ABSTRACT
BACKGROUND: Obesity poses a significant global health challenge, given its association with the excessive accumulation of adipose tissue (AT) and various systemic disruptions. Within the adipose microenvironment, expansion and enrichment with immune cells trigger the release of inflammatory mediators and growth factors, which can disrupt tissues, including bones. While obesity's contribution to bone loss is well established, the direct impact of obese AT on osteoblast maturation remains uncertain. This study aimed to explore the influence of the secretomes from obese and lean AT on osteoblast differentiation and activity. METHODS: SAOS-2 cells were exposed to the secretomes obtained by culturing human subcutaneous AT from individuals with obesity (OATS) or lean patients, and their effects on osteoblasts were evaluated. RESULTS: In the presence of the OATS, mature osteoblasts underwent dedifferentiation, showing an increased proliferation accompanied by a morphological shift towards a mesenchymal phenotype, with detrimental effects on osteogenic markers and the calcification capacity. Concurrently, the OATS promoted the expression of mesenchymal and adipogenic markers, inducing the formation of cytoplasmic lipid droplets in SAOS-2 cells exposed to an adipogenic differentiation medium. Additionally, TGF-ß1 emerged as a key mediator of these effects, as the OATS was enriched with this growth factor. CONCLUSIONS: Our findings demonstrate that obese subcutaneous AT promotes the dedifferentiation of osteoblasts and increases the adipogenic profile in these cells.
Subject(s)
Adipogenesis , Adipose Tissue , Cell Dedifferentiation , Obesity , Osteoblasts , Phenotype , Signal Transduction , Transforming Growth Factor beta1 , Humans , Osteoblasts/metabolism , Osteoblasts/pathology , Obesity/pathology , Obesity/metabolism , Transforming Growth Factor beta1/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Secretome/metabolism , Cell Differentiation , Cell Proliferation , Osteogenesis , MaleABSTRACT
Skeletal muscle fibrosis is defined as the excessive accumulation of extracellular matrix (ECM) components and is a hallmark of muscular dystrophies. Fibro-adipogenic progenitors (FAPs) are the main source of ECM, and thus have been strongly implicated in fibrogenesis. In skeletal muscle fibrotic models, including muscular dystrophies, FAPs undergo dysregulations in terms of proliferation, differentiation, and apoptosis, however few studies have explored the impact of FAPs migration. Here, we studied fibroblast and FAPs migration and identified lysophosphatidic acid (LPA), a signaling lipid central to skeletal muscle fibrogenesis, as a significant migration inductor. We identified LPA receptor 1 (LPA1) mediated signaling as crucial for this effect through a mechanism dependent on the Hippo pathway, another pathway implicated in fibrosis across diverse tissues. This cross-talk favors the activation of the Yes-associated protein 1 (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ), leading to increased expression of fibrosis-associated genes. This study reveals the role of YAP in LPA-mediated fibrotic responses as inhibition of YAP transcriptional coactivator activity hinders LPA-induced migration in fibroblasts and FAPs. Moreover, we found that FAPs derived from the mdx4cv mice, a murine model of Duchenne muscular dystrophy, display a heightened migratory phenotype due to enhanced LPA signaling compared to wild-type FAPs. Remarkably, we found that the inhibition of LPA1 or YAP transcriptional coactivator activity in mdx4cv FAPs reverts this phenotype. In summary, the identified LPA-LPA1-YAP pathway emerges as a critical driver of skeletal muscle FAPs migration and provides insights into potential novel targets to mitigate fibrosis in muscular dystrophies.
Subject(s)
Adaptor Proteins, Signal Transducing , Cell Movement , Fibroblasts , Fibrosis , Lysophospholipids , Muscle, Skeletal , Receptors, Lysophosphatidic Acid , Signal Transduction , YAP-Signaling Proteins , Lysophospholipids/metabolism , Animals , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Mice , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Hippo Signaling Pathway , Mice, Inbred mdx , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Adipogenesis/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/pathologyABSTRACT
BACKGROUND: Adipocytokines play a pivotal role in maintaining adipose tissue homeostasis by regulating cellular metabolism, proliferation, differentiation, and secretory activity. These soluble factors are relevant components for healthy adipose tissue, while their deficiency is closely associated with the development of obesity and related metabolic diseases, e.g., chronic inflammation. In human adipose tissue, inter-α-trypsin inhibitor heavy chain 5 (ITIH5) is expressed in proportion to the development of adipose tissue, i.e., the individual's BMI. Thus, ITIH5 has been proposed to be an inert marker of human obesity. However, when applied to adipose stem cells in vitro, recombinant (r)ITIH5 protein inhibited proliferation and adipogenesis, suggesting that ITIH5 negatively affects the development of fat mass. We now tested the role of ITIH5 in vivo and compared ITIH5+/+ wildtype with ITIH5-/- knockout mice. RESULTS: Genetic deletion of ITIH5 significantly increased adipose tissue mass relative to animal bodyweight (p < 0.05). Next, we characterized adipose stem cells (ASCs) from both genotypes in vitro. ITIH5-/- cells exhibited increased proliferation and adipogenic differentiation (p < 0.001), which could explain the increase in adipose tissue in vivo. Furthermore, ASCs from ITIH5-/- animals were more responsive to stimulation with inflammatory mediators, i.e., these cells released greater amounts of IL-6 and MCP-1 (p < 0.001). Importantly, the application of the rITIH5 protein reversed the observed knockout effects in ASCs. CONCLUSIONS: Our data suggest that ITIH5 potently regulates adipose tissue development and homeostasis by modulating ASC biology in mice. In addition, the effect of the rITIH5 protein underscores its potential as a therapeutic agent to correct the adipose tissue dysregulation often associated with obesity and metabolic disorders.
Subject(s)
Adipogenesis , Adipose Tissue , Mice, Knockout , Animals , Male , Mice , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue/metabolism , Cell Differentiation , Cell Proliferation/genetics , Gene Deletion , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Proteinase Inhibitory Proteins, Secretory/geneticsABSTRACT
Malva parviflora has shown anti-inflammatory, antihypertensive, antihyperlipidemic, and hypoglycemic effects. This study is aimed to evaluate the anti-adipogenic effect of M. parviflora on 3T3-L1 adipocytes. Fibroblast differentiation was induced either in the absence or presence of M. parviflora fractions (F3, F4, F7, F12, F13, F17, F18 and F19) for 4 days; F17 and 18 were the most effective fractions in reducing intracellular lipid accumulation (by 25.6% and 23.1%, respectively). EC50 of F17 and F18 (14 µg/mL and 17 µg/mL, respectively) were used to evaluate their anti adipogenic effect. After 10 days of inducing differentiation in the absence or presence of the extracts at the EC50 of F17 and F18, lipid accumulation, the concentration of interleukin 6 (IL-6) were measured in the culture medium; the presence of PPAR-γ, AKT, and p-AKT was also determined. In differentiated adipocytes (C2), F17 maintained intracellular lipid concentration at levels comparable to metformin, while decreasing PPAR-γ and increasing p-AKT presence; it also prevented IL-6 expression. F17 consists of alanine, valine, phenylalanine, and proline. On the other hand, F18 reduced intracellular lipid concentrations, prevented the increase of PPAR-γ and p-AKT, and maintained IL-6 expression at similar levels as metformin. F18 is mainly constituted by alanine, valine, proline, and sucrose. In conclusion, M. parviflora fractions (F17 and F18) control the process of adipogenesis, lipogenesis, and cellular dysfunction.
Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , PPAR gamma , Plant Extracts , Animals , Mice , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Adipogenesis/drug effects , Plant Extracts/pharmacology , PPAR gamma/metabolism , Interleukin-6/metabolism , Cell Differentiation/drug effects , Lipid Metabolism/drug effects , Proto-Oncogene Proteins c-akt/metabolismABSTRACT
White adipocytes store energy, while brown and brite adipocytes release heat via nonshivering thermogenesis. In this study, we characterized two murine embryonic clonal preadipocyte lines, EB5 and EB7, each displaying unique gene marker expression profiles. EB5 cells differentiate into brown adipocytes, whereas EB7 cells into brite (also known as beige) adipocytes. To draw a comprehensive comparison, we contrasted the gene expression patterns, adipogenic capacity, as well as carbohydrate and lipid metabolism of these cells to that of F442A, a well-known white preadipocyte and adipocyte model. We found that commitment to differentiation in both EB5 and EB7 cells can be induced by 3-Isobutyl-1-methylxanthine/dexamethasone (Mix/Dex) and staurosporine/dexamethasone (St/Dex) treatments. Additionally, the administration of rosiglitazone significantly enhances the brown and brite adipocyte phenotypes. Our data also reveal the involvement of a series of genes in the transcriptional cascade guiding adipogenesis, pinpointing GSK3ß as a critical regulator for both EB5 and EB7 adipogenesis. In a developmental context, we observe that, akin to brown fat progenitors, brite fat progenitors make their appearance in murine development by 11-12 days of gestation or potentially earlier. This result contributes to our understanding of adipocyte lineage specification during embryonic development. In conclusion, EB5 and EB7 cell lines are valuable for research into adipocyte biology, providing insights into the differentiation and development of brown and beige adipocytes. Furthermore, they could be useful for the characterization of drugs targeting energy balance for the treatment of obesity and metabolic diseases.
Subject(s)
Adipocytes, Beige , Adipocytes, Brown , Adipogenesis , Cell Differentiation , Animals , Mice , Adipocytes, Brown/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipocytes, Beige/metabolism , Adipocytes, Beige/cytology , Adipogenesis/genetics , Adipogenesis/drug effects , Cell Differentiation/genetics , Cell Differentiation/drug effects , Cell LineABSTRACT
Osteoporosis is the most common metabolic bone disorder and is associated with a high incidence of fractures. Angiogenesis and adequate blood flow are important during bone repair and maintenance. Estrogens play a key role in bone formation, in the prevention of bone resorption and vasculature maintenance. Hormone replacement therapy (HRT) has been used with great benefits for bone fracture prevention but has been linked to the development of serious important side effects, including cancer and stroke. Phytoestrogens are an attractive alternative to HRT because their chemical structure is similar to estradiol but, they could behave as selective modulators: acting as antagonists of estrogen receptors in the breast and endometrium and as agonists in the vascular endothelium and bone. Hops contain a wide variety of phytoestrogens that have individually been shown to possess estrogenic activity by either blocking or mimicking. In this study we have to evaluate the in vitro effects and mechanisms of action of hops extracts on the osteogenic and adipogenic capacity of bone marrow progenitor cells (BMPCs), and the angiogenic potential of EA.hy926 endothelial cells. We show that hops extracts increase the proliferative capacity of BMPCs and promote their osteogenic differentiation while decreasing their pro-osteoclastogenic capacity; and that these effects are mediated by the MAPK pathway. Additionally, hops extracts prevent the adipogenic differentiation of BMPCs and promote endothelial cell activity, by mechanisms also partially mediated by MAPK.
Subject(s)
Bone Marrow Cells , Cell Differentiation , Cell Proliferation , Endothelial Cells , Humulus , Osteogenesis , Plant Extracts , Humulus/chemistry , Osteogenesis/drug effects , Humans , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Plant Extracts/pharmacology , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Neovascularization, Physiologic/drug effects , Phytoestrogens/pharmacology , Adipogenesis/drug effects , Mice , MAP Kinase Signaling System/drug effects , Cells, Cultured , Cell LineABSTRACT
The study evaluated the effects of Arthrospira maxima phycobiliproteins (PBPs), rosiglitazone (RSG), and 17ß-estradiol (E) on the differentiation process of 3T3-L1 cells and on their regulation of lipogenic and inflammatory gene expression at different stages of the process. The results showed that phycobiliproteins promoted cell proliferation after 24 h of treatment. Furthermore, for all three treatments, the regulation of the highest number of markers occurred on days 6 and 12 of differentiation, regardless of when the treatment was applied. Phycobiliproteins reduced lipid droplet accumulation on days 3, 6, 10, and 13 of the adipogenic process, while rosiglitazone showed no differences compared to the control. On day 6, both phycobiliproteins and rosiglitazone positively regulated Acc1 mRNA. Meanwhile, all three treatments negatively regulated Pparγ and C/ebpα. Phycobiliproteins and estradiol also negatively regulated Ucp1 and Glut4 mRNAs. Rosiglitazone and estradiol, on the other hand, negatively regulated Ppara and Il-6 mRNAs. By day 12, phycobiliproteins and rosiglitazone upregulated Pparγ mRNA and negatively regulated Tnfα and Il-1ß. Additionally, phycobiliproteins and estradiol positively regulated Il-6 and negatively regulated Ppara, Ucp2, Acc1, and Glut4. Rosiglitazone and estradiol upregulate C/ebpα and Ucp1 mRNAs. The regulation exerted by phycobiliproteins on the mRNA expression of the studied markers was dependent on the phase of cell differentiation. The results of this study highlight that phycobiliproteins have an anti-adipogenic and anti-inflammatory effect by reducing the expression of adipogenic, lipogenic, and inflammatory genes in 3T3-L1 cells at different stages of the differentiation process.
Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Differentiation , Estradiol , Phycobiliproteins , Rosiglitazone , Animals , Mice , Estradiol/pharmacology , Rosiglitazone/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/cytology , Cell Differentiation/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Phycobiliproteins/pharmacology , Phycobiliproteins/metabolism , Phycobiliproteins/genetics , Gene Expression Regulation/drug effects , Lipogenesis/drug effects , Lipogenesis/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Cell Proliferation/drug effects , Inflammation/metabolism , Inflammation/genetics , SpirulinaABSTRACT
BACKGROUND: Human Adenovirus D-36 (HAdV-D36) promotes adipogenesis in cellular and animal models and may contribute to the development of human obesity. Induction of PPARγ by HAdV-D36 seems to have a central role in the maintenance of adipogenic status. There is limited information about epigenetic mechanisms contributing to this process in human adipose tissue. This study evaluated the expression of lncRNAs (ADINR, GAS5 and MEG3) and miRNAs (miR-18a and miR-140) involved in the adipogenic process in visceral adipose tissue (VAT) of subjects with obesity with previous HAdV-D36 infection (seropositive) and unexposed (seronegative) subjects with obesity. METHODS: Individuals with obesity were grouped according to the presence of antibodies against HAdV-D36 (Seropositive: HAdV-D36[+], n = 29; and Seronegative: HAdV-D36[-], n = 28). Additionally, a group of individuals without obesity (n = 17) was selected as a control group. The HAdV-D36 serology was carried out by ELISA. Biopsies of VAT were obtained during an elective and clinically indicated surgery (bariatric or cholecystectomy). RNA extraction from VAT was performed and the expression of PPARG and non-coding RNAs was evaluated by qPCR. RESULTS: HAdV-D36[+] individuals had lower expression of anti-adipogenic lncRNAs GAS5 (p = 0.016) and MEG3 (p = 0.035) compared with HAdV-D36[-] subjects with obesity. HAdV-D36[+] subjects also presented increased expression of the adipogenic miRNA miR-18a (p = 0.042), which has been reported to be modulated by GAS5 through a RNA sponging mechanism during adipogenic differentiation. Additionally, an inverse correlation of GAS5 with PPARG expression was observed (r = -0.917, p = 0.01). CONCLUSION: Our results suggest that HAdV-D36 is related to non-coding RNAs implicated in adipogenesis, representing a potential mechanism by which previous HAdV-D36 infection could be associated with the long-term maintenance of adipogenic status, probably through the GAS5/miR-18a axis.
Subject(s)
Adipogenesis , Obesity , RNA, Long Noncoding , Humans , RNA, Long Noncoding/metabolism , Male , Female , Obesity/metabolism , Obesity/genetics , Adult , Middle Aged , Adipogenesis/genetics , Adenoviruses, Human/genetics , Adipose Tissue/metabolism , Intra-Abdominal Fat/metabolism , Adenovirus Infections, Human/metabolismABSTRACT
Adipose tissue metabolism is actively involved in the regulation of energy balance. Adipose-derived stem cells (ASCs) play a critical role in maintaining adipose tissue function through their differentiation into mature adipocytes (Ad). This study aimed to investigate the impact of an obesogenic environment on the epigenetic landscape of ASCs and its impact on adipocyte differentiation and its metabolic consequences. Our results showed that ASCs from rats on a high-fat sucrose (HFS) diet displayed reduced adipogenic capacity, increased fat accumulation, and formed larger adipocytes than the control (C) group. Mitochondrial analysis revealed heightened activity in undifferentiated ASC-HFS but decreased respiratory and glycolytic capacity in mature adipocytes. The HFS diet significantly altered the H3K4me3 profile in ASCs on genes related to adipogenesis, mitochondrial function, inflammation, and immunomodulation. After differentiation, adipocytes retained H3K4me3 alterations, confirming the upregulation of genes associated with inflammatory and immunomodulatory pathways. RNA-seq confirmed the upregulation of genes associated with inflammatory and immunomodulatory pathways in adipocytes. Overall, the HFS diet induced significant epigenetic and transcriptomic changes in ASCs, impairing differentiation and causing dysfunctional adipocyte formation.NEW & NOTEWORTHY Obesity is associated with the development of chronic diseases like metabolic syndrome and type 2 diabetes, and adipose tissue plays a crucial role. In a rat model, our study reveals how an obesogenic environment primes adipocyte precursor cells, leading to epigenetic changes that affect inflammation, adipogenesis, and mitochondrial activity after differentiation. We highlight the importance of histone modifications, especially the trimethylation of histone H3 to lysine 4 (H3K4me3), showing its influence on adipocyte expression profiles.
Subject(s)
Adipocytes , Adipogenesis , Adipose Tissue , Diet, High-Fat , Epigenesis, Genetic , Histones , Transcriptome , Animals , Rats , Adipocytes/metabolism , Diet, High-Fat/adverse effects , Histones/metabolism , Male , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue/metabolism , Cell Differentiation/genetics , Stem Cells/metabolism , Obesity/metabolism , Obesity/genetics , Cellular Reprogramming/physiology , Cells, Cultured , Rats, Wistar , Rats, Sprague-DawleyABSTRACT
Periodontal ligament stem cells (PDLSCs) show plasticity towards the adipogenic lineage; however, little has been done on the participation of epigenetic mechanisms. Histone acetylation is a dynamic process, though balanced by histone acetyltransferases (HATs) and histone deacetylases (HDACs) activities. This process can be halted by HDACs inhibitors, such as trichostatin A (TSA) and valproic acid (VPA). This study aimed to determine the role of HDACs class I in adipogenic differentiation of PDL cells. PDLSCs were treated with TSA at concentrations of 100, 200, and 250 nM, or VPA at 1, 4 and 8 mM. Cell viability was assessed using MTT assays. Gene expression of pluripotency markers (NANOG, OCT4, SOX2), HAT genes (p300, GCN5), and HDACs genes (HDAC1-3) was analyzed by RT-qPCR. Adipogenic differentiation was evaluated via oil red O staining, and acetylation of histone H3 lysine 9 (H3K9ac) was examined by Western blot. VPA treatment resulted in a 60% reduction in cell proliferation, compared to a 50% when using TSA. Cell viability was not affected by either inhibitor. Furthermore, both TSA and VPA induced adipogenic differentiation, through an increase in the deposition of lipid droplets and in GCN5 and p300 expression were observed. Western blot analysis showed that TSA increased H3K9ac levels on adipogenic differentiation of PDLSCs. These findings highlight the potential of HDAC inhibitors as a tool for modulating H3K9 acetylation status and thus influencing adipogenic differentiation of PDLCs.
Subject(s)
Adipogenesis , Cell Differentiation , Cell Survival , Histone Deacetylase Inhibitors , Periodontal Ligament , Valproic Acid , Humans , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Histone Deacetylase Inhibitors/pharmacology , Adipogenesis/drug effects , Adipogenesis/genetics , Valproic Acid/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Acetylation/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Cells, Cultured , Histones/metabolism , Cell Proliferation/drug effects , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolismABSTRACT
Cellular Communication Network Factor 2, CCN2, is a profibrotic cytokine implicated in physiological and pathological processes in mammals. The expression of CCN2 is markedly increased in dystrophic muscles. Interestingly, diminishing CCN2 genetically or inhibiting its function improves the phenotypes of chronic muscular fibrosis in rodent models. Elucidating the cell-specific mechanisms behind the induction of CCN2 is a fundamental step in understanding its relevance in muscular dystrophies. Here, we show that the small lipids LPA and 2S-OMPT induce CCN2 expression in fibro/adipogenic progenitors (FAPs) through the activation of the LPA1 receptor and, to a lower extent, by also the LPA6 receptor. These cells show a stronger induction than myoblasts or myotubes. We show that the LPA/LPARs axis requires ROCK kinase activity and organized actin cytoskeleton upstream of YAP/TAZ signaling effectors to upregulate CCN2 levels, suggesting that mechanical signals are part of the mechanism behind this process. In conclusion, we explored the role of the LPA/LPAR axis on CCN2 expression, showing a strong cytoskeletal-dependent response in muscular FAPs.
Subject(s)
Adipogenesis , Connective Tissue Growth Factor , Lysophospholipids , Animals , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Mice , Lysophospholipids/metabolism , Cell Communication , Signal Transduction , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Stem Cells/metabolism , Stem Cells/cytology , Gene Expression Regulation , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Cell Differentiation , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Humans , Actin Cytoskeleton/metabolismABSTRACT
Maternal obesity is a well-known risk factor for developing premature obesity, metabolic syndrome, cardiovascular disease and type 2 diabetes in the progeny. The development of white adipose tissue is a dynamic process that starts during prenatal life: fat depots laid down in utero are associated with the proportion of fat in children later on. How early this programming takes place is still unknown. However, recent evidence shows that mesenchymal stem cells (MSC), the embryonic adipocyte precursor cells, show signatures of the early setting of an adipogenic committed phenotype when exposed to maternal obesity. This review aims to present current findings on the cellular adaptations of MSCs from the offspring of women with obesity and how the metabolic environment of MSCs could affect the early commitment towards adipocytes. In conclusion, maternal obesity can induce early programming of fetal adipose tissue by conditioning MSCs. These cells have higher expression of adipogenic markers, altered insulin signalling and mitochondrial performance, compared to MSCs of neonates from lean pregnancies. Fetal MSCs imprinting by maternal obesity could help explain the increased risk of childhood obesity and development of further noncommunicable diseases.
Subject(s)
Mesenchymal Stem Cells , Obesity, Maternal , Prenatal Exposure Delayed Effects , Humans , Female , Pregnancy , Obesity, Maternal/metabolism , Adipose Tissue , Pediatric Obesity , Adipogenesis/physiology , Infant, Newborn , AdipocytesABSTRACT
White adipose tissue (WAT) regulates energy balance through energy storage, adipokines secretion and the thermogenesis process. Beige adipocytes are responsible for WAT thermogenesis. They are generated by adipogenesis or transdifferentiation during cold or ß3-adrenergic agonist stimulus through a process called browning. Browning has gained significant interest for to its preventive effect on obesity. Glucocorticoids (GCs) have several functions in WAT biology; however, their role in beige adipocyte generation and WAT browning is not fully understood. The aim of our study was to determine the effect of dexamethasone (DXM) on WAT thermogenesis. For this purpose, rats were treated with DXM at room temperature (RT) or cold conditions to determine different thermogenic markers. Furthermore, the effects of DXM on the adipogenic potential of beige precursors and on mature beige adipocytes were evaluated in vitro. Our results showed that DXM decreased UCP-1 mRNA and protein levels, mainly after cold exposure. In vitro studies showed that DXM decreased the expression of a beige precursor marker (Ebf2), affecting their ability to differentiate into beige adipocytes, and inhibited the thermogenic response of mature beige adipocytes (Ucp-1, Dio2 and Pgc1α gene expressions and mitochondrial respiration). Overall, our data strongly suggest that DXM can inhibit the thermogenic program of both retroperitoneal and inguinal WAT depots, an effect that could be exerted, at least partially, by inhibiting de novo cell generation and the thermogenic response in beige adipocytes.
Subject(s)
Adipose Tissue, Brown , Adipose Tissue, White , Rats , Animals , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Obesity/metabolism , Adipogenesis , Dexamethasone/pharmacology , ThermogenesisABSTRACT
Obesity, a chronic condition marked by the excessive accumulation of adipose tissue, not only affects individual well-being but also significantly inflates healthcare costs. The physiological excess of fat manifests as triglyceride (TG) deposition within adipose tissue, with white adipose tissue (WAT) expansion via adipocyte hyperplasia being a key adipogenesis mechanism. As efforts intensify to address this global health crisis, understanding the complex interplay of contributing factors becomes critical for effective public health interventions and improved patient outcomes. In this context, gut microbiota-derived metabolites play an important role in orchestrating obesity modulation. Microbial lipopolysaccharides (LPS), secondary bile acids (BA), short-chain fatty acids (SCFAs), and trimethylamine (TMA) are the main intestinal metabolites in dyslipidemic states. Emerging evidence highlights the microbiota's substantial role in influencing host metabolism and subsequent health outcomes, presenting new avenues for therapeutic strategies, including polyphenol-based manipulations of these microbial populations. Among various agents, caffeine emerges as a potent modulator of metabolic pathways, exhibiting anti-inflammatory, antioxidant, and obesity-mitigating properties. Notably, caffeine's anti-adipogenic potential, attributed to the downregulation of key adipogenesis regulators, has been established. Recent findings further indicate that caffeine's influence on obesity may be mediated through alterations in the gut microbiota and its metabolic byproducts. Therefore, the present review summarizes the anti-adipogenic effect of caffeine in modulating obesity through the intestinal microbiota and its metabolites.
Subject(s)
Adipogenesis , Gastrointestinal Microbiome , Humans , Caffeine/pharmacology , Caffeine/therapeutic use , Obesity/drug therapy , Obesity/metabolism , Adipose Tissue/metabolism , Diet, High-FatABSTRACT
Understanding the intricate molecular mechanisms governing the fate of human adipose-derived stem cells (hASCs) is essential for elucidating the delicate balance between adipogenic and osteogenic differentiation in both healthy and pathological conditions. Long non-coding RNAs (lncRNAs) have emerged as key regulators involved in lineage commitment and differentiation of stem cells, operating at various levels of gene regulation, including transcriptional, post-transcriptional, and post-translational processes. To gain deeper insights into the role of lncRNAs' in hASCs' differentiation, we conducted a comprehensive analysis of the lncRNA transcriptome (RNA-seq) and translatome (polysomal-RNA-seq) during a 24 h period of adipogenesis and osteogenesis. Our findings revealed distinct expression patterns between the transcriptome and translatome during both differentiation processes, highlighting 90 lncRNAs that are exclusively regulated in the polysomal fraction. These findings underscore the significance of investigating lncRNAs associated with ribosomes, considering their unique expression patterns and potential mechanisms of action, such as translational regulation and potential coding capacity for microproteins. Additionally, we identified specific lncRNA gene expression programs associated with adipogenesis and osteogenesis during the early stages of cell differentiation. By shedding light on the expression and potential functions of these polysome-associated lncRNAs, we aim to deepen our understanding of their involvement in the regulation of adipogenic and osteogenic differentiation, ultimately paving the way for novel therapeutic strategies and insights into regenerative medicine.
Subject(s)
Adipogenesis , RNA, Long Noncoding , Humans , Adipogenesis/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Osteogenesis/genetics , Cell Differentiation/genetics , Stem Cells/metabolism , Polyribosomes/metabolismABSTRACT
Perfluorooctanoic acid (PFOA) is a synthetic organofluoride surfactant associated with several toxic effects in humans and animals. Particularly, it has been observed that PFOA treatment of mice results in weight loss associated with recruited brown adipose tissue (BAT), including an increased amount of uncoupling protein 1 (UCP1). The molecular mechanism behind this BAT recruitment is presently unknown. To investigate the existence of possible cell-autonomous effects of PFOA, we treated primary cultures of brown and white (inguinal) adipocytes with PFOA, or with the non-fluorinated equivalent octanoate, or with vehicle, for 48 h (from day 5 to day 7 of differentiation). PFOA in itself increased the gene expression (mRNA levels) of UCP1 and carnitine palmitoyltransferase 1A (CPT1α) (thermogenesis-related genes) in both brown and white adipocytes. In addition, PFOA increased the expression of fatty acid binding protein 4 (FABP4) and peroxisome proliferator-activated receptor α (PPARα) (adipogenesis-related genes). Also the protein levels of UCP1 were increased in brown adipocytes exposed to PFOA. This increase was more due to an increase in the fraction of cells that expressed UCP1 than to an increase in UCP1 levels per cell. The PFOA-induced changes were even more pronounced under simultaneous adrenergic stimulation. Octanoate induced less pronounced effects on adipocytes than did PFOA. Thus, PFOA in itself increased the levels of thermogenic markers in brown and white adipocytes. This could enhance the energy metabolism of animals (and humans) exposed to the compound, resulting in a negative energy balance, leading to diminished fitness.
Subject(s)
Adipogenesis , Caprylates , Fluorocarbons , Humans , Mice , Animals , Caprylates/toxicity , Adipocytes, White , Thermogenesis/geneticsABSTRACT
Aim: Obesity is a chronic pathology of epidemic proportions. Mature adipocytes from a 3T3-L1 cell line were used as in vitro obesity model to test different bioactive compounds. We aim to evaluate cassis (Ribes nigrum) extract antioxidant activity and its antiadipogenic effect on mature adipocytes. Results: We produced an extract by using enzyme that combines cellulase and pectinase; we obtained high yield of the bioactive compound anthocyanin. Extract showed high antioxidant capacity. We conducted in vitro assays by adding the extract to adipocytes culture medium. Extract reduced intracellular levels of triglyceride by 62% and cholesterol by 32%. Conclusion: Enzymatic extract's high antioxidant activity was likely attributable to its high concentration of anthocyanin. This extract inhibits lipid accumulation in adipocytes.
Obesity is a disease all over the world. By 2030, nearly 20% of adults are predicted to be obese. The consumption of processed foods is related to obesity in some countries such as Argentina. More natural food is needed. There are many different anti-obesity medicines but there is no good one to lose weight. We took extracts from cassis fruits and tested whether they could decrease fats like cholesterol within fat cells. We found that these extracts could successfully reduce the fat levels in the cells. Our results indicate that natural compounds like cassis fruit extract may be helpful in preventing future obesity epidemics.
Subject(s)
Anti-Obesity Agents , Ribes , Triglycerides/metabolism , Triglycerides/pharmacology , Anthocyanins/pharmacology , Adipogenesis , Anti-Obesity Agents/metabolism , Anti-Obesity Agents/pharmacology , Antioxidants/pharmacology , Antioxidants/metabolism , Plant Extracts/pharmacology , Adipocytes/metabolism , Obesity/metabolism , CholesterolABSTRACT
BACKGROUND: The search for nutritional intervention strategies against obesity has grown, highlighting the low-carbohydrate diet model. However, little is known about the impact of the quality of fatty acids consumed in this diet. Thus, we aim to investigate the influence of fatty acid quality on dietary strategy on obesity. METHODS: Male Swiss mice were diet-induced to obesity. Afterward, mice consume a low-carb diet with different types of fat: saturated, polyunsaturated ω-3, ω-6, and monounsaturated ω-9 fatty acids. Weight gain and food consumption were monitored weekly. An oral glucose tolerance test was performed and blood and tissue samples were collected for analysis of insulin resistance markers. Protein expression of insulin signaling pathway molecules, lipid metabolism, mitochondrial function, macrophage polarization, and cytokine production were analyzed. RESULTS: The high-fat diet was able to induce obesity and glucose intolerance. The switch to a low-carbohydrate dietary pattern reversed the glucose intolerance, with better results in the ω-3 and ω-9 groups. After the low-carbohydrate diet, groups ω-3 and ω-9 presented improved fasting serum glucose, insulin, and HOMA indexes. The low-carbohydrate diet also increased the activity of insulin pathway proteins such as IR, IRS1, and AKT. Furthermore, the ω-3 diet group showed greater activity of mitochondrial complexes and AMPK signaling pathway proteins. The ω-6 and ω-9 -rich diet induced M2-type macrophage polarization, as well as cytokine production modulation by the low-carbohydrate diet in the ω-3 and ω-9 groups. CONCLUSIONS: Consuming a low-carbohydrate diet pattern promotes weight loss and improves glucose intolerance in obesity. Also, the quality of lipids has a direct influence, demonstrating that the consumption of ω-3 polyunsaturated and ω-9 monounsaturated lipids can lead to more favorable outcomes for the improvement of glucose intolerance, lipid metabolism, and anti-inflammatory effects.
Subject(s)
Fatty Acids, Omega-3 , Glucose Intolerance , Insulin Resistance , Male , Mice , Animals , Fatty Acids/analysis , Adipogenesis , Obesity/metabolism , Fatty Acids, Omega-3/pharmacology , Insulin , Diet, High-Fat/adverse effects , Fatty Acids, Monounsaturated , Diet, Carbohydrate-Restricted , Cytokines , Blood Glucose/metabolismABSTRACT
Maternal obesity predisposes offspring to obesity in adulthood. Since the perinatal period is a critical window for adipose organogenesis, we evaluated if maternal obesity affects the perinatal offspring adipogenesis. Female mice were fed a standard diet (eutrophic dam, ED) or a high-fat diet supplemented with condensed milk (obese dam, OD) for 6 weeks before mating, and the diets were maintained until the end of the protocol. Inguinal adipose tissue of offspring at gestational day 16.5 (E16.5), postnatal day 0 (P0), and P2 was collected to analyze morphological and molecular features. In OD offspring, the number of preadipocytes increased at E16.5 and P0 compared to ED offspring. The cell cycle-related elements Ccnd1 and Ki67 were also upregulated in these groups. In parallel, lipid accumulation started at E16.5 in OD offspring, while ED offspring preadipocytes only accumulated lipids after P0. Peroxisome proliferator-activated receptor gamma (PPARγ) levels and activity were decreased in OD offspring due to impaired nuclear migration. Increased Hdac1 expression, which negatively regulates PPAR-responsive elements in the genome, was also detected. At P2, OD adipocytes presented abnormal features, including a clustered distribution and decreased expression of PPARγ target genes and Adbr3 and Slc2a4, which are highly expressed in mature functional adipocytes. The abnormal adipose tissue is one of the major factors promoting metabolic abnormalities in adulthood. This study demonstrates for the first time the morphological and molecular alterations induced by maternal obesity in vivo in the perinatal adipogenesis in murine inguinal adipose tissue.
Subject(s)
Adipogenesis , Obesity, Maternal , Animals , Female , Humans , Mice , Pregnancy , 3T3-L1 Cells , Adipogenesis/genetics , Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Obesity/metabolism , Obesity, Maternal/metabolism , PPAR gamma/genetics , PPAR gamma/metabolismABSTRACT
Human mesenchymal stem cells (hMSC) represent a unique and promising platform because of their ability to promote soft tissue regeneration, particularly their ability to differentiate into adipocytes, which are important for adipose tissue regeneration. In this context, type I collagen is the most abundant extracellular matrix component of adipose tissue and can act as a natural spheroid source to support the differentiation process of stem cells. However, spheroids based on collagen and hMSCs without numerous pro-adipogenic factors that can induce adipogenesis have not yet been investigated. In this study, we focused on developing collagen-hMSC spheroids capable of differentiating into adipocyte-like cells in a short time (eight culture days) without adipogenic factors, with potential applications in adipose tissue repair. The physical and chemical properties of the spheroids indicated successful cross-linking of collagen. Upon spheroid development, stability, cell viability, and metabolic activity of the constructs were maintained. During adipogenesis, cell morphology shows significant changes, in which cells change from a fibroblast-like shape to an adipocyte-like shape, and adipogenic gene expression after eight days of cell culture. These results support the utility of collagen-hMSC 3 mg ml-1collagen concentration spheroids to differentiate into adipocyte-like cells in a short time without adverse effects on biocompatibility, metabolic activity, or cell morphology, suggesting that this construct may be used in soft tissue engineering.