Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (113)

Search Parameters:
Keywords = lipid droplet-associated genes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 8196 KiB  
Article
Dual Modulation of Adipogenesis and Apoptosis by PPARG Agonist Rosiglitazone and Antagonist Betulinic Acid in 3T3-L1 Cells
by Patsawee Sriboonaied, Pornwipa Phuangbubpha, Puretat Saetan, Purin Charoensuksai and Adisri Charoenpanich
Biomedicines 2025, 13(6), 1340; https://doi.org/10.3390/biomedicines13061340 - 30 May 2025
Viewed by 726
Abstract
Background/Objectives: Disruptions in adipose tissue dynamics contribute to obesity-related metabolic disorders, emphasizing the need for targeted therapies focusing on adipose tissue cells, including progenitor cells and adipocytes. Peroxisome proliferator-activated receptor gamma (PPARG) ligands are potent insulin sensitizers used in type 2 diabetes treatment. [...] Read more.
Background/Objectives: Disruptions in adipose tissue dynamics contribute to obesity-related metabolic disorders, emphasizing the need for targeted therapies focusing on adipose tissue cells, including progenitor cells and adipocytes. Peroxisome proliferator-activated receptor gamma (PPARG) ligands are potent insulin sensitizers used in type 2 diabetes treatment. This study investigated the effects of rosiglitazone, a PPARG agonist, and betulinic acid, a PPARG antagonist, on adipogenesis and apoptosis in 3T3-L1 pre-adipocytes. Method: 3T3-L1 pre-adipocytes were treated with rosiglitazone or betulinic acid during adipogenic differentiation. Lipid droplet formation was used to evaluate adipogenesis. Cell growth and cell death were assessed using the resazurin-based cell viability assay, trypan blue exclusion assay, LDH assay, and Annexin V/PI staining. Quantitative PCR was conducted to examine the expression of genes associated with adipogenesis and apoptosis. Results: Betulinic acid reduced adipogenesis only when administered daily for eight days. Rosiglitazone did not alter the overall lipid quantity; however, it promoted a shift toward fewer but larger lipid droplets. Both compounds increased Adipoq and Cfd expression, and betulinic acid also elevated Fabp4. Rosiglitazone induced stronger cell aggregation. Despite increased cell death, overall viability was maintained. Apoptotic cell death was enhanced by both compounds and confirmed via Annexin V/PI staining and flow cytometry, accompanied by downregulation of Ccnd1 and Bcl2. Additionally, rosiglitazone markedly increased the expression of Cebpa, a key regulator that can modulate lipid droplet formation and the balance between cell growth and death. Conclusions: Rosiglitazone and betulinic acid differentially modulate adipogenesis and apoptosis in 3T3-L1 cells, revealing a complex interplay between lipid accumulation and programmed cell death. Together, the findings underscore the potential of dual PPARG-targeting approaches for metabolic disease interventions. Full article
(This article belongs to the Special Issue PPARs in Health and Disease, 2nd Edition)
Show Figures

Figure 1

16 pages, 4010 KiB  
Article
Lipidomic Profiling Reveals HSD17B13 Deficiency-Associated Dysregulated Hepatic Phospholipid Metabolism in Aged Mice
by Cong Zhang, Yingxin Feng, Xiaoyan Zhang, Youfei Guan and Wen Su
Metabolites 2025, 15(6), 353; https://doi.org/10.3390/metabo15060353 - 27 May 2025
Viewed by 757
Abstract
Objectives: HSD17B13 (17β-hydroxysteroid dehydrogenase 13), a lipid droplet-associated enzyme, has emerged as a key regulator of hepatic lipid metabolism and a potential therapeutic target for metabolic-associated fatty liver disease (MAFLD). While its role in lipid homeostasis and liver inflammation has been partially revealed, [...] Read more.
Objectives: HSD17B13 (17β-hydroxysteroid dehydrogenase 13), a lipid droplet-associated enzyme, has emerged as a key regulator of hepatic lipid metabolism and a potential therapeutic target for metabolic-associated fatty liver disease (MAFLD). While its role in lipid homeostasis and liver inflammation has been partially revealed, the impact of HSD17B13 deficiency on lipid metabolism in aged mice remains poorly understood. In this study, we performed comprehensive lipidomic profiling of liver tissues from aged Hsd17b13 gene knockout (Hsd17b13 KO) mice to investigate the effects of Hsd17b13 deletion on hepatic lipid composition and metabolic pathways. Methods: Changes in hepatic lipid profiles were assessed through a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipidomic analysis. Results: The lipid profiles, including triglycerides (TGs), diglycerides (DGs), phosphatidylcholines (PCs), phosphatidylethanolamines (PEs), phosphatidylglycerols (PGs), and ceramides (Cers), exhibited notable alterations in the Hsd17b13 KO mice. Conclusions: HSD17B13 plays a pivotal role in liver lipid metabolism during aging, and it is involved in the regulation of hepatic phospholipid metabolism. Our study highlights the importance of HSD17B13 in maintaining liver lipid homeostasis and its potential as a therapeutic target for age-related liver diseases. Full article
(This article belongs to the Special Issue Metabolic Dysregulation in Fatty Liver Disease)
Show Figures

Figure 1

18 pages, 15168 KiB  
Article
Impact of Drp1 Loss on Organelle Interaction, Metabolism, and Inflammation in Mouse Liver
by Lixiang Wang, Seiji Nomura, Nao Hasuzawa, Sadaki Yokota, Ayako Nagayama, Kenji Ashida, Junjiro Rikitake, Yoshinori Moriyama, Masatoshi Nomura and Ken Yamamoto
Cells 2025, 14(10), 679; https://doi.org/10.3390/cells14100679 - 8 May 2025
Viewed by 761
Abstract
Dynamin-related protein 1 (Drp1) is a crucial player in mitochondrial fission and liver function. The interactions between mitochondria, endoplasmic reticulum (ER), and lipid droplets (LDs) are fundamental for lipid metabolism. This study utilized liver-specific Drp1 knockout (Drp1LiKO) mice to investigate the [...] Read more.
Dynamin-related protein 1 (Drp1) is a crucial player in mitochondrial fission and liver function. The interactions between mitochondria, endoplasmic reticulum (ER), and lipid droplets (LDs) are fundamental for lipid metabolism. This study utilized liver-specific Drp1 knockout (Drp1LiKO) mice to investigate the effects of Drp1 deficiency on organelle interactions, metabolism, and inflammation. Our analysis revealed disrupted interactions between mitochondria and LDs, as well as altered interactions among ER, mitochondria, and LDs in Drp1LiKO mice. Through mass spectrometry and microarray analysis, we identified changes in lipid profiles and perturbed expression of lipid metabolism genes in the livers of Drp1LiKO mice. Further in vitro experiments using primary hepatocytes from Drp1LiKO mice confirmed disturbances in lipid metabolism and increased inflammation. These findings highlight the critical involvement of Drp1 in regulating organelle interactions for efficient lipid metabolism and overall liver health. Targeting Drp1-mediated organelle interactions may offer potential for developing therapies for liver diseases associated with disrupted lipid metabolism. Full article
Show Figures

Graphical abstract

25 pages, 31779 KiB  
Article
Transcriptomic Analysis of Muscle Satellite Cell Regulation on Intramuscular Preadipocyte Differentiation in Tan Sheep
by Xiaochun Xu, Cong Zhan, Jiaqi Qiao, Yuxuan Yang, Changyuan Li, Pan Li and Sen Ma
Int. J. Mol. Sci. 2025, 26(7), 3414; https://doi.org/10.3390/ijms26073414 - 5 Apr 2025
Cited by 2 | Viewed by 626
Abstract
Intramuscular fat (IMF) content is a key factor influencing meat properties including tenderness, flavor, and marbling. However, the complex molecular mechanisms regulating IMF deposition, especially the interactions between intramuscular preadipocytes (IMAdCs) and skeletal muscle satellite cells (SMSCs), remain unclear. In this study, a [...] Read more.
Intramuscular fat (IMF) content is a key factor influencing meat properties including tenderness, flavor, and marbling. However, the complex molecular mechanisms regulating IMF deposition, especially the interactions between intramuscular preadipocytes (IMAdCs) and skeletal muscle satellite cells (SMSCs), remain unclear. In this study, a direct co-culture system of sheep IMAdCs and SMSCs was used to elucidate their intercellular interactions. RNA sequencing and bioinformatics analyses were performed under monoculture and co-culture conditions for later stages of differentiation. The obtained results showed that SMSCs significantly inhibited the adipogenic capacity of IMAdCs. This was reflected in the co-culture markedly altered gene expression and observations of lipid droplets in our studies, i.e., the PPARG, ACOX2, PIK3R1, FABP5, FYN, ALDOC, PFKM, PFKL, HADH, and HADHB genes were down-regulated in the co-cultured IMAdCs in association with the inhibition of fat deposition, whereas ACSL3, ACSL4, ATF3, EGR1, and IGF1R within the genes upregulated in co-culture IMAdCs were associated with the promotion of lipid metabolism. In addition, GO, KEGG, and ligand–receptor pairing analyses further elucidated the molecular mechanisms of intercellular communication. These findings emphasize the regulatory role of SMSCs on intramuscular preadipocyte differentiation and lipid metabolism, providing a theoretical framework for targeted molecular strategies to improve sheep meat quality. Full article
Show Figures

Figure 1

23 pages, 5019 KiB  
Article
Core Molecular Clock Factors Regulate Osteosarcoma Stem Cell Survival and Behavior via CSC/EMT Pathways and Lipid Droplet Biogenesis
by Sukanya Bhoumik and Yool Lee
Cells 2025, 14(7), 517; https://doi.org/10.3390/cells14070517 - 31 Mar 2025
Cited by 1 | Viewed by 1075
Abstract
The circadian clock, an intrinsic 24 h cellular timekeeping system, regulates fundamental biological processes, including tumor physiology and metabolism. Cancer stem cells (CSCs), a subpopulation of cancer cells with self-renewal and tumorigenic capacities, are implicated in tumor initiation, recurrence, and metastasis. Despite growing [...] Read more.
The circadian clock, an intrinsic 24 h cellular timekeeping system, regulates fundamental biological processes, including tumor physiology and metabolism. Cancer stem cells (CSCs), a subpopulation of cancer cells with self-renewal and tumorigenic capacities, are implicated in tumor initiation, recurrence, and metastasis. Despite growing evidence for the circadian clock’s involvement in regulating CSC functions, its precise regulatory mechanisms remain largely unknown. Here, using a human osteosarcoma (OS) model (143B), we have shown that core molecular clock factors are critical for OS stem cell survival and behavior via direct modulation of CSC and lipid metabolic pathways. In single-cell-derived spheroid formation assays, 143B OS cells exhibited robust spheroid-forming capacity under 3D culture conditions. Furthermore, siRNA-mediated depletion of core clock components (i.e., BMAL1, CLOCK, CRY1/2, PER1/2)—essential positive and negative elements of the circadian clock feedback loop—significantly reduced spheroid formation in 143B CSCs isolated from in vivo OS xenografts. In contrast, knockdown of the secondary clock-stabilizing factor genes NR1D1 and NR1D2 had little effect. We also found that knockdown of BMAL1, CLOCK, or CRY1/2 markedly impaired the migration and invasion capacities of 143B CSCs. At the molecular level, silencing of BMAL1, CLOCK, or CRY1/2 distinctly altered the expression of genes associated with stem cell properties and the epithelial–mesenchymal transition (EMT) in 143B CSCs. In addition, disruption of BMAL1, CLOCK, or CRY1/2 expression significantly reduced lipid droplet formation by downregulating the expression of genes involved in lipogenesis (e.g., DGAT1, FASN, ACSL4, PKM2, CHKA, SREBP1), which are closely linked to CSC/EMT processes. Furthermore, transcriptomic analysis of human OS patient samples revealed that compared with other core clock genes, CRY1 was highly expressed in OS tumors relative to controls, and its expression exhibited strong positive correlations with patient prognosis, survival, and LD biogenesis gene expression. These findings highlight the critical role of the molecular circadian clock in regulating CSC properties and metabolism, underscoring the therapeutic potential of targeting the core clock machinery to enhance OS treatment outcomes. Full article
(This article belongs to the Special Issue The Role of Stem Cells and Circadian Clock in Cancer Immunotherapy)
Show Figures

Figure 1

14 pages, 2648 KiB  
Review
CSN-CRL Complexes: New Regulators of Adipogenesis
by Dawadschargal Dubiel, Michael Naumann and Wolfgang Dubiel
Biomolecules 2025, 15(3), 372; https://doi.org/10.3390/biom15030372 - 5 Mar 2025
Viewed by 957
Abstract
Recent discoveries revealed mechanistic insights into the control of adipogenesis by the Constitutive Photomorphogenesis 9 Signalosome (CSN) and its variants, CSNCSN7A and CSNCSN7B, which differ in the paralog subunits, CSN7A and CSN7B. CSNCSN7A and CSNCSN7B variants form permanent [...] Read more.
Recent discoveries revealed mechanistic insights into the control of adipogenesis by the Constitutive Photomorphogenesis 9 Signalosome (CSN) and its variants, CSNCSN7A and CSNCSN7B, which differ in the paralog subunits, CSN7A and CSN7B. CSNCSN7A and CSNCSN7B variants form permanent complexes with cullin-RING-ubiquitin ligases 3 and 4A (CRL3 and CRL4A), respectively. These complexes can be found in most eukaryotic cells and represent a critical reservoir for cellular functions. In an early stage of adipogenesis, mitotic clonal expansion (MCE), CSN-CRL1, and CSNCSN7B-CRL4A are blocked to ubiquitinate the cell cycle inhibitor p27KIP, leading to cell cycle arrest. In addition, in MCE CSN-CRL complexes rearrange the cytoskeleton for adipogenic differentiation and CRL3KEAP1 ubiquitylates the inhibitor of adipogenesis C/EBP homologous protein (CHOP) for degradation by the 26S proteasome, an adipogenesis-specific proteolysis. During terminal adipocyte differentiation, the CSNCSN7A-CRL3 complex is recruited to a lipid droplet (LD) membrane by RAB18. Currently, the configuration of the substrate receptors of CSNCSN7A-CRL3 on LDs is unclear. CSNCSN7A-CRL3 is activated by neddylation on the LD membrane, an essential adipogenic step. Damage to CSN/CUL3/CUL4A genes is associated with diverse diseases, including obesity. Due to the tremendous impact of CSN-CRLs on adipogenesis, we need strategies for adequate treatment in the event of malfunctions. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

24 pages, 7291 KiB  
Article
ACAA2 Protects Against Cardiac Dysfunction and Lipid Peroxidation in Renal Insufficiency with the Treatment of S-Nitroso-L-Cysteine
by Zhengqi Xu, Feng Jiang, Xiaofan Wu, Bowen Ren, Cuntai Zhang, Li Lin and Sheng Li
Biomolecules 2025, 15(3), 364; https://doi.org/10.3390/biom15030364 - 3 Mar 2025
Cited by 1 | Viewed by 1256
Abstract
The key fatty acid β-oxidation protein acetyl-CoA acyltransferase 2 (ACAA2) plays a significant role in myocardial lipid peroxidation and cardiac dysfunction induced by renal insufficiency. However, the mechanisms of lipid metabolism related to renal insufficiency-associated cardiac dysfunction remain poorly understood, and current clinical [...] Read more.
The key fatty acid β-oxidation protein acetyl-CoA acyltransferase 2 (ACAA2) plays a significant role in myocardial lipid peroxidation and cardiac dysfunction induced by renal insufficiency. However, the mechanisms of lipid metabolism related to renal insufficiency-associated cardiac dysfunction remain poorly understood, and current clinical treatments have been largely ineffective. Through analysis of the Gene Expression Omnibus (GEO) database, we identified that the cardiac functional changes caused by renal insufficiency were primarily centered around the fatty acid β-oxidation signaling pathway, where ACAA2 plays a pivotal role in fatty acid β-oxidation, the tricarboxylic acid cycle, and ketone body metabolism. In an adenine-induced renal insufficiency mouse model, further examination with hematoxylin-eosin staining, Masson staining, and Oil Red O staining revealed alterations in the heart and kidney as well as the accumulation of lipid. Non-invasive blood pressure measurements and ultrasound images demonstrated improvements of peripheral vascular and right ventricular hemodynamic parameters with S-nitroso-L-cysteine (CSNO) inhalation therapy. In cell experiments, knocking down ACAA2 led to accumulation of lipid droplets and exacerbation of oxidative stress in cardiomyocytes, while overexpression of ACAA2 reversed these effects. The transcription factor FOXO4 was found to regulate lipid peroxidation by modulating ACAA2, and knocking down FOXO4 partially restored the expression of ACAA2, reducing oxidative stress in cardiomyocytes. Furthermore, exogenous CSNO effectively restored the expression of ACAA2 and reduced the level of FOXO4, thereby mitigating lipid peroxidation and improving cardiac function. Therefore, in the context of renal insufficiency, regulating the FOXO4–ACAA2 axis through CSNO inhalation therapy may provide a novel therapeutic strategy for alleviating myocardial lipid peroxidation and improving cardiac function. Full article
(This article belongs to the Special Issue Recent Trends in Kidney and Cardiovascular Diseases)
Show Figures

Graphical abstract

15 pages, 5221 KiB  
Article
Phosphatidylcholine Cytidine Transferase α (CCTα) Affects LD Formation Through Fusion and Lipophagy in Bovine Mammary Epithelial Cells
by Jingna Yang, Yuxin Fan, Fangyuan Kang, Yanbin Yang, Yueying Wang, Yang Liu and Liqiang Han
Int. J. Mol. Sci. 2025, 26(5), 2135; https://doi.org/10.3390/ijms26052135 - 27 Feb 2025
Viewed by 660
Abstract
Phosphatidylcholine cytidine transferase α (CCTα) is a key rate-limiting enzyme in the CDP–choline pathway, the primary pathway for phosphatidylcholine (PC) synthesis in mammals. This study investigated the role of CCTα in lipid droplet (LD) formation, phospholipid synthesis, LD fusion, and lipophagy in bovine [...] Read more.
Phosphatidylcholine cytidine transferase α (CCTα) is a key rate-limiting enzyme in the CDP–choline pathway, the primary pathway for phosphatidylcholine (PC) synthesis in mammals. This study investigated the role of CCTα in lipid droplet (LD) formation, phospholipid synthesis, LD fusion, and lipophagy in bovine mammary epithelial cells (BMECs) through CCTα gene knockout (CCT-KO) and overexpression (CCT-OE). CCTα mRNA expression was significantly increased in bovine mammary gland tissue after lactation. In BMECs, CCTα was transferred from the nucleus to the endoplasmic reticulum and localized on LD surfaces in the presence of linoleic acid. Compared with normal BMECs (NC), CCTα knockout (CCT-KO) cells had significantly greater LD diameters (1.53 μm vs. 1.68 μm, p < 0.05), lower proportions of small LDs (<1 µm; 11.39% vs. 5.42%), and higher proportions of large LDs (>3 µm; 0.67% vs. 2.88%). In contrast, CCTα overexpression (CCT-OE) decreased the diameter of LDs to 1.18 μm (p < 0.01), increased the proportion of small LDs to 35.48%, and decreased the proportion of large LDs to 0.24%. CCTα knockout significantly decreased the PC content and the ratio of PC to PE, whereas CCTα overexpression increased the PC content and the ratio of PC to phosphatidyl ethanolamine (PE) (p < 0.05). The lipidomics analysis indicated that PC synthesis was significantly influenced by CCTα gene expression. Live cell observations showed that CCTα knockout promoted the fusion of small LDs into large LDs. In cells with CCT α overexpression, the expression of the microtubule-associated protein 1 light chain 3 (LC3) protein and the number of lysosomes was elevated, and the lysosomal phagocytosis of LDs was observed through transmission electron microscopy, thus indicating that CCTα overexpression enhanced lipophagy. In conclusion, these results suggest that CCTα plays a role in regulating LD formation by influencing PC synthesis, LD fusion, and lipophagy in BMECs. Full article
(This article belongs to the Special Issue Lipid Droplet Proteins Function and Metabolism)
Show Figures

Graphical abstract

11 pages, 2102 KiB  
Article
STEAP3 Inhibits Porcine Reproductive and Respiratory Syndrome Virus Replication by Regulating Fatty Acid and Lipid Droplet Synthesis
by Chenyang Yuan, Kaifeng Guan and Gaiping Zhang
Vet. Sci. 2025, 12(2), 147; https://doi.org/10.3390/vetsci12020147 - 8 Feb 2025
Viewed by 1361
Abstract
Porcine Reproductive and Respiratory Syndrome (PRRS) is a contagious disease that impacts swine health worldwide. Lipid metabolism plays a vital role in energy production and is regulated by various genes involved in lipogenesis and lipolysis. In this study, we found that PRRSV infection [...] Read more.
Porcine Reproductive and Respiratory Syndrome (PRRS) is a contagious disease that impacts swine health worldwide. Lipid metabolism plays a vital role in energy production and is regulated by various genes involved in lipogenesis and lipolysis. In this study, we found that PRRSV infection significantly reduced the protein expression of STEAP3. The overexpression of STEAP3 can notably inhibit PRRSV replication. Additionally, we utilized transcriptomics and metabolomics to examine the effects of STEAP3 on PRRSV replication, identifying important pathways associated with energy metabolism and lipogenesis. We subsequently found that STEAP3 can suppress PRRSV replication by regulating fatty acid synthesis and enhancing lipid droplet formation. Overall, these findings indicate that STEAP3 could be a potential target for developing strategies to manage PRRSV infection by modulating lipid metabolism. Full article
Show Figures

Figure 1

19 pages, 3696 KiB  
Article
Coriandrum sativum L. Leaf Extract Ameliorates Metabolic Dysfunction-Associated Steatotic Liver Disease by Modulating the AMPK Pathway in High Fat-Fed C57BL/6 Mice
by Min Ji Gu, Yejin Ahn, Yu Ra Lee, Guijae Yoo, Yoonsook Kim, Inwook Choi, Sang Keun Ha and Donghwan Kim
Nutrients 2024, 16(23), 4165; https://doi.org/10.3390/nu16234165 - 30 Nov 2024
Cited by 1 | Viewed by 1256
Abstract
Background: Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. In recent times, the term NAFLD has been modified to metabolic dysfunction-associated steatotic liver disease (MASLD), reflecting its comprehensive scope encompassing a range of metabolic abnormalities. Coriandrum sativum L. (CS) [...] Read more.
Background: Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. In recent times, the term NAFLD has been modified to metabolic dysfunction-associated steatotic liver disease (MASLD), reflecting its comprehensive scope encompassing a range of metabolic abnormalities. Coriandrum sativum L. (CS) is a traditional medicine, although the preventive mechanism of CS extracts remains unclear. Objective: This study evaluated the preventive effects of CS in high-fat diet (HFD)-induced MASLD mice by oral administration of 100 or 200 mg/kg/day of CS extracts for 12 weeks. Results: The major CS extract compounds were chlorogenic acid, caffeic acid, rutin, and isoquercetin. The administration of CS extract suppressed HFD-induced weight gain, liver weight, and the liver/body weight ratio. It improved the mice’s serum biological profiles and suppressed HFD-induced lipid droplet and lipid accumulation by inhibiting lipid accumulation-related gene expression in the liver. It modulated HFD-induced Ampk-Srebp1c pathways and suppressed HFD-induced NF-κB pathway activation in the liver. It regulated inflammation and the AMPK alpha signaling pathway in HFD-fed mice by reducing the accumulation of specific amino acids, leading to the amelioration of fatty liver. Conclusions: The CS extract prevents HFD-induced MASLD and may help prevent or treat MASLD. Full article
(This article belongs to the Special Issue Natural Products and Human Health)
Show Figures

Figure 1

16 pages, 5425 KiB  
Article
Integrative Transcriptomics and Proteomics Analysis Reveals THRSP’s Role in Lipid Metabolism
by Yujie Li, Ke Xu, Ao Zhou, Zhong Xu, Junjing Wu, Xianwen Peng, Shuqi Mei and Hongbo Chen
Genes 2024, 15(12), 1562; https://doi.org/10.3390/genes15121562 - 30 Nov 2024
Viewed by 1914
Abstract
Background/Objectives: Abnormalities in lipid metabolism and endoplasmic reticulum (ER) stress are strongly associated with the development of a multitude of pathological conditions, including nonalcoholic fatty liver disease (NAFLD), diabetes mellitus, and obesity. Previous studies have indicated a potential connection between thyroid hormone responsive [...] Read more.
Background/Objectives: Abnormalities in lipid metabolism and endoplasmic reticulum (ER) stress are strongly associated with the development of a multitude of pathological conditions, including nonalcoholic fatty liver disease (NAFLD), diabetes mellitus, and obesity. Previous studies have indicated a potential connection between thyroid hormone responsive (THRSP) and lipid metabolism and that ER stress may participate in the synthesis of key regulators of adipogenesis. However, the specific mechanisms remain to be investigated. Methods: In this study, we explored the roles of THRSP in lipid metabolism by interfering with THRSP gene expression in mouse mesenchymal stem cells, comparing the effects on adipogenesis between control and interfered groups, and by combining transcriptomic and proteomic analysis. Results: Our results showed that the number of lipid droplets was significantly reduced after interfering with THRSP, and the expression levels of key regulators of adipogenesis, such as LPL, FABP4, PLIN1, and CIDEC, were significantly downregulated. Both transcriptomic and proteomic results showed that the differential genes (proteins) were enriched in the processes of lipolytic regulation, ER stress, cholesterol metabolism, sphingolipid metabolism, PPAR signaling pathway, and glycerophospholipid metabolism. The ER stress marker gene, ATF6, was the most significantly downregulated transcription factor. In addition, RT-qPCR validation indicated that the expression levels of PPAR signaling pathway gene SCD1; key genes of lipid droplet generation including LIPE, DGAT1, and AGPAT2; and ER stress marker gene ATF6 were significantly downregulated. Conclusions: These suggest that THRSP is involved in regulating ER stress and the PPAR signaling pathway, which is closely related to lipid synthesis and metabolism. Interfering with the expression of THRSP may be helpful in ameliorating the occurrence of diseases related to abnormalities in lipid metabolism. Full article
(This article belongs to the Special Issue Functional Genomics and Breeding of Animals)
Show Figures

Figure 1

15 pages, 5181 KiB  
Article
Deciphering the Role of the SREBF1 Gene in the Transcriptional Regulation of Porcine Adipogenesis Using CRISPR/Cas9 Editing
by Mehmet Onur Aksoy, Adrianna Bilinska, Monika Stachowiak, Tatiana Flisikowska and Izabela Szczerbal
Int. J. Mol. Sci. 2024, 25(23), 12677; https://doi.org/10.3390/ijms252312677 - 26 Nov 2024
Cited by 2 | Viewed by 1793
Abstract
Sterol regulatory element-binding protein 1 (SREBP1) is an important transcription factor that controls lipid metabolism and adipogenesis. Two isoforms, SREBP1a and SREBP1c, are generated by alternative splicing of the first exon of the SREBF1 gene. The porcine SREBF1 gene has mainly been studied [...] Read more.
Sterol regulatory element-binding protein 1 (SREBP1) is an important transcription factor that controls lipid metabolism and adipogenesis. Two isoforms, SREBP1a and SREBP1c, are generated by alternative splicing of the first exon of the SREBF1 gene. The porcine SREBF1 gene has mainly been studied for its role in lipid metabolism in adipose tissues, but little is known about its involvement, and the role of its two isoforms, in adipogenesis. The aim of the present study was to introduce a deletion in the 5′-regulatory region of the SREBF1c gene, considered crucial for adipogenesis, using the Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9 (CRISPR/Cas9) method. This approach allows for the evaluation of how inhibiting SREBF1c transcription affects the expression of other genes essential for adipocyte differentiation, particularly PPARG, CEBPA, CEBPB, CEBPD, GATA2, and FABP4. It was observed that disrupting the SREBF1c isoform had no effect on the GATA2 gene but did result in a decrease in the expression of the CEBPA and CEBPD genes, an increase in the expression of CEBPB, and an inhibition in the expression of the PPARG and FABP4 genes. These changes in gene expression blocked adipogenesis, as could be seen by the failure of lipid droplets to accumulate. Our results provide evidence highlighting the pivotal role of the SREBP1c isoform in the regulation of porcine adipogenesis. Full article
Show Figures

Figure 1

16 pages, 5113 KiB  
Article
Analysis of Lipid Metabolism in Adipose Tissue and Liver of Chinese Soft-Shelled Turtle Pelodiscus sinensis During Hibernation
by Feng Jin, Yunfei You, Junliang Wan, Huaiyi Zhu, Kou Peng, Zhenying Hu, Qi Zeng, Beijuan Hu, Junhua Wang, Jingjing Duan and Yijiang Hong
Int. J. Mol. Sci. 2024, 25(22), 12124; https://doi.org/10.3390/ijms252212124 - 12 Nov 2024
Cited by 2 | Viewed by 1310
Abstract
Hibernation serves as an energy-conserving strategy that enables animals to withstand harsh environments by reducing their metabolic rate significantly. However, the mechanisms underlying energy adaptation in hibernating ectotherms, such as Pelodiscus sinensis, remain contentious. This paper first reports the decrease in lipid [...] Read more.
Hibernation serves as an energy-conserving strategy that enables animals to withstand harsh environments by reducing their metabolic rate significantly. However, the mechanisms underlying energy adaptation in hibernating ectotherms, such as Pelodiscus sinensis, remain contentious. This paper first reports the decrease in lipid levels and the expression of metabolism-related genes in P. sinensis during hibernation. The results of physiological and biochemical analysis showed that adipocyte cell size was reduced and liver lipid droplet (LD) contents were decreased during hibernation in P. sinensis. Concurrently, serum levels of triglycerides (TGs), total cholesterol (TC), non-esterified fatty acids (NEFAs), high-density lipoprotein cholesterol (HDLC), and low-density lipoprotein cholesterol (LDLC) were diminished (n = 8, p < 0.01), while an increase in serum glucose (Glu) (n = 8, p < 0.01) was noted among hibernating P. sinensis. These observations suggest a shift in energy metabolism during hibernation. To gain insights into the molecular mechanisms, we performed integrated transcriptomic and lipidomic analyses of adipose tissue and livers from summer-active versus overwintering P. sinensis, which revealed downregulation of free fatty acids (FFAs), triglycerides (TGs), diglycerides (DGs), and ceramides (Cers) during hibernation. The results of GSEA analysis showed that metabolic pathways associated with lipid metabolism, including glycerolipid metabolism and regulation of lipolysis in adipocytes, were suppressed significantly. Notably, acute cold exposure induced significant downregulation of genes related to lipolysis such as PNPLA2, ABHD5, LPL, CPT1A, and PPARα. The results indicate that lipolysis is suppressed during hibernation in P. sinensis. Collectively, these findings deepen our understanding of survival mechanisms and elucidate the unique energy adaptation strategies employed by hibernating ectotherms. Future research should explore the implications of these findings for the conservation of ectotherms and the applications for artificially inducing hibernation. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

15 pages, 17178 KiB  
Article
The IL-33/ST2 Axis Affects Adipogenesis Through Regulating the TRAF6/RelA Pathway
by Shujun Cao, Xuyong Qin, Chengping Li, Lichun Zhang, Shizhong Ren, Wenhao Zhou, Meiman Zhao and Guoli Zhou
Int. J. Mol. Sci. 2024, 25(22), 12005; https://doi.org/10.3390/ijms252212005 - 8 Nov 2024
Viewed by 1287
Abstract
Understanding the regulatory mechanisms of adipogenesis is essential for preventing obesity. Interleukin-33 (IL-33) has recently attracted increasing attention for its role in adipogenesis. The purpose of this study was to explore the function and regulatory mechanism of IL-33 and its receptor suppression of [...] Read more.
Understanding the regulatory mechanisms of adipogenesis is essential for preventing obesity. Interleukin-33 (IL-33) has recently attracted increasing attention for its role in adipogenesis. The purpose of this study was to explore the function and regulatory mechanism of IL-33 and its receptor suppression of tumorigenicity 2 (ST2) on adipogenesis. Here, Oil Red O staining was used to detect the accumulation of intracellular lipid droplets. Molecular techniques such as qRT-PCR and Western blotting were used to detect the expression of pivotal genes and adipogenic marker genes. Gains and losses of function experiments were used to explore the potential regulatory mechanism of the IL-33/ST2 axis in adipogenesis. Functionally, IL-33 is negatively associated with adipogenesis in 3T3-L1 preadipocytes, while ST2 is positively associated with it, encompassing both the trans-membrane receptor ST2 (ST2L) and the soluble ST2 (sST2). Mechanistically, the IL-33/ST2 axis affects adipogenesis by regulating the expression of the TRAF6/RelA pathway in 3T3-L1 preadipocytes. Downregulating the expression of ST2 suppressed the activation of the IL-33/ST2 axis, which subsequently inhibits the expression of TRAF6. This further attenuates the expression of RelA, ultimately resulting in the suppression of adipogenesis in 3T3-L1 preadipocytes. This study reveals a new mechanism by which the IL-33/ST2 axis regulates the differentiation of preadipocytes and provides a new idea for improving obesity prevention. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

17 pages, 4138 KiB  
Article
Apolipoprotein E Induces Lipid Accumulation Through Dgat2 That Is Prevented with Time-Restricted Feeding in Drosophila
by Ruan C. M. Moraes, Jonathan R. Roth, Hailey Mao, Savannah R. Crawley, Brittney P. Xu, John C. Watson and Girish C. Melkani
Genes 2024, 15(11), 1376; https://doi.org/10.3390/genes15111376 - 25 Oct 2024
Cited by 2 | Viewed by 1874
Abstract
Background: Apolipoprotein E (ApoE) is the leading genetic risk factor for late-onset Alzheimer’s disease (AD), which is the leading cause of dementia worldwide. Most people have two ApoE-ε3 (ApoE3) alleles, while ApoE-ε2 (ApoE2) is protective from AD, and ApoE-ε4 (ApoE4) confers AD risk. [...] Read more.
Background: Apolipoprotein E (ApoE) is the leading genetic risk factor for late-onset Alzheimer’s disease (AD), which is the leading cause of dementia worldwide. Most people have two ApoE-ε3 (ApoE3) alleles, while ApoE-ε2 (ApoE2) is protective from AD, and ApoE-ε4 (ApoE4) confers AD risk. How these alleles modulate AD risk is not clearly defined, and ApoE’s role in lipid metabolism is also not fully known. Lipid droplets increase in AD. However, how ApoE contributes to lipid accumulation in the brain remains unknown. Methods: Here, we use Drosophila to study the effects of ApoE alleles on lipid accumulation in the brain and muscle in a cell-autonomous and non-cell-autonomous manner. Results: We report that pan-neuronal expression of each ApoE allele induces lipid accumulation specifically in the brain, but not in the muscle. However, this was not the case when expressed with muscle-specific drivers. ApoE2- and ApoE3-induced lipid accumulation is dependent on the expression of Dgat2, a key regulator of triacylglycerol production, while ApoE4 still induces lipid accumulation even with knock-down of Dgat2. Additionally, we find that implementation of time-restricted feeding (TRF), a dietary intervention in which food access only occurs in the active period (day), prevents ApoE-induced lipid accumulation in the brain of flies and modulates lipid metabolism genes. Conclusions: Altogether, our results demonstrate that ApoE induces lipid accumulation in the brain, that ApoE4 is unique in causing lipid accumulation independent of Dgat2, and that TRF prevents ApoE-induced lipid accumulation. These results support the idea that lipid metabolism is critical in AD, and that TRF could be a promising therapeutic approach to prevent ApoE-associated dysfunction in lipid metabolism. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Graphical abstract

Back to TopTop