Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Vis Exp ; (207)2024 May 03.
Article in English | MEDLINE | ID: mdl-38767365

ABSTRACT

Intermuscular adipose tissue (IMAT) is a relatively understudied adipose depot located between muscle fibers. IMAT content increases with age and BMI and is associated with metabolic and muscle degenerative diseases; however, an understanding of the biological properties of IMAT and its interplay with the surrounding muscle fibers is severely lacking. In recent years, single-cell and nuclei RNA sequencing have provided us with cell type-specific atlases of several human tissues. However, the cellular composition of human IMAT remains largely unexplored due to the inherent challenges of its accessibility from biopsy collection in humans. In addition to the limited amount of tissue collected, the processing of human IMAT is complicated due to its proximity to skeletal muscle tissue and fascia. The lipid-laden nature of the adipocytes makes it incompatible with single-cell isolation. Hence, single nuclei RNA sequencing is optimal for obtaining high-dimensional transcriptomics at single-cell resolution and provides the potential to uncover the biology of this depot, including the exact cellular composition of IMAT. Here, we present a detailed protocol for nuclei isolation and library preparation of frozen human IMAT for single nuclei RNA sequencing. This protocol allows for the profiling of thousands of nuclei using a droplet-based approach, thus providing the capacity to detect rare and low-abundant cell types.


Subject(s)
Adipose Tissue , Cell Nucleus , Sequence Analysis, RNA , Humans , Adipose Tissue/cytology , Sequence Analysis, RNA/methods , Cell Nucleus/chemistry , Cell Nucleus/genetics , Single-Cell Analysis/methods , Muscle, Skeletal/cytology , Muscle, Skeletal/chemistry
2.
Am J Physiol Cell Physiol ; 326(4): C1248-C1261, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38581663

ABSTRACT

Adipose-derived stem cells (ADSCs) play an important role in the differential capacity for excess energy storage between upper body abdominal (ABD) adipose tissue (AT) and lower body gluteofemoral (GF) AT. We cultured ADSCs from subcutaneous ABD AT and GF AT isolated from eight women with differential body fat distribution and performed single-cell RNA sequencing. Six populations of ADSCs were identified and segregated according to their anatomical origin. The three ADSC subpopulations in GF AT were characterized by strong cholesterol/fatty acid (FA) storage and proliferation signatures. The two ABD subpopulations, differentiated by higher expression of committed preadipocyte marker genes, were set apart by differential expression of extracellular matrix and ribosomal genes. The last population, identified in both depots, was similar to smooth muscle cells and when individually isolated and cultured in vitro they differentiated less than the other subpopulations. This work provides important insight into the use of ADSC as an in vitro model of adipogenesis and suggests that specific subpopulations of GF-ADSCs contribute to the more robust capacity for GF-AT to expand and grow compared with ABD-AT in women.NEW & NOTEWORTHY Identification of distinct subpopulations of adipose-derived stem cells (ADSCs) in upper body abdominal subcutaneous (ABD) and lower body gluteofemoral subcutaneous (GF) adipose tissue depots. In ABD-ADSCs, subpopulations are more committed to adipocyte lineage. GF-ADSC subpopulations are enriched for genes involved in lipids and cholesterol metabolism. Similar depot differences were found in stem cell population identified in freshly isolated stoma vascular fraction. The repertoire of ADSCs subpopulations was different in apple-shaped versus pear-shaped women.


Subject(s)
Adipose Tissue , Subcutaneous Fat , Humans , Female , Adipose Tissue/metabolism , Adipocytes/metabolism , Sequence Analysis, RNA , Cholesterol/metabolism
3.
Am J Physiol Endocrinol Metab ; 325(4): E291-E302, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37584609

ABSTRACT

Insulin resistance and blunted mitochondrial capacity in skeletal muscle are often synonymous, however, this association remains controversial. The aim of this study was to perform an in-depth multifactorial comparison of skeletal muscle mitochondrial capacity between individuals who were lean and active (Active, n = 9), individuals with obesity (Obese, n = 9), and individuals with obesity, insulin resistance, and type 2 diabetes (T2D, n = 22). Mitochondrial capacity was assessed by ex vivo mitochondrial respiration with fatty-acid and glycolytic-supported protocols adjusted for mitochondrial content (mtDNA and citrate synthase activity). Supercomplex assembly was measured by Blue Native (BN)-PAGE and immunoblot. Tricarboxylic (TCA) cycle intermediates were assessed with targeted metabolomics. Exploratory transcriptomics and DNA methylation analyses were performed to uncover molecular differences affecting mitochondrial function among the three groups. We reveal no discernable differences in skeletal muscle mitochondrial content, mitochondrial capacity, supercomplex assembly, TCA cycle intermediates, and mitochondrial molecular profiles between obese individuals with and without T2D that had comparable levels of confounding factors (body mass index, age, and aerobic capacity). We highlight that lean, active individuals have greater mitochondrial content, mitochondrial capacity, supercomplex assembly, and TCA cycle intermediates. These phenotypical changes are reflected at the level of DNA methylation and gene transcription. The collective observation of comparable muscle mitochondrial capacity in individuals with obesity and T2D (vs. individuals without T2D) underscores a dissociation from skeletal muscle insulin resistance. Clinical trial number: NCT01911104.NEW & NOTEWORTHY Whether impaired mitochondrial capacity contributes to skeletal muscle insulin resistance is debated. Our multifactorial analysis shows no differences in skeletal muscle mitochondrial content, mitochondrial capacity, and mitochondrial molecular profiles between obese individuals with and without T2D that had comparable levels of confounding factors (BMI, age, aerobic capacity). We highlight that lean, active individuals have enhanced skeletal muscle mitochondrial capacity that is also reflected at the level of DNA methylation and gene transcription.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Insulin Resistance/physiology , Diabetes Mellitus, Type 2/metabolism , Mitochondria , Muscle, Skeletal/metabolism , Obesity/metabolism , Mitochondria, Muscle/metabolism
4.
Res Sq ; 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37503028

ABSTRACT

White adipose tissue (WAT) is a robust energy storage and endocrine organ critical for maintaining metabolic health as we age. Our aim was to identify cell-specific transcriptional aberrations that occur in WAT with aging. We leveraged full-length snRNA-Seq to characterize the cellular landscape of human subcutaneous WAT in a prospective cohort of 10 Younger (≤ 30 years) and 10 Older individuals (≥ 65 years) balanced for sex and body mass index (BMI). We highlight that aging WAT is associated with adipocyte hypertrophy, increased proportions of resident macrophages (M2), an upregulated innate immune response and senescence profiles in specific adipocyte populations, highlighting CXCL14 as a biomarker of this process. We also identify novel markers of pre-adipocytes and track their expression levels through pre-adipocyte differentiation. We propose that aging WAT is associated with low-grade inflammation that is managed by a foundation of innate immunity to preserve the metabolic health of the WAT.

5.
STAR Protoc ; 4(1): 102054, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36853719

ABSTRACT

Automated single-cell dispensing is incompatible with white adipose tissue (WAT) due to lipid-laden adipocytes. Single-nuclei RNA-Seq permits transcriptional profiling of all cells from WAT. Human WAT faces unique technical challenges in isolating nuclei compared to rodent tissue due to greater extra-cellular matrix content and larger lipid droplets. In this protocol, we detail how to isolate nuclei from frozen subcutaneous human WAT for single-nuclei RNA-Seq. For complete information on the generation and use of this protocol, please refer to Whytock et al. (2022).1.


Subject(s)
Adipose Tissue, White , Subcutaneous Fat , Humans , Cell Nucleus/genetics , Adipocytes , RNA-Seq
6.
Int J Mol Sci ; 25(1)2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38203607

ABSTRACT

The genome of human adipose-derived stem cells (ADSCs) from abdominal and gluteofemoral adipose tissue depots are maintained in depot-specific stable epigenetic conformations that influence cell-autonomous gene expression patterns and drive unique depot-specific functions. The traditional approach to explore tissue-specific transcriptional regulation has been to correlate differential gene expression to the nearest-neighbor linear-distance regulatory region defined by associated chromatin features including open chromatin status, histone modifications, and DNA methylation. This has provided important information; nonetheless, the approach is limited because of the known organization of eukaryotic chromatin into a topologically constrained three-dimensional network. This network positions distal regulatory elements in spatial proximity with gene promoters which are not predictable based on linear genomic distance. In this work, we capture long-range chromatin interactions using HiChIP to identify remote genomic regions that influence the differential regulation of depot-specific genes in ADSCs isolated from different adipose depots. By integrating these data with RNA-seq results and histone modifications identified by ChIP-seq, we uncovered distal regulatory elements that influence depot-specific gene expression in ADSCs. Interestingly, a subset of the HiChIP-defined chromatin loops also provide previously unknown connections between waist-to-hip ratio GWAS variants with genes that are known to significantly influence ADSC differentiation and adipocyte function.


Subject(s)
Adipocytes , Ascomycota , Humans , Promoter Regions, Genetic , Adipose Tissue , Chromatin/genetics , Stem Cells
7.
Cells ; 13(1)2023 12 30.
Article in English | MEDLINE | ID: mdl-38201289

ABSTRACT

Determining the mechanism driving body fat distribution will provide insights into obesity-related health risks. We used functional genomics tools to profile the epigenomic landscape to help infer the differential transcriptional potential of apple- and pear-shaped women's subcutaneous adipose-derived stem cells (ADSCs). We found that CCCTC-binding factor (CTCF) expression and its chromatin binding were increased in ADSCs from pear donors compared to those from apple donors. Interestingly, the pear enriched CTCF binding sites were located predominantly at the active transcription start sites (TSSs) of genes with active histone marks and YY1 motifs and were also associated with pear enriched RNAPII binding. In contrast, apple enriched CTCF binding sites were mainly found at intergenic regions and when identified at TSS, they were enriched with the bivalent chromatin signatures. Altogether, we provide evidence that CTCF plays an important role in differential regulation of subcutaneous ADSCs gene expression and may influence the development of apple vs. pear body shape.


Subject(s)
Gene Expression Regulation , Transcription Factors , Female , Humans , CCCTC-Binding Factor , Chromatin , Subcutaneous Fat
8.
iScience ; 25(8): 104772, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35992069

ABSTRACT

White adipose tissue (WAT) is a complex mixture of adipocytes and non-adipogenic cells. Characterizing the cellular composition of WAT is critical for identifying where potential alterations occur that impact metabolism. Most single-cell (sc) RNA-Seq studies focused on the stromal vascular fraction (SVF) which does not contain adipocytes and have used technology that has a 3' or 5' bias. Using full-length sc/single-nuclei (sn) RNA-Seq technology, we interrogated the transcriptional composition of WAT using: snRNA-Seq of whole tissue, snRNA-Seq of isolated adipocytes, and scRNA-Seq of SVF. Whole WAT snRNA-Seq provided coverage of major cell types, identified three distinct adipocyte clusters, and was capable of tracking adipocyte differentiation with pseudotime. Compared to WAT, adipocyte snRNA-Seq was unable to match adipocyte heterogeneity. SVF scRNA-Seq provided greater resolution of non-adipogenic cells. These findings provide critical evidence for the utility of sc full-length transcriptomics in WAT and SVF in humans.

9.
Genes Dev ; 36(9-10): 566-581, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35618313

ABSTRACT

Accumulation of fat above the waist is an important risk factor in developing obesity-related comorbidities independently of BMI or total fat mass. Deciphering the gene regulatory programs of the adipose tissue precursor cells within upper body or abdominal (ABD) and lower body or gluteofemoral (GF) depots is important to understand their differential capacity for lipid accumulation, maturation, and disease risk. Previous studies identified the HOX transcript antisense intergenic RNA (HOTAIR) as a GF-specific lncRNA; however, its role in adipose tissue biology is still unclear. Using three different approaches (silencing of HOTAIR in GF human adipose-derived stem cells [GF hASCs], overexpression of HOTAIR in ABD hASCs, and ChIRP-seq) to localize HOTAIR binding in GF hASC chromatin, we found that HOTAIR binds and modulates expression, both positively and negatively, of genes involved in adipose tissue-specific pathways, including adipogenesis. We further demonstrate a direct interaction between HOTAIR and genes with high RNAPII binding in their gene bodies, especially at their 3' ends or transcription end sites. Computational analysis suggests HOTAIR binds preferentially to the 3' ends of genes containing predicted strong RNA-RNA interactions with HOTAIR. Together, these results reveal a unique function for HOTAIR in hASC depot-specific regulation of gene expression.


Subject(s)
RNA, Long Noncoding , Adipocytes/metabolism , Adipose Tissue/metabolism , Gene Expression , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Stem Cells/metabolism
10.
Cells ; 11(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35269469

ABSTRACT

Polycystic ovary syndrome (PCOS) is often associated with metabolic syndrome features, including central obesity, suggesting that adipose tissue (AT) is a key organ in PCOS pathobiology. In this study, we compared both abdominal (ABD) and gluteofemoral (GF) subcutaneous AT in women with and without PCOS. ABD and GF subcutaneous ATs from PCOS and BMI/WHR-matched control women were analyzed by RT-qPCR, FACS and histology. ABD and GF adipose-derived stem cell (ASC) transcriptome and methylome were analyzed by RNA-seq and DNA methylation array. Similar to the control group with abdominal obesity, the GF AT of PCOS women showed lower expression of genes involved in lipid accumulation and angiogenesis compared to ABD depot. FACS analysis revealed an increase in preadipocytes number in both AT depots from PCOS. Further pathway analysis of RNA-seq comparisons demonstrated that the ASCs derived from PCOS are pro-inflammatory and exhibit a hypoxic signature in the ABD depot and have lower expression of adipogenic genes in GF depot. We also found a higher CpG methylation level in PCOS compared to control exclusively in GF-ASCs. Our data suggest that ASCs play an important role in the etiology of PCOS, potentially by limiting expansion of the healthy lower-body AT.


Subject(s)
Polycystic Ovary Syndrome , Adipose Tissue , DNA Methylation/genetics , Female , Humans , Obesity/genetics , Obesity, Abdominal , Polycystic Ovary Syndrome/genetics , Stem Cells , Subcutaneous Fat
11.
Front Genet ; 12: 709342, 2021.
Article in English | MEDLINE | ID: mdl-34394195

ABSTRACT

Preferential accumulation of fat in the gluteo-femoral (GF) depot (pear shape) rather than in the abdominal (A) depot (apple shape), protects against the development of metabolic diseases but the underlying molecular mechanism is still unknown. Recent data, including our work, suggest that differential epigenetic marking is associated with regulation of genes attributed to distinct fat distribution. Here, we aimed to compare the genomic DNA methylation signatures between apple and pear-shaped premenopausal women. To investigate the contribution of upper and lower body fat, we used paired samples of A-FAT and GF-FAT, analyzed on the BeadChip Methylation Array and quantified the differentially methylated sites between the 2 groups of women. We found unique DNA methylation patterns within both fat depots that are significantly different depending on the body fat distribution. Around 60% of the body shape specific DNA methylation sites identified in adipose tissue are maintained ex vivo in cultured preadipocytes. As it has been reported before in other cell types, we found only a hand full of genes showing coordinated differential methylation and expression levels. Finally, we determined that more than 50% of the body shape specific DNA methylation sites could also be detected in whole blood derived DNA. These data reveal a strong DNA methylation program associated with adipose tissue distribution with the possibility that a simple blood test could be used as a predictive diagnostic indicator of young women who are at increased risk for progressing to the apple body shape with a higher risk of developing obesity related complications. Clinical Trial Registration:https://clinicaltrials.gov/ct2/show/NCT02728635 and https://clinicaltrials.gov/ct2/show/NCT02226640, identifiers NCT02728635 and NCT02226640.

12.
J Endocr Soc ; 4(6): bvaa042, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32500109

ABSTRACT

BACKGROUND: Preferential accumulation of fat in the upper body (apple shape) is associated with higher risk of developing metabolic syndrome relative to lower body fat (pear shape). We previously discovered that chromatin openness partially defined the transcriptome of preadipocytes isolated from abdominal and gluteofemoral fat. However, the molecular mechanisms underlying interindividual variation in body shape are unknown. METHODS: Adipocyte fraction was isolated from abdominal and gluteofemoral fat biopsies of premenopausal women (age and body mass index matched) segregated initially only by their waist-to-hip ratio. We evaluated transcriptomic and chromatin accessibility using RNA sequencing and assay for transposase-accessible chromatin using sequencing (ATAC-seq) along with key clinical parameters. RESULTS: Our data showed that higher lower body fat mass was associated with better lipid profile and free fatty acid decrease after glucose administration. Lipid and glucose metabolic pathways genes were expressed at higher levels in gluteofemoral adipocyte fraction in pears, whereas genes associated with inflammation were higher both in abdominal and gluteofemoral apple adipocyte fraction. Gluteofemoral adipocyte chromatin from pear-shaped women contained a significantly higher number of differentially open ATAC-seq peaks relative to chromatin from the apple-shaped gluteofemoral adipocytes. In contrast, abdominal adipocyte chromatin openness showed few differences between apple- and pear-shaped women. We revealed a correlation between gene transcription and open chromatin at the proximity of the transcriptional start site of some of the differentially expressed genes. CONCLUSIONS: Integration of data from all 3 approaches suggests that chromatin openness partially governs the transcriptome of gluteofemoral adipocytes and may be involved in the early metabolic syndrome predisposition associated with body shape.

13.
Clin Epigenetics ; 10(1): 148, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30477572

ABSTRACT

BACKGROUND: Increased lower body fat is associated with reduced cardiometabolic risk. The molecular basis for depot-specific differences in gluteofemoral (GF) compared with abdominal (A) subcutaneous adipocyte function is poorly understood. In the current report, we used a combination of Assay for Transposase-Accessible Chromatin followed by sequencing (ATAC-seq), RNA-seq, and chromatin immunoprecipitation (ChIP)-qPCR analyses that provide evidence that depot-specific gene expression patterns are associated with differential epigenetic chromatin signatures. METHODS: Preadipocytes cultured from A and GF adipose tissue obtained from premenopausal apple-shaped women were used to perform transcriptome analysis by RNA-seq and assess accessible chromatin regions by ATAC-seq. We measured mRNA expression and performed ChIP-qPCR experiments for histone modifications of active (H3K4me3) and repressed chromatin (H3K27me3) regions respectively on the promoter regions of differentially expressed genes. RESULTS: RNA-seq experiments revealed an A-fat and GF-fat selective gene expression signature, with 126 genes upregulated in abdominal preadipocytes and 90 genes upregulated in GF cells. ATAC-seq identified almost 10-times more A-specific chromatin-accessible regions. Using a combined analysis of ATAC-seq and global gene expression data, we identified 74 of the 126 abdominal-specific genes (59%) with A-specific accessible chromatin sites within 200 kb of the transcription start site (TSS), including HOXA3, HOXA5, IL8, IL1b, and IL6. Interestingly, only 14 of the 90 GF-specific genes (15%) had GF-specific accessible chromatin sites within 200 kb of the corresponding TSS, including HOXC13 and HOTAIR, whereas 25 of them (28%) had abdominal-specific accessible chromatin sites. ChIP-qPCR experiments confirmed that the active H3K4me3 chromatin mark was significantly enriched at the promoter regions of HOXA5 and HOXA3 genes in abdominal preadipocytes, while H3K27me3 was less abundant relative to chromatin from GF. This is consistent with their A-fat specific gene expression pattern. Conversely, analysis of the promoter regions of the GF specific HOTAIR and HOXC13 genes exhibited high H3K4me3 and low H3K27me3 levels in GF chromatin compared to A chromatin. CONCLUSIONS: Global transcriptome and open chromatin analyses of depot-specific preadipocytes identified their gene expression signature and differential open chromatin profile. Interestingly, A-fat-specific open chromatin regions can be observed in the proximity of GF-fat genes, but not vice versa. TRIAL REGISTRATION: Clinicaltrials.gov, NCT01745471 . Registered 5 December 2012.


Subject(s)
Chromatin/genetics , Gene Expression Profiling/methods , Menopause/genetics , Sequence Analysis, RNA/methods , Subcutaneous Fat/cytology , Adipocytes/cytology , Adult , Cells, Cultured , Chromatin Immunoprecipitation , DNA Methylation , Epigenesis, Genetic , Female , Histone Code , Humans , Promoter Regions, Genetic , Young Adult
14.
Obesity (Silver Spring) ; 26(1): 135-140, 2018 01.
Article in English | MEDLINE | ID: mdl-29178600

ABSTRACT

OBJECTIVE: Human brown adipose tissue (BAT) activity decreases with age and obesity. In addition to uncoupling protein 1 (UCP1), several genetic markers of BAT in humans have been published. However, the link between human BAT activity and genetic markers has been inadequately explored. METHODS: White adipose tissue (WAT) and BAT biopsies were obtained from 16 patients undergoing deep neck surgery. In vitro differentiated adipocytes were used to measure norepinephrine-stimulated mitochondrial uncoupling as a measure of in vitro BAT activity. Gene expression was determined in adipose tissue biopsies. RESULTS: Norepinephrine increased in vitro BAT activity in adipocytes derived from human BAT, and this increase was abolished by propranolol. Furthermore, in vitro BAT activity showed a negative correlation to age and BMI. UCP1 messenger RNA (mRNA) expression showed a positive correlation to in vitro BAT activity, while zinc finger protein of cerebellum 1 (ZIC1) mRNA showed a negative correlation to in vitro BAT activity. In human BAT biopsies, UCP1 mRNA showed negative correlations to age and BMI, while ZIC1 mRNA showed positive correlations to age and BMI. CONCLUSIONS: Differentiated adipocytes derived from human BAT maintain intrinsic characteristics of the donor. High ZIC1 mRNA does not necessarily reflect high BAT activity.


Subject(s)
Adipose Tissue, Brown/metabolism , Genetic Markers/genetics , Adult , Aged , Animals , Cell Differentiation , Female , Humans , Male , Middle Aged , Young Adult
15.
Obesity (Silver Spring) ; 25(8): 1375-1383, 2017 08.
Article in English | MEDLINE | ID: mdl-28649807

ABSTRACT

OBJECTIVE: Lower body fat is associated with diminishing cardiometabolic risk. Physiological differences between gluteofemoral and abdominal subcutaneous adipocyte functions are known, but the molecular basis for depot differences in adipocyte function is poorly understood. The objective of this study was to identify depot differences in microRNA (miRNA) expression in human abdominal and gluteofemoral subcutaneous adipose tissues and their implication in gene regulation. METHODS: Abdominal and gluteofemoral adipose tissue aspirates obtained from 18 participants (9 male and 9 female, age 30 ± 1.5 y, BMI 27.3 ± 1.23 kg/m2 ) were analyzed for miRNA expression profiles by next-generation DNA sequencing. The raw reads were mapped to miRBase 17, and differentially expressed miRNAs were confirmed by qRT-PCR. The hsa-mimic-miR196a was transfected into cultured abdominal preadipocytes isolated from five women with obesity. Target gene expression was evaluated by RT-qPCR. RESULTS: Among the 640 miRNAs detected in adipose tissue, miR196a2, miR196a1, miR196b, and miR204 showed a higher expression in the gluteofemoral depot (fold change = 2.7, 2.3, 1.7, and 2.3, respectively) independent of sex. Bioinformatic analyses and human primary preadipocyte transfection with miR196 suggested that the differentially expressed miRNAs could directly or indirectly modulate homeobox (HOX) gene expression. CONCLUSIONS: The miR196 gene family could play an important role in the regulation of HOX gene expression in subcutaneous adipose tissue and in fat distribution variation.


Subject(s)
Buttocks , Homeodomain Proteins/genetics , MicroRNAs/genetics , Subcutaneous Fat/metabolism , Adipocytes/metabolism , Adult , Body Mass Index , Computational Biology , Female , Gene Expression Regulation , Homeodomain Proteins/metabolism , Humans , Male , MicroRNAs/metabolism , Obesity/genetics , Obesity/metabolism , Sequence Analysis, DNA
16.
Proc Natl Acad Sci U S A ; 113(42): E6418-E6426, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27708159

ABSTRACT

We describe a metabolic disorder characterized by lipodystrophy, hepatic steatosis, insulin resistance, severe diabetes, and growth retardation observed in mice carrying N-ethyl-N-nitrosourea (ENU)-induced mutations. The disorder was ascribed to a mutation of kelch repeat and BTB (POZ) domain containing 2 (Kbtbd2) and was mimicked by a CRISPR/Cas9-targeted null allele of the same gene. Kbtbd2 encodes a BTB-Kelch family substrate recognition subunit of the Cullin-3-based E3 ubiquitin ligase. KBTBD2 targeted p85α, the regulatory subunit of the phosphoinositol-3-kinase (PI3K) heterodimer, causing p85α ubiquitination and proteasome-mediated degradation. In the absence of KBTBD2, p85α accumulated to 30-fold greater levels than in wild-type adipocytes, and excessive p110-free p85α blocked the binding of p85α-p110 heterodimers to IRS1, interrupting the insulin signal. Both transplantation of wild-type adipose tissue and homozygous germ line inactivation of the p85α-encoding gene Pik3r1 rescued diabetes and hepatic steatosis phenotypes of Kbtbd2-/- mice. Kbtbd2 was down-regulated in diet-induced obese insulin-resistant mice in a leptin-dependent manner. KBTBD2 is an essential regulator of the insulin-signaling pathway, modulating insulin sensitivity by limiting p85α abundance.


Subject(s)
Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Diet/adverse effects , Genetic Predisposition to Disease , Insulin Resistance , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/transplantation , Animals , Blood Glucose , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Cullin Proteins/metabolism , Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation , Genetic Association Studies , Genotype , Insulin/blood , Insulin Resistance/genetics , Lipodystrophy/etiology , Lipodystrophy/metabolism , Lipodystrophy/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Obesity/etiology , Obesity/pathology , Phenotype , Protein Binding , Protein Transport , Signal Transduction , Ubiquitination
17.
Obesity (Silver Spring) ; 22(8): 1781-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24862299

ABSTRACT

OBJECTIVE: Peripheral lower body fat is associated with lower cardiometabolic risk. Physiological differences in gluteal compared with abdominal subcutaneous (sc) adipocyte functions are known but the molecular basis for depot differences in adipocyte function is poorly understood. Our goal is to identify novel gene regulatory pathways that underlie the heterogeneity of human fat distribution. METHODS: Abdominal and gluteal adipose tissue aspirates obtained from 35 subjects (age = 30 ± 1.6 years; BMI = 27.3 ± 1.3 kg/m(2) ) were analyzed using Illumina microarrays and confirmed by RT-PCR. The HOTAIR gene was stably transfected into primary cultured human abdominal sc preadipocytes using a lentivirus and effects on adipogenic differentiation were analyzed. RESULTS: A long noncoding RNA, HOTAIR that was expressed in gluteal but not in Abd sc adipose tissue was identified. This difference was retained throughout in vitro differentiation and was maximal at day 4. Ectopic expression of HOTAIR in abdominal preadipocytes produced an increase in differentiation as reflected by a higher percentage of differentiated cells, and increased expression of key adipogenic genes including PPARγ and LPL. CONCLUSIONS: HOTAIR is expressed in gluteal adipose and may regulate key processes in adipocyte differentiation. The role of this lncRNA in determining the metabolic properties of gluteal compared with abdominal adipocytes merits further study.


Subject(s)
Adipocytes/cytology , Cell Differentiation , RNA, Long Noncoding/genetics , Subcutaneous Fat/metabolism , Adult , Body Fat Distribution , Body Mass Index , Cells, Cultured , Female , Humans , Male , PPAR gamma/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
J Clin Endocrinol Metab ; 98(1): 362-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23150689

ABSTRACT

CONTEXT: Fat distribution differs in men and women, but in both sexes, a predominantly gluteal-femoral compared with abdominal (central) fat distribution is associated with lower metabolic risk. Differences in cellular characteristics and metabolic functions of these depots have been described, but the molecular mechanisms involved are not understood. OBJECTIVE: Our objective was to identify depot- and sex-dependent differences in gene expression in human abdominal and gluteal sc adipose tissues. DESIGN AND METHODS: Abdominal and gluteal adipose tissue aspirates were obtained from 14 premenopausal women [age 27.5 ± 7.0 yr, body mass index (BMI) 27.3 ± 6.2 kg/m(2), and waist-to-hip ratio 0.82 ± 0.04] and 21 men (age 29.7±7.4 yr, BMI 27.2 ± 4.5 kg/m(2), and waist-to-hip ratio 0.91 ± 0.07) and transcriptomes were analyzed using Illumina microarrays. Expression of selected genes was determined in isolated adipocytes and stromal vascular fractions from each depot, and in in vitro cultures before and after adipogenic differentiation. RESULTS: A total of 284 genes were differentially expressed between the abdominal and gluteal depot, either specifically in males (n = 66) or females (n = 159) or in both sexes (n = 59). Most notably, gene ontology and pathway analysis identified homeobox genes (HOXA2, HOXA3, HOXA4, HOXA5, HOXA9, HOXB7, HOXB8, HOXC8, and IRX2) that were down-regulated in the gluteal depot in both sexes (P = 2 × 10(-10)). Conversely, HOXA10 was up-regulated in gluteal tissue and HOXC13 was detected exclusively in this depot. These differences were independent of BMI, were present in both adipocytes and stromal vascular fractions of adipose tissue, and were retained throughout in vitro differentiation. CONCLUSIONS: We conclude that developmentally programmed differences may contribute to the distinct phenotypic characteristics of peripheral fat.


Subject(s)
Abdominal Fat/metabolism , Subcutaneous Fat/metabolism , Transcriptome , Abdominal Fat/chemistry , Adolescent , Adult , Body Composition/genetics , Buttocks , Female , Gene Expression Regulation, Developmental , Humans , Male , Obesity/genetics , Obesity/metabolism , Sex Characteristics , Subcutaneous Fat/chemistry , Young Adult
19.
J Clin Endocrinol Metab ; 97(9): E1677-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22745246

ABSTRACT

AIMS AND HYPOTHESIS: Mast cells are immune cells known for their role in several inflammatory and fibrotic diseases. Recent works in mice suggest that mast cells could be cellular actors involved in the pathophysiology of obesity, a disease characterized by white adipose tissue (WAT) and systemic inflammation. The aim of the study was to better characterize mast cells in WAT of obese with or without type 2 diabetes and lean subjects as well as to explore the relationship with WAT inflammation and fibrosis. METHODS: Subcutaneous and omental adipose tissue from six lean subjects, 10 obese nondiabetic, and 10 diabetic patients was analyzed by immunohistochemistry and real-time PCR for inflammatory and fibrosis markers. Cytokines secretion of mast cells isolated from WAT and cultured in different conditions was estimated by cytokine array kit. RESULTS: We found that mast cells are activated in human adipose tissue and localized preferentially in fibrosis depots, a local condition that stimulates their inflammatory state. Mast cells with tryptase(+) chymase(+) staining tended to be higher in obese omental adipose tissue. We found positive links between mast cell number and several characteristics of obese WAT including fibrosis, macrophage accumulation, and endothelial cell inflammation. Mast cell number and their inflammatory phenotype are associated with diabetes parameters. CONCLUSION AND INTERPRETATION: Mast cells are cellular actors of WAT inflammation and possibly fibrotic state found in obesity and diabetes. Whether mast cells could be involved in the pathophysiology of diabetes needs additional study as well as the positioning of these cells in driving pathological alterations of WAT in these chronic metabolic diseases.


Subject(s)
Adipose Tissue/pathology , Diabetes Mellitus, Type 2/pathology , Inflammation/pathology , Mast Cells/pathology , Obesity, Morbid/pathology , Adipose Tissue, White/pathology , Adult , Biomarkers/analysis , Blood Glucose/metabolism , Cell Count , Cell Separation , Chymases/chemistry , Endothelial Cells/pathology , Female , Fibrosis/pathology , Homeostasis/physiology , Humans , Immunohistochemistry , Lipids/blood , Liver Function Tests , Male , Middle Aged , Obesity, Morbid/metabolism , Phenotype , Real-Time Polymerase Chain Reaction , Tryptases/chemistry
20.
J Clin Endocrinol Metab ; 97(4): 1320-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22259057

ABSTRACT

CONTEXT: Extracellular matrix (ECM) remodeling is essential for adipose tissue growth and expansion in high fat-fed mice, and there is evidence of fibrosis in adipose tissue in human obesity. OBJECTIVE: The aim of the study was to explore the role of ECM remodeling in adipose tissue in healthy, growing children. RESEARCH DESIGN, SETTING, AND PARTICIPANTS: Abdominal sc adipose biopsies were obtained from 65 otherwise healthy children [57 boys; age, 5.3 ± 3.8 yr (mean ± sd)] having elective surgery (cross-sectional study). Twenty percent of the participants were classified as overweight/obese based on body mass index (BMI) z score. MAIN OUTCOME MEASURES: We examined collagen (total and pericellular), HAM56+ macrophages, CD206+ M2 phenotype macrophages, and CD3+ T cells measured by immunohistochemistry and ECM gene expression markers. RESULTS: Overweight children had significantly less total collagen compared to normal weight children (median, 3.4 vs. 9.1%; P = 0.001). However, collagen areas were not positive for COL6 and showed little evidence of collagen surrounding adipocytes. Fat cell size was negatively correlated with the percentage of total (r = -0.398; P = 0.003) and pericellular collagen (r = -0.462; P < 0.001) but positively correlated with HAM56+ macrophages (r = 0.541; P < 0.001). The percentage of total collagen was inversely associated with BMI z score (r = -0.345; P = 0.01) and age (r = -0.348; P = 0.005), with older (>11 yr old) children in the top BMI z tertile having less collagen (3.8%) than younger (2-5 yr old) children in the bottom BMI z tertile (12.6%). Adipose tissue in overweight children showed little evidence of crown-like structures or T cells. CONCLUSION: In healthy, growing children, increased collagen in adipose tissue is associated with decreased fat cell size and BMI z score and increased M2+ phenotype macrophages, suggesting dynamic interaction between ECM remodeling and immune cells even at an early age.


Subject(s)
Adipogenesis , Child Development , Collagen/metabolism , Extracellular Matrix/metabolism , Overweight/metabolism , Subcutaneous Fat, Abdominal/growth & development , Biomarkers/metabolism , Biopsy , Body Mass Index , Cell Count , Cell Size , Child , Child, Preschool , Cohort Studies , Collagen/genetics , Cross-Sectional Studies , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Gene Expression Regulation, Developmental , Humans , Macrophages/metabolism , Macrophages/pathology , Male , New South Wales , Overweight/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Subcutaneous Fat, Abdominal/immunology , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...