Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 47(9): e49, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30793190

RESUMEN

In most cells, transcriptionally inactive heterochromatin is preferentially localized in the nuclear periphery and transcriptionally active euchromatin is localized in the nuclear interior. Different cell types display characteristic chromatin distribution patterns, which change dramatically during cell differentiation, proliferation, senescence and different pathological conditions. Chromatin organization has been extensively studied on a cell population level, but there is a need to understand dynamic reorganization of chromatin at the single cell level, especially in live cells. We have developed a novel image analysis tool that we term Fluorescence Ratiometric Imaging of Chromatin (FRIC) to quantitatively monitor dynamic spatiotemporal distribution of euchromatin and total chromatin in live cells. A vector (pTandemH) assures stoichiometrically constant expression of the histone variants Histone 3.3 and Histone 2B, fused to EGFP and mCherry, respectively. Quantitative ratiometric (H3.3/H2B) imaging displayed a concentrated distribution of heterochromatin in the periphery of U2OS cell nuclei. As proof of concept, peripheral heterochromatin responded to experimental manipulation of histone acetylation. We also found that peripheral heterochromatin depended on the levels of the inner nuclear membrane protein Samp1, suggesting an important role in promoting peripheral heterochromatin. Taken together, FRIC is a powerful and robust new tool to study dynamic chromatin redistribution in live cells.


Asunto(s)
Cromatina/genética , Proteínas de la Membrana/genética , Imagen Molecular/métodos , Proteínas Nucleares/genética , Acetilación , Línea Celular , Núcleo Celular/genética , Eucromatina/genética , Heterocromatina/genética , Histonas/genética , Humanos , Membrana Nuclear/genética , Procesamiento Proteico-Postraduccional/genética
2.
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
3.
Diabetes Obes Metab ; 20(10): 2416-2425, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29862627

RESUMEN

AIM: To examine the cell membrane transporters involved in mediating the antilipolytic effect of biguanides in human fat cells. MATERIALS AND METHODS: Gene expression of biguanide transporters was mapped in human subcutaneous adipose tissue and in adipocytes before and after differentiation. Those expressed in mature fat cells were knocked down by RNA interference (RNAi) and the antilipolytic effects of metformin and two novel, highly potent biguanides, NT1014 and NT1044, were examined. RESULTS: Analysis of the transporter affinity of biguanides in HEK293 cells overexpressing individual transporters showed that NT1014 and NT1044 had >10 times higher affinity than metformin. Animal studies showed that NT1014 was >5 times more potent than metformin in lowering plasma glucose in mice. In human fat cells, the novel biguanides displayed higher AMP-activated protein kinase activation and antilipolytic efficacy than metformin. Five transporters, organic cation transporter (OCT)1 (SLC22A1), organic cation transporter novel type 1 (OCTN1; SLC22A4), OCT3 (SLC22A3), plasma membrane monoamine transporter (PMAT; SLC29A4) and multidrug and toxin extrusion transporter (MATE1; SLC47A1), were detectable in fat cells but only OCT3, PMAT and MATE1 increased during adipogenesis in vitro and were enriched in fat cells compared with other adipose cell types. Gene knockdown by RNAi showed that MATE1 and PMAT reduction attenuated the antilipolytic effect of metformin but only PMAT knockdown decreased the effect of all three biguanides. CONCLUSIONS: While human fat cells primarily express three biguanide transporters, our data suggest that PMAT is the primary target for development of fat cell-specific antilipolytic biguanides with high sensitivity and potency.


Asunto(s)
Adipocitos/metabolismo , Biguanidas/metabolismo , Lipólisis/genética , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Adipocitos/patología , Adulto , Anciano , Animales , Biguanidas/uso terapéutico , Transporte Biológico/genética , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Distribución Tisular
4.
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
5.
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
6.
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.

7.
Front Endocrinol (Lausanne) ; 14: 1166961, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37361522

RESUMEN

Background and purpose: Napping is a widespread practice worldwide and has in recent years been linked to increased abdominal adiposity. Lipase E or LIPE encodes the protein hormone-sensitive lipase (HSL), an enzyme that plays an important role in lipid mobilization and exhibits a circadian expression rhythm in human adipose tissue. We hypothesized that habitual napping may impact the circadian expression pattern of LIPE, which in turn may attenuate lipid mobilization and induce abdominal fat accumulation. Methods: Abdominal adipose tissue explants from participants with obesity (n = 17) were cultured for a 24-h duration and analyzed every 4 h. Habitual nappers (n = 8) were selected to match non-nappers (n = 9) in age, sex, BMI, adiposity, and metabolic syndrome traits. Circadian LIPE expression rhythmicity was analyzed using the cosinor method. Results: Adipose tissue explants exhibited robust circadian rhythms in LIPE expression in non-nappers. In contrast, nappers had a flattened rhythm. LIPE amplitude was decreased in nappers as compared with non-nappers (71% lower). The decrease in amplitude among nappers was related to the frequency of napping (times per week) where a lower rhythm amplitude was associated with a higher napping frequency (r = -0.80; P = 0.018). Confirmatory analyses in the activity of LIPE's protein (i.e., HSL) also showed a significant rhythm in non-nappers, whereas significance in the activity of HSL was lost among nappers. Conclusion: Our results suggest that nappers display dysregulated circadian LIPE expression as well as dysregulated circadian HSL activity, which may alter lipid mobilization and contribute to increased abdominal obesity in habitual nappers.


Asunto(s)
Tejido Adiposo , Lipasa , Esterol Esterasa , Humanos , Grasa Abdominal/metabolismo , Tejido Adiposo/metabolismo , Ritmo Circadiano , Obesidad/metabolismo , Esterol Esterasa/metabolismo
8.
Sci Rep ; 12(1): 3666, 2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35256633

RESUMEN

Total body fat and central fat distribution are heritable traits and well-established predictors of adverse metabolic outcomes. Lipolysis is the process responsible for the hydrolysis of triacylglycerols stored in adipocytes. To increase our understanding of the genetic regulation of body fat distribution and total body fat, we set out to determine if genetic variants associated with body mass index (BMI) or waist-hip-ratio adjusted for BMI (WHRadjBMI) in genome-wide association studies (GWAS) mediate their effect by influencing adipocyte lipolysis. We utilized data from the recent GWAS of spontaneous and isoprenaline-stimulated lipolysis in the unique GENetics of Adipocyte Lipolysis (GENiAL) cohort. GENiAL consists of 939 participants who have undergone abdominal subcutaneous adipose biopsy for the determination of spontaneous and isoprenaline-stimulated lipolysis in adipocytes. We report 11 BMI and 15 WHRadjBMI loci with SNPs displaying nominal association with lipolysis and allele-dependent gene expression in adipose tissue according to in silico analysis. Functional evaluation of candidate genes in these loci by small interfering RNAs (siRNA)-mediated knock-down in adipose-derived stem cells identified ZNF436 and NUP85 as intrinsic regulators of lipolysis consistent with the associations observed in the clinical cohorts. Furthermore, candidate genes in another BMI-locus (STX17) and two more WHRadjBMI loci (NID2, GGA3, GRB2) control lipolysis alone, or in conjunction with lipid storage, and may hereby be involved in genetic control of body fat. The findings expand our understanding of how genetic variants mediate their impact on the complex traits of fat storage and distribution.


Asunto(s)
Estudio de Asociación del Genoma Completo , Lipólisis , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Sitios Genéticos , Humanos , Isoproterenol/metabolismo , Lipólisis/genética , Factores de Transcripción/metabolismo
9.
Diabetes ; 71(6): 1350-1362, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35320353

RESUMEN

Interindividual differences in generation of new fat cells determine body fat and type 2 diabetes risk. In the GENetics of Adipocyte Lipolysis (GENiAL) cohort, which consists of participants who have undergone abdominal adipose biopsy, we performed a genome-wide association study (GWAS) of fat cell number (n = 896). Candidate genes from the genetic study were knocked down by siRNA in human adipose-derived stem cells. We report 318 single nucleotide polymorphisms (SNPs) and 17 genetic loci displaying suggestive (P < 1 × 10-5) association with fat cell number. Two loci pass threshold for GWAS significance, on chromosomes 2 (lead SNP rs149660479-G) and 7 (rs147389390-deletion). We filtered for fat cell number-associated SNPs (P < 1.00 × 10-5) using evidence of genotype-specific expression. Where this was observed we selected genes for follow-up investigation and hereby identified SPATS2L and KCTD18 as regulators of cell proliferation consistent with the genetic data. Furthermore, 30 reported type 2 diabetes-associated SNPs displayed nominal and consistent associations with fat cell number. In functional follow-up of candidate genes, RPL8, HSD17B12, and PEPD were identified as displaying effects on cell proliferation consistent with genetic association and gene expression findings. In conclusion, findings presented herein identify SPATS2L, KCTD18, RPL8, HSD17B12, and PEPD of potential importance in controlling fat cell numbers (plasticity), the size of body fat, and diabetes risk.


Asunto(s)
Diabetes Mellitus Tipo 2 , Estudio de Asociación del Genoma Completo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Recuento de Células , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Sitios Genéticos , Humanos , Polimorfismo de Nucleótido Simple
10.
Physiol Rep ; 8(16): e14538, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32812397

RESUMEN

BACKGROUND: In obesity, the expression level of thyroid stimulating hormone receptor in adipose tissue is reduced and the levels of thyroid stimulating hormone (TSH) are often elevated within the normal range. PURPOSE/AIM: To investigate the role of TSHR in brown and white adipose tissue (AT) using TSHR knockout (KO) mice and the physiological phenotypes affected by the TSHR knockout. METHODS: AT-specific TSHR KO male mice and wild type (WT) controls were given a high-fat diet (HFD) or a control diet (CD). Body weights and food consumption were recorded for 20 weeks and body temperatures for the first 3 weeks. At termination, white and brown adipocytes were isolated. Gene expressios was investigated using real-time PCR. In a subgroup of female KO mice, glucose tolerance was investigated. RESULTS: TSHR were partially knocked out in KO mice, which gained more weight than WT mice when fed both a CD (p = .03) and HFD (p = .003). Body temperatures were lower in KO mice on CD (p <.001) and on HFD (p <.001) than WT controls. This was in agreement with reduced gene expression of UCP1 in brown adipocytes in the KO mice. Glucose tolerance was significantly impaired in KO mice on CD mice before termination (p <.01). Expression of adipogenic and lipolytic genes were reduced in KO mice, which was exacerbated by HFD. The mRNA levels of adipokines including ADIPOQ and LEP were altered in white adipocytes of KO mice. CONCLUSIONS: TSHR KO led to dysfunction of both white and brown AT and predisposition to excess body weight gain in mice. Our data show that TSHR in AT regulates glucose tolerance, lipid metabolism, adipokine profile, and thermogenesis.


Asunto(s)
Adipocitos/metabolismo , Temperatura Corporal , Peso Corporal , Receptores de Tirotropina/metabolismo , Animales , Células Cultivadas , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Tirotropina/genética , Transcriptoma , Aumento de Peso
11.
Cells ; 9(5)2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32349335

RESUMEN

An increased adipocyte size relative to the size of fat depots, also denoted hypertrophic adipose morphology, is a strong risk factor for the future development of insulin resistance and type 2 diabetes. The regulation of adipose morphology is poorly understood. We set out to identify genetic loci associated with adipose morphology and functionally evaluate candidate genes for impact on adipocyte development. We performed a genome-wide association study (GWAS) in the unique GENetics of Adipocyte Lipolysis (GENiAL) cohort comprising 948 participants who have undergone abdominal subcutaneous adipose biopsy with a determination of average adipose volume and morphology. The GWAS identified 31 genetic loci displaying suggestive association with adipose morphology. Functional evaluation of candidate genes by small interfering RNAs (siRNA)-mediated knockdown in adipose-derived precursor cells identified six genes controlling adipocyte renewal and differentiation, and thus of potential importance for adipose hypertrophy. In conclusion, genetic and functional studies implicate a regulatory role for ATL2, ARHGEF10, CYP1B1, TMEM200A, C17orf51, and L3MBTL3 in adipose morphology by their impact on adipogenesis.


Asunto(s)
Adipocitos/citología , Diabetes Mellitus Tipo 2/genética , Obesidad/genética , Adipocitos/fisiología , Adipogénesis/genética , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Adiposidad , Adulto , Diferenciación Celular , Estudios de Cohortes , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Lipólisis/fisiología , Masculino , Persona de Mediana Edad , Grasa Subcutánea
12.
J Clin Endocrinol Metab ; 105(12)2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32725188

RESUMEN

BACKGROUND: Fat mobilization in adipose tissue (AT) has a specific timing. However, circadian rhythms in the activity of the major enzyme responsible for fat mobilization, hormone-sensitive lipase (HSL), have not been demonstrated in humans. OBJECTIVE: To analyze in a cross-sectional study whether there is an endogenous circadian rhythm in HSL activity in human AT ex vivo and whether rhythm characteristics are related to food timing or fasting duration. METHODS: Abdominal AT biopsies were obtained from 18 severely obese participants (age: 46 ± 11 years; body mass index 42 ± 6 kg/m2) who underwent laparoscopic gastric bypass. Twenty-four-hour rhythms of HSL activity and LIPE (HSL transcript in humans) expression in subcutaneous AT were analyzed together with habitual food timing and night fasting duration. RESULTS: HSL activity exhibited a circadian rhythm (P = .023) and reached the maximum value at circadian time 16 (CT) that corresponded to around midnight (relative local clock time. Similarly, LIPE displayed a circadian rhythm with acrophase also at night (P = .0002). Participants with longer night fasting duration >11.20 hours displayed almost double the amplitude (1.91 times) in HSL activity rhythm than those with short duration (P = .013); while habitual early diners (before 21:52 hours) had 1.60 times higher amplitude than late diners (P = .035). CONCLUSIONS: Our results demonstrate circadian rhythms in HSL activity and may lead to a better understanding of the intricate relationships between food timing, fasting duration and body fat regulation.


Asunto(s)
Tejido Adiposo/metabolismo , Ritmo Circadiano/fisiología , Ayuno/metabolismo , Obesidad/metabolismo , Esterol Esterasa/metabolismo , Adulto , Estudios Transversales , Femenino , Derivación Gástrica , Humanos , Estilo de Vida , Masculino , Persona de Mediana Edad , Obesidad/cirugía
13.
Mol Metab ; 34: 85-96, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32180562

RESUMEN

OBJECTIVES: Lipolysis, hydrolysis of triglycerides to fatty acids in adipocytes, is tightly regulated, poorly understood, and, if perturbed, can lead to metabolic diseases including obesity and type 2 diabetes. The goal of this study was to identify the genetic regulators of lipolysis and elucidate their molecular mechanisms. METHODS: Adipocytes from abdominal subcutaneous adipose tissue biopsies were isolated and were incubated without (spontaneous lipolysis) or with a catecholamine (stimulated lipolysis) to analyze lipolysis. DNA was extracted and genome-wide genotyping and imputation conducted. After quality control, 939 samples with genetic and lipolysis data were available. Genome-wide association studies of spontaneous and stimulated lipolysis were conducted. Subsequent in vitro gene expression analyses were used to identify candidate genes and explore their regulation of adipose tissue biology. RESULTS: One locus on chromosome 19 demonstrated genome-wide significance with spontaneous lipolysis. 60 loci showed suggestive associations with spontaneous or stimulated lipolysis, of which many influenced both traits. In the chromosome 19 locus, only HIF3A was expressed in the adipocytes and displayed genotype-dependent gene expression. HIF3A knockdown in vitro increased lipolysis and the expression of key lipolysis-regulating genes. CONCLUSIONS: In conclusion, we identified a genetic regulator of spontaneous lipolysis and provided evidence of HIF3A as a novel key regulator of lipolysis in subcutaneous adipocytes as the mechanism through which the locus influences adipose tissue biology.


Asunto(s)
Adipocitos/metabolismo , Estudio de Asociación del Genoma Completo , Lipólisis/genética , Tejido Adiposo/metabolismo , Adulto , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Cromosomas Humanos Par 19/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética
14.
Sci Rep ; 9(1): 13891, 2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31554889

RESUMEN

MicroRNAs (miRNA) modulate gene expression through feed-back and forward loops. Previous studies identified miRNAs that regulate transcription factors, including Peroxisome Proliferator Activated Receptor Gamma (PPARG), in adipocytes, but whether they influence adipogenesis via such regulatory loops remain elusive. Here we predicted and validated a novel feed-forward loop regulating adipogenesis and involved miR-27a/b-3p, PPARG and Secretory Carrier Membrane Protein 3 (SCAMP3). In this loop, expression of both PPARG and SCAMP3 was independently suppressed by miR-27a/b-3p overexpression. Knockdown of PPARG downregulated SCAMP3 expression at the late phase of adipogenesis, whereas reduction of SCAMP3 mRNA levels increased PPARG expression at early phase in differentiation. The latter was accompanied with upregulation of adipocyte-enriched genes, including ADIPOQ and FABP4, suggesting an anti-adipogenic role for SCAMP3. PPARG and SCAMP3 exhibited opposite behaviors regarding correlations with clinical phenotypes, including body mass index, body fat mass, adipocyte size, lipolytic and lipogenic capacity, and secretion of pro-inflammatory cytokines. While adipose PPARG expression was associated with more favorable metabolic phenotypes, SCAMP3 expression was linked to increased fat mass and insulin resistance. Together, we identified a feed-forward loop through which miR-27a/b-3p, PPARG and SCAMP3 cooperatively fine tune the regulation of adipogenesis, which potentially may impact whole body metabolism.


Asunto(s)
Adipogénesis/genética , Proteínas Portadoras/genética , Proteínas de la Membrana/genética , MicroARNs/genética , PPAR gamma/genética , Adipocitos/fisiología , Adipogénesis/fisiología , Tejido Adiposo/fisiología , Índice de Masa Corporal , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Regulación hacia Abajo/genética , Regulación hacia Abajo/fisiología , Femenino , Humanos , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Lipogénesis/genética , Lipogénesis/fisiología , Fenotipo , ARN Mensajero/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
15.
J Clin Endocrinol Metab ; 104(10): 4552-4562, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31132124

RESUMEN

OBJECTIVE: Although IL-10 is generally considered as an anti-inflammatory cytokine, it was recently shown to have detrimental effects on insulin sensitivity and fat cell metabolism in rodents. Whether this also pertains to human white adipose tissue (hWAT) is unclear. We therefore determined the main cellular source and effects of IL-10 on human adipocytes and hWAT-resident immune cells and its link to insulin resistance. METHODS: Associations between hWAT IL-10 production and metabolic parameters were investigated in 216 participants with large interindividual variations in body mass index and insulin sensitivity. Adipose cells expressing or secreting IL-10 and the cognate IL-10 receptor α (IL10RA) were identified by flow cytometry sorting. Effects on adipogenesis, lipolysis, and inflammatory/metabolic gene expression were measured in two human primary adipocyte models. Secretion of inflammatory cytokines was investigated in cultures of IL-10-treated hWAT macrophages and leukocytes by Luminex analysis (Luminex Corp.). RESULTS: IL-10 gene expression and protein secretion in hWAT correlated positively with body mass index (BMI) and homeostasis model assessment-insulin resistance (HOMA-IR). Gene expression analyses in mature fat cells and flow cytometry-sorted hWAT-resident adipocyte progenitors, macrophages, and leukocytes demonstrated that the expression of IL-10 and the IL10RA were significantly enriched in proinflammatory M1 macrophages. In contrast to murine data, functional studies showed that recombinant IL-10 had no effect on adipocyte phenotype. In hWAT-derived macrophages and leukocytes, it induced an anti-inflammatory profile. CONCLUSION: In hWAT, IL-10 is upregulated in proinflammatory macrophages of obese and insulin-resistant persons. However, in contrast to findings in mice, IL-10 does not directly affect human adipocyte function.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Resistencia a la Insulina , Interleucina-10/metabolismo , Macrófagos/metabolismo , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo/metabolismo , Adulto , Estudios de Casos y Controles , Células Cultivadas , Femenino , Citometría de Flujo , Expresión Génica , Humanos , Técnicas In Vitro , Inflamación/genética , Interleucina-10/farmacología , Subunidad alfa del Receptor de Interleucina-10/metabolismo , Grasa Intraabdominal , Lipólisis/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Persona de Mediana Edad , Obesidad Metabólica Benigna , ARN Mensajero , Grasa Subcutánea , Células THP-1 , Adulto Joven
16.
Metabolism ; 101: 153999, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31672447

RESUMEN

BACKGROUND: Adipose tissue plays a crucial role in diet- and obesity-related insulin resistance, with implications for several metabolic diseases. Identification of novel target genes and mechanisms that regulate adipocyte function could lead to improved treatment strategies. RND3 (RhoE/Rho8), a Rho-related GTP-binding protein that inhibits Rho kinase (ROCK) signaling, has been linked to diverse diseases such as apoptotic cardiomyopathy, heart failure, cancer and type 2 diabetes, in part by regulating cytoskeleton dynamics and insulin-mediated glucose uptake. RESULTS: We here investigated the expression of RND3 in adipose tissue in human obesity, and discovered a role for RND3 in regulating adipocyte metabolism. In cross-sectional and prospective studies, we observed 5-fold increased adipocyte levels of RND3 mRNA in obesity, reduced levels after surgery-induced weight loss, and positive correlations of RND3 mRNA with adipocyte size and surrogate measures of insulin resistance (HOMA2-IR and circulating triglyceride/high-density lipoprotein cholesterol (TAG/HDL-C) ratio). By screening for RND3-dependent gene expression following siRNA-mediated RND3 knockdown in differentiating human adipocytes, we found downregulation of inflammatory genes and upregulation of genes related to adipocyte ipolysis and insulin signaling. Treatment of adipocytes with tumor necrosis factor alpha (TNFα), lipopolysaccharide (LPS), hypoxia or cAMP analogs increased RND3 mRNA levels 1.5-2-fold. Functional assays in primary human adipocytes confirmed that RND3 knockdown reduces cAMP- and isoproterenol-induced lipolysis, which were mimicked by treating cells with ROCK inhibitor. This effect could partly be explained by reduced protein expression of adipose triglyceride lipase (ATGL) and phosphorylated hormone-sensitive lipase (HSL). CONCLUSION: We here uncovered a novel differential expression of adipose RND3 in obesity and insulin resistance, which may at least partly depend on a causal effect of RND3 on adipocyte lipolysis.


Asunto(s)
Adipocitos/metabolismo , Lipólisis/efectos de los fármacos , Proteínas de Unión al GTP rho/fisiología , Animales , Células Cultivadas , Estudios Transversales , Regulación de la Expresión Génica , Humanos , Resistencia a la Insulina , Obesidad/metabolismo , Estudios Prospectivos , ARN Mensajero/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
17.
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
18.
Mol Cell Endocrinol ; 472: 50-56, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-29191698

RESUMEN

Reduced adipose expression of the transcription factor Early B cell factor 1 (EBF1) is linked to white adipose tissue (WAT) hypertrophy. We aimed to identify microRNAs (miRNAs) associated with WAT hypertrophy and EBF1 regulation. We mapped WAT miRNA expression from 26 non-obese women discordant in WAT morphology and determined EBF1 activity in the non-obese and 30 obese women. Expression of 15 miRNAs was higher in hypertrophy and 10 were predicted to target EBF1. Binding of miR-365-5p/miR-574-5p were validated with 3'-UTR assay. Overexpression of miR-365-5p or miR-574-5p reduced EBF1 while inhibition of miR-574 increased EBF1 expression in human adipocytes in vitro. Additive effects on EBF1 were observed when concomitantly overexpressing both miRNAs. EBF1 targets were affected by over expression/inhibition of either miRNAs. Finally, miR-365-5p/miR-574-5p expression in 56 individuals correlated significantly with EBF1 activity. Our results suggest that miR-365-5p and miR-574-5p may be linked to WAT hypertrophy via effects on EBF1 expression.


Asunto(s)
Tejido Adiposo Blanco/anatomía & histología , Tejido Adiposo Blanco/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Transactivadores/genética , Adipocitos/metabolismo , Adipocitos/patología , Diferenciación Celular/genética , Femenino , Humanos , Hipertrofia , MicroARNs/genética , Reproducibilidad de los Resultados , Transactivadores/metabolismo
19.
Cell Rep ; 25(3): 551-560.e5, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332637

RESUMEN

White adipose tissue (WAT) mass is determined by adipocyte size and number. While adipocytes are continuously turned over, the mechanisms controlling fat cell number in WAT upon weight changes are unclear. Herein, prospective studies of human subcutaneous WAT demonstrate that weight gain increases both adipocyte size and number, but the latter remains unaltered after weight loss. Transcriptome analyses associate changes in adipocyte number with the expression of 79 genes. This gene set is enriched for growth factors, out of which one, transforming growth factor-ß3 (TGFß3), stimulates adipocyte progenitor proliferation, resulting in a higher number of cells undergoing differentiation in vitro. The relevance of these observations was corroborated in vivo where Tgfb3+/- mice, in comparison with wild-type littermates, display lower subcutaneous adipocyte progenitor proliferation, WAT hypertrophy, and glucose intolerance. TGFß3 is therefore a regulator of subcutaneous adipocyte number and may link WAT morphology to glucose metabolism.


Asunto(s)
Adipogénesis , Tejido Adiposo Blanco/patología , Intolerancia a la Glucosa/etiología , Obesidad/complicaciones , Grasa Subcutánea/patología , Factor de Crecimiento Transformador beta3/fisiología , Tejido Adiposo Blanco/metabolismo , Adolescente , Animales , Estudios de Casos y Controles , Diferenciación Celular , Femenino , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estudios Prospectivos , Grasa Subcutánea/metabolismo
20.
Mol Biol Cell ; 13(12): 4195-205, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12475945

RESUMEN

The biochemical properties and spatial localization of the protein alpha-dystrobrevin and other isoforms were investigated in cells of the human promyelocytic leukemia line HL-60 granulocytic differentiation as induced by retinoic acid (RA). Alpha-dystrobrevin was detected both in the cytosol and the nuclei of these cells, and a short isoform (gamma-dystrobrevin) was modified by tyrosine phosphorylation soon after the onset of the RA-triggered differentiation. Varying patterns of distribution of alpha-dystrobrevin and its isoforms could be discerned in HL-60 promyelocytes, RA-differentiated mature granulocytes, and human neutrophils. Moreover, the gamma-dystrobrevin isoform was found in association with actin and myosin light chain. The results provide new information about potential involvement of alpha-dystrobrevin and its splice isoforms in signal transduction in myeloid cells during induction of granulocytic differentiation and/or at the commitment stage of differentiation or phagocytic cells.


Asunto(s)
Proteínas del Citoesqueleto/química , Proteínas Asociadas a la Distrofina , Granulocitos/citología , Proteínas de la Membrana/química , Actinas/metabolismo , Diferenciación Celular , Núcleo Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Citosol/metabolismo , Electroforesis en Gel de Poliacrilamida , Granulocitos/metabolismo , Células HL-60 , Humanos , Immunoblotting , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Cadenas Ligeras de Miosina/metabolismo , Neutrófilos/metabolismo , Fagocitosis , Fosforilación , Pruebas de Precipitina , Isoformas de Proteínas , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Tiempo , Tripsina/farmacología , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA