Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 340
Filter
1.
Int J Obes (Lond) ; 48(4): 523-532, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38135702

ABSTRACT

BACKGROUND AND AIMS: Obesity predisposes to metabolic and cardiovascular diseases. Adipose tissue inflammation and systemic inflammation contribute to these complications. There are strong sex differences in adipose tissue distribution and in systemic inflammation. Women have more subcutaneous adipose tissue (SAT) and less visceral adipose tissue (VAT) than men. We explored the sex differences in the association between the different adipose compartments and inflammatory markers that are important in cardiometabolic disease pathophysiology. METHODS: Single-center observational cohort study with 302 individuals with a BMI ≥ 27 kg/m2. We were unable to acquire MRI data from seven individuals and from another 18 the MRI data were not usable, resulting in 277 people (155 men, 122 women), aged 55-81 years. INTERVENTION: We performed the following measurements: abdominal magnetic resonance imaging to measure VAT, and SAT (deep and superficial) volumes; circulating leukocyte counts and cytokine production capacity of peripheral blood mononuclear cells (PBMCs), circulating cytokines, adipokines, and targeted proteomics; abdominal sSAT biopsies for histology and gene expression. RESULTS: Only in women, (s)SAT volume was associated with circulating leukocytes, monocytes, and neutrophils. Circulating IL-6 and IL-18BP were associated with SAT volume in women and VAT in men. Several circulating proteins, including monocyte-colony-stimulating factor 1 and hepatocyte growth factor, are associated with sSAT in women and VAT in men. Only in women, SAT volume is associated with SAT expression of inflammatory proteins, including leptin, CD68, TNFα and IL-1α. CONCLUSION: In women living with obesity, abdominal SAT volume, especially sSAT, is associated with circulating leukocytes and inflammatory proteins. In men, these parameters mainly show associations with VAT volume. This could be because only in women, sSAT volume is associated with sSAT expression of inflammatory proteins. These findings underscore that future research on adipose tissue in relation to cardiometabolic and cardiovascular disease should take sex differences into account.


Subject(s)
Cardiovascular Diseases , Leukocytes, Mononuclear , Humans , Female , Male , Leukocytes, Mononuclear/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Inflammation/metabolism , Adipose Tissue/metabolism , Subcutaneous Fat, Abdominal/metabolism , Cardiovascular Diseases/complications , Immunity, Innate , Intra-Abdominal Fat/metabolism
2.
J Clin Endocrinol Metab ; 108(10): 2537-2549, 2023 09 18.
Article in English | MEDLINE | ID: mdl-37014796

ABSTRACT

CONTEXT: Adipose tissue (AT) inflammation predisposes to insulin resistance and metabolic syndrome in obesity. OBJECTIVE: To investigate the association between adipocyte size, AT inflammation, systemic inflammation, and metabolic and atherosclerotic complications of obesity in a sex-specific manner. DESIGN: Cross-sectional cohort study. SETTING: University hospital in the Netherlands. PARTICIPANTS: A total of 302 adult subjects with a body mass index (BMI) ≥ 27 kg/m2. MAIN OUTCOME MEASURES: We obtained subcutaneous abdominal fat biopsies and systematically assessed, in a sex-specific manner, associations of several parameters of AT inflammation (including adipocyte size, macrophage content, crown-like structures, and gene expression) to biomarkers of systemic inflammation, leukocyte number and function, and to the presence of metabolic syndrome, insulin resistance, and carotid atherosclerotic plaques, assessed with ultrasound. RESULTS: Adipocyte size was associated with metabolic syndrome and AT macrophage content with insulin resistance. In contrast, none of the AT parameters was associated with carotid atherosclerosis, although mRNA expression of the anti-inflammatory IL-37 was associated with a lower intima-media thickness. We revealed profound sex-specific differences, with an association between BMI and adipocyte size, and between adipocyte size and metabolic syndrome in men only. Also, only men showed an association between adipocyte size, AT expression of leptin and MCP-1, and AT macrophage numbers, and between AT inflammation (crown-like structure number) and several circulating inflammatory proteins, including high specificity C-reactive protein, and IL-6. CONCLUSIONS: Inflammation in abdominal subcutaneous adipose tissue is more related to the metabolic than the atherosclerotic complications of obesity, and there are profound sex-specific differences in the association between BMI, adipocyte size, AT inflammation, and systemic inflammation, which are much stronger in men than women.


Subject(s)
Insulin Resistance , Metabolic Syndrome , Male , Adult , Humans , Female , Metabolic Syndrome/complications , Insulin Resistance/genetics , Carotid Intima-Media Thickness , Cross-Sectional Studies , Adipose Tissue/metabolism , Obesity/metabolism , Inflammation/pathology , Subcutaneous Fat, Abdominal/metabolism
3.
Front Endocrinol (Lausanne) ; 14: 1077604, 2023.
Article in English | MEDLINE | ID: mdl-36909330

ABSTRACT

Introduction: Polycystic ovary syndrome (PCOS) is a complex endocrine disorder that often coexists with a metabolic disorder. Studies have demonstrated that the malfunction of adipose tissue, particularly abdominal adipose tissue, could exacerbate reproductive and metabolic problems in PCOS patients. Adipose tissue-secreted signaling mediators (e.g., lipids and metabolites) would then interact with other body organs, including the ovary, to maintain the systemic equilibrium. Methods: In this study, we examined adipose samples from PCOS patients and unaffected individuals using a liquid chromatography-mass spectrometry-based metabonomics approach (LC-MS/MS). PCOS biomarkers were selected using multivariate statistical analysis. Results: Our pathway analysis revealed that these differential metabolites could be engaged in inflammatory diseases and mitochondrial beta-oxidation. We further developed an in vitro PCOS cell model to examine the effects of hyperandrogenism on granulosa cells and related metabolic disorders. We noted that isoleucine recovered the promotive effect on cell apoptosis, inhibitory effect on cell proliferation, sex hormone secretion, and mitochondrial function induced by dehydroepiandrosterone. Our gas chromatography-mass spectrometry targeted analysis (GC-MS/MS) revealed that isoleucine was significantly decreased in PCOS patients. Discussion: Based on these results, we speculate that metabolome alterations are vital in ameliorating PCOS symptoms. This may be a novel therapeutic target for PCOS treatment. Our study provides preliminary evidence that these findings will enhance our ability to accurately diagnose and intervene in PCOS.


Subject(s)
Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , Subcutaneous Fat, Abdominal/metabolism , Isoleucine , Metabolomics
4.
Sci Rep ; 12(1): 18408, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36319747

ABSTRACT

The mechanisms that underlie exercise-induced adaptations in adipose tissue have not been elucidated, yet, accumulating studies suggest an important role for microRNAs (miRNAs). This study aimed to investigate miRNA expression in gluteal subcutaneous adipose tissue (GSAT) in response to a 12-week exercise intervention in South African women with obesity, and to assess depot-specific differences in miRNA expression in GSAT and abdominal subcutaneous adipose tissue (ASAT). In addition, the association between exercise-induced changes in miRNA expression and metabolic risk was evaluated. Women underwent 12-weeks of supervised aerobic and resistance training (n = 19) or maintained their regular physical activity during this period (n = 12). Exercise-induced miRNAs were identified in GSAT using Illumina sequencing, followed by analysis of differentially expressed miRNAs in GSAT and ASAT using quantitative real-time PCR. Associations between the changes (pre- and post-exercise training) in miRNA expression and metabolic parameters were evaluated using Spearman's correlation tests. Exercise training significantly increased the expression of miR-155-5p (1.5-fold, p = 0.045), miR-329-3p (2.1-fold, p < 0.001) and miR-377-3p (1.7-fold, p = 0.013) in GSAT, but not in ASAT. In addition, a novel miRNA, MYN0617, was identified in GSAT, with low expression in ASAT. The exercise-induced differences in miRNA expression were correlated with each other and associated with changes in high-density lipoprotein concentrations. Exercise training induced adipose-depot specific miRNA expression within subcutaneous adipose tissue depots from South African women with obesity. The significance of the association between exercise-induced miRNAs and metabolic risk warrants further investigation.


Subject(s)
MicroRNAs , Subcutaneous Fat , Humans , Female , Subcutaneous Fat/metabolism , Obesity/metabolism , Exercise , Subcutaneous Fat, Abdominal/metabolism , MicroRNAs/genetics , Adipose Tissue/metabolism
5.
Sci Rep ; 12(1): 11771, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35817784

ABSTRACT

We investigated gluteal (GSAT) and abdominal subcutaneous adipose tissue (ASAT) DNA methylation of FKBP5 in response to a 12-week intervention in African women with obesity, as well as the effect of the rs1360780 single nucleotide polymorphism (SNP) on FKBP5 methylation, gene expression and post-exercise training adaptations in obesity and metabolic related parameters. Exercise (n = 19) participants underwent 12-weeks of supervised aerobic and resistance training while controls (n = 12) continued their usual behaviours. FKBP5 methylation was measured in GSAT and ASAT using pyrosequencing. SNP and gene expression analyses were conducted using quantitative real-time PCR. Exercise training induced FKBP5 hypermethylation at two CpG dinucleotides within intron 7. When stratified based on the rs1360780 SNP, participants with the CT genotype displayed FKBP5 hypermethylation in GSAT (p < 0.05), and ASAT displayed in both CC and CT carriers. CC allele carriers displayed improved cardiorespiratory fitness, insulin sensitivity, gynoid fat mass, and waist circumference (p < 0.05) in response to exercise training, and these parameters were attenuated in women with the CT genotype. These findings provide a basis for future studies in larger cohorts, which should assess whether FKBP5 methylation and/or genetic variants such as the rs1360780 SNP could have a significant impact on responsiveness to exercise interventions.


Subject(s)
Resistance Training , Tacrolimus Binding Proteins , Epigenesis, Genetic , Exercise , Female , Genotype , Humans , Obesity/genetics , Obesity/metabolism , Obesity/therapy , Pilot Projects , Polymorphism, Single Nucleotide , Subcutaneous Fat, Abdominal/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
6.
J Acquir Immune Defic Syndr ; 90(2): 175-183, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35125474

ABSTRACT

OBJECTIVE: Fat redistribution from subcutaneous adipose tissue (SAT) to the abdominal viscera, pericardium, liver, and skeletal muscle contributes to the rising burden of cardiometabolic disease among persons with HIV (PWH). Previous studies found SAT inflammation in PWH impairs lipid storage and persists despite plasma viral suppression on antiretroviral therapy (ART). In this study, we identified SAT immune-related genes associated with ectopic fat deposition in PWH on long-term ART. DESIGN AND METHODS: A total of 92 PWH with well-controlled viremia underwent computed tomography imaging and abdominal SAT biopsy for gene expression analysis. SAT gene expression was measured using a NanoString panel of 255 immune-related genes. Associations between gene expression and computed tomography measurements of the volume and attenuation (radiodensity) of metabolically relevant ectopic fat depots were assessed using multivariable linear regression and network analysis. RESULTS: Greater SAT volume was associated with higher visceral and pericardial adipose tissue volume, but lower skeletal muscle attenuation. Lower SAT attenuation, a measure of lipid content, was associated with lower visceral adipose tissue attenuation. Hierarchical clustering identified a subset of macrophage-related genes in SAT, including CCL2, CCL22, CCL13, CCR1, CD86, CD163, IL-6, IL-10, MRC1, and TREM2, which were associated with an increased lipid deposition in multiple ectopic depots. CONCLUSION: Altered expression of macrophage-related genes in SAT is associated with differences in ectopic fat depot morphometrics among PWH on long-term ART, including in the pericardial and visceral compartments. These findings provide basis for future studies to assess host, virus, and treatment factors shaping the SAT immune environment and its effects on morphometric changes and metabolic comorbidities in PWH.


Subject(s)
HIV Infections , Adipose Tissue/metabolism , Gene Expression , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/genetics , Humans , Inflammation/complications , Intra-Abdominal Fat/metabolism , Lipids , Subcutaneous Fat , Subcutaneous Fat, Abdominal/metabolism
7.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216509

ABSTRACT

The metabolic syndrome (MetS) is a cluster of the most dangerous heart attack risk factors: diabetes or raised fasting plasma glucose, abdominal obesity, high cholesterol and high blood pressure. The goal of this study is to compare the state of the main features of obesity-associated white adipose tissue (WAT) dysfunction in 66 women with severe obesity without (MetS-) or with MetS (MetS+). Fat cell area, adipocyte size distribution and histological fibrosis were analysed in visceral (VAT) and abdominal subcutaneous WAT (SAT) in 33 age- and BMI-matched pairs of MetS- and MetS+ subjects. The mRNA expression of 93 genes implicated in obesity-associated WAT dysfunction was analysed by RT-qPCR in both fat depots. MetS+ females showed higher adipocyte hypertrophy in both fat depots and increased fibrosis and expression of macrophage and hypoxia markers in SAT. Transcriptional data suggest increased fatty acid oxidation in SAT and impaired thermogenesis and extracellular matrix remodelling in VAT from MetS+ subjects. A sPLS-DA model, including SAT expression of PPARA and LEPR genes identified MetS with an AUC = 0.87. Despite equal age, BMI and body composition, MetS+ females display morphological and transcriptional differences in both WAT depots, especially in SAT. These factors may contribute to the transition to MetS.


Subject(s)
Metabolic Syndrome/pathology , Obesity, Morbid/pathology , Subcutaneous Fat, Abdominal/pathology , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Body Composition/physiology , Body Mass Index , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Female , Humans , Hypertension/metabolism , Hypertension/pathology , Intra-Abdominal Fat/metabolism , Metabolic Syndrome/metabolism , Middle Aged , Obesity, Abdominal/metabolism , Obesity, Abdominal/pathology , Obesity, Morbid/metabolism , Subcutaneous Fat, Abdominal/metabolism , Thermogenesis/physiology
8.
J Clin Endocrinol Metab ; 107(3): 755-775, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34669916

ABSTRACT

CONTEXT: Adipose tissue distribution is a key factor influencing metabolic health and risk in obesity-associated comorbidities. OBJECTIVE: Here we aim to compare the proteomic profiles of mature adipocytes from different depots. METHODS: Abdominal subcutaneous (SA) and omental visceral adipocytes (VA) were isolated from paired adipose tissue biopsies obtained during bariatric surgery on 19 severely obese women (body mass index > 30 kg/m2) and analyzed using state-of-the-art mass spectrometry. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were performed to investigate proteome signature properties and to examine a possible association of the protein expression with the clinical data. RESULTS: We identified 3686 protein groups and found 1140 differentially expressed proteins (adj. P value < 0.05), of which 576 proteins were upregulated in SA and 564 in VA samples. We provide a global protein profile of abdominal SA and omental VA, present the most differentially expressed pathways and processes distinguishing SA from VA, and correlate them with clinical and body composition data. We show that SA are significantly more active in processes linked to vesicular transport and secretion, and to increased lipid metabolism activity. Conversely, the expression of proteins involved in the mitochondrial energy metabolism and translational or biosynthetic activity is higher in VA. CONCLUSION: Our analysis represents a valuable resource of protein expression profiles in abdominal SA and omental VA, highlighting key differences in their role in obesity.


Subject(s)
Adipocytes/metabolism , Intra-Abdominal Fat/metabolism , Obesity, Morbid/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adult , Bariatric Surgery , Female , Gene Regulatory Networks , Humans , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/pathology , Middle Aged , Obesity, Morbid/pathology , Obesity, Morbid/surgery , Omentum/cytology , Omentum/metabolism , Omentum/pathology , Omentum/surgery , Proteomics , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/pathology
9.
J Clin Endocrinol Metab ; 107(3): e1057-e1073, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34718616

ABSTRACT

CONTEXT: The upregulation of TRIB3 (Tribbles homolog 3), a stress-inducible gene encoding a pseudokinase, has been implicated in the development of insulin resistance in the skeletal muscle and liver of patients with obesity and type 2 diabetes. However, there is little information regarding TRIB3 expression in human adipose tissue. OBJECTIVE: To investigate whether TRIB3 expression is dysregulated in human adipose tissue in the context of obesity and type 2 diabetes and whether TRIB3 expression in adipose tissues is associated with insulin resistance. METHODS: We measured metabolic parameters and TRIB3 expression in abdominal subcutaneous and visceral adipose tissue in obese (with or without type 2 diabetes) and normal-weight women. Regulation of TRIB3 expression was studied in human adipocytes. RESULTS: TRIB3 expression in both fat depots was higher in patients with obesity and/or type 2 diabetes; in addition, the expression level was significantly associated with insulin resistance. Incubating adipocytes under conditions mimicking the microenvironment of obese adipose tissue, including increased endoplasmic reticulum (ER) stress, induced TRIB3 expression. In human adipocytes, the overexpression of TRIB3 impaired insulin-stimulated protein kinase B (AKT) phosphorylation and caused dysregulation of the transcription of genes encoding bioactive molecules released from adipocytes, such as proinflammatory cytokines, adiponectin, and leptin. Pioglitazone, an insulin-sensitizing agent, reduced both these effects of TRIB3 and the ER stressor-induced expression of TRB3. CONCLUSION: Our data indicate that TRIB3 expression in adipose tissue is enhanced in patients with obesity and suggest that increased TRIB3 dysregulates adipocyte function, which may contribute to the development of insulin resistance.


Subject(s)
Cell Cycle Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adult , Cell Cycle Proteins/analysis , Cells, Cultured , Coculture Techniques , Endoplasmic Reticulum Stress , Female , Humans , Insulin Resistance , Intra-Abdominal Fat/cytology , Macrophages , Middle Aged , Pioglitazone/pharmacology , Primary Cell Culture , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/analysis , Subcutaneous Fat, Abdominal/cytology , Up-Regulation/drug effects
10.
Rev. cuba. med. gen. integr ; 38(3): e1973, 2022. tab, graf
Article in Spanish | LILACS, CUMED | ID: biblio-1408718

ABSTRACT

Introducción: La adiposidad central como factor desencadenante de resistencia a la insulina precoz constituye una amenaza potencial de riesgo metabólico y cardiovascular en el embarazo. Objetivo: Determinar la capacidad discriminante de las grasas abdominales sobre la resistencia a la insulina, diagnosticada por el índice triglicéridos/glucosa-IMC al finalizar el primer trimestre del embarazo. Métodos: Se realizó un estudio observacional analítico de 526 gestantes con embarazo simple y edad gestacional entre 12 y 13 semanas, entre los años 2016 y 2020. Se estudió el test de triglicéridos/glucosa-IMC y las grasas abdominales por ultrasonido. Se utilizaron las curvas ROC (Receiver operating characteristic Curve) para discriminar la resistencia a la insulina al finalizar el primer trimestre de la gestación, cuando aumentan las grasas abdominales. Resultados: La grasa subcutánea fue la que presentó mayor área bajo la curva en la discriminación de la resistencia a la insulina, con un nivel de sensibilidad y especificidad aceptable. Conclusiones: La grasa subcutánea, aunque con bajo valor discriminativo, puede considerarse como augurio de resistencia a la insulina y de diabetes gestacional. Se requiere profundizar en el estudio de las grasas abdominales dado el conocimiento de su impacto en los desórdenes metabólicos en el curso avanzado de la gestación(AU)


Introduction: Central adiposity as a triggering factor for early insulin resistance is a potential threat of metabolic and cardiovascular risk in pregnancy. Objective: To determine the discriminating capacity of abdominal fat over insulin resistance, diagnosed by the triglyceride/glucose-BMI index at the end of the first trimester of pregnancy. Methods: An analytical and observational study was carried out with 526 pregnant women of singleton pregnancy and gestational age between twelve and thirteen weeks, between 2016 and 2020. The triglyceride/glucose-BMI test was studied, together with abdominal fats by ultrasound. ROC (receiver operating characteristic) curves were used to discriminate insulin resistance at the end of the first trimester of gestation, when abdominal fats increase. Results: Subcutaneous fat presented the highest area under the curve in the discrimination of insulin resistance, with an acceptable level of sensitivity and specificity. Conclusions: Subcutaneous fat, although with low discriminative value, can be considered as a harbinger of insulin resistance and gestational diabetes. Further study of abdominal fat is required, given the knowledge of its impact on metabolic disorders in late gestation(AU)


Subject(s)
Humans , Female , Pregnancy , Pregnancy Trimester, First , Insulin Resistance/physiology , Subcutaneous Fat, Abdominal/metabolism , Obesity, Abdominal/metabolism , Triglycerides/blood , Blood Glucose/analysis , Body Mass Index , Cross-Sectional Studies , Predictive Value of Tests , ROC Curve
11.
Nutrients ; 13(12)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34960108

ABSTRACT

The study evaluated the association between nutrient patterns with body fat and regional adiposity in middle-aged black South African (SA) men and women and determined if this differed by sex. Body fat and regional adiposity (dual-energy x-ray absorptiometry), and dietary intake (7-day quantified food frequency questionnaire) were measured in black SA men (n = 414) and women (n = 346). Using principal component analysis, nutrient patterns were computed from 25 nutrients in the combined sample. Four nutrient patterns were extracted, explaining 67% of the variance in nutrient intake. Animal and fat, as well as the vitamin C, sugar, and potassium driven patterns, were positively associated with total adiposity. In contrast, the retinol and vitamin B12 pattern was associated with the centralisation of fat. Notably, the strength of the association between the animal-driven nutrient pattern and BMI was greater in men (1.14 kg/m2, 95% CI (0.63-1.66)) than in women (0.81 kg/m2, 95% CI (0.25-1.36)) (Pint = 0.017). In contrast, the plant-driven pattern was associated with higher abdominal subcutaneous adipose tissue (SAT) in women (44 cm2, 95% CI (22-67)) but not men (Pint = 1.54 × 10-4). These differences suggest that although men and women have similar nutrient patterns, their associations with the whole body and regional body fat are different.


Subject(s)
Adipose Tissue/metabolism , Adiposity , Black People , Feeding Behavior , Nutrients/administration & dosage , Absorptiometry, Photon/methods , Animals , Body Mass Index , Cross-Sectional Studies , Diet/statistics & numerical data , Energy Intake , Female , Humans , Male , Middle Aged , Nutritional Status , Obesity/epidemiology , Sex Characteristics , Sex Factors , South Africa/epidemiology , Subcutaneous Fat, Abdominal/metabolism
12.
Obesity (Silver Spring) ; 29(12): 2068-2080, 2021 12.
Article in English | MEDLINE | ID: mdl-34672413

ABSTRACT

OBJECTIVE: This study investigated whether variations in cell death-inducing DNA fragmentation factor alpha subunit-like effector A (CIDEA) mRNA expression and protein levels are modulated by the pattern of abdominal fat distribution in adolescent girls with obesity. METHODS: This study recruited 35 adolescent girls with obesity and characterized their abdominal fat distribution by magnetic resonance imaging. Participants had only a periumbilical/abdominal (n = 14) or a paired abdominal and gluteal subcutaneous adipose tissue (SAT) biopsy (n = 21). CIDEA expression was determined by reverse transcription-polymerase chain reaction, CIDEA protein level by Western blot, and the turnover of adipose lipids and adipocytes by 2 H2 O labeling. In six girls, a second abdominal SAT biopsy was performed (after ~34.2 months) to explore the weight gain effect on CIDEA expression in abdominal SAT. RESULTS: CIDEA expression decreased in abdominal SAT from participants with high visceral adipose tissue (VAT)/(VAT+SAT); CIDEA inversely correlated with number of small adipocytes, with the increase in preadipocyte proliferation, and with adipogenesis. A strong inverse correlation was found between CIDEA protein level with the newly synthetized glycerol (r = -0.839, p = 0.0047). Following weight gain, an increase in adipocytes' cell diameter with a decrease in CIDEA expression and RNA-sequencing transcriptomic profile typical of adipocyte dysfunction was observed. CONCLUSIONS: Reduced expression of CIDEA in girls with high VAT/(VAT+SAT) is associated with adipocyte hypertrophy and insulin resistance.


Subject(s)
Apoptosis Regulatory Proteins , Obesity , Subcutaneous Fat , Abdominal Fat/metabolism , Adolescent , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Female , Humans , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Subcutaneous Fat, Abdominal/metabolism
13.
Nutrients ; 13(9)2021 Sep 21.
Article in English | MEDLINE | ID: mdl-34579170

ABSTRACT

This study aimed to investigate relationships between infant abdominal visceral and subcutaneous adiposity and human milk (HM) components and maternal body composition (BC) during first year of lactation. Subcutaneous-abdominal depth (SAD), subcutaneous-abdominal fat area (SFA), visceral depth (VD) and preperitoneal fat area of 20 breastfed infants were assessed at 2, 5, 9 and 12 months using ultrasound. Maternal BC was determined with bioimpedance spectroscopy. HM macronutrients and bioactive components concentrations and infant 24-h milk intake were measured and calculated daily intakes (CDI) determined. Maternal adiposity associated with infant SFA (negatively at 2, 5, 12, positively at 9 months, all overall p < 0.05). 24-h milk intake positively associated with infant SAD (p = 0.007) and VD (p = 0.013). CDI of total protein (p = 0.013), total carbohydrates (p = 0.004) and lactose (p = 0.013) positively associated with SFA. Lactoferrin concentration associated with infant VD (negatively at 2, 12, positively at 5, 9 months, overall p = 0.003). CDI of HM components and maternal adiposity have differential effects on development of infant visceral and subcutaneous abdominal adiposity. Maintaining healthy maternal BC and continuing breastfeeding to 12 months and beyond may facilitate favourable BC development reducing risk of obesity.


Subject(s)
Body Composition , Breast Feeding/methods , Intra-Abdominal Fat/metabolism , Milk, Human/chemistry , Subcutaneous Fat, Abdominal/metabolism , Adiponectin/analysis , Adiposity , Body Weight , Dietary Carbohydrates/analysis , Female , Humans , Infant , Lactation/metabolism , Leptin/analysis , Longitudinal Studies , Male , Nutrients/analysis , Obesity/epidemiology
14.
BMC Pregnancy Childbirth ; 21(1): 490, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34233642

ABSTRACT

BACKGROUND: It was reported that steroid-related gene expressions in the adipose tissue (AT) of women differ between women affected with polycystic ovary syndrome (PCOS) and non-PCOS. Although association between PCOS in mother and offspring's health is a crucial issue, there are few studies focusing on AT of pregnant women suffering from PCOS. Our objectives were to determine the differences between mRNA expression levels of key steroid-converting enzymes in abdominal subcutaneous AT of pregnant women afflicted with PCOS and non-PCOS. METHODS: Twelve pregnant women with PCOS (case) and thirty six non-PCOS pregnant women (control) (1:3 ratio; age- and BMI-matched) undergoing cesarean section were enrolled for the present study. Expressions of fifteen genes related to steriodogenesis in abdominal subcutaneous AT were investigated using quantitative real-time PCR. RESULTS: No significant differences were detected with respect to age, BMI (prior pregnancy and at delivery day), gestational period and parity among pregnant women with PCOS and non-PCOS. Most of the sex steroid-converting genes except 17ß-Hydroxysteroid dehydrogenases2 (17BHSD2), were highly expressed on the day of delivery in subcutaneous AT. Women with PCOS showed significantly higher mRNA levels of steroidgenic acute regulator (STAR; P < 0.001), cytochrome P450 monooxygenase (CYP11A1; P < 0.05), 17α-hydroxylase (CYP17A1; P < 0.05), and 11ß-Hydroxysteroid dehydrogenase (11BHSD1 and 11BHSD2; P < 0.05). The expression of steroid 21-hydroxylase (CYP21) in non-PCOS was fourfold higher than those of women with PCOS (P < 0.001). There were no significant differences between relative expression of aromatase cytochrome P450 (CYP19A1), 3ß-hydroxysteroid dehydrogenase (3BHSD1 and 3BHSD2), and 17BHSD family (1, 3, 5, 7, and 12) between the two groups. CONCLUSION: The expression levels of genes related to sex steroids metabolism were similar to age-matched and BMI- matched pregnant non-PCOS and pregnant women with PCOS at delivery day. However, the alterations in gene expressions involved in glucocorticoids and mineralocorticoids metabolism were shown. It is necessary to point out that further studies regarding functional activity are required. More attention should be given to AT of pregnant women with PCOS that was previously ignored.


Subject(s)
Gonadal Steroid Hormones/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Polycystic Ovary Syndrome/genetics , Steroid Hydroxylases/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adult , Case-Control Studies , Cesarean Section , Female , Gene Expression/genetics , Glucocorticoids/metabolism , Humans , Mineralocorticoids/metabolism , Phosphoproteins/metabolism , Pregnancy , RNA, Messenger/metabolism
15.
Sci Rep ; 11(1): 14750, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285301

ABSTRACT

Menopause is associated with a redistribution of adipose tissue towards central adiposity, known to cause insulin resistance. In this cross-sectional study of 33 women between 45 and 60 years, we assessed adipose tissue inflammation and morphology in subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) across menopause and related this to menopausal differences in adipose tissue distribution and insulin resistance. We collected paired SAT and VAT biopsies from all women and combined this with anthropometric measurements and estimated whole-body insulin sensitivity. We found that menopause was associated with changes in adipose tissue phenotype related to metabolic dysfunction. In SAT, postmenopausal women showed adipocyte hypertrophy, increased inflammation, hypoxia and fibrosis. The postmenopausal changes in SAT was associated with increased visceral fat accumulation. In VAT, menopause was associated with adipocyte hypertrophy, immune cell infiltration and fibrosis. The postmenopausal changes in VAT phenotype was associated with decreased insulin sensitivity. Based on these findings we suggest, that menopause is associated with changes in adipose tissue phenotype related to metabolic dysfunction in both SAT and VAT. Whereas increased SAT inflammation in the context of menopause is associated with VAT accumulation, VAT morphology is related to insulin resistance.


Subject(s)
Intra-Abdominal Fat/pathology , Subcutaneous Fat, Abdominal/pathology , Aging , Body Fat Distribution , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Inflammation/pathology , Insulin/metabolism , Insulin Resistance , Intra-Abdominal Fat/metabolism , Menopause , Metabolic Syndrome/pathology , Middle Aged , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Phenotype , Subcutaneous Fat, Abdominal/metabolism
16.
J Clin Endocrinol Metab ; 106(10): e3881-e3889, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34137897

ABSTRACT

CONTEXT: Subcutaneous adipose tissue (SAT) is not homogeneous, as the fascia scarpa separates the deep SAT (dSAT) from the superficial SAT (sSAT). OBJECTIVE: The aim of this study is to evaluate the sex-specific associations of sSAT and dSAT with hepatic steatosis and metabolic syndrome in overweight individuals. METHODS: We recruited 285 individuals with a body mass index (BMI) greater than or equal to 27 and aged 55 to 81 years. Abdominal magnetic resonance imaging was performed around level L4 to L5 to measure visceral adipose tissue (VAT), dSAT, and sSAT volumes. The amount of hepatic fat was quantified by MR spectroscopy. RESULTS: Men had significantly higher volumes of VAT (122.6 cm3 vs 98.7 cm3, P < .001) and had only half the volume of sSAT compared to women adjusted for BMI (50.3 cm3 in men vs 97.0 cm3 in women, P < .001). dSAT correlated significantly with hepatic fat content in univariate analysis (standardized ß = .190, P < .05), while VAT correlated significantly with hepatic steatosis in a multivariate model, adjusted for age, alcohol use, and other abdominal fat compartments (standardized ß = .184, P = .037). Moreover, dSAT in men correlated negatively with HDL cholesterol (standardized ß = -0.165, P = .038) in multivariate analyses. In women with a BMI between 30 and 40, in a multivariate model adjusted for age, alcohol use, and other abdominal fat compartments, VAT correlated positively (standardized ß = -.404, P = .003), and sSAT negatively (standardized ß = -.300, P = .04) with hepatic fat content. CONCLUSION: In men, dSAT is associated with hepatic steatosis and adverse metabolic traits, such as lower HDL cholesterol levels, whereas in women with obesity sSAT shows a beneficial relation with respect to hepatic fat content.


Subject(s)
Fatty Liver/etiology , Intra-Abdominal Fat/pathology , Metabolic Syndrome/etiology , Subcutaneous Fat, Abdominal/pathology , Aged , Aged, 80 and over , Cohort Studies , Fatty Liver/diagnosis , Fatty Liver/epidemiology , Fatty Liver/pathology , Female , Humans , Intra-Abdominal Fat/diagnostic imaging , Intra-Abdominal Fat/metabolism , Magnetic Resonance Imaging , Male , Metabolic Syndrome/diagnosis , Metabolic Syndrome/epidemiology , Metabolic Syndrome/pathology , Middle Aged , Obesity/diagnosis , Obesity/epidemiology , Obesity/metabolism , Obesity/pathology , Organ Size/physiology , Overweight/diagnosis , Overweight/epidemiology , Overweight/metabolism , Overweight/pathology , Risk Factors , Sex Characteristics , Sex Factors , Subcutaneous Fat/diagnostic imaging , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology , Subcutaneous Fat, Abdominal/diagnostic imaging , Subcutaneous Fat, Abdominal/metabolism
17.
Gastroenterology ; 161(3): 968-981.e12, 2021 09.
Article in English | MEDLINE | ID: mdl-34004161

ABSTRACT

BACKGROUND AND AIMS: Insulin resistance is a key factor in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). We evaluated the importance of subcutaneous abdominal adipose tissue (SAAT) inflammation and both plasma and SAAT-derived exosomes in regulating insulin sensitivity in people with obesity and NAFLD. METHODS: Adipose tissue inflammation (macrophage and T-cell content and expression of proinflammatory cytokines), liver and whole-body insulin sensitivity (assessed using a hyperinsulinemic-euglycemic clamp and glucose tracer infusion), and 24-hour serial plasma cytokine concentrations were evaluated in 3 groups stratified by adiposity and intrahepatic triglyceride (IHTG) content: (1) lean with normal IHTG content (LEAN; N = 14); (2) obese with normal IHTG content (OB-NL; N = 28); and (3) obese with NAFLD (OB-NAFLD; N = 28). The effect of plasma and SAAT-derived exosomes on insulin-stimulated Akt phosphorylation in human skeletal muscle myotubes and mouse primary hepatocytes was assessed in a subset of participants. RESULTS: Proinflammatory macrophages, proinflammatory CD4 and CD8 T-cell populations, and gene expression of several cytokines in SAAT were greater in the OB-NAFLD than the OB-NL and LEAN groups. However, with the exception of PAI-1, which was greater in the OB-NAFLD than the LEAN and OB-NL groups, 24-hour plasma cytokine concentration areas-under-the-curve were not different between groups. The percentage of proinflammatory macrophages and plasma PAI-1 concentration areas-under-the-curve were inversely correlated with both hepatic and whole-body insulin sensitivity. Compared with exosomes from OB-NL participants, plasma and SAAT-derived exosomes from the OB-NAFLD group decreased insulin signaling in myotubes and hepatocytes. CONCLUSIONS: Systemic insulin resistance in people with obesity and NAFLD is associated with increased plasma PAI-1 concentrations and both plasma and SAAT-derived exosomes. ClinicalTrials.gov number: NCT02706262 (https://clinicaltrials.gov/ct2/show/NCT02706262).


Subject(s)
Cytokines/blood , Exosomes/metabolism , Insulin Resistance , Macrophages/metabolism , Memory T Cells/metabolism , Non-alcoholic Fatty Liver Disease/blood , Obesity/blood , Plasminogen Activator Inhibitor 1/blood , Subcutaneous Fat, Abdominal/metabolism , Adult , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cells, Cultured , Exosomes/immunology , Female , Hepatocytes/metabolism , Humans , Insulin/blood , Liver/metabolism , Macrophages/immunology , Male , Memory T Cells/immunology , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/diagnosis , Obesity/immunology , Obesity/physiopathology , Subcutaneous Fat, Abdominal/immunology , Tissue Culture Techniques
18.
Mol Cell Endocrinol ; 533: 111323, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34000351

ABSTRACT

BACKGROUND: Obesity is characterized by the excess accumulation of white adipose tissue (WAT). Src family kinases (SFKs) are non-receptor tyrosine kinases consisting of eight members (SRC, FYN, YES1, HCK, LCK, LYN, FGR and BLK) that have been studied extensively in mammalian cells. Although individual members in murine cells provide some clues that are associated with the regulation of adipogenesis, the specific role of this family in adipocyte differentiation has rarely been assessed, especially in human adipocytes. METHODS: Herein, we first explored the expression profiles of SFKs during human preadipocyte differentiation. Then, we used the pyrazolo-pyrimidinyl-amine compound PP1, a potent SFK inhibitor, to evaluate the function of SFKs during adipocyte differentiation. Furthermore, we adopted a loss-of-function strategy with siRNAs to determine the role of FGR in adipocyte differentiation. RESULTS: Here, we found that SRC, FYN, YES1, LYN and FGR were expressed in human preadipocytes and induced after the initiation of differentiation. Furthermore, the SFK inhibitor PP1 suppressed adipocyte differentiation. We also found that PP1 significantly suppressed the SFK activity in preadipocytes and decreased the expression of adipogenic genes in early and late differentiation. Given that FGR exhibited the most expression enhancement in mature adipocytes, we focused on FGR and found that its knockdown reduced lipid accumulation and adipogenic gene expression. CONCLUSIONS: Collectively, these findings suggest that SFKs, especially FGR, are involved in the differentiation of human preadipocytes. Our results lay a foundation for further understanding the role of SFKs in adipocyte differentiation and provide new clues for anti-obesity therapies.


Subject(s)
Adipocytes/cytology , Adipogenesis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , src-Family Kinases/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/metabolism , Subcutaneous Fat, Abdominal/cytology , Subcutaneous Fat, Abdominal/metabolism
19.
Hepatology ; 74(3): 1287-1299, 2021 09.
Article in English | MEDLINE | ID: mdl-33743554

ABSTRACT

BACKGROUND AND AIMS: It is proposed that impaired expansion of subcutaneous adipose tissue (SAT) and an increase in adipose tissue (AT) fibrosis causes ectopic lipid accumulation, insulin resistance (IR), and metabolically unhealthy obesity. We therefore evaluated whether a decrease in SAT expandability, assessed by measuring SAT lipogenesis (triglyceride [TG] production), and an increase in SAT fibrogenesis (collagen production) are associated with NAFLD and IR in persons with obesity. APPROACH AND RESULTS: In vivo abdominal SAT lipogenesis and fibrogenesis, expression of SAT genes involved in extracellular matrix (ECM) formation, and insulin sensitivity were assessed in three groups of participants stratified by adiposity and intrahepatic TG (IHTG) content: (1) healthy lean with normal IHTG content (Lean-NL; n = 12); (2) obese with normal IHTG content and normal glucose tolerance (Ob-NL; n = 25); and (3) obese with NAFLD and abnormal glucose metabolism (Ob-NAFLD; n = 25). Abdominal SAT TG synthesis rates were greater (P < 0.05) in both the Ob-NL (65.9 ± 4.6 g/wk) and Ob-NAFLD groups (71.1 ± 6.7 g/wk) than the Lean-NL group (16.2 ± 2.8 g/wk) without a difference between the Ob-NL and Ob-NAFLD groups. Abdominal SAT collagen synthesis rate and the composite expression of genes encoding collagens progressively increased from the Lean-NL to the Ob-NL to the Ob-NAFLD groups and were greater in the Ob-NAFLD than the Ob-NL group (P < 0.05). Composite expression of collagen genes was inversely correlated with both hepatic and whole-body insulin sensitivity (P < 0.001). CONCLUSIONS: AT expandability is not impaired in persons with obesity and NAFLD. However, SAT fibrogenesis is greater in persons with obesity and NAFLD than in those with obesity and normal IHTG content, and is inversely correlated with both hepatic and whole-body insulin sensitivity.


Subject(s)
Collagen/metabolism , Glucose Intolerance/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Subcutaneous Fat, Abdominal/metabolism , Triglycerides/metabolism , Adipose Tissue/metabolism , Adult , Extracellular Matrix/metabolism , Female , Fibrosis , Glucose Intolerance/complications , Humans , Insulin Resistance , Lipogenesis , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/complications , Obesity/complications , Subcutaneous Fat/metabolism
20.
Cell Rep ; 34(9): 108804, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33657384

ABSTRACT

Intermittent fasting is a beneficial dietary treatment for obesity. But the response of each distinct adipose depot is currently poorly defined. Here we explore the response of key adipose depots to every-other-day fasting (EODF) in mice using proteomics. A key change in subcutaneous white adipose tissue (scWAT) and visceral WAT (vWAT) depots is an increase in mitochondrial protein content after EODF. This effect is correlated with increased fatty acid synthesis enzymes in both WAT depots but not in brown adipose tissue. Strikingly, EODF treatment downregulates lipolysis specifically in vWAT, mediated by a large decrease in the abundance of the catecholamine receptor (ADRB3). Together, these changes are important for preservation of the visceral lipid store during EODF. Enrichment analysis highlights downregulation of inflammatory collagen IV specifically in vWAT, allowing improved insulin sensitivity. This resource for adipose-depot-specific fasting adaptations in mice is available using a web-based interactive visualization.


Subject(s)
Energy Metabolism , Fasting/metabolism , Intra-Abdominal Fat/metabolism , Lipid Metabolism , Proteome , Proteomics , Subcutaneous Fat, Abdominal/metabolism , Adaptation, Physiological , Animals , Extracellular Matrix/metabolism , Fatty Acids/metabolism , Lipolysis , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Receptors, Adrenergic, beta-3/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...