Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Trends Mol Med ; 30(4): 295-296, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38604129
2.
Trends Mol Med ; 30(1): 1-2, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38211558
3.
Nat Commun ; 14(1): 4162, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443109

ABSTRACT

The current obesity epidemic and high prevalence of metabolic diseases necessitate efficacious and safe treatments. Brown adipose tissue in this context is a promising target with the potential to increase energy expenditure, however no pharmacological treatments activating brown adipose tissue are currently available. Here, we identify AXL receptor tyrosine kinase as a regulator of adipose function. Pharmacological and genetic inhibition of AXL enhance thermogenic capacity of brown and white adipocytes, in vitro and in vivo. Mechanistically, these effects are mediated through inhibition of PI3K/AKT/PDE signaling pathway, resulting in induction of nuclear FOXO1 localization and increased intracellular cAMP levels via PDE3/4 inhibition and subsequent stimulation of the PKA-ATF2 pathway. In line with this, both constitutive Axl deletion as well as inducible adipocyte-specific Axl deletion protect animals from diet-induced obesity concomitant with increases in energy expenditure. Based on these data, we propose AXL receptor as a target for the treatment of obesity.


Subject(s)
Adipose Tissue, Brown , Axl Receptor Tyrosine Kinase , Mice , Animals , Adipose Tissue, Brown/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Obesity/metabolism , Adipocytes, White/metabolism , Energy Metabolism , Adipose Tissue, White/metabolism , Thermogenesis/genetics , Adipocytes, Brown/metabolism , Mice, Inbred C57BL , Adipose Tissue/metabolism
4.
Nat Med ; 28(12): 2537-2546, 2022 12.
Article in English | MEDLINE | ID: mdl-36536256

ABSTRACT

Serotonin reuptake inhibitors and receptor agonists are used to treat obesity, anxiety and depression. Here we studied the role of the serotonin 2C receptor (5-HT2CR) in weight regulation and behavior. Using exome sequencing of 2,548 people with severe obesity and 1,117 control individuals without obesity, we identified 13 rare variants in the gene encoding 5-HT2CR (HTR2C) in 19 unrelated people (3 males and 16 females). Eleven variants caused a loss of function in HEK293 cells. All people who carried variants had hyperphagia and some degree of maladaptive behavior. Knock-in male mice harboring a human loss-of-function HTR2C variant developed obesity and reduced social exploratory behavior; female mice heterozygous for the same variant showed similar deficits with reduced severity. Using the 5-HT2CR agonist lorcaserin, we found that depolarization of appetite-suppressing proopiomelanocortin neurons was impaired in knock-in mice. In conclusion, we demonstrate that 5-HT2CR is involved in the regulation of human appetite, weight and behavior. Our findings suggest that melanocortin receptor agonists might be effective in treating severe obesity in individuals carrying HTR2C variants. We suggest that HTR2C should be included in diagnostic gene panels for severe childhood-onset obesity.


Subject(s)
Obesity, Morbid , Receptor, Serotonin, 5-HT2C , Animals , Child , Female , Humans , Male , Mice , HEK293 Cells , Obesity/genetics , Receptor, Serotonin, 5-HT2C/genetics , Serotonin , Serotonin 5-HT2 Receptor Agonists/pharmacology , Adaptation, Psychological
5.
Open Biol ; 12(3): 210345, 2022 03.
Article in English | MEDLINE | ID: mdl-35291877

ABSTRACT

Obesity, defined as an excess of adipose tissue that adversely affects health, is a major cause of morbidity and mortality. However, to date, understanding the structure and function of human adipose tissue has been limited by the inability to visualize cellular components due to the innate structure of adipocytes, which are characterized by large lipid droplets. Combining the iDISCO and the CUBIC protocols for whole tissue staining and optical clearing, we developed a protocol to enable immunostaining and clearing of human subcutaneous white adipose tissue (WAT) obtained from individuals with severe obesity. We were able to perform immunolabelling of sympathetic nerve terminals in whole WAT and subsequent optical clearing by eliminating lipids to render the opaque tissue completely transparent. We then used light sheet confocal microscopy to visualize sympathetic innervation of human WAT from obese individuals in a three-dimensional manner. We demonstrate the visualization of sympathetic nerve terminals in human WAT. This protocol can be modified to visualize other structures such as blood vessels involved in the development, maintenance and function of human adipose tissue in health and disease.


Subject(s)
Adipose Tissue, White , Adipose Tissue , Adipocytes , Adipose Tissue, White/innervation , Humans , Obesity , Sympathetic Nervous System/physiology
6.
J Clin Endocrinol Metab ; 107(6): e2532-e2544, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35137184

ABSTRACT

CONTEXT: Genetic variants affecting the nuclear hormone receptor coactivator steroid receptor coactivator, SRC-1, have been identified in people with severe obesity and impair melanocortin signaling in cells and mice. As a result, obese patients with SRC-1 deficiency are being treated with a melanocortin 4 receptor agonist in clinical trials. OBJECTIVE: Here, our aim was to comprehensively describe and characterize the clinical phenotype of SRC-1 variant carriers to facilitate diagnosis and clinical management. METHODS: In genetic studies of 2462 people with severe obesity, we identified 23 rare heterozygous variants in SRC-1. We studied 29 adults and 18 children who were SRC-1 variant carriers and performed measurements of metabolic and endocrine function, liver imaging, and adipose tissue biopsies. Findings in adult SRC-1 variant carriers were compared to 30 age- and body mass index (BMI)-matched controls. RESULTS: The clinical spectrum of SRC-1 variant carriers included increased food intake in children, normal basal metabolic rate, multiple fractures with minimal trauma (40%), persistent diarrhea, partial thyroid hormone resistance, and menorrhagia. Compared to age-, sex-, and BMI-matched controls, adult SRC-1 variant carriers had more severe adipose tissue fibrosis (46.2% vs 7.1% respectively, P = .03) and a suggestion of increased liver fibrosis (5/13 cases vs 2/13 in controls, odds ratio = 3.4), although this was not statistically significant. CONCLUSION: SRC-1 variant carriers exhibit hyperphagia in childhood, severe obesity, and clinical features of partial hormone resistance. The presence of adipose tissue fibrosis and hepatic fibrosis in young patients suggests that close monitoring for the early development of obesity-associated metabolic complications is warranted.


Subject(s)
Nuclear Receptor Coactivator 1 , Obesity, Morbid , Female , Fibrosis , Humans , Male , Nuclear Receptor Coactivator 1/genetics , Obesity, Morbid/complications , Obesity, Morbid/genetics
7.
N Engl J Med ; 385(17): 1581-1592, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34614324

ABSTRACT

BACKGROUND: GNAS encodes the Gαs (stimulatory G-protein alpha subunit) protein, which mediates G protein-coupled receptor (GPCR) signaling. GNAS mutations cause developmental delay, short stature, and skeletal abnormalities in a syndrome called Albright's hereditary osteodystrophy. Because of imprinting, mutations on the maternal allele also cause obesity and hormone resistance (pseudohypoparathyroidism). METHODS: We performed exome sequencing and targeted resequencing in 2548 children who presented with severe obesity, and we unexpectedly identified 22 GNAS mutation carriers. We investigated whether the effect of GNAS mutations on melanocortin 4 receptor (MC4R) signaling explains the obesity and whether the variable clinical spectrum in patients might be explained by the results of molecular assays. RESULTS: Almost all GNAS mutations impaired MC4R signaling. A total of 6 of 11 patients who were 12 to 18 years of age had reduced growth. In these patients, mutations disrupted growth hormone-releasing hormone receptor signaling, but growth was unaffected in carriers of mutations that did not affect this signaling pathway (mean standard-deviation score for height, -0.90 vs. 0.75, respectively; P = 0.02). Only 1 of 10 patients who reached final height before or during the study had short stature. GNAS mutations that impaired thyrotropin receptor signaling were associated with developmental delay and with higher thyrotropin levels (mean [±SD], 8.4±4.7 mIU per liter) than those in 340 severely obese children who did not have GNAS mutations (3.9±2.6 mIU per liter; P = 0.004). CONCLUSIONS: Because pathogenic mutations may manifest with obesity alone, screening of children with severe obesity for GNAS deficiency may allow early diagnosis, improving clinical outcomes, and melanocortin agonists may aid in weight loss. GNAS mutations that are identified by means of unbiased genetic testing differentially affect GPCR signaling pathways that contribute to clinical heterogeneity. Monogenic diseases are clinically more variable than their classic descriptions suggest. (Funded by Wellcome and others.).


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Mutation , Pediatric Obesity/genetics , Receptor, Melanocortin, Type 4/metabolism , Adolescent , Body Height , Child , Chromogranins/genetics , Female , GTP-Binding Protein alpha Subunits, Gs/deficiency , Humans , Male , Mutation, Missense , Receptors, Thyrotropin/metabolism , Signal Transduction , Exome Sequencing
8.
Cell Metab ; 31(6): 1107-1119.e12, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492392

ABSTRACT

Obesity is genetically heterogeneous with monogenic and complex polygenic forms. Using exome and targeted sequencing in 2,737 severely obese cases and 6,704 controls, we identified three genes (PHIP, DGKI, and ZMYM4) with an excess burden of very rare predicted deleterious variants in cases. In cells, we found that nuclear PHIP (pleckstrin homology domain interacting protein) directly enhances transcription of pro-opiomelanocortin (POMC), a neuropeptide that suppresses appetite. Obesity-associated PHIP variants repressed POMC transcription. Our demonstration that PHIP is involved in human energy homeostasis through transcriptional regulation of central melanocortin signaling has potential diagnostic and therapeutic implications for patients with obesity and developmental delay. Additionally, we found an excess burden of predicted deleterious variants involving genes nearest to loci from obesity genome-wide association studies. Genes and gene sets influencing obesity with variable penetrance provide compelling evidence for a continuum of causality in the genetic architecture of obesity, and explain some of its missing heritability.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Pediatric Obesity/genetics , Pro-Opiomelanocortin/genetics , Adult , Animals , Cells, Cultured , Child , Chlorocebus aethiops , Exome , Female , Genetic Variation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
9.
Article in English | MEDLINE | ID: mdl-32595605

ABSTRACT

When exposed to cold temperatures, mice increase their thermogenic capacity by an expansion of brown adipose tissue mass and the formation of brite/beige adipocytes in white adipose tissue depots. However, the process of the transcriptional changes underlying the conversion of a phenotypic white to brite/beige adipocytes is only poorly understood. By analyzing transcriptome profiles of inguinal adipocytes during cold exposure and in mouse models with a different propensity to form brite/beige adipocytes, we identified ESRRG and PERM1 as modulators of this process. The production of heat by mitochondrial uncoupled respiration is a key feature of brite/beige compared to white adipocytes and we show here that both candidates are involved in PGC1α transcriptional network to positively regulate mitochondrial capacity. Moreover, we show that an increased expression of ESRRG or PERM1 supports the formation of brown or brite/beige adipocytes in vitro and in vivo. These results reveal that ESRRG and PERM1 are early induced in and important regulators of brite/beige adipocyte formation.


Subject(s)
Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Mitochondria/metabolism , Muscle Proteins/metabolism , Receptors, Estrogen/metabolism , Thermogenesis , Animals , Cell Line , Cold Temperature , Male , Mice, Inbred C57BL , Transcriptome
10.
Cell Rep ; 25(3): 784-797.e4, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332656

ABSTRACT

Recruitment and activation of thermogenic adipocytes have received increasing attention as a strategy to improve systemic metabolic control. The analysis of brown and brite adipocytes is complicated by the complexity of adipose tissue biopsies. Here, we provide an in-depth analysis of pure brown, brite, and white adipocyte transcriptomes. By combining mouse and human transcriptome data, we identify a gene signature that can classify brown and white adipocytes in mice and men. Using a machine-learning-based cell deconvolution approach, we develop an algorithm proficient in calculating the brown adipocyte content in complex human and mouse biopsies. Applying this algorithm, we can show in a human weight loss study that brown adipose tissue (BAT) content is associated with energy expenditure and the propensity to lose weight. This online available tool can be used for in-depth characterization of complex adipose tissue samples and may support the development of therapeutic strategies to increase energy expenditure in humans.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Biomarkers/analysis , Computational Biology/methods , Obesity/physiopathology , Software , Adipogenesis , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Adult , Aged , Animals , Cohort Studies , Energy Metabolism , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Thermogenesis , Young Adult
11.
Nat Med ; 24(11): 1776, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30087435

ABSTRACT

In the version of this article originally published, the months on the axis labeled projected month of conception in Fig. 1a were out of order. April and March should have been the first and last months listed, respectively. The error has been corrected in the print, PDF and HTML versions of this article.

12.
Nat Med ; 24(11): 1777, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30087436

ABSTRACT

In the version of this article originally published, the bars in the mean temperature graph in Fig. 1a were incorrectly aligned. The left-most bar should have been aligned with the Apr label on the projected month of conception axis. The error has been corrected in the print, PDF and HTML versions of this article.

13.
Nat Med ; 24(9): 1372-1383, 2018 09.
Article in English | MEDLINE | ID: mdl-29988127

ABSTRACT

Recent research has focused on environmental effects that control tissue functionality and systemic metabolism. However, whether such stimuli affect human thermogenesis and body mass index (BMI) has not been explored. Here we show retrospectively that the presence of brown adipose tissue (BAT) and the season of conception are linked to BMI in humans. In mice, we demonstrate that cold exposure (CE) of males, but not females, before mating results in improved systemic metabolism and protection from diet-induced obesity of the male offspring. Integrated analyses of the DNA methylome and RNA sequencing of the sperm from male mice revealed several clusters of co-regulated differentially methylated regions (DMRs) and differentially expressed genes (DEGs), suggesting that the improved metabolic health of the offspring was due to enhanced BAT formation and increased neurogenesis. The conclusions are supported by cell-autonomous studies in the offspring that demonstrate an enhanced capacity to form mature active brown adipocytes, improved neuronal density and more norepinephrine release in BAT in response to cold stimulation. Taken together, our results indicate that in humans and in mice, seasonal or experimental CE induces an epigenetic programming of the sperm such that the offspring harbor hyperactive BAT and an improved adaptation to overnutrition and hypothermia.


Subject(s)
Adipose Tissue, Brown/metabolism , Cold Temperature , Epigenesis, Genetic , Spermatozoa/metabolism , Adipocytes, Brown/metabolism , Animals , DNA Methylation/genetics , Diet, High-Fat , Female , HEK293 Cells , Humans , Insulin Resistance , Male , Mice, Inbred C57BL , Neurogenesis , Obesity/metabolism , Oxygen Consumption , Pregnancy , Principal Component Analysis , Receptors, Adrenergic, beta-3/metabolism , Uncoupling Protein 1/metabolism
14.
Sci Signal ; 10(466)2017 02 14.
Article in English | MEDLINE | ID: mdl-28196906

ABSTRACT

Brown adipose tissue (BAT) is responsible for thermogenesis that is not associated with shivering through the process of converting chemical energy into heat through uncoupling protein 1 (UCP1) in the mitochondria. Thus, expanding or activating BAT could be a potential tool against obesity. To analyze the effect of kinase signaling on brown adipocyte formation, a process that describes the acquisition of the ability to dissipate energy as heat, we performed lentiviral-mediated short hairpin knockdown or used pharmacological inhibitors in a high-content and high-throughput in vitro image-based screen. We identified 190 kinases that either stimulated or inhibited brown adipocyte proliferation, differentiation, or formation. Among these kinases, we found that 5' AMP-activated protein kinase (AMPK) promoted the formation of brown adipocytes abundant inUCP1. Together, our results provide insight into the kinases, particularly AMPK, that regulate brown adipocyte formation.


Subject(s)
Adipocytes, Brown/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , High-Throughput Screening Assays/methods , Phosphotransferases/genetics , RNA Interference , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Blotting, Western , Cells, Cultured , Mice , Phosphotransferases/classification , Phosphotransferases/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Proteomics/methods
15.
Biochim Biophys Acta ; 1861(9 Pt A): 1121-1131, 2016 09.
Article in English | MEDLINE | ID: mdl-27317982

ABSTRACT

Visceral and subcutaneous adipose tissue depots have distinct features and contribute differentially to the development of metabolic dysfunction. We show here that adipocyte differentiation in subcutaneous stromal-vascular fraction (SVF) is increased compared to visceral SVF, however this increased differentiation capacity seems not to be due to changes in the number of adipocyte precursor cells. Rather, we demonstrate that secreted heat-sensitive factors from the SVF can inhibit adipocyte differentiation and that this effect is higher in visceral than in subcutaneous SVF, suggesting that visceral SVF is a source of secreted factors that can inhibit adipocyte formation. In order to explore secreted proteins that potentially inhibit differentiation in visceral preadipocytes we analyzed the secretome of both SVFs which led to the identification of 113 secreted proteins with an overlap of 42%. Further expression analysis in both depots revealed 16 candidates that were subsequently analyzed in a differentiation screen using an adenoviral knockdown system. From this analysis we were able to identify two potential inhibitory candidates, namely decorin (Dcn) and Sparc-like 1 (Sparcl1). We could show that ablation of either candidate enhanced adipogenesis in visceral preadipocytes, while treatment of primary cultures with recombinant Sparcl1 and Dcn blocked adipogenesis in a dose dependent manner. In conclusion, our data suggests that the differences in adipogenesis between depots might be due to paracrine and autocrine feedback mechanisms which could in turn contribute to metabolic homeostasis.


Subject(s)
Adipogenesis/genetics , Calcium-Binding Proteins/biosynthesis , Decorin/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Obesity/genetics , Adipose Tissue/growth & development , Adipose Tissue/metabolism , Animals , Calcium-Binding Proteins/genetics , Cell Differentiation/genetics , Decorin/genetics , Extracellular Matrix Proteins/genetics , Humans , Intra-Abdominal Fat/growth & development , Intra-Abdominal Fat/metabolism , Mice , Obesity/metabolism , Paracrine Communication/genetics , Subcutaneous Fat/growth & development , Subcutaneous Fat/metabolism
16.
Nat Cell Biol ; 15(6): 659-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624403

ABSTRACT

Brown adipose tissue helps to maintain body temperature in hibernators, rodents and neonatal mammals by converting lipids and glucose into heat, thereby increasing energy expenditure. In addition to classical brown adipocytes, adult rodents-like adult humans-harbour brown-like adipocytes in the predominantly white adipose tissue. The formation of these brite (brown-in-white) adipocytes is a physiological response to chronic cold and their cellular origin is under debate. We show here that cold-induced formation of brite adipocytes in mice is reversed within 5 weeks of warm adaptation, but the brite adipocytes formed by cold stimulation are not eliminated. Genetic tracing and transcriptional characterization of isolated adipocytes demonstrates that they are converted into cells with the morphology and gene expression pattern of white adipocytes. Moreover, these white-typical adipocytes can convert into brite adipocytes on additional cold stimulation. Shifting the balance of this interconversion from the white towards the brite phenotype might provide a new means of counteracting obesity by increasing energy expenditure.


Subject(s)
Adipocytes, Brown/physiology , Adipocytes, White/cytology , Adipocytes, White/physiology , Energy Metabolism , Adipocytes, Brown/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Apoptosis , Cell Differentiation , Cold Temperature , Green Fluorescent Proteins , Hot Temperature , Ion Channels/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Obesity , Phenotype , T-Box Domain Proteins/metabolism , Transcription, Genetic , Uncoupling Protein 1
17.
Curr Protoc Mouse Biol ; 3(4): 205-16, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-26069094

ABSTRACT

The Western world is in the midst of an epidemic of obesity, which is the cause of severe clinical complications such as type 2 diabetes, hypertension, and cardiovascular disease. Obesity develops when energy intake chronically exceeds energy expenditure; thus, either reducing the energy intake and/or increasing the energy expenditure has been used in the treatment and prevention of obesity. On a cellular level, energy storage is mediated by white adipocyte tissue (WAT). In contrast, brown adipose tissue (BAT) contributes to body temperature and metabolic homeostasis by metabolizing lipids and glucose. Adipose tissue is a notoriously difficult tissue to work with, due to the high content of triglycerides and the fragility of the cells. In this unit, several approaches to analysis of BAT and WAT are described that overcome these limitations. Curr. Protoc. Mouse Biol. 3:205-216 © 2013 by John Wiley & Sons, Inc.

SELECTION OF CITATIONS
SEARCH DETAIL
...