAT impact tissue expansion.


Asunto(s)
Peso Corporal , Obesidad , Grasa Subcutánea Abdominal/metabolismo , Transcriptoma/genética , Adulto , Peso Corporal/genética , Peso Corporal/fisiología , Femenino , Humanos , Inflamación/genética , Lipogénesis/genética , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo , Estudios Prospectivos
4.
Nucleic Acids Res ; 45(6): 3266-3279, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28334807

RESUMEN

Genome-wide association studies identified numerous disease risk loci. Delineating molecular mechanisms influenced by cis-regulatory variants is essential to understand gene regulation and ultimately disease pathophysiology. Combining bioinformatics and public domain chromatin information with quantitative proteomics supports prediction of cis-regulatory variants and enabled identification of allele-dependent binding of both, transcription factors and coregulators at the type 2 diabetes associated PPARG locus. We found rs7647481A nonrisk allele binding of Yin Yang 1 (YY1), confirmed by allele-specific chromatin immunoprecipitation in primary adipocytes. Quantitative proteomics also found the coregulator RING1 and YY1 binding protein (RYBP) whose mRNA levels correlate with improved insulin sensitivity in primary adipose cells carrying the rs7647481A nonrisk allele. Our findings support a concept with diverse cis-regulatory variants contributing to disease pathophysiology at one locus. Proteome-wide identification of both, transcription factors and coregulators, can profoundly improve understanding of mechanisms underlying genetic associations.


Asunto(s)
Alelos , PPAR gamma/genética , Proteómica , Elementos Reguladores de la Transcripción , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Sitios Genéticos , Variación Genética , Humanos , Resistencia a la Insulina/genética , Ratones , Ratas , Factores de Transcripción/metabolismo , Transcripción Genética , Factor de Transcripción YY1/metabolismo
5.
Diabetologia ; 61(5): 1112-1123, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29487953

RESUMEN

AIMS/HYPOTHESIS: By genome-wide association meta-analysis, 17 genetic loci associated with fasting serum insulin (FSI), a marker of systemic insulin resistance, have been identified. To define potential culprit genes in these loci, in a cross-sectional study we analysed white adipose tissue (WAT) expression of 120 genes in these loci in relation to systemic and adipose tissue variables, and functionally evaluated genes demonstrating genotype-specific expression in WAT (eQTLs). METHODS: Abdominal subcutaneous adipose tissue biopsies were obtained from 114 women. Basal lipolytic activity was measured as glycerol release from adipose tissue explants. Adipocytes were isolated and insulin-stimulated incorporation of radiolabelled glucose into lipids was used to quantify adipocyte insulin sensitivity. Small interfering RNA-mediated knockout in human mesenchymal stem cells was used for functional evaluation of genes. RESULTS: Adipose expression of 48 of the studied candidate genes associated significantly with FSI, whereas expression of 24, 17 and 2 genes, respectively, associated with adipocyte insulin sensitivity, lipolysis and/or WAT morphology (i.e. fat cell size relative to total body fat mass). Four genetic loci contained eQTLs. In one chromosome 4 locus (rs3822072), the FSI-increasing allele associated with lower FAM13A expression and FAM13A expression associated with a beneficial metabolic profile including decreased WAT lipolysis (regression coefficient, R = -0.50, p = 5.6 × 10-7). Knockdown of FAM13A increased lipolysis by ~1.5-fold and the expression of LIPE (encoding hormone-sensitive lipase, a rate-limiting enzyme in lipolysis). At the chromosome 7 locus (rs1167800), the FSI-increasing allele associated with lower POM121C expression. Consistent with an insulin-sensitising function, POM121C expression associated with systemic insulin sensitivity (R = -0.22, p = 2.0 × 10-2), adipocyte insulin sensitivity (R = 0.28, p = 3.4 × 10-3) and adipose hyperplasia (R = -0.29, p = 2.6 × 10-2). POM121C knockdown decreased expression of all adipocyte-specific markers by 25-50%, suggesting that POM121C is necessary for adipogenesis. CONCLUSIONS/INTERPRETATION: Gene expression and adipocyte functional studies support the notion that FAM13A and POM121C control adipocyte lipolysis and adipogenesis, respectively, and might thereby be involved in genetic control of systemic insulin sensitivity.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Estudio de Asociación del Genoma Completo , Insulina/metabolismo , Glicoproteínas de Membrana/genética , Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/metabolismo , Adiposidad , Adulto , Ayuno , Femenino , Estudios de Seguimiento , Genotipo , Glucosa/metabolismo , Humanos , Resistencia a la Insulina , Lipólisis , Persona de Mediana Edad , Obesidad/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Sitios de Carácter Cuantitativo , Suecia
6.
Int J Obes (Lond) ; 42(12): 2047-2056, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29453465

RESUMEN

BACKGROUND: Adipokines are peptides secreted from white adipose tissue (WAT), which have been linked to WAT dysfunction and metabolic complications of obesity. We set out to identify novel adipokines in subcutaneous WAT (sWAT) linked to insulin resistance (IR). METHODS: Gene expression was determined by microarray and qPCR in obese and non-obese subjects with varying degree of IR. WAT-secreted and circulating protein levels were measured by ELISA. RESULTS: In sWAT of 80 obese women discordant for IR, 44 genes encoding potential adipose-secreted proteins were differentially expressed. Among these, merely two proteins, S100A4 and MXRA5 were released from sWAT in a time-dependent manner (criterion for true adipokines) but only the circulating levels of S100A4 were higher in IR. In two additional cohorts (n = 29 and n = 56), sWAT S100A4 secretion was positively and BMI-independently associated with IR (determined by clamp or HOMA-IR), ATP-III risk score and adipocyte size (hypertrophy). In non-obese (n = 20) and obese subjects before and after bariatric surgery (n = 21), circulating and sWAT-secreted levels were highest in the obese and normalized following weight loss. Serum S100A4 concentrations were higher in subjects with type 2 diabetes. S100A4 sWAT expression associated positively with genes involved in inflammation/extracellular matrix formation and inversely with genes in metabolic pathways. Although S100A4 was expressed in both stromal cells and adipocytes, only the expression in adipocytes associated with BMI. CONCLUSIONS: S100A4 is a novel adipokine associated with IR and sWAT inflammation/adipocyte hypertrophy independently of BMI. Its value as a circulating marker for dysfunctional WAT and IR needs to be validated in larger cohorts.


Asunto(s)
Adipoquinas/metabolismo , Tejido Adiposo Blanco/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Proteína de Unión al Calcio S100A4/metabolismo , Adipoquinas/sangre , Tejido Adiposo Blanco/química , Adulto , Biomarcadores , Estudios de Cohortes , Femenino , Humanos , Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Obesidad/sangre , Obesidad/epidemiología , Proteína de Unión al Calcio S100A4/sangre
7.
Proc Natl Acad Sci U S A ; 112(14): 4363-8, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25831505

RESUMEN

Genome-wide association studies have implicated PLEXIN D1 (PLXND1) in body fat distribution and type 2 diabetes. However, a role for PLXND1 in regional adiposity and insulin resistance is unknown. Here we use in vivo imaging and genetic analysis in zebrafish to show that Plxnd1 regulates body fat distribution and insulin sensitivity. Plxnd1 deficiency in zebrafish induced hyperplastic morphology in visceral adipose tissue (VAT) and reduced lipid storage. In contrast, subcutaneous adipose tissue (SAT) growth and morphology were unaffected, resulting in altered body fat distribution and a reduced VAT:SAT ratio in zebrafish. A VAT-specific role for Plxnd1 appeared conserved in humans, as PLXND1 mRNA was positively associated with hypertrophic morphology in VAT, but not SAT. In zebrafish plxnd1 mutants, the effect on VAT morphology and body fat distribution was dependent on induction of the extracellular matrix protein collagen type V alpha 1 (col5a1). Furthermore, after high-fat feeding, zebrafish plxnd1 mutant VAT was resistant to expansion, and excess lipid was disproportionately deposited in SAT, leading to an even greater exacerbation of altered body fat distribution. Plxnd1-deficient zebrafish were protected from high-fat-diet-induced insulin resistance, and human VAT PLXND1 mRNA was positively associated with type 2 diabetes, suggesting a conserved role for PLXND1 in insulin sensitivity. Together, our findings identify Plxnd1 as a novel regulator of VAT growth, body fat distribution, and insulin sensitivity in both zebrafish and humans.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Colágeno Tipo V/biosíntesis , Insulina/metabolismo , Grasa Intraabdominal/patología , Glicoproteínas de Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Composición Corporal , Proliferación Celular , Células Endoteliales/citología , Matriz Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lípidos/química , Ratones , Mutación , Obesidad , ARN Mensajero/metabolismo , Transducción de Señal , Pez Cebra
8.
Am J Physiol Endocrinol Metab ; 312(6): E482-E494, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28270439

RESUMEN

The key pathological link between obesity and type 2 diabetes is insulin resistance, but the molecular mechanisms are not entirely identified. micro-RNAs (miRNA) are dysregulated in obesity and may contribute to insulin resistance. Our objective was to detect and functionally investigate miRNAs linked to insulin sensitivity in human subcutaneous white adipose tissue (scWAT). Subjects were selected based on the insulin-stimulated lipogenesis response of subcutaneous adipocytes. Global miRNA profiling was performed in abdominal scWAT of 18 obese insulin-resistance (OIR), 21 obese insulin-sensitive (OIS), and 9 lean women. miRNAs demonstrating differential expression between OIR and OIS women were overexpressed in human in vitro-differentiated adipocytes followed by assessment of lipogenesis and identification of miRNA targets by measuring mRNA/protein expression and 3'-untranslated region analysis. Eleven miRNAs displayed differential expression between OIR and OIS states. Overexpression of miR-143-3p and miR-652-3p increased insulin-stimulated lipogenesis in human in vitro differentiated adipocytes and directly or indirectly affected several genes/proteins involved in insulin signaling at transcriptional or posttranscriptional levels. Adipose expression of miR-143-3p and miR-652-3p was positively associated with insulin-stimulated lipogenesis in scWAT independent of body mass index. In conclusion, miR-143-3p and miR-652-3p are linked to scWAT insulin resistance independent of obesity and influence insulin-stimulated lipogenesis by interacting at different steps with insulin-signaling pathways.


Asunto(s)
Regulación de la Expresión Génica , Resistencia a la Insulina , MicroARNs/metabolismo , Obesidad Mórbida/metabolismo , Obesidad/metabolismo , Grasa Subcutánea Abdominal/metabolismo , Regiones no Traducidas 3' , Adulto , Biopsia , Índice de Masa Corporal , Células Cultivadas , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica , Humanos , Lipogénesis , Masculino , MicroARNs/agonistas , Persona de Mediana Edad , Obesidad/patología , Obesidad Mórbida/patología , ARN/metabolismo , Grasa Subcutánea Abdominal/patología
9.
Diabetologia ; 59(4): 799-812, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26750116

RESUMEN

AIMS/HYPOTHESIS: Individuals who had a low birthweight (LBW) are at an increased risk of insulin resistance and type 2 diabetes when exposed to high-fat overfeeding (HFO). We studied genome-wide mRNA expression and DNA methylation in subcutaneous adipose tissue (SAT) after 5 days of HFO and after a control diet in 40 young men, of whom 16 had LBW. METHODS: mRNA expression was analysed using Affymetrix Human Gene 1.0 ST arrays and DNA methylation using Illumina 450K BeadChip arrays. RESULTS: We found differential DNA methylation at 53 sites in SAT from LBW vs normal birthweight (NBW) men (false discovery rate <5%), including sites in the FADS2 and CPLX1 genes previously associated with type 2 diabetes. When we used reference-free cell mixture adjustments to potentially adjust for cell composition, 4,323 sites had differential methylation in LBW vs NBW men. However, no differences in SAT gene expression levels were identified between LBW and NBW men. In the combined group of all 40 participants, 3,276 genes (16.5%) were differentially expressed in SAT after HFO (false discovery rate <5%) and there was no difference between LBW men and controls. The most strongly upregulated genes were ELOVL6, FADS2 and NNAT; in contrast, INSR, IRS2 and the SLC27A2 fatty acid transporter showed decreased expression after HFO. Interestingly, SLC27A2 expression correlated negatively with diabetes- and obesity-related traits in a replication cohort of 142 individuals. DNA methylation at 652 CpG sites (including in CDK5, IGFBP5 and SLC2A4) was altered in SAT after overfeeding in this and in another cohort. CONCLUSIONS/INTERPRETATION: Young men who had a LBW exhibit epigenetic alterations in their adipose tissue that potentially influence insulin resistance and risk of type 2 diabetes. Short-term overfeeding influences gene transcription and, to some extent, DNA methylation in adipose tissue; there was no major difference in this response between LBW and control participants.


Asunto(s)
Tejido Adiposo/metabolismo , Transcriptoma/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Adulto , Metilación de ADN/genética , Diabetes Mellitus Tipo 2/genética , Dieta Alta en Grasa/efectos adversos , Epigenómica , Ácido Graso Desaturasas/genética , Humanos , Recién Nacido de Bajo Peso/fisiología , Masculino , Proteínas del Tejido Nervioso/genética , Adulto Joven
10.
Diabetologia ; 59(11): 2393-2405, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27535281

RESUMEN

AIMS/HYPOTHESIS: Insulin resistance (IR) links obesity to type 2 diabetes. The aim of this study was to explore whether white adipose tissue (WAT) epigenetic dysregulation is associated with systemic IR by genome-wide CG dinucleotide (CpG) methylation and gene expression profiling in WAT from insulin-resistant and insulin-sensitive women. A secondary aim was to determine whether the DNA methylation signature in peripheral blood mononuclear cells (PBMCs) reflects WAT methylation and, if so, can be used as a marker for systemic IR. METHODS: From 220 obese women, we selected a total of 80 individuals from either of the extreme ends of the distribution curve of HOMA-IR, an indirect measure of systemic insulin sensitivity. Genome-wide transcriptome and DNA CpG methylation profiling by array was performed on subcutaneous (SAT) and visceral (omental) adipose tissue (VAT). CpG methylation in PBMCs was assayed in the same cohort. RESULTS: There were 647 differentially expressed genes (false discovery rate [FDR] 10%) in SAT, all of which displayed directionally consistent associations in VAT. This suggests that IR is associated with dysregulated expression of a common set of genes in SAT and VAT. The average degree of DNA methylation did not differ between the insulin-resistant and insulin-sensitive group in any of the analysed tissues/cells. There were 223 IR-associated genes in SAT containing a total of 336 nominally significant differentially methylated sites (DMS). The 223 IR-associated genes were over-represented in pathways related to integrin cell surface interactions and insulin signalling and included COL5A1, GAB1, IRS2, PFKFB3 and PTPRJ. In VAT there were a total of 51 differentially expressed genes (FDR 10%); 18 IR-associated genes contained a total of 29 DMS. CONCLUSIONS/INTERPRETATION: In individuals discordant for insulin sensitivity, the average DNA CpG methylation in SAT and VAT is similar, although specific genes, particularly in SAT, display significantly altered expression and DMS in IR, possibly indicating that epigenetic regulation of these genes influences metabolism.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética/genética , Resistencia a la Insulina/fisiología , Obesidad/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Tejido Adiposo Blanco/metabolismo , Adulto , Colágeno Tipo V/genética , Femenino , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Resistencia a la Insulina/genética , Grasa Intraabdominal/metabolismo , Leucocitos Mononucleares/metabolismo , Fosfofructoquinasa-2/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
11.
Mol Genet Metab ; 119(1-2): 174-85, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27386756

RESUMEN

Fibrillin-1 is a large glycoprotein encoded by the FBN1 gene in humans. It provides strength and elasticity to connective tissues and is involved in regulating the bioavailability of the growth factor TGFß. Mutations in FBN1 may be associated with depleted or abnormal adipose tissue, seen in some patients with Marfan syndrome and lipodystrophies. As this lack of adipose tissue does not result in high morbidity or mortality, it is generally under-appreciated, but is a cause of psychosocial problems particularly to young patients. We examined the role of fibrillin-1 in adipogenesis. In inbred mouse strains we found significant variation in the level of expression in the Fbn1 gene that correlated with variation in several measures of body fat, suggesting that mouse fibrillin-1 is associated with the level of fat tissue. Furthermore, we found that FBN1 mRNA was up-regulated in the adipose tissue of obese women compared to non-obese, and associated with an increase in adipocyte size. We used human mesenchymal stem cells differentiated in culture to adipocytes to show that fibrillin-1 declines after the initiation of differentiation. Gene expression results from a similar experiment (available through the FANTOM5 project) revealed that the decline in fibrillin-1 protein was paralleled by a decline in FBN1 mRNA. Examination of the FBN1 gene showed that the region commonly affected in FBN1-associated lipodystrophy is highly conserved both across the three human fibrillin genes and across genes encoding fibrillin-1 in vertebrates. These results suggest that fibrillin-1 is involved as the undifferentiated mesenchymal stem cells transition to adipogenesis but then declines as the developing adipocytes take on their final phenotype. Since the C-terminal peptide of fibrillin-1 is a glucogenic hormone, individuals with low fibrillin-1 (for example with FBN1 mutations associated with lipodystrophy) may fail to differentiate adipocytes and/or to accumulate adipocyte lipids, although this still needs to be shown experimentally.


Asunto(s)
Adipogénesis/genética , Diferenciación Celular/genética , Fibrilina-1/genética , Células Madre Mesenquimatosas/metabolismo , Animales , Femenino , Regulación de la Expresión Génica , Humanos , Lipodistrofia/genética , Lipodistrofia/fisiopatología , Masculino , Síndrome de Marfan/genética , Síndrome de Marfan/fisiopatología , Ratones , Mutación , Fenotipo , ARN Mensajero/genética , Caracteres Sexuales
12.
Lipids Health Dis ; 14: 42, 2015 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-25934644

RESUMEN

BACKGROUND: Visceral fat accumulation is associated with metabolic disease. It is therefore relevant to study factors that regulate adipose tissue distribution. Recent data shows that overeating saturated fatty acids promotes greater visceral fat storage than overeating unsaturated fatty acids. Visceral adiposity is observed in states of hypercortisolism, and the enzyme 11-ß-hydroxysteroid-dehydrogenase type 1 (11ß-hsd1) is a major regulator of cortisol activity by converting inactive cortisone to cortisol in adipose tissue. We hypothesized that tissue fatty acid composition regulates body fat distribution through local effects on the expression of 11ß-hsd1 and its corresponding gene (HSD11B1) resulting in altered cortisol activity. FINDINGS: Visceral- and subcutaneous adipose tissue biopsies were collected during Roux-en-Y gastric bypass surgery from 45 obese women (BMI; 41±4 kg/m2). The fatty acid composition of each biopsy was measured and correlated to the mRNA levels of HSD11B1. 11ß-hsd1 protein levels were determined in a subgroup (n=12) by western blot analysis. Our main finding was that tissue saturated fatty acids (e.g. palmitate) were associated with increased 11ß-hsd1 gene- and protein-expression in visceral but not subcutaneous adipose tissue. CONCLUSIONS: The present study proposes a link between HSD11B1 and saturated fatty acids in visceral, but not subcutaneous adipose tissue. Nutritional regulation of visceral fat mass through HSD11B1 is of interest for the modulation of metabolic risk and warrants further investigation.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Ácidos Grasos/análisis , Grasa Intraabdominal/química , Adulto , Western Blotting , Femenino , Expresión Génica , Humanos , Grasa Intraabdominal/enzimología
13.
Am J Physiol Endocrinol Metab ; 306(3): E267-74, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24326420

RESUMEN

Cancer cachexia is associated with pronounced adipose tissue loss due to, at least in part, increased fat cell lipolysis. MicroRNAs (miRNAs) have recently been implicated in controlling several aspects of adipocyte function. To gain insight into the possible impact of miRNAs on adipose lipolysis in cancer cachexia, global miRNA expression was explored in abdominal subcutaneous adipose tissue from gastrointestinal cancer patients with (n = 10) or without (n = 11) cachexia. Effects of miRNA overexpression or inhibition on lipolysis were determined in human in vitro differentiated adipocytes. Out of 116 miRNAs present in adipose tissue, five displayed distinct cachexia-associated expression according to both microarray and RT-qPCR. Four (miR-483-5p/-23a/-744/-99b) were downregulated, whereas one (miR-378) was significantly upregulated in cachexia. Adipose expression of miR-378 associated strongly and positively with catecholamine-stimulated lipolysis in adipocytes. This correlation is most probably causal because overexpression of miR-378 in human adipocytes increased catecholamine-stimulated lipolysis. In addition, inhibition of miR-378 expression attenuated stimulated lipolysis and reduced the expression of LIPE, PLIN1, and PNPLA2, a set of genes encoding key lipolytic regulators. Taken together, increased miR-378 expression could play an etiological role in cancer cachexia-associated adipose tissue loss via effects on adipocyte lipolysis.


Asunto(s)
Tejido Adiposo/metabolismo , Caquexia/etiología , Lipólisis/genética , MicroARNs/fisiología , Neoplasias/complicaciones , Adolescente , Anciano , Caquexia/metabolismo , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , MicroARNs/genética , Análisis por Micromatrices , Persona de Mediana Edad , Neoplasias/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-38967296

RESUMEN

OBJECTIVE: The objective of this study was to identify the transcriptional landscape of insulin resistance (IR) in subcutaneous adipose tissue (SAT) in humans across the spectrum of obesity. METHODS: We used SAT RNA sequencing in 220 individuals with metabolic phenotyping. RESULTS: We identified a 35-gene signature with high predictive accuracy for homeostatic model of IR that was expressed across a variety of non-immune cell populations. We observed primarily "protective" IR associations for adipocyte transcripts and "deleterious" associations for macrophage transcripts, as well as a high concordance between SAT and visceral adipose tissue (VAT). Multiple SAT genes exhibited dynamic expression 5 years after weight loss surgery and with insulin stimulation. Using available expression quantitative trait loci in SAT and/or VAT, we demonstrated similar genetic effect sizes of SAT and VAT on type 2 diabetes and BMI. CONCLUSIONS: SAT is conventionally viewed as a metabolic buffer for lipid deposition during positive energy balance, whereas VAT is viewed as a dominant contributor to and prime mediator of IR and cardiometabolic disease risk. Our results implicate a dynamic transcriptional architecture of IR that resides in both immune and non-immune populations in SAT and is shared with VAT, nuancing the current VAT-centric concept of IR in humans.

15.
BMC Endocr Disord ; 13: 5, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23360456

RESUMEN

BACKGROUND: Genetic studies have implicated the NPC1 gene (Niemann Pick type C1) in susceptibility to obesity. METHODS: To assess the potential function of NPC1 in obesity, we determined its expression in abdominal white adipose tissue (WAT) in relation to obesity. NPC1 mRNA was measured by RT-qPCR in lean and obese individuals, paired samples of subcutaneous (sc) and omental (om) WAT, before and after weight loss, in isolated adipocytes and intact adipose pieces, and in primary adipocyte cultures during adipocyte differentiation. NPC1 protein was examined in isolated adipocytes. RESULTS: NPC1 mRNA was significantly increased in obese individuals in scWAT and omWAT and downregulated by weight loss. NPC1 mRNA was enriched in isolated fat cells of WAT, in scWAT versus omWAT but not modified during adipocyte differentiation. NPC1 protein mirrored expression of mRNA in lean and obese individuals. CONCLUSIONS: NPC1 is highly expressed in human WAT adipocytes with increased levels in obese. These results suggest that NPC1 may play a role in adipocyte processes underlying obesity.

16.
BMC Endocr Disord ; 13: 54, 2013 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-24267103

RESUMEN

BACKGROUND: Allograft inflammatory factor 1 (AIF-1) is a putative obesity gene. Our aim was to examine the expression of AIF-1 in human white adipose tissue (WAT) in relation to obesity and metabolic phenotypes in women. METHODS: WAT secretion of AIF-1 was determined in subcutaneous adipose tissue pieces in vitro by ELISA from 5 subjects. mRNA expression of AIF-1 was determined by RT-qPCR in the isolated cell fractions of adipose tissue (n = 5-6 per group), in subcutaneous and visceral WAT pieces from non-obese (n = 12) and obese women (n = 23), and in some subcutaneous WAT also before and after weight reduction (n = 10). Finally, adipose AIF-1 mRNA was related to metabolic phenotypes in 96 subjects with a wide range of BMI. RESULTS: AIF-1 was secreted in a time dependent fashion from WAT. The major source of AIF-1 was WAT resident macrophages. Expression of AIF-1 was similar in visceral and subcutaneous WAT and was two-fold increased in obese women (P < 0.01). AIF-1 mRNA expression levels were normalized after weight reduction (P < 0.01). Expression of AIF-1 was inversely correlated with insulin sensitivity as assessed by insulin tolerance test (KITT), and circulating levels of adiponectin (P = 0.02), and positively correlated with insulin resistance as estimated by HOMA (=0.0042). CONCLUSIONS: AIF-1 is a novel adipokine produced mainly by macrophages within human WAT. Its expression is increased in obese women and associates with unfavourable metabolic phenotypes. AIF-1 may play a paracrine role in the regulation of WAT function through cross-talk between macrophages and other cell types within the adipose tissue.

17.
Nat Genet ; 30(2): 149-50, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11799396

RESUMEN

It is increasingly apparent that the identification of true genetic associations in common multifactorial disease will require studies comprising thousands rather than the hundreds of individuals employed to date. Using 2,873 families, we were unable to confirm a recently published association of the interleukin 12B gene in 422 type I diabetic families. These results emphasize the need for large datasets, small P values and independent replication if results are to be reliable.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Regiones no Traducidas 3' , Bases de Datos Genéticas , Diabetes Mellitus Tipo 1/inmunología , Genética de Población , Humanos , Interleucina-12/genética , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple
18.
Diabetes ; 72(3): 316-325, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36445942

RESUMEN

Sedentary people have insulin resistance in their skeletal muscle, but whether this also occurs in fat cells was unknown. Insulin inhibition of hydrolysis of triglycerides (antilipolysis) and stimulation of triglyceride formation (lipogenesis) were investigated in subcutaneous fat cells from 204 sedentary and 336 physically active subjects. Insulin responsiveness (maximum hormone effect) and sensitivity (half-maximal effective concentration) were determined. In 69 women, hyperinsulinemia-induced circulating fatty acid levels were measured. In 128 women, adipose gene expression was analyzed. Responsiveness of insulin for antilipolysis (60% inhibition) and lipogenesis (twofold stimulation) were similar between sedentary and active subjects. Sensitivity for both measures decreased ˜10-fold in sedentary subjects (P < 0.01). However, upon multiple regression analysis, only the association between antilipolysis sensitivity and physical activity remained significant when adjusting for BMI, age, sex, waist-to-hip ratio, fat-cell size, and cardiometabolic disorders. Fatty acid levels decreased following hyperinsulinemia but remained higher in sedentary compared with active women (P = 0.01). mRNA expression of insulin receptor and its substrates 1 and 2 was decreased in sedentary subjects. In conclusion, while the maximum effect is preserved, sensitivity to insulin's antilipolytic effect in subcutaneous fat cells is selectively lower in sedentary subjects.


Asunto(s)
Hiperinsulinismo , Resistencia a la Insulina , Humanos , Femenino , Resistencia a la Insulina/fisiología , Glucemia/metabolismo , Conducta Sedentaria , Obesidad/metabolismo , Insulina/metabolismo , Hiperinsulinismo/metabolismo , Ácidos Grasos/metabolismo , Tejido Adiposo/metabolismo
19.
United European Gastroenterol J ; 11(1): 79-91, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36454055

RESUMEN

BACKGROUND: Post-pancreatitis diabetes mellitus (PPDM) is a common consequence of chronic pancreatitis (CP). We aimed to determine the incidence and predictors of PPDM after CP onset, as well as complications and antidiabetic therapy requirements, in a high-volume tertiary center. METHODS: We did a cohort study with retrospectively collected data from patients with definite CP seen at the Karolinska University Hospital between January 1999 and December 2020. Cause-specific Cox regression analysis was used to assess PPDM predictors. To estimate risk of complications and need for therapy the Fine-Gray subdistribution hazard model was employed, accounting for death as a competing risk. RESULTS: We identified 481 patients with CP. The cumulative incidence of PPDM was 5.1%, 13.2%, 27.5% and 38.9% at 5, 10, 15 and 20 years, respectively. Compared to CP patients without diabetes, patients with PPDM were predominantly male (55% vs. 75%), had more frequently alcoholic etiology (44% vs. 62%) and previous acute pancreatitis. The only independent predictor of PPDM was presence of pancreatic calcifications (aHR = 2.45, 95% CI 1.30-4.63). Patients with PPDM had higher rates of microangiopathy (aSHR = 1.59, 95% CI 1.02-2.52) and infection (aSHR = 4.53, 95% CI 2.60-9.09) compared to CP patients who had type 2 diabetes (T2DM). The rate of insulin use was three-fold higher, whereas metformin use rate was two-fold higher in the same comparison. CONCLUSIONS: Patients with PPDM have a higher frequency of clinically significant complications and were more commonly prescribed insulin and metformin, suggesting a more aggressive phenotype than that of T2DM. Greater PPDM awareness is needed to optimize disease management.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insulinas , Metformina , Pancreatitis Crónica , Masculino , Femenino , Humanos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/epidemiología , Estudios de Cohortes , Estudios Retrospectivos , Enfermedad Aguda , Pancreatitis Crónica/complicaciones , Pancreatitis Crónica/diagnóstico , Pancreatitis Crónica/epidemiología , Metformina/uso terapéutico
20.
J Biol Chem ; 286(1): 370-9, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21030586

RESUMEN

The Liver X receptor (LXR) is an important regulator of carbohydrate and lipid metabolism in humans and mice. We have recently shown that activation of LXR regulates cellular fuel utilization in adipocytes. In contrast, the role of LXR in human adipocyte lipolysis, the major function of human white fat cells, is not clear. In the present study, we stimulated in vitro differentiated human and murine adipocytes with the LXR agonist GW3965 and observed an increase in basal lipolysis. Microarray analysis of human adipocyte mRNA following LXR activation revealed an altered gene expression of several lipolysis-regulating proteins, which was also confirmed by quantitative real-time PCR. We show that expression and intracellular localization of perilipin1 (PLIN1) and hormone-sensitive lipase (HSL) are affected by GW3965. Although LXR activation does not influence phosphorylation status of HSL, HSL activity is required for the lipolytic effect of GW3965. This effect is abolished by PLIN1 knockdown. In addition, we demonstrate that upon activation, LXR binds to the proximal regions of the PLIN1 and HSL promoters. By selective knock-down of either LXR isoform, we show that LXRα is the major isoform mediating the lipolysis-related effects of LXR. In conclusion, the present study demonstrates that activation of LXRα up-regulates basal human adipocyte lipolysis. This is at least partially mediated through LXR binding to the PLIN1 promoter and down-regulation of PLIN1 expression.


Asunto(s)
Adipocitos/metabolismo , Lipólisis , Receptores Nucleares Huérfanos/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos Blancos/citología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Animales , Proteínas Portadoras , Regulación hacia Abajo/efectos de los fármacos , Humanos , Resistencia a la Insulina , Lipólisis/efectos de los fármacos , Receptores X del Hígado , Ratones , Receptores Nucleares Huérfanos/agonistas , Perilipina-1 , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptores X Retinoide/metabolismo , Transducción de Señal/efectos de los fármacos , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA