Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Publication year range
1.
Brain Behav Immun ; 117: 330-346, 2024 03.
Article in English | MEDLINE | ID: mdl-38309640

ABSTRACT

Nutrient composition in obesogenic diets may influence the severity of disorders associated with obesity such as insulin-resistance and chronic inflammation. Here we hypothesized that obesogenic diets rich in fat and varying in fatty acid composition, particularly in omega 6 (ω6) to omega 3 (ω3) ratio, have various effects on energy metabolism, neuroinflammation and behavior. Mice were fed either a control diet or a high fat diet (HFD) containing either low (LO), medium (ME) or high (HI) ω6/ω3 ratio. Mice from the HFD-LO group consumed less calories and exhibited less body weight gain compared to other HFD groups. Both HFD-ME and HFD-HI impaired glucose metabolism while HFD-LO partly prevented insulin intolerance and was associated with normal leptin levels despite higher subcutaneous and perigonadal adiposity. Only HFD-HI increased anxiety and impaired spatial memory, together with increased inflammation in the hypothalamus and hippocampus. Our results show that impaired glucose metabolism and neuroinflammation are uncoupled, and support that diets with a high ω6/ω3 ratio are associated with neuroinflammation and the behavioral deterioration coupled with the consumption of diets rich in fat.


Subject(s)
Insulins , Neuroinflammatory Diseases , Animals , Mice , Obesity/metabolism , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Inflammation , Glucose
2.
Int J Mol Sci ; 25(3)2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38338999

ABSTRACT

Plant-based food interventions are promising therapeutic approaches for non-alcoholic fatty liver disease (NAFLD) treatment, and microRNAs (miRNAs) have emerged as functional bioactive components of dietary plants involved in cross-kingdom communication. Deeper investigations are needed to determine the potential impact of plant miRNAs in NAFLD. This study aimed to identify plant miRNAs that could eventually modulate the expression of human metabolic genes and protect against the progression of hepatic steatosis. Plant miRNAs from the miRBase were used to predict human target genes, and miR8126-3p and miR8126-5p were selected as candidates for their potential role in inhibiting glucose and lipid metabolism-related genes. Human HepG2 cells were transfected with plant miRNA mimics and then exposed to a mixture of oleic and palmitic acids to mimic steatosis. miR8126-3p and miR8126-5p transfections inhibited the expression of the putative target genes QKI and MAPKAPK2, respectively, and had an impact on the expression profile of key metabolic genes, including PPARA and SREBF1. Quantification of intrahepatic triglycerides revealed that miR8126-3p and miR8126-5p attenuated lipid accumulation. These findings suggest that plant miR8126-3p and miR8126-5p would induce metabolic changes in human hepatocytes eventually protecting against lipid accumulation, and thus, they could be potential therapeutic tools for preventing and alleviating lipid accumulation.


Subject(s)
MicroRNAs , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Hepatocytes/metabolism , MicroRNAs/metabolism , Lipid Metabolism/genetics , Lipids , Liver/metabolism
3.
Genes Dev ; 29(1): 7-22, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25504365

ABSTRACT

Long-term exposure to peroxisome proliferator-activated receptor γ (PPARγ) agonists such as rosiglitazone induces browning of rodent and human adipocytes; however, the transcriptional mechanisms governing this phenotypic switch in adipocytes are largely unknown. Here we show that rosiglitazone-induced browning of human adipocytes activates a comprehensive gene program that leads to increased mitochondrial oxidative capacity. Once induced, this gene program and oxidative capacity are maintained independently of rosiglitazone, suggesting that additional browning factors are activated. Browning triggers reprogramming of PPARγ binding, leading to the formation of PPARγ "superenhancers" that are selective for brown-in-white (brite) adipocytes. These are highly associated with key brite-selective genes. Based on such an association, we identified an evolutionarily conserved metabolic regulator, Kruppel-like factor 11 (KLF11), as a novel browning transcription factor in human adipocytes that is required for rosiglitazone-induced browning, including the increase in mitochondrial oxidative capacity. KLF11 is directly induced by PPARγ and appears to cooperate with PPARγ in a feed-forward manner to activate and maintain the brite-selective gene program.


Subject(s)
Adipocytes/metabolism , Cell Cycle Proteins/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Repressor Proteins/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes, Brown/cytology , Apoptosis Regulatory Proteins , Cell Cycle Proteins/genetics , Cellular Reprogramming , Chromatin/metabolism , Gene Expression Regulation , Humans , Hypoglycemic Agents/pharmacology , Mitochondria/drug effects , Oxidation-Reduction , Protein Binding , Repressor Proteins/genetics , Rosiglitazone , Thiazolidinediones/pharmacology , Transcriptional Activation/drug effects
4.
Int J Mol Sci ; 24(11)2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37298701

ABSTRACT

Oxytocin (OT), a neuropeptide best known for its role in emotional and social behaviors, has been linked to osteoarthritis (OA). This study aimed to investigate the serum OT level in hip and/or knee OA patients and to study its association with disease progression. Patients from the KHOALA cohort with symptomatic hip and/or knee OA (Kellgren and Lawrence (KL) scores of 2 and 3) and follow-up at 5 years were included in this analysis. The primary endpoint was structural radiological progression, which was defined as an increase of at least one KL point at 5 years. Logistic regression models were used to estimate the associations between OT levels and KL progression while controlling for gender, age, BMI, diabetes and leptin levels. Data from 174 hip OA patients and 332 knee OA patients were analyzed independently. No differences in OT levels were found between the 'progressors' and 'non-progressors' groups among the hip OA patients and knee OA patients, respectively. No statistically significant associations were found between the OT levels at baseline and KL progression at 5 years, the KL score at baseline or the clinical outcomes. Higher structural damage at baseline and severe structural progression of hip and knee osteoarthritis did not appear to be associated with a low serum OT level at baseline.


Subject(s)
Osteoarthritis, Hip , Osteoarthritis, Knee , Humans , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Hip/diagnostic imaging , Oxytocin , Prospective Studies , Radiography , Disease Progression
5.
Am J Physiol Cell Physiol ; 320(5): C822-C841, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33439778

ABSTRACT

Adipocytes are specialized cells with pleiotropic roles in physiology and pathology. Several types of fat cells with distinct metabolic properties coexist in various anatomically defined fat depots in mammals. White, beige, and brown adipocytes differ in their handling of lipids and thermogenic capacity, promoting differences in size and morphology. Moreover, adipocytes release lipids and proteins with paracrine and endocrine functions. The intrinsic properties of adipocytes pose specific challenges in culture. Mature adipocytes float in suspension culture due to high triacylglycerol content and are fragile. Moreover, a fully differentiated state, notably acquirement of the unilocular lipid droplet of white adipocyte, has so far not been reached in two-dimensional culture. Cultures of mouse and human-differentiated preadipocyte cell lines and primary cells have been established to mimic white, beige, and brown adipocytes. Here, we survey various models of differentiated preadipocyte cells and primary mature adipocyte survival describing main characteristics, culture conditions, advantages, and limitations. An important development is the advent of three-dimensional culture, notably of adipose spheroids that recapitulate in vivo adipocyte function and morphology in fat depots. Challenges for the future include isolation and culture of adipose-derived stem cells from different anatomic location in animal models and humans differing in sex, age, fat mass, and pathophysiological conditions. Further understanding of fat cell physiology and dysfunction will be achieved through genetic manipulation, notably CRISPR-mediated gene editing. Capturing adipocyte heterogeneity at the single-cell level within a single fat depot will be key to understanding diversities in cardiometabolic parameters among lean and obese individuals.


Subject(s)
Adipocytes/physiology , Adipose Tissue/physiology , Adipogenesis , Adipose Tissue/cytology , Animals , Cell Communication , Cell Culture Techniques , Cell Line , Cell Survival , Humans , Phenotype , Species Specificity , Spheroids, Cellular , Tissue Culture Techniques
6.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769215

ABSTRACT

Oxytocin (OT) is involved in breastfeeding and childbirth and appears to play a role in regulating the bone matrix. OT is synthesized in the supraoptic and paraventricular nuclei of the hypothalamus and is released in response to numerous stimuli. It also appears to be produced by osteoblasts in the bone marrow, acting as a paracrine-autocrine regulator of bone formation. Osteoarthritis (OA) is a disease of the whole joint. Different tissues involved in OA express OT receptors (OTRs), such as chondrocytes and osteoblasts. This hormone, which levels are reduced in patients with OA, appears to have a stimulatory effect on chondrogenesis. OT involvement in bone biology could occur at both the osteoblast and chondrocyte levels. The relationships between metabolic syndrome, body weight, and OA are well documented, and the possible effects of OT on different parameters of metabolic syndrome, such as diabetes and body weight, are important. In addition, the effects of OT on adipokines and inflammation are also discussed, especially since recent data have shown that low-grade inflammation is also associated with OA. Furthermore, OT also appears to mediate endogenous analgesia in animal and human studies. These observations provide support for the possible interest of OT in OA and its potential therapeutic treatment.


Subject(s)
Osteoarthritis/metabolism , Oxytocin/metabolism , Adipokines/metabolism , Animals , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrogenesis , Humans , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Osteoblasts/metabolism , Osteoblasts/pathology , Receptors, Oxytocin/metabolism
7.
Int J Mol Sci ; 21(11)2020 May 31.
Article in English | MEDLINE | ID: mdl-32486506

ABSTRACT

This study investigated the relationship of oxytocin (OT) to chondrogenesis and osteoarthritis (OA). Human bone marrow and multipotent adipose-derived stem cells were cultured in vitro in the absence or presence of OT and assayed for mRNA transcript expression along with histological and immunohistochemical analyses. To study the effects of OT in OA in vivo, a rat model and a human cohort of 63 men and 19 women with hand OA and healthy controls, respectively, were used. The baseline circulating OT, interleukin-6, leptin, and oestradiol levels were measured, and hand X-ray examinations were performed for each subject. OT induced increased aggrecan, collagen (Col) X, and cartilage oligomeric matrix protein mRNA transcript levels in vitro, and the immunolabelling experiments revealed a normalization of Sox9 and Col II protein expression levels. No histological differences in lesion severity were observed between rat OA groups. In the clinical study, a multivariate analysis adjusted for age, body mass index, and leptin levels revealed a significant association between OA and lower levels of OT (odds ratio = 0.77; p = 0.012). Serum OT levels are reduced in patients with hand OA, and OT showed a stimulatory effect on chondrogenesis. Thus, OT may contribute to the pathophysiology of OA.


Subject(s)
Chondrogenesis/drug effects , Osteoarthritis/drug therapy , Oxytocin/pharmacology , Aged , Animals , Body Mass Index , Bone Marrow/metabolism , Cell Culture Techniques , Chondrocytes/metabolism , Collagen Type II/blood , Estradiol/blood , Extracellular Matrix/metabolism , Female , Humans , Immunohistochemistry , Interleukin-1beta/metabolism , Interleukin-6/blood , Leptin/blood , Male , Middle Aged , Multivariate Analysis , Osteoarthritis/metabolism , Oxytocin/blood , RNA, Messenger/metabolism , Rats , SOX9 Transcription Factor/blood , SOX9 Transcription Factor/metabolism , Stem Cells/cytology
8.
Handb Exp Pharmacol ; 251: 183-214, 2019.
Article in English | MEDLINE | ID: mdl-30141101

ABSTRACT

Fatty acids are essential contributors to adipocyte-based non-shivering thermogenesis by acting as activators of uncoupling protein 1 and serving as fuel for mitochondrial heat production. Novel evidence suggests a contribution to this thermogenic mechanism by their conversion to bioactive compounds. Mammalian cells produce a plethora of oxylipins and endocannabinoids, some of which have been identified to affect the abundance or thermogenic activity of brown and brite adipocytes. These effectors are produced locally or at distant sites and signal toward thermogenic adipocytes via a direct interaction with these cells or indirectly via secondary mechanisms. These interactions are evoked by the activation of receptor-mediated pathways. The endogenous production of these compounds is prone to modulation by the dietary intake of the respective precursor fatty acids. The effect of nutritional interventions on uncoupling protein 1-derived thermogenesis may thus at least in part be conferred by the production of a supportive oxylipin and endocannabinoid profile. The manipulation of this system in future studies will help to elucidate the physiological potential of these compounds as novel, endogenous regulators of non-shivering thermogenesis.


Subject(s)
Adipocytes , Fatty Acids/metabolism , Mitochondria/metabolism , Thermogenesis , Uncoupling Protein 1/metabolism , Animals , Mitochondria/physiology , Uncoupling Protein 1/genetics
9.
J Lipid Res ; 59(3): 452-461, 2018 03.
Article in English | MEDLINE | ID: mdl-29343538

ABSTRACT

The recent characterization of functional brown adipose tissue in adult humans has opened new perspectives for regulation of energy expenditure with respect to obesity and diabetes. Furthermore, dietary recommendations have taken into account the insufficient dietary intake of ω3 PUFAs and the concomitant excessive intake of ω6 PUFA associated with the occurrence of overweight/obesity. We aimed to study whether ω3 PUFAs could play a role in the recruitment and function of energy-dissipating brown/brite adipocytes. We show that ω3 PUFA supplementation has a beneficial effect on the thermogenic function of adipocytes. In vivo, a low dietary ω6:ω3 ratio improved the thermogenic response of brown and white adipose tissues to ß3-adrenergic stimulation. This effect was recapitulated in vitro by PUFA treatment of hMADS adipocytes. We pinpointed the ω6-derived eicosanoid prostaglandin (PG)F2α as the molecular origin because the effects were mimicked with a specific PGF2α receptor agonist. PGF2α level in hMADS adipocytes was reduced in response to ω3 PUFA supplementation. The recruitment of thermogenic adipocytes is influenced by the local quantity of individual oxylipins, which is controlled by the ω6:ω3 ratio of available lipids. In human nutrition, energy homeostasis may thus benefit from the implementation of a more balanced dietary ω6:ω3 ratio.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/pharmacology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cells, Cultured , Humans , Oxylipins/metabolism , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/metabolism
10.
Circ J ; 82(12): 2954-2961, 2018 11 24.
Article in English | MEDLINE | ID: mdl-30282882

ABSTRACT

BACKGROUND: Severe abdominal aortic calcification (AAC) points to high cardiovascular risk and leptin stimulates arterial calcification; however, clinical data on their association are scarce. We studied the link between serum leptin and AAC severity and progression, and the effect of smoking and lipid levels, on this association in men. Methods and Results: At baseline, 548 community-dwelling men aged 50-85 years underwent blood collection and lateral lumbar spine radiography. In 448 men, X-ray was repeated after 3 and 7.5 years. AAC was assessed using Kauppila's semiquantitative score. In multivariable models, high leptin was associated with higher odds of severe AAC (odds ratio [OR]=1.71 per SD, 95% confidence interval [CI]: 1.22-2.40). The odds of severe AAC were the highest in men who had elevated leptin levels and either were ever-smokers (OR=9.22, 95% CI: 3.43-24.78) or had hypertriglyceridemia (vs. men without these characteristics). Higher leptin was associated with greater AAC progression (OR=1.34 per SD, 95% CI: 1.04-1.74). The risk of AAC progression was the highest in men who had elevated leptin levels and either were current smokers or had high low-density lipoprotein-cholesterol levels (OR=5.91, 95% CI: 2.46-14.16 vs. men without these characteristics). These links remained significant after adjustment for baseline AAC and in subgroups defined according to smoking and low-density lipoprotein-cholesterol levels. CONCLUSIONS: In older men, high leptin levels are associated with greater severity and rapid progression of AAC independent of smoking, low-density lipoprotein-cholesterol or triglycerides.


Subject(s)
Aorta, Abdominal , Aortic Diseases/blood , Leptin/blood , Vascular Calcification/blood , Age Factors , Aged , Aged, 80 and over , Aortic Diseases/diagnostic imaging , Humans , Male , Middle Aged , Prospective Studies , Severity of Illness Index , Vascular Calcification/diagnostic imaging
11.
Biochim Biophys Acta ; 1861(4): 285-93, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26775637

ABSTRACT

Brite adipocytes recently discovered in humans are of considerable importance in energy expenditure by converting energy excess into heat. This property could be useful in the treatment of obesity, and nutritional aspects are relevant to this important issue. Using hMADS cells as a human cell model which undergoes a white to a brite adipocyte conversion, we had shown previously that arachidonic acid, the major metabolite of the essential nutrient Ω6-linoleic acid, plays a major role in this process. Its metabolites PGE2 and PGF2 alpha inhibit this process via a calcium-dependent pathway, whereas in contrast carbaprostacyclin (cPGI2), a stable analog of prostacyclin, activates white to brite adipocyte conversion. Herein, we show that cPGI2 generates via its cognate cell-surface receptor IP-R, a cyclic AMP-signaling pathway involving PKA activity which in turn induces the expression of UCP1. In addition, cPGI2 activates the pathway of nuclear receptors of the PPAR family, i.e. PPARα and PPARγ, which act separately from IP-R to up-regulate the expression of key genes involved in the function of brite adipocytes. Thus dual pathways are playing in concert for the occurrence of a browning process of human white adipocytes. These results make prostacyclin analogs as a new class of interesting molecules to treat obesity and associated diseases.


Subject(s)
Adipocytes, Brown/drug effects , Adipocytes, White/drug effects , Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Epoprostenol/analogs & derivatives , PPAR alpha/agonists , PPAR gamma/agonists , Receptors, Prostaglandin/agonists , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Enzyme Activation , Epoprostenol/pharmacology , Humans , Infant , Ion Channels/genetics , Ion Channels/metabolism , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/metabolism , Phenotype , RNA Interference , Receptors, Epoprostenol , Receptors, Prostaglandin/metabolism , Signal Transduction/drug effects , Thermogenesis/drug effects , Time Factors , Transfection , Uncoupling Protein 1
12.
Hum Mol Genet ; 24(2): 471-9, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25205110

ABSTRACT

Andersen's syndrome (AS) is a rare and dominantly inherited pathology, linked to the inwardly rectifying potassium channel Kir2.1. AS patients exhibit a triad of symptoms that include periodic paralysis, cardiac dysrhythmia and bone malformations. Some progress has been made in understanding the contribution of the Kir2.1 channel to skeletal and cardiac muscle dysfunctions, but its role in bone morphogenesis remains unclear. We isolated myoblast precursors from muscle biopsies of healthy individuals and typical AS patients with dysmorphic features. Myoblast cultures underwent osteogenic differentiation that led to extracellular matrix mineralization. Osteoblastogenesis was monitored through the activity of alkaline phosphatase, and through the hydroxyapatite formation using Alizarin Red and Von Kossa staining techniques. Patch-clamp recordings revealed the presence of an inwardly rectifying current in healthy cells that was absent in AS osteoblasts, showing the dominant-negative effect of the Kir2.1 mutant allele in osteoblasts. We also found that while control cells actively synthesize hydroxyapatite, AS osteoblasts are unable to efficiently form any extracellular matrix. To further demonstrate the role of the Kir2.1 channels during the osteogenesis, we inhibited Kir2.1 channel activity in healthy patient cells by applying extracellular Ba(2+) or using adenoviruses carrying mutant Kir2.1 channels. In both cases, cells were no longer able to produce extracellular matrixes. Moreover, osteogenic activity of AS osteoblasts was restored by rescue experiments, via wild-type Kir2.1 channel overexpression. These observations provide a proof that normal Kir2.1 channel function is essential during osteoblastogenesis.


Subject(s)
Glycogen Storage Disease Type IV/metabolism , Myoblasts/metabolism , Osteogenesis , Potassium Channels, Inwardly Rectifying/metabolism , Cells, Cultured , Extracellular Matrix/metabolism , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/physiopathology , Humans , Myoblasts/cytology , Osteoblasts/cytology , Osteoblasts/metabolism , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics
13.
FASEB J ; 30(2): 909-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26527067

ABSTRACT

Brown adipose tissue (BAT) is essential for adaptive thermogenesis and dissipation of caloric excess through the activity of uncoupling protein (UCP)-1. BAT in humans is of great interest for the treatment of obesity and related diseases. In this study, the expression of Twik-related acid-sensitive K(+) channel (TASK)-1 [a pH-sensitive potassium channel encoded by the potassium channel, 2-pore domain, subfamily K, member 3 (Kcnk3) gene] correlated highly with Ucp1 expression in obese and cold-exposed mice. In addition, Task1-null mice, compared with their controls, became overweight, mainly because of an increase in white adipose tissue mass and BAT whitening. Task1(-/-)-mouse-derived brown adipocytes, compared with wild-type mouse-derived brown adipocytes, displayed an impaired ß3-adrenergic receptor response that was characterized by a decrease in oxygen consumption, Ucp1 expression, and lipolysis. This phenotype was thought to be caused by an exacerbation of mineralocorticoid receptor (MR) signaling, given that it was mimicked by corticoids and reversed by an MR inhibitor. We concluded that the K(+) channel TASK1 controls the thermogenic activity in brown adipocytes through modulation of ß-adrenergic receptor signaling.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Receptors, Adrenergic, beta-3/metabolism , Receptors, Mineralocorticoid/metabolism , Signal Transduction/physiology , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Animals , Female , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Oxygen Consumption/physiology , Potassium Channels, Tandem Pore Domain/genetics , Receptors, Mineralocorticoid/genetics , Thermogenesis/physiology
14.
Stem Cells ; 32(6): 1578-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24375761

ABSTRACT

Adipose tissue contains thermogenic adipocytes (i.e., brown and brite/beige) that oxidize nutrients at exceptionally high rates via nonshivering thermogenesis. Its recent discovery in adult humans has opened up new avenues to fight obesity and related disorders such as diabetes. Here, we identified miR-26a and -26b as key regulators of human white and brite adipocyte differentiation. Both microRNAs are upregulated in early adipogenesis, and their inhibition prevented lipid accumulation while their overexpression accelerated it. Intriguingly, miR-26a significantly induced pathways related to energy dissipation, shifted mitochondrial morphology toward that seen in brown adipocytes, and promoted uncoupled respiration by markedly increasing the hallmark protein of brown fat, uncoupling protein 1. By combining in silico target prediction, transcriptomics, and an RNA interference screen, we identified the sheddase ADAM metallopeptidase domain 17 (ADAM17) as a direct target of miR-26 that mediated the observed effects on white and brite adipogenesis. These results point to a novel, critical role for the miR-26 family and its downstream effector ADAM17 in human adipocyte differentiation by promoting characteristics of energy-dissipating thermogenic adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Adipogenesis/genetics , MicroRNAs/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Adipocytes, Brown/cytology , Adipocytes, Brown/ultrastructure , Adipose Tissue, White/metabolism , Adipose Tissue, White/ultrastructure , Adult , Cell Differentiation/genetics , Child, Preschool , Cold Temperature , Computer Simulation , Humans , Infant , Ion Channels , Male , MicroRNAs/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins , Signal Transduction/genetics , Transcriptome/genetics , Uncoupling Protein 1 , Up-Regulation/genetics
15.
Cell Mol Life Sci ; 71(9): 1741-59, 2014 May.
Article in English | MEDLINE | ID: mdl-24026398

ABSTRACT

Chromosomally separated, co-expressed genes can be in spatial proximity, but there is still debate about how this nuclear organization is achieved. Proposed mechanisms include global genome organization, preferential positioning of chromosome territories, or gene-gene sharing of various nuclear bodies. To investigate this question, we selected a set of genes that were co-expressed upon differentiation of human multipotent stem cells. We applied a novel multi-dimensional analysis procedure which revealed that prior to gene expression, the relative position of these genes was conserved in nuclei. Upon stem cell differentiation and concomitant gene expression, we found that co-expressed genes were closer together. In addition, we found that genes in the same 1-µm-diameter neighborhood associated with either the same splicing speckle or to a lesser extent with the same transcription factory. Dispersal of speckles by overexpression of the serine-arginine (SR) protein kinase cdc2-like kinase Clk2 led to a significant drop in the number of genes in shared neighborhoods. We demonstrate quantitatively that the frequencies of speckle and factory sharing can be explained by assuming stochastic selection of a nuclear body within a restricted sub-volume defined by the original global gene positioning present prior to gene expression. We conclude that the spatial organization of these genes is a two-step process in which transcription-induced association with nuclear bodies enhances and refines a pre-existing global organization.


Subject(s)
Nuclear Proteins/metabolism , RNA Polymerase II/metabolism , Ribonucleoproteins/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Chromosomes/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , HEK293 Cells , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA Polymerase II/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Ribonucleoproteins/genetics , Serine-Arginine Splicing Factors , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
16.
Biochim Biophys Acta ; 1831(5): 905-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23146742

ABSTRACT

Brown adipose tissue (BAT) has long been thought to be absent or very scarce in human adults so that its contribution to energy expenditure was not considered as relevant. The recent discovery of thermogenic BAT in human adults opened the field for innovative strategies to combat overweight/obesity and associated diseases. This energy-dissipating function of BAT is responsible for adaptive thermogenesis in response to cold stimulation. In this context, adipocytes can be converted, within white adipose tissue (WAT), into multilocular adipocytes expressing UCP1, a mitochondrial protein that plays a key role in heat production by uncoupling the activity of the respiratory chain from ATP synthesis. These adipocytes have been named "brite" or "beige" adipocytes. Whereas BAT has been studied for a long time in murine models both in vivo and in vitro, there is now a strong demand for human cellular models to validate and/or identify critical factors involved in the induction of a thermogenic program within adipocytes. In this review we will discuss the different human cellular models described in the literature and what is known regarding the regulation of their differentiation and/or activation process. In addition, the role of microRNAs as novel regulators of brown/"brite" adipocyte differentiation and conversion will be depicted. Finally, investigation of both the conversion and the metabolism of white-to-brown converted adipocytes is required for the development of therapeutic strategies targeting overweight/obesity and associated diseases. This article is part of a Special Issue entitled Brown and White Fat: From Signaling to Disease.


Subject(s)
Adipogenesis , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Cell Differentiation , Disease Models, Animal , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Humans , Signal Transduction
17.
Biochem Biophys Res Commun ; 440(4): 786-91, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24134848

ABSTRACT

Chondrogenesis has been widely investigated in vitro using bone marrow-derived mesenchymal stromal cells (BM-MSCs) or primary chondrocytes. However, their use raises some issues partially circumvented by the availability of Adipose tissue-derived MSCs. Herein; we characterized the chondrogenic potential of human Multipotent Adipose-Derived Stem (hMADS) cells, and their potential use as pharmacological tool. hMADS cells are able to synthesize matrix proteins including COMP, Aggrecan and type II Collagen. Furthermore, hMADS cells express BMP receptors in a similar manner to BM-MSC, and BMP6 treatment of differentiated cells prevents expression of the hypertrophic marker type X Collagen. We tested whether IL-1ß and nicotine could impact chondrocyte differentiation. As expected, IL-1ß induced ADAMTS-4 gene expression and modulated negatively chondrogenesis while these effects were reverted in the presence of the IL-1 receptor antagonist. Nicotine, at concentrations similar to those observed in blood of smokers, exhibited a dose dependent increase of Aggrecan expression, suggesting an unexpected protective effect of the drug under these conditions. Therefore, hMADS cells represent a valuable tool for the analysis of in vitro chondrocyte differentiation and to screen for potentially interesting pharmacological drugs.


Subject(s)
Adipose Tissue/cytology , Chondrocytes/cytology , Chondrogenesis/physiology , Multipotent Stem Cells/cytology , ADAM Proteins/genetics , ADAMTS4 Protein , Aggrecans/biosynthesis , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein Receptors/metabolism , Cell Separation , Chondrogenesis/genetics , Collagen Type X/metabolism , Gene Expression/drug effects , Humans , Interleukin-1beta/pharmacology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Nicotine/pharmacology , Procollagen N-Endopeptidase/genetics
18.
Med Sci (Paris) ; 39(12): 937-944, 2023 Dec.
Article in French | MEDLINE | ID: mdl-38108724

ABSTRACT

Animal models remain important for the study of human pathologies. The most widely used model (mouse) is an endothermic mammal like humans, maintained at ambient temperatures (22 °C). Its energy metabolism is overactivated, a situation rarely observed in humans thanks to various adaptations (clothing, heating…). The thermoneutral zone is defined as a range of ambient temperatures that allows an organism to regulate body temperature without using additional thermoregulatory processes. There are many examples of divergent results between studies conducted at 22 °C or at 30 °C (thermoneutrality for mice). Therefore, it seems essential to take into account the housing temperature both for animal welfare and for the relevance of the results.


Title: Thermoneutralité chez la souris et expérimentation animale. Abstract: Les modèles animaux demeurent une nécessité pour l'étude des maladies humaines. Le modèle le plus utilisé, la souris, est, comme les êtres humains, un mammifère endotherme maintenu à des températures ambiantes (22 °C). Son métabolisme énergétique est donc suractivé, une situation rarement observée chez les êtres humains grâce à diverses adaptations (vêtements, chauffage, etc.). La zone de thermoneutralité est définie comme une plage de températures ambiantes qui permet à un organisme de réguler sa température corporelle sans recourir à des processus de thermorégulation supplémentaires. Il existe de nombreux exemples de résultats divergents entre des études menées à 22 °C et celles réalisées à 30 °C (thermoneutralité chez la souris). Il semble donc essentiel de prendre en compte la température d'hébergement tant pour le bien-être animal que pour la pertinence des résultats des expériences réalisées.


Subject(s)
Body Temperature Regulation , Energy Metabolism , Humans , Animals , Mice , Models, Animal , Temperature , Mammals
19.
Front Nutr ; 10: 1287312, 2023.
Article in English | MEDLINE | ID: mdl-38099184

ABSTRACT

Background: Edible plants can exert anti-inflammatory activities in humans, being potentially useful in the treatment of inflammatory diseases. Plant-derived microRNAs have emerged as cross-kingdom gene expression regulators and could act as bioactive molecules involved in the beneficial effects of some edible plants. We investigated the role of edible plant-derived microRNAs in the modulation of pro-inflammatory human genes. Methods: MicroRNAs from plant-derived foods were identified by next-generation sequencing. MicroRNAs with inflammatory putative targets were selected, after performing in silico analyses. The expression of candidate plant-derived miRNAs was analyzed by qPCR in edible plant-derived foods and their effects were evaluated in THP-1 monocytes differentiated to macrophages. The bioavailability of candidate plant miRNAs in humans was evaluated in feces and serum samples by qPCR. Results: miR482f and miR482c-5p are present in several edible plant-derived foods, such as fruits, vegetables, and cooked legumes and cereals, and fats and oils. Transfections with miR482f and miR482c-5p mimics decreased the gene expression of CLEC7A and NFAM1, and TRL6, respectively, in human THP-1 monocytes differentiated to macrophages, which had an impact on gene expression profile of inflammatory biomarkers. Both microRNAs (miR482f and miR482c-5p) resisted degradation during digestion and were detected in human feces, although not in serum. Conclusion: Our findings suggest that miR482f and miR482c-5p can promote an anti-inflammatory gene expression profile in human macrophages in vitro and their bioavailability in humans can be achieved through diet, but eventually restricted at the gut level.

20.
Cells ; 12(3)2023 01 30.
Article in English | MEDLINE | ID: mdl-36766790

ABSTRACT

Obesity is a complex disease highly related to diet and lifestyle and is associated with low amount of thermogenic adipocytes. Therapeutics that regulate brown adipocyte recruitment and activity represent interesting strategies to fight overweight and associated comorbidities. Recent studies suggest a role for several fatty acids and their metabolites, called lipokines, in the control of thermogenesis. The purpose of this work was to analyze the role of several lipokines in the control of brown/brite adipocyte formation. We used a validated human adipocyte model, human multipotent adipose-derived stem cell model (hMADS). In the absence of rosiglitazone, hMADS cells differentiate into white adipocytes, but convert into brite adipocytes upon rosiglitazone or prostacyclin 2 (PGI2) treatment. Gene expression was quantified using RT-qPCR and protein levels were assessed by Western blotting. We show here that lipokines such as 12,13-diHOME, 12-HEPE, 15dPGJ2 and 15dPGJ3 were not able to induce browning of white hMADS adipocytes. However, both fatty acid esters of hydroxy fatty acids (FAHFAs), 9-PAHPA and 9-PAHSA potentiated brown key marker UCP1 mRNA levels. Interestingly, CTA2, the stable analog of thromboxane A2 (TXA2), but not its inactive metabolite TXB2, inhibited the rosiglitazone and PGI2-induced browning of hMADS adipocytes. These results pinpoint TXA2 as a lipokine inhibiting brown adipocyte formation that is antagonized by PGI2. Our data open new horizons in the development of potential therapies based on the control of thromboxane A2/prostacyclin balance to combat obesity and associated metabolic disorders.


Subject(s)
Fatty Acids , Thromboxane A2 , Humans , Thromboxane A2/metabolism , Rosiglitazone/pharmacology , Fatty Acids/metabolism , Adipocytes, Brown/metabolism , Obesity/metabolism , Prostaglandins I/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL