RESUMO
Brown adipose tissue (BAT) functions as a thermogenic organ and is negatively associated with cardiometabolic diseases. N6 -methyladenosine (m6 A) modulation regulates the fate of stem cells. Here, we show that the prostaglandin E2 (PGE2 )-E-prostanoid receptor 3 (EP3) axis was activated during mouse interscapular BAT development. Disruption of EP3 impaired the browning process during adipocyte differentiation from pre-adipocytes. Brown adipocyte-specific depletion of EP3 compromised interscapular BAT formation and aggravated high-fat diet-induced obesity and insulin resistance in vivo. Mechanistically, activation of EP3 stabilized the Zfp410 mRNA via WTAP-mediated m6 A modification, while knockdown of Zfp410 abolished the EP3-induced enhancement of brown adipogenesis. EP3 prevented ubiquitin-mediated degradation of WTAP by eliminating PKA-mediated ERK1/2 inhibition during brown adipocyte differentiation. Ablation of WTAP in brown adipocytes abrogated the protective effect of EP3 overexpression in high-fat diet-fed mice. Inhibition of EP3 also retarded human embryonic stem cell differentiation into mature brown adipocytes by reducing the WTAP levels. Thus, a conserved PGE2 -EP3 axis promotes BAT development by stabilizing WTAP/Zfp410 signaling in a PKA/ERK1/2-dependent manner.
Assuntos
Tecido Adiposo Marrom , Dinoprostona , Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Dinoprostona/metabolismo , Humanos , Metiltransferases/metabolismo , Camundongos , RNA/metabolismo , Fatores de Processamento de RNA/metabolismo , Receptores de Prostaglandina E Subtipo EP3 , TermogêneseRESUMO
The TP53 gene is mutated in approximately 30% of all breast cancer cases. Adipocytes and preadipocytes, which constitute a substantial fraction of the stroma of normal mammary tissue and breast tumors, undergo transcriptional, metabolic, and phenotypic reprogramming during breast cancer development and play an important role in tumor progression. We report here that p53 loss in breast cancer cells facilitates the reprogramming of preadipocytes, inducing them to acquire a unique transcriptional and metabolic program that combines impaired adipocytic differentiation with augmented cytokine expression. This, in turn, promotes the establishment of an inflammatory tumor microenvironment, including increased abundance of Ly6C+ and Ly6G+ myeloid cells and elevated expression of the immune checkpoint ligand PD-L1. We also describe a potential gain-of-function effect of common p53 missense mutations on the inflammatory reprogramming of preadipocytes. Altogether, our study implicates p53 deregulation in breast cancer cells as a driver of tumor-supportive adipose tissue reprogramming, expanding the network of non-cell autonomous mechanisms whereby p53 dysfunction may promote cancer. Further elucidation of the interplay between p53 and adipocytes within the tumor microenvironment may suggest effective therapeutic targets for the treatment of breast cancer patients.
Assuntos
Neoplasias da Mama , Proteína Supressora de Tumor p53 , Humanos , Feminino , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Mama/patologia , Genes p53 , Tecido Adiposo/metabolismo , Adipócitos/metabolismo , Microambiente Tumoral/genéticaRESUMO
Leptin can indirectly regulate fatty-acid metabolism and synthesis in muscle in vivo and directly in incubated muscle ex vivo. In addition, non-synonymous mutations in the bovine leptin gene (LEP) are associated with carcass intramuscular fat (IMF) content. However, the effects of LEP on lipid synthesis of adipocytes have not been clearly studied at the cellular level. Therefore, this study focused on bovine primary intramuscular preadipocytes to investigate the effects of LEP on the proliferation and differentiation of intramuscular preadipocytes, as well as its regulatory mechanism in lipid synthesis. The results showed that both the LEP and leptin receptor gene (LEPR) were highly expressed in IMF tissues, and their mRNA expression levels were positively correlated at different developmental stages of intramuscular preadipocytes. The overexpression of LEP inhibited the proliferation and differentiation of intramuscular preadipocytes, while interference with LEP had the opposite effect. Additionally, LEP significantly promoted the phosphorylation level of AMPKα by promoting the protein expression of CAMKK2. Meanwhile, rescue experiments showed that the increasing effect of AMPK inhibitors on the number of intramuscular preadipocytes was significantly weakened by the overexpression of LEP. Furthermore, the overexpression of LEP could weaken the promoting effect of AMPK inhibitor on triglyceride content and droplet accumulation, and prevent the upregulation of adipogenic protein expression (SREBF1, FABP4, FASN, and ACCα) caused by AMPK inhibitor. Taken together, LEP acted on the AMPK signaling pathway by regulating the protein expression of CAMKK2, thereby downregulating the expression of proliferation-related and adipogenic-related genes and proteins, ultimately reducing intramuscular adipogenesis.
Assuntos
Proteínas Quinases Ativadas por AMP , Adipócitos , Adipogenia , Leptina , Transdução de Sinais , Animais , Adipogenia/fisiologia , Bovinos , Adipócitos/metabolismo , Adipócitos/citologia , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Leptina/metabolismo , Leptina/genética , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Receptores para Leptina/metabolismo , Receptores para Leptina/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/citologiaRESUMO
MicroRNAs (miRNAs) are widely involved in various lipogenic processes, including adipocyte proliferation and differentiation, lipid droplet formation, and adipocyte-specific gene activation. The present study aimed to investigate the gene expression profiles of bovine preadipocytes under high miR-10167-3p expression using the RNA-seq technique and to verify the functions of its downstream target genes on the proliferation and differentiation of bovine preadipocytes. First, RNA-seq identified 573 differentially expressed genes (DEGs), of which 243 were downregulated and 330 were upregulated. Then, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that 15.19% of the DEGs were enriched in pathways related to lipid metabolism. Meanwhile, dual-luciferase reporter gene assay verified the target-binding relationship between miR-10167-3p and TCF7L1. The function of TCF7L1 was assessed using several experiments in adipocytes with high TCF7L1 expression and RNA interference. The mRNA and protein expression of proliferation, differentiation, and apoptosis marker genes were detected using qPCR and western blot, respectively; lipid droplet synthesis was detected using oil red O, Nile red, and bodipy staining; adipocyte proliferation was detected by EdU; and apoptosis was detected using flow cytometry. The results revealed that TCF7L1 overexpression inhibited bovine preadipocyte differentiation and apoptosis and promoted their proliferation, with opposite results obtained with its RNA interference. These results may provide a reference for the subsequent investigation of the molecular mechanism of bovine fat deposition.
Assuntos
Adipócitos , Diferenciação Celular , Proliferação de Células , MicroRNAs , Animais , Bovinos , MicroRNAs/genética , MicroRNAs/metabolismo , Adipócitos/metabolismo , Adipócitos/citologia , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Adipogenia/genética , Células Cultivadas , ApoptoseRESUMO
BACKGROUND: Skeletal muscle development and fat deposition have important effects on meat quality. The study of regulating skeletal muscle development and fat deposition is of great significance in improving the quality of carcass and meat. In the present study, whole transcriptome sequencing (including RNA-Seq and miRNA-Seq) was performed on the longissimus dorsi muscle (LDM) of Jinfen White pigs at 1, 90, and 180 days of age. RESULTS: The results showed that a total of 245 differentially expressed miRNAs were screened in any two comparisons, which may be involved in the regulation of myogenesis. Among them, compared with 1-day-old group, miR-22-5p was significantly up-regulated in 90-day-old group and 180-day-old group. Functional studies demonstrated that miR-22-5p inhibited the proliferation and differentiation of porcine skeletal muscle satellite cells (PSCs). Pearson correlation coefficient analysis showed that long non-coding RNA (lncRNA) LOC106505926 and CXXC5 gene had strong negative correlations with miR-22-5p. The LOC106505926 and CXXC5 were proven to promote the proliferation and differentiation of PSCs, as opposed to miR-22-5p. In terms of mechanism, LOC106505926 functions as a molecular sponge of miR-22-5p to modulate the expression of CXXC5, thereby inhibits the differentiation of PSCs. In addition, LOC106505926 regulates the differentiation of porcine preadipocytes through direct binding with FASN. CONCLUSIONS: Collectively, our results highlight the multifaceted regulatory role of LOC106505926 in controlling skeletal muscle and adipose tissue development in pigs and provide new targets for improving the quality of livestock products by regulating skeletal muscle development and fat deposition.
Assuntos
Diferenciação Celular , Lipogênese , MicroRNAs , Desenvolvimento Muscular , RNA Longo não Codificante , Animais , RNA Longo não Codificante/genética , Desenvolvimento Muscular/genética , Suínos , MicroRNAs/genética , MicroRNAs/metabolismo , Lipogênese/genética , Diferenciação Celular/genética , Proliferação de Células , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/crescimento & desenvolvimento , Células CultivadasRESUMO
Melanoma, the deadliest skin cancer, originates from epidermal melanocytes. The influence of preadipocytes on melanoma is less understood. We co-cultured mouse melanoma B16 cells with 3T3L1 preadipocytes to form mixed spheroids and observed increased melanoma proliferation and growth compared to B16-only spheroids. Metastasis-related proteins YAP, TAZ, and PD-L1 levels were also higher in mixed spheroids. Treatment with exosome inhibitor GW4869 halted melanoma growth and reduced expression of these proteins, suggesting exosomal crosstalk between B16 and 3T3L1 cells. MiR-155 expression was significantly higher in mixed spheroids, and GW4869 reduced its levels. Additionally, co-culturing with Raw264.7 macrophage cells increased M2 markers IL-4 and CD206 in Raw264.7 cells, effects that were diminished by GW4869. These results indicate that preadipocytes may enhance melanoma progression and metastasis via exosomal interactions.
Assuntos
Adipócitos , Exossomos , Macrófagos , Melanoma Experimental , Microambiente Tumoral , Animais , Camundongos , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/efeitos dos fármacos , Adipócitos/metabolismo , Adipócitos/patologia , Adipócitos/efeitos dos fármacos , Melanoma Experimental/patologia , Melanoma Experimental/metabolismo , Células RAW 264.7 , Exossomos/metabolismo , Técnicas de Cocultura , Progressão da Doença , Células 3T3-L1 , Compostos de Benzilideno/farmacologia , Compostos de Anilina/farmacologia , Proliferação de Células/efeitos dos fármacos , Melanoma/patologia , Melanoma/metabolismo , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/metabolismo , Linhagem Celular Tumoral , MicroRNAs/metabolismo , MicroRNAs/genéticaRESUMO
Adipogenesis is tightly regulated by various factors, including genes and microRNAs. Excessive fat deposition is the key feature of obesity, which is a low-grade chronic inflammatory disease. Follistatin-like 1 (FSTL1) has been reported to be an important mediator involved in various inflammatory diseases. However, the underlying mechanism of FSTL1 in preadipocyte differentiation and inflammatory response is still unclear. The current study was designed to explore the biological function and potential mechanism of FSTL1 in mouse subcutaneous preadipocyte differentiation. We found that FSTL1 was highly expressed in the early stage of differentiation and subsequently decreased sharply, suggesting that FSTL1 played a possible role in adipogenesis. Meanwhile, the gain- and loss-of-function assays showed that FSTL1 was not only involved in the inflammatory response by inducing the expression of pro-inflammatory factors IL-1ß and CCL2 but also significantly attenuated preadipocyte differentiation, as evidenced by the reduction of lipid accumulation and the levels of adipogenic genes, including PPARγ and FABP4. In addition, the target gene prediction and luciferase reporter assay validated that miR-125a-3p targeted the 3' UTR region of FSTL1. These results demonstrated that miR-125a-3p negatively regulated the expression of FSTL1 at the mRNA and protein levels. Furthermore, overexpressing miR-125a-3p in preadipocytes dramatically accelerated adipogenic differentiation and downregulated the levels of IL-1ß and CCL2, which were in accordance with the knockdown of FSTL1. On the contrary, treatment with miR-125a-3p inhibitors attenuated adipogenesis but induced the expression of inflammatory genes. In summary, this study suggests a positive function of FSTL1 in adipocyte-induced inflammation and negatively regulates preadipocyte differentiation. Further studies demonstrated that miR-125a-3p could reverse the effect by targeting FSTL1, which might provide a better understanding of treating obesity-related inflammatory diseases.
Assuntos
Adipogenia , MicroRNAs , Animais , Camundongos , Adipócitos/metabolismo , Adipogenia/genética , MicroRNAs/metabolismo , Obesidade/genética , Obesidade/metabolismo , RNA Mensageiro/metabolismoRESUMO
Obesity is one of the more complicated diseases, it can induce numerous life-threatening diseases mainly diabetes mellitus, cardiovascular disease, hypertension, and certain cancers. In this study, we assessed the efficacy of bacoside-A (a dammarane-type triterpenoid saponin derived from the plant Bacopa monniera Linn.) on the adipogenesis of 3T3-L1 preadipocytes. Results of this study illustrated that bacoside-A decreased the differentiation of 3T3-L1 cell, as evidenced by diminution of lipid droplets, which contains triglycerides and other lipids. During the differentiation process, transcription factors, which are mainly participating in adipogenesis such us CCAAT/enhancer-binding protein α (C/EBPα) and C/EBPß, peroxisome proliferator-activated receptor-γ (PPARγ), and sterol regulatory element-binding protein-1c (SREBP-1c), expressions were significantly suppressed by bacoside-A. In addition, bacoside-A showed a potent reduction in genes precise to adipocytes such as lipoprotein lipase (LPL), fatty acid synthase (FAS), adipocyte fatty acid-binding protein (FABP4), and leptin expressions. Further, bacoside-A stimulated the phosphorylation of acetyl CoA carboxylase (ACC) and AMP-activated protein kinase (AMPK). These results demonstrated that bacoside-A has anti-adipogenic effects by regulating the transcription factors involved in adipocyte differentiation. Therefore, bacoside-A might be considered as a potent therapeutic agent for alleviating obesity and hyperlipidemia.
Assuntos
Células 3T3-L1 , Adipócitos , Adipogenia , Diferenciação Celular , Metabolismo dos Lipídeos , Saponinas , Triterpenos , Animais , Camundongos , Saponinas/farmacologia , Saponinas/química , Triterpenos/farmacologia , Triterpenos/metabolismo , Triterpenos/química , Adipogenia/efeitos dos fármacos , Adipócitos/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/citologia , Diferenciação Celular/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacosRESUMO
Tetrabromobisphenol A (TBBPA), a widely-used brominated flame retardant, has been revealed to exert endocrine disrupting effects and induce adipogenesis. Given the high structural similarities of TBBPA analogues and their increasing exposure risks, their effects on lipid metabolism are necessary to be explored. Herein, 9 representative TBBPA analogues were screened for their interference on 3T3-L1 preadipocyte adipogenesis, differentiation of C3H10T1/2 mesenchymal stem cells (MSCs) to brown adipocytes, and lipid accumulation of HepG2 cells. TBBPA bis(2-hydroxyethyl ether) (TBBPA-BHEE), TBBPA mono(2-hydroxyethyl ether) (TBBPA-MHEE), TBBPA bis(glycidyl ether) (TBBPA-BGE), and TBBPA mono(glycidyl ether) (TBBPA-MGE) were found to induce adipogenesis in 3T3-L1 preadipocytes to different extends, as evidenced by the upregulated intracellular lipid generation and expressions of adipogenesis-related biomarkers. TBBPA-BHEE exhibited a stronger obesogenic effect than did TBBPA. In contrast, the test chemicals had a weak impact on the differentiation process of C3H10T1/2 MSCs to brown adipocytes. As for hepatic lipid formation test, only TBBPA mono(allyl ether) (TBBPA-MAE) was found to significantly promote triglyceride (TG) accumulation in HepG2 cells, and the effective exposure concentration of the chemical under oleic acid (OA) co-exposure was lower than that without OA co-exposure. Collectively, TBBPA analogues may perturb lipid metabolism in multiple tissues, which varies with the test tissues. The findings highlight the potential health risks of this kind of emerging chemicals in inducing obesity, non-alcoholic fatty liver disease (NAFLD) and other lipid metabolism disorders, especially under the conditions in conjunction with high-fat diets.
Assuntos
Células 3T3-L1 , Adipogenia , Retardadores de Chama , Metabolismo dos Lipídeos , Bifenil Polibromatos , Bifenil Polibromatos/toxicidade , Metabolismo dos Lipídeos/efeitos dos fármacos , Animais , Camundongos , Adipogenia/efeitos dos fármacos , Humanos , Retardadores de Chama/toxicidade , Células Hep G2 , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismoRESUMO
Recently, the trend of obesity is becoming increasingly prevalent, and the underlying pathogenesis of obesity is complex and needs to be researched further. In this study, we report a decreased expression of let-7a-5p in the white adipose tissue (WAT) of animals with obesity. Using the RNA oligo, let-7a-5p over-expression or suppression-expression is achieved, impacting the proliferation and differentiation of preadipocytes in vitro. Srebf2 mechanistically interacts with the metabolic effect of let-7a-5p and participates in lipid accumulation by regulating Srebf2 downstream signaling. Moreover, let-7a-5p binds to Thbs1 to interact with the PI3K-AKT-mTOR pathway, down-regulating the phosphorylation levels of AKT, mTOR, and S6K1 to decrease lipid accumulation. In conclusion, our study highlights the physiological significance of let-7a-5p in lipid accumulation and suggests that the let-7a-5p/Srebf2 and let-7a-5p/Thbs1/PI3K-AKT-mTOR axes may represent potential mechanisms for controlling lipid accumulation in obesity.
Assuntos
Metabolismo dos Lipídeos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Animais , Obesidade , Serina-Treonina Quinases TORRESUMO
1. Intramuscular fat (IMF) is a key parameter for chicken meat quality. IMF deposition is driven by genetic, nutritional and management factors, with genetics being the determining factor. Previous whole transcriptome sequencing revealed that microRNA gga-let-7a-3p was related to lipid metabolism in breast muscle. This study further investigated the potential role of gga-let-7a-3p in IMF deposition.2. The mimic and inhibitor of gga-let-7a-3p were individually transfected into chicken intramuscular preadipocytes. Subsequently, the proliferation and differentiation states of the cells were detected. Transcriptome sequencing was performed on cells transfected with gga-let-7a-3p mimic.3. The results indicated that gga-let-7a-3p suppressed the mRNA levels of proliferation and differentiation-related genes, as well as the protein levels. EdU and Oil Red O assays revealed that gga-let-7a-3p restrained preadipocyte proliferation and differentiation. In addition, a total of 333 up-regulated genes and 807 down-regulated genes were identified in cells transfected with gga-let-7a-3p mimic. Using Kyoto Encyclopaedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis, differential genes were found to be enriched in processes such as the peroxisome proliferator activated receptor (PPAR) signalling pathway and oxidative phosphorylation.4. The study demonstrated that gga-let-7a-3p inhibits the proliferation and differentiation of chicken intramuscular preadipocytes, which provides new understanding to further unravel the function of gga-let-7a-3p.
Assuntos
Galinhas , MicroRNAs , Animais , Galinhas/genética , Diferenciação Celular , Metabolismo dos Lipídeos , MicroRNAs/genética , Proliferação de CélulasRESUMO
Green tea possesses a range of beneficial effects, including anti-obesity, antioxidant, and anti-inflammatory properties, owing to its biologically active components, primarily catechins such as epicatechin (EC), epicatechin gallate (ECG), epigallocatechin (EGC), and epigallocatechin gallate (EGCG). However, few studies have investigated the four catechin monomers simultaneously, and the molecular mechanisms of their anti-obesity effects have not been fully elucidated. In this study, we investigated the effects of four catechin monomers on the differentiation of 3T3-L1 preadipocytes of mice. Our findings demonstrated that four catechin monomers EC/ECG/EGC/EGCG (12, 25, 50 µM) dose-dependently inhibited the differentiation of 3T3-L1 preadipocytes and reduced triglyceride content. EGCG exhibited the most potent inhibitory effect with an optimal concentration of 50 µM. In addition, transcriptome sequencing and lipidomic analysis of EGCG-treated 3T3-L1 preadipocytes revealed that Ptgs2 and Pim1 were the most differentially expressed genes involved in regulating adipocyte differentiation. The results suggested that EGCG up-regulated the expression of the Pla2g2e gene and down-regulated the expression of the Pla2g4a and Pla2g2a genes via the glycerophospholipid metabolic pathway, which subsequently elevated lysophosphatidylcholine (LPC) levels, influencing the differentiation process of 3T3-L1 preadipocytes.
RESUMO
As one of the important traits in pig production, meat quality has important research significance and value. Intramuscular fat (IMF) content is one of the most important factors affecting pork quality. Many experimental studies have shown that IMF content is closely related to the flavor, tenderness, and juiciness of pork. Therefore, it is of great significance to study the mechanism of porcine IMF deposition. Previous research indicated that miR-149-5p promoted the proliferation of porcine intramuscular (IM) preadipocytes and decreased their ability to differentiate, albeit the exact mechanism of action is unknown. In vitro, foreign pigs showed increased miR-149-5p expression and reduced fat deposition when compared to Queshan Black pigs. This study conducted metabolomics and transcriptomics analyses of porcine IM preadipocytes overexpressing miR-149-5p to verify their effects on lipid formation. According to metabolomics analysis, the overexpression of miR-149-5p has significantly altered the lipid, organic acid, and organic oxygen metabolites of porcine IM preadipocytes. Specially speaking, it has changed 115 metabolites, including 105 up-regulated and 10 down-regulated ones, as well as the composition of lipid, organic acid, and organic oxygen metabolism-related metabolites. RNA-seq analysis showed that overexpression of miR-149-5p significantly altered 857 genes, of which 442 were up-regulated, and 415 were down-regulated, with enrichment to MAPK, IL-17, PI3K-Akt, and ErbB signaling pathways. We found that overexpression of miR-149-5p inhibited adipogenic differentiation by changing cAMP signaling pathway in porcine IM preadipocytes. In addition, the overexpression of miR-149-5p may affect the transport of Cu2+ by targeting ATP7A and inhibiting adipogenic differentiation. These findings elucidate the regulatory function of miR-149-5p in porcine IM preadipocytes, which may be a key target for controlling pork quality.
Assuntos
Adipócitos , MicroRNAs , Suínos , Animais , Adipócitos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Transcriptoma , Fosfatidilinositol 3-Quinases/metabolismo , Adipogenia/genética , Lipídeos , Diferenciação Celular/genéticaRESUMO
The preadipocytes differentiation is a vital process of lipogenesis; exploring the molecular mechanisms of lipogenesis contributes to improve the meat quality and final commercial income. Lipogenesis has been widely reported in other livestock, but little is known about the gene expression profiles at different stages during preadipocytes differentiation in sheep. In this study, ovine preadipocytes were cultured in vitro and then induced to begin differentiation. Then, the gene expression profiles of preadipocytes collected on day 0 (D0), day 2 (D2), and day 8 (D8) of differentiation were analyzed by RNA-seq technology. According to the findings, 2254 differentially expressed genes (DEGs) were found in D2 vs D0; 1817 DEGs and 1902 DEGs were found in D8 vs D0 and D8 vs D2, respectively. The DEGs were found to be enriched in several biological processes, including focal adhesion, ECM-receptor interaction, PI3K-Akt signaling pathway, steroid biosynthesis, and MAPK signaling pathway, according to Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. The regulatory network of the DEGs related to ovine preadipocytes differentiation was systematically constructed, which showed that hub genes might modulate ovine preadipocytes differentiation. In summary, preadipocyte differentiation is regulated by several key genes, including ACACB, CXCL6, SREBF1, INSIG1, APOE, GJA1, CDH11, SYNE1, PCSK1, S100A4, FN1, PLIN2, CXCL6, FN1, PTX3, and FABP3. This study provides a deeper knowledge of the roles of genes in sheep lipogenesis by revealing global gene expression profiles during preadipocyte differentiation.
Assuntos
Perfilação da Expressão Gênica , Fosfatidilinositol 3-Quinases , Animais , Ovinos/genética , Transcriptoma , Análise em Microsséries , RNA-Seq , Redes Reguladoras de Genes , Biologia Computacional , Ontologia GenéticaRESUMO
In the pilot GWAS of children obesity, DNAJC6 gene was found as a regulator for resting metabolic rate (RMR) and obesity in children aged 8-9 years. To investigate whether DNAJC6 gene regulated obesity and energy metabolism, the physiological mechanisms during adipogenesis of 3T3-L1 preadipocytes were confirmed after DNAJC6 gene was overexpressed or inhibited. Overexpressing DNAJC6 gene maintained a 3T3-L1 preadipocyte status during cell differentiation (MTT, ORO, DAPI/BODIPY). It suppressed adipogenesis and adipokine production (leptin, adiponectin), insulin signaling with IRS-GLUT4 system (RT-PCR, Western blotting), and mitochondrial function (Mito Stress Test). DNAJC6 overexpressed cells inhibited mTOR expression, but maintained LC3 expression at a high level, indicating that autophagy occurred and energy was obtained. However, when DNAJC6 gene was inhibited, fat synthesis factor was highly expressed during differentiation (PPARr, C/EBPa, aP2, etc) and the intracellular stress level increased accordingly, which affected the reduction of reserve respiratory capacity during mitochondrial respiration. Our study confirmed gene regulation of DNAJC6, overexpression or inhibition, affects adipogenesis with energy metabolism and mitochondrial functions. This basic data can be used for clinic obesity studies to control an energy imbalance.
Assuntos
Adipogenia , Obesidade Infantil , Animais , Criança , Humanos , Camundongos , Células 3T3-L1 , Adipogenia/genética , Diferenciação Celular/genética , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP40/genética , MitocôndriasRESUMO
In this study, methionine sulfoxide (MetO) was identified as an active metabolite that suppresses adipogenesis after screening obese individuals versus the normal population. MetO suppressed the gene and protein expression of CCAAT/enhancer binding protein (C/EBP) α, adipocyte fatty acid binding protein 4 (FABP4), and the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) during human preadipocyte (HPA) differentiation. Adipogenesis decreased following MetO treatment; however, the preadipocyte number, proliferation, and apoptosis were unaffected. The activity of phosphorylated extracellular signal-related kinase (P-ERK) of the mitogen-activated protein kinase (MAPK) pathway was significantly inhibited in HPA after MetO treatment. Furthermore, treatment of preadipocytes with the selective P-ERK1/2 agonist Ro 67-7476 abolished the effect of MetO against adipogenesis suggesting that MetO function is dependent on the MAPK pathway. The mechanistic insights of adipogenesis suppression by MetO presented in this study shows its potential as an antiobesity drug.
Assuntos
Adipócitos , Adipogenia , Humanos , Camundongos , Animais , Adipócitos/metabolismo , Transdução de Sinais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/farmacologia , PPAR gama/metabolismo , Células 3T3-L1 , Diferenciação CelularRESUMO
BACKGROUND: Previous studies demonstrated that mast cells with their degranulated component heparin are the major endogenous factors that stimulate preadipocyte differentiation and promote fascial adipogenesis, and this effect is related to the structure of heparin. Regarding the structural and physiological properties of the negatively charged polymers, hexasulfonated suramin, a centuries-old medicine that is still used for treating African trypanosomiasis and onchocerciasis, is assumed to be a heparin-related analog or heparinoid. This investigation aims to elucidate the influence of suramin on the adipogenesis. METHODS: To assess the influence exerted by suramin on adipogenic differentiation of primary white adipocytes in rats, this exploration was conducted both in vitro and in vivo. Moreover, it was attempted to explore the role played by the sulfonic acid groups present in suramin in mediating this adipogenic process. RESULTS: Suramin demonstrated a dose- and time-dependent propensity to stimulate the adipogenic differentiation of rat preadipocytes isolated from the superficial fascia tissue and from adult adipose tissue. This stimulation was concomitant with a notable upregulation in expression levels of pivotal adipogenic factors as the adipocyte differentiation process unfolded. Intraperitoneal injection of suramin into rats slightly increased adipogenesis in the superficial fascia and in the epididymal and inguinal fat depots. PPADS, NF023, and NF449 are suramin analogs respectively containing 2, 6, and 8 sulfonic acid groups, among which the last two moderately promoted lipid droplet formation and adipocyte differentiation. The number and position of sulfonate groups may be related to the adipogenic effect of suramin. CONCLUSIONS: Suramin emerges as a noteworthy pharmaceutical agent with the unique capability to significantly induce adipocyte differentiation, thereby fostering adipogenesis.
Assuntos
Adipogenia , Suramina , Ratos , Animais , Suramina/farmacologia , Antiparasitários/farmacologia , Diferenciação Celular , Adipócitos Brancos , Heparina/farmacologiaRESUMO
Lanthanum (La) as a rare earth element is widely used in agriculture, industry, and medicine. It has been suggested in several studies that La might influence glycolipid metabolism in vivo. In this study, we used 3T3-L1 preadipocytes as in vitro cell model to elucidate the effects of La(NO3 )3 on adipogenesis and the underlying mechanisms. The results showed that La(NO3 )3 could inhibit the adipogenic differentiation of 3T3-L1 preadipocytes, which showed a decrease in lipid accumulation and the downregulation of specific adipogenic transcription factors. La(NO3 )3 exerted its inhibitory effect mainly at the early differentiation stage. Furthermore, La(NO3 )3 influenced the S-phase entry and cell cycle process during the mitotic clonal expansion and regulated the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and expressions of the proteins in phosphatidylinositol 3-kinase (PI3K)/Akt pathway at the early stage of differentiation. Besides, La(NO3 )3 upregulated the expressions of wnt10b mRNA and ß-catenin protein and promoted the nucleus translocation of ß-catenin. Additionally, we found that La(NO3 )3 could promote the growth of 3T3-L1 preadipocytes both with and without MDI (3-isobutyl-1-methylxanthine [IBMX], dexamethasone [Dex], and insulin) stimulation. Collectively, these results indicated that La(NO3 )3 could inhibit adipogenesis in 3T3-L1 preadipocytes and influence cell proliferation.
Assuntos
Adipogenia , Lantânio , Animais , Camundongos , Lantânio/toxicidade , Células 3T3-L1 , Fosfatidilinositol 3-Quinases , Diferenciação CelularRESUMO
Preadipocytes can give rise to either white adipocytes or beige adipocytes. Owing to their distinct abilities in nutrient storage and energy expenditure, strategies that specifically promote "beiging" of adipocytes hold great promise for counterbalancing obesity and metabolic diseases. Yet, factors dictating the differentiation fate of adipocyte progenitors remain to be elucidated. We found that stearoyl-coenzyme A desaturase 1 (Scd1)-deficient mice, which resist metabolic stress, possess augmentation in beige adipocytes under basal conditions. Deletion of Scd1 in mature adipocytes expressing Fabp4 or Ucp1 did not affect thermogenesis in mice. Rather, Scd1 deficiency shifted the differentiation fate of preadipocytes from white adipogenesis to beige adipogenesis. Such effects are dependent on succinate accumulation in adipocyte progenitors, which fuels mitochondrial complex II activity. Suppression of mitochondrial complex II by Atpenin A5 or oxaloacetic acid reverted the differentiation potential of Scd1-deficient preadipocytes to white adipocytes. Furthermore, supplementation of succinate was found to increase beige adipocyte differentiation both in vitro and in vivo. Our data reveal an unappreciated role of Scd1 in determining the cell fate of adipocyte progenitors through succinate-dependent regulation of mitochondrial complex II.
Assuntos
Complexo II de Transporte de Elétrons/metabolismo , Gorduras/metabolismo , Obesidade/enzimologia , Estearoil-CoA Dessaturase/genética , Ácido Succínico/metabolismo , Adipócitos Bege/citologia , Adipócitos Bege/metabolismo , Adipogenia , Animais , Metabolismo Energético , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Estearoil-CoA Dessaturase/metabolismo , TermogêneseRESUMO
KLF7 belongs to the Krüppel-like factors (KLFs) family, which function as transcriptional regulators controlling a number of basic cellular processes, involving proliferation, differentiation, and migration. Here, we reveal insights into the differentiated expression of KLF7 in different goat tissues and different stages of growth, and the inhibition role of KLF7 knockdown to differentiation by using goat intramuscular and subcutaneous preadipocytes. We demonstrate that KLF7 expression is obviously changed during the differentiation of preadipocytes into mature adipocytes. Knockdown of KLF7 inhibited lipid droplet accumulation, reduced the expression of adipogenic markers both in intramuscular and subcutaneous preadipocytes in goats, suggesting that KLF7 is a novel regulator of adipogenesis. KLF7 expression changed also up or down-regulation the other KLF family members, but there were differences between these two types of cells. Investigation into the mechanism that KLF7 regulates preadipocyte differentiation revealed that KLF family members KLF1, KLF5, KLF6, KLF8, KLF11, KLF12, KLF16, KLF17 and adipogenic markers C/EBPα and SREBP1 promoter region present KLF7 transcriptional binding sites. Altogether, the data here identify KLF7 as a novel regulator of adipogenesis.