Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,328)

Search Parameters:
Keywords = adipogenesis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 2432 KB  
Review
N6-Methyladenosine (m6A)-Mediated Regulation of Lipid Metabolism: Molecular Mechanisms, Pathological Implications, and Therapeutic Perspectives
by Qingjun Zhu, Yunyi Hu, Minhao Li, Haili Yang, Le Zhao and Yongju Zhao
Biomolecules 2026, 16(1), 101; https://doi.org/10.3390/biom16010101 - 7 Jan 2026
Viewed by 198
Abstract
Dysregulated lipid metabolism constitutes the fundamental etiology underlying the global burden of obesity and its associated metabolic disorders. N6-methyladenosine (m6A) is the most abundant reversible chemical modification on messenger RNA and influences virtually every aspect of RNA metabolism. Recent [...] Read more.
Dysregulated lipid metabolism constitutes the fundamental etiology underlying the global burden of obesity and its associated metabolic disorders. N6-methyladenosine (m6A) is the most abundant reversible chemical modification on messenger RNA and influences virtually every aspect of RNA metabolism. Recent studies demonstrate that m6A mediates regulatory networks governing lipid metabolism and contributes to the pathogenesis of multiple metabolic diseases. However, the precise roles of m6A in lipid metabolism and related metabolic disorders remain incompletely understood. This review positions m6A modification as a central epigenetic switch that governs lipid homeostasis. We first summarize the molecular components of the dynamic m6A regulatory machinery and delineate the mechanisms by which it controls key lipid metabolic processes, with an emphasis on adipogenesis, thermogenesis and lipolysis. Building on this, we further discuss how dysregulated m6A acts as a shared upstream driver linking obesity, type 2 diabetes (T2D), metabolic dysfunction-associated steatotic liver disease (MASLD), and insulin resistance through tissue-specific and inter-organ communication mechanisms. We also evaluate the potential of targeting m6A regulators as therapeutic strategies for precision intervention in metabolic diseases. Ultimately, deciphering the complex interplay between m6A modification and lipid homeostasis offers a promising frontier for the development of epitranscriptome-targeted precision medicine against obesity and its associated metabolic disorders. Full article
(This article belongs to the Special Issue Obesity-Related Diseases: Molecular Basis and Therapeutic Approaches)
Show Figures

Graphical abstract

14 pages, 960 KB  
Review
A Comprehensive Review on Medium- and Long-Chain Fatty Acid-Derived Metabolites: From Energy Sources to Metabolic Signals
by Jin-Byung Park, Sungyun Cho and Sung-Joon Lee
Metabolites 2026, 16(1), 45; https://doi.org/10.3390/metabo16010045 - 4 Jan 2026
Viewed by 216
Abstract
Medium- and long-chain fatty acids (MLFAs) are increasingly recognized not only as metabolic substrates but also as precursors of diverse bioactive metabolites generated through host and microbial transformations. Recent advances in analytical chemistry and microbiome research have revealed that gut microorganisms catalyze extensive [...] Read more.
Medium- and long-chain fatty acids (MLFAs) are increasingly recognized not only as metabolic substrates but also as precursors of diverse bioactive metabolites generated through host and microbial transformations. Recent advances in analytical chemistry and microbiome research have revealed that gut microorganisms catalyze extensive modifications of dietary MLFAs—producing hydroxylated, conjugated, and keto-fatty acids with enhanced potency toward host receptors. These metabolites exhibit dual activity on classical metabolic receptors, including FFAR1/4 and PPARα/γ, as well as ectopically expressed chemosensory receptors such as olfactory receptors (ORs) and bitter taste receptors (TAS2Rs). This expanded receptor landscape establishes a previously unrecognized chemosensory–metabolic axis that integrates dietary signals, microbial metabolism, and host physiology. Microbial MLFA derivatives such as 10-hydroxyoctadecenoic acid and conjugated linoleic acid regulate incretin secretion, adipogenesis, macrophage polarization, and intestinal barrier function through coordinated activation of FFARs and PPARs. Concurrently, dicarboxylic acids such as azelaic acid activate Olfr544 to modulate lipolysis, ketogenesis, GLP-1 release, and feeding behavior. TAS2Rs also sense oxidized lipids, linking lipid metabolism to immune regulation and enteroendocrine signaling. Collectively, these pathways highlight the microbiome as a metabolic transducer that converts dietary lipids into signaling molecules influencing endocrine, immune, and gut–brain circuits. Understanding the mechanisms governing MLFA bioconversion and receptor engagement provides new opportunities for therapeutic and nutritional intervention. Targeting ORs and TAS2Rs, engineering probiotics to enhance beneficial FA-derived metabolites, and developing receptor-selective synthetic analogs represent promising strategies. Future progress will require integrative approaches combining physiology, biochemistry, metabolomics, and microbial genomics to elucidate receptor specificity and host variability. Full article
Show Figures

Figure 1

22 pages, 3584 KB  
Article
Effects of Chlorogenic Acid on Cellular Senescence in an In Vitro Model of 3T3-L1 Murine Adipocytes
by Maria Sofia Molonia, Federica Lina Salamone, Santi Trischitta, Antonella Saija, Francesco Cimino and Antonio Speciale
Molecules 2026, 31(1), 167; https://doi.org/10.3390/molecules31010167 - 1 Jan 2026
Viewed by 196
Abstract
Cellular senescence is a stress-induced process that contributes to adipose tissue dysfunction by promoting inflammation, impaired adipogenesis, and insulin resistance, alterations that are closely associated with age-related cellular dysfunction and metabolic disorders. In this study, we evaluated the protective role of chlorogenic acid [...] Read more.
Cellular senescence is a stress-induced process that contributes to adipose tissue dysfunction by promoting inflammation, impaired adipogenesis, and insulin resistance, alterations that are closely associated with age-related cellular dysfunction and metabolic disorders. In this study, we evaluated the protective role of chlorogenic acid (CGA), a polyphenol with known antioxidant and anti-inflammatory properties, against oxidative stress-induced senescence in murine 3T3-L1 adipocytes. The results obtained showed that CGA treatment significantly alleviated the senescent phenotype by restoring Lamin B1 levels and the Bcl-2/Bax ratio. Additionally, CGA downregulated key senescence-related cell cycle progression markers, modulating p53, p21, and MAPK signaling. CGA also restored insulin signaling through the PI3K-AKT-GLUT4 axis and improved glucose uptake, while attenuating oxidative stress, inflammatory cytokine expression, and extracellular matrix remodeling factors associated with SASP. Collectively, these findings support the role of CGA as a promising senotherapeutic nutraceutical able to reduce adipocyte senescence and its metabolic consequences, offering novel insights for the development of dietary supplements targeting age-related cellular dysfunction. Full article
Show Figures

Figure 1

19 pages, 7672 KB  
Article
GATA-3 Suppression by DNAzyme Modulates Interleukin-10 and Liver Injury Markers in db/db Mice
by Layla Al-Mansoori, Asma A. Elashi, Laila Hedaya, Maha Alser, Shamma Almuraikhy, Najeha Anwardeen, Hend Al-Jaber, Suhad Hussain, Hamda A. Al-Naemi, Vijay Govindharajan, Rafif Mahmood Al-Saady, Mohammed Imad Malki, Khaled Naja and Mohamed A. Elrayess
Biology 2026, 15(1), 89; https://doi.org/10.3390/biology15010089 - 31 Dec 2025
Viewed by 229
Abstract
Obesity plays a crucial role in the progression of insulin resistance and type 2 diabetes which are related to inflammation and liver disease. GATA-3 is a transcription factor that is involved in adipogenesis and inflammation. Therefore, it could be a potential therapeutic target [...] Read more.
Obesity plays a crucial role in the progression of insulin resistance and type 2 diabetes which are related to inflammation and liver disease. GATA-3 is a transcription factor that is involved in adipogenesis and inflammation. Therefore, it could be a potential therapeutic target for obesity-associated metabolic disorders. This study aimed to examine the effects of GATA-3 suppression on body weight, fat depot redistribution, liver histopathology, and inflammatory markers in transgenic db/db obese mice. Male db/db mice received subcutaneous injections of GATA-3-specific DNAzyme (hgd40; 10 or 100 µg/mL), pioglitazone (as a positive control), or vehicle only (as a negative control), twice weekly for two weeks. Body weight, organ weights, liver histopathology, mRNA expression of selected genes and serum cytokine levels were assessed. GATA-3 expression was not region specific, and its suppression did not significantly affect fat depot distribution or organ weights. However, the low dose of hgd40 accelerated body weight gain transiently. It also increased Il10 mRNA expression in the liver and significantly increased IL-10 protein concentration in the serum. In addition, a high dose of hgd40 resulted in a marked decrease in hepatocyte ballooning degeneration. These findings suggest that GATA-3 suppression may modulate inflammation and liver injury in obesity, warranting further investigation into its therapeutic potential for obesity-related metabolic disorders. Full article
(This article belongs to the Special Issue Molecular Basis of Metabolic Homeostasis)
Show Figures

Figure 1

24 pages, 9054 KB  
Article
Toward Efficient Beige Adipogenesis: Protocol Optimization Using Adipose-Derived Stem Cells
by Klaudia Simka-Lampa, Agnieszka Kosowska, Wojciech Garczorz, Małgorzata Kimsa-Furdzik, Grzegorz Wystrychowski, Celina Kruszniewska-Rajs, Małgorzata Muc-Wierzgoń and Tomasz Francuz
Cells 2026, 15(1), 54; https://doi.org/10.3390/cells15010054 - 28 Dec 2025
Viewed by 359
Abstract
Brown adipose tissue (BAT) has emerged as a promising therapeutic target for metabolic disorders such as type 2 diabetes and obesity. To advance research on BAT activation and elucidate the mechanisms underlying adipogenesis, it is crucial to develop a reliable in vitro model. [...] Read more.
Brown adipose tissue (BAT) has emerged as a promising therapeutic target for metabolic disorders such as type 2 diabetes and obesity. To advance research on BAT activation and elucidate the mechanisms underlying adipogenesis, it is crucial to develop a reliable in vitro model. This study aimed to optimize the differentiation of adipose-derived stem cells (ADSCs) into beige adipocytes and to validate the protocol using primary human ADSCs obtained from eight donors. Protocol optimization was first performed with commercial ADSCs, testing more than 30 combinations of adipogenic conditions. Differentiation was assessed by microscopy, Oil Red O staining, and uncoupling protein 1 (UCP1) expression via reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot. Among the key adipogenic factors, rosiglitazone proved more effective than indomethacin. Extending the induction phase from 4 to 8 days and maintaining dexamethasone throughout the culture markedly enhanced differentiation efficiency. Serum concentration above 5% was inhibitory, while optimal conditions were identified as 5 μM rosiglitazone and 20 μg/mL insulin. The optimized protocol successfully induced beige adipogenesis in ADSCs from eight independent donors, though efficiency varied considerably which could be attributed to individual donor variability. These findings provide a robust in vitro model for studying beige fat biology and highlight the relevance of personalized approaches in metabolic research. Full article
Show Figures

Figure 1

16 pages, 3017 KB  
Article
Nobiletin Attenuates Adipogenesis and Promotes Browning in 3T3-L1 Adipocytes Through Exosomal miRNA-Mediated AMPK Activation
by Shweta Chauhan, Hana Baek, Varun Jaiswal, Miey Park and Hae-Jeung Lee
Curr. Issues Mol. Biol. 2026, 48(1), 36; https://doi.org/10.3390/cimb48010036 - 26 Dec 2025
Viewed by 252
Abstract
Nobiletin, a citrus-derived polymethoxylated flavone, has been reported to exert anti-obesity effects, but its molecular mechanisms remain poorly understood. This study aimed to investigate whether nobiletin suppresses adipogenesis and promotes browning in 3T3-L1 adipocytes by modulating exosomal microRNAs (miRNAs) and AMPK signaling. To [...] Read more.
Nobiletin, a citrus-derived polymethoxylated flavone, has been reported to exert anti-obesity effects, but its molecular mechanisms remain poorly understood. This study aimed to investigate whether nobiletin suppresses adipogenesis and promotes browning in 3T3-L1 adipocytes by modulating exosomal microRNAs (miRNAs) and AMPK signaling. To this end, we treated 3T3-L1 adipocytes with various concentrations of nobiletin and evaluated gene and protein expression by RT-qPCR and Western blotting. Nobiletin significantly reduced intracellular lipid accumulation at 50 μM (p < 0.001) and downregulated key adipogenic transcription factors, PPARγ, C/EBPα, and SREBP-1c, and suppressed the lipogenic enzyme FAS, while activating the AMPK/ACC signaling pathway. Concomitantly, it enhanced the expression of thermogenic markers UCP-1, PRDM16, and PGC-1α, indicating a metabolic shift toward energy expenditure. Exosomal RNA-seq revealed 10 differentially expressed miRNAs, of which miR-181d-5p (3.1-fold) and miR-221-3p (2.4-fold) were upregulated, whereas miR-205-5p (−2.9-fold), miR-331-3p (−3.2-fold), miR-130b-3p (−2.6-fold), miR-143-5p (−2.9-fold), miR-183-3p (−2.8-fold), miR-196b-5p (−2.4-fold), miR-26b-3p (−2.2-fold), and miR-378d (−2.7-fold) were verified by RT-qPCR after nobiletin treatment (50 μM). These miRNAs are functionally associated with adipogenic and thermogenic pathways, supporting a regulatory role of the exosomal miRNA network in nobiletin’s action. Collectively, our results identify a novel exosome–miRNA–AMPK axis underlying the anti-adipogenic and browning-inducing activities of nobiletin, highlighting its potential as a therapeutic phytochemical for obesity prevention. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

11 pages, 2567 KB  
Article
Effects of Luteolin and Apigenin on Adipogenesis Markers PPARγ and FABP4 and Thermogenesis Marker UCP1 in 3T3-L1 Preadipocyte Cell Line
by Gülcan Uysal Yeler, Ayşegül Sivaslıoğlu, Tuğba Gülsün and Zeynep Göktaş
Int. J. Mol. Sci. 2026, 27(1), 139; https://doi.org/10.3390/ijms27010139 - 22 Dec 2025
Viewed by 230
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) plays a crucial role in the differentiation and maturation of preadipocytes. PPARγ promotes adipogenesis by inducing the expression of fatty acid-binding protein 4 (FABP4). Uncoupling protein 1 (UCP1) is involved in non-shivering thermogenesis and adipocyte browning. The present [...] Read more.
Peroxisome proliferator-activated receptor γ (PPARγ) plays a crucial role in the differentiation and maturation of preadipocytes. PPARγ promotes adipogenesis by inducing the expression of fatty acid-binding protein 4 (FABP4). Uncoupling protein 1 (UCP1) is involved in non-shivering thermogenesis and adipocyte browning. The present study aimed to examine the effects of luteolin and apigenin on the gene expression levels and protein concentrations of PPARγ and FABP4, which are involved in adipogenesis, and their effect on UCP1, a thermogenic protein, in the 3T3-L1 preadipocyte cell line. Luteolin and apigenin were prepared at concentrations of 10, 20, and 40 µM and applied to 3T3-L1 preadipocytes during differentiation and maturation. Gene expression levels were measured by real-time PCR, and protein concentrations were measured by ELISA. It was found that the doses used did not cause cytotoxicity in the cells. Luteolin treatment during differentiation and maturation resulted in a decrease in PPARγ and FABP4 gene expression, although the protein concentrations remained unchanged. Additionally, while luteolin treatment did not significantly alter UCP1 gene expression or protein levels during differentiation, it led to a decrease in UCP1 protein concentration during maturation. Apigenin treatment also tended to decrease PPARγ and FABP4 gene expression compared to the control, although no statistical difference was observed. These results suggest that luteolin and apigenin may have regulatory effects on adipogenesis by modulating PPARγ, FABP4, and UCP1 gene expression. Full article
Show Figures

Figure 1

25 pages, 1376 KB  
Review
Mollugin: A Comprehensive Review of Its Multifaceted Pharmacological Properties and Therapeutic Potential
by Sandra Ross Olakkengil Shajan, Bushra Zia, Charu Sharma, Sandeep B. Subramanya and Shreesh Ojha
Int. J. Mol. Sci. 2025, 26(24), 12003; https://doi.org/10.3390/ijms262412003 - 13 Dec 2025
Viewed by 426
Abstract
The substantial interest in plant-based drugs or plant-derived phytocompounds drives researchers to conduct comprehensive investigations on their therapeutic properties. Mollugin, one of the major active constituents of Rubia cardifolia, has been well-studied for its pharmacological properties, demonstrating potent anti-inflammatory properties by suppressing [...] Read more.
The substantial interest in plant-based drugs or plant-derived phytocompounds drives researchers to conduct comprehensive investigations on their therapeutic properties. Mollugin, one of the major active constituents of Rubia cardifolia, has been well-studied for its pharmacological properties, demonstrating potent anti-inflammatory properties by suppressing the TAK-1-mediated activation of NF-κB/MAPK and enhancing the Nrf2/HO-1-mediated antioxidant response. It exhibits strong anticancer effects through ferroptosis via IGF2BP3/GPX4 pathways, induces mitochondrial apoptosis, and targets NF-κB, ERK, and PI3K/Akt/mTOR to suppress tumor progression. Mollugin also inhibits JAK2/STAT and PARP1 pathways, suppressing IL-1β expression via the modulation of ZFP91. Moreover, it regulates the MAPK/p38 pathway, promotes neuroprotection, and improves cognitive performance through GLP-1 receptor activation. Mollugin promotes osteogenesis by activating the BMP-2/Smad1/5/8 signaling pathway and downregulates MAPK, Akt, and GSK3β expression, leading to the inhibition of osteoclastogenesis. It overcomes multidrug resistance by downregulating MDR1/P-gp, CREB, NF-κB, and COX-2 through AMPK activation. Its antibacterial effect is mediated by strong binding to FUR, UDP, and IpxB proteins in Enterobacter xiangfangensis. Mollugin mitigates Klebsiella pneumoniae infection, suppresses adipogenesis without causing cytotoxicity, and protects endothelial cells via the BDNF/TrkB-Akt signaling pathway. Synthetic derivatives of mollugin, such as oxomollugin and azamollugin, have shown enhanced anticancer and anti-inflammatory effects by regulating EGFR, PKM2, TLR4/MyD88/IRAK/TRAF6, and NF-κB/IRF3 pathways with improved solubility and stability. Collectively, these findings emphasize the broad-spectrum activity of mollugin. This review provides a critical interpretation of the mechanistic pathways regulated by mollugin and its derivatives, emphasizing their pharmacological significance and exploring their potential for future translation as multitarget drug candidates. Full article
(This article belongs to the Special Issue Plant-Derived Bioactive Compounds for Pharmacological Applications)
Show Figures

Graphical abstract

21 pages, 4258 KB  
Article
Caspase-1 Mediated Cleavage of BMP Type I Receptor Drives BMP2-Induced Differentiation of Bone Marrow Mesenchymal Stem Cells into Adipocytes
by Kelechi Chukwuocha, Venu Pandit, Daniel Halloran and Anja Nohe
Appl. Sci. 2025, 15(24), 13105; https://doi.org/10.3390/app152413105 - 12 Dec 2025
Viewed by 266
Abstract
Bone Morphogenetic Protein-2 (BMP2) is a growth factor that maintains bone homeostasis through the BMP receptor type Ia (BMPRIa) and type II (BMPRII). BMP2 promotes osteogenesis by inducing the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts; however, it can also [...] Read more.
Bone Morphogenetic Protein-2 (BMP2) is a growth factor that maintains bone homeostasis through the BMP receptor type Ia (BMPRIa) and type II (BMPRII). BMP2 promotes osteogenesis by inducing the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts; however, it can also trigger BMSC differentiation into adipocytes. BMP2’s osteo-inductive ability has made it a potential treatment for osteoporosis, yet its dual role in BMSC differentiation complicates its efficacy. High BMP2 levels cause BMPRIa cleavage, but the downstream effects and the mechanisms governing BMP2-induced osteogenesis or adipogenesis are unresolved. Here, we identify Caspase-1 as a key mediator of BMPRIa cleavage and its downstream effects on adipogenesis. We used primary BMSCs from C57BL/6 mice, stimulated with varying BMP2 concentrations, to explore BMP2-induced BMPRIa cleavage and its impact on PPARγ—a key regulator of adipogenesis. Western blotting and immunostaining using antibodies against BMPRIa and PPARγ uncovered BMPRIa cleavage and revealed the nuclear translocation of the cleaved segment, colocalizing with PPARγ. Caspase-1 inhibition significantly reduced BMPRIa cleavage and PPARγ expression, highlighting its pivotal role in adipogenic differentiation. Understanding the molecular mechanisms of BMP2-induced adipogenesis and Caspase-1 inhibition could improve BMP2 therapeutic efficacy for osteoporosis by promoting osteogenesis over adipogenesis. Full article
(This article belongs to the Section Applied Biosciences and Bioengineering)
Show Figures

Figure 1

18 pages, 2295 KB  
Article
Integrated Transcriptomic and Proteomic Analysis Associated with Knockdown and Overexpression Studies Revealed ECHDC1 as a Regulator of Intramuscular Fat Deposition in Cattle
by Ruiying He, Li Liu, Xianya Kong, Nanfei Wang, Jianbing Tan, Zhangqing Wu, Linsen Zan and Wucai Yang
Animals 2025, 15(24), 3558; https://doi.org/10.3390/ani15243558 - 11 Dec 2025
Viewed by 332
Abstract
Intramuscular fat (IMF) content is a critical determinant of beef quality and is regulated by various factors. However, the molecular mechanisms underlying IMF deposition in cattle remain poorly understood, particularly with regard to multi-omics integrated analyses. In this study, an integrative analysis of [...] Read more.
Intramuscular fat (IMF) content is a critical determinant of beef quality and is regulated by various factors. However, the molecular mechanisms underlying IMF deposition in cattle remain poorly understood, particularly with regard to multi-omics integrated analyses. In this study, an integrative analysis of transcriptomic and proteomic profiles was performed on intramuscular fat tissue of Jiaxian Red cattle with different marbling grades to identify key genes and pathways involved in intramuscular fat deposition in beef cattle. Integrated analysis showed that 4532 genes were co-expressed at both mRNA and protein level, among which 21 genes exhibited significant differential expression at mRNA and protein level. These genes showed significant enrichment in lipid biosynthesis and metabolic processes. ECHDC1 was selected for in-depth functional studies on bovine intramuscular preadipocyte adipogenesis as a candidate gene via RNA interference and overexpression techniques. The results indicated that knockdown of ECHDC1 inhibited the adipogenesis of bovine intramuscular preadipocytes and significantly upregulated the expression of HSL. Conversely, overexpression of ECHDC1 promoted the adipogenesis of bovine intramuscular preadipocytes, decreased the expression of HSL and increased the expression of FABP4. In summary, these results suggested that ECHDC1 promoted intramuscular fat deposition by attenuating lipolysis rate by downregulating the expression of the lipolytic gene HSL, providing a potential molecular target for enhancing IMF content in beef cattle. Full article
Show Figures

Graphical abstract

21 pages, 4445 KB  
Article
The SCD5 Gene Modulates Adipogenic Differentiation via the WNT5B Signaling Pathway in Xinjiang Brown Cattle
by Yiran Wang, Wanping Ren, Wei Shao, Yuxin Zhou, Yili Liu, Junwei Cao, Fengju Wang, Jingdong Bi and Liang Yang
Animals 2025, 15(24), 3547; https://doi.org/10.3390/ani15243547 - 10 Dec 2025
Viewed by 343
Abstract
This study investigated the role of SCD5 in bovine preadipocyte proliferation and adipogenic differentiation. SCD5 overexpression suppressed proliferation, reducing the percentage of EdU-positive cells and overall cell viability (p < 0.05). It also downregulated proliferative factors CDK1 and CDK2 (p < [...] Read more.
This study investigated the role of SCD5 in bovine preadipocyte proliferation and adipogenic differentiation. SCD5 overexpression suppressed proliferation, reducing the percentage of EdU-positive cells and overall cell viability (p < 0.05). It also downregulated proliferative factors CDK1 and CDK2 (p < 0.05), and reduced lipid accumulation (p < 0.001) along with key adipogenic markers PPARγ, C/EBPα, and FABP4 (p < 0.01). Conversely, SCD5 knockdown promoted these processes. Mechanistically, SCD5 overexpression downregulated WNT5B (p < 0.05), while knockdown had the opposite effect. Silencing WNT5B significantly decreased β-catenin transcription (p < 0.05), total protein (p < 0.05), and phosphorylation (p < 0.001). In conclusion, our findings identify SCD5 as a novel negative regulator of bovine preadipocyte proliferation and differentiation, which exerts its function through the WNT5B/β-catenin signaling axis. This discovery elucidates previously uncharacterized regulatory mechanisms underlying adipogenesis in livestock species. Full article
(This article belongs to the Special Issue Advances in Cattle Genetics and Breeding)
Show Figures

Figure 1

13 pages, 2241 KB  
Article
An ATF3 Inducer Ameliorates Metabolic Dysfunction-Associated Steatotic Liver Disease Through the AMPK and PKA Pathways
by Ching-Feng Cheng, Ruey-Bing Yang, Wen-Ting Chen, Jia-Fang Chung and Hui-Chen Ku
Int. J. Mol. Sci. 2025, 26(24), 11877; https://doi.org/10.3390/ijms262411877 - 9 Dec 2025
Viewed by 377
Abstract
Obesity is linked to metabolic dysfunction-associated steatotic liver disease (MASLD), but the molecular mechanisms and effective treatments remain unclear. This study investigated whether ST32db, an inducer of activating transcription factor 3 (ATF3), affects lipid metabolism in MASLD. An in vitro model was established [...] Read more.
Obesity is linked to metabolic dysfunction-associated steatotic liver disease (MASLD), but the molecular mechanisms and effective treatments remain unclear. This study investigated whether ST32db, an inducer of activating transcription factor 3 (ATF3), affects lipid metabolism in MASLD. An in vitro model was established involving the treatment of HepG2 cells with 1 mM oleic acid (OA) with or without 20 µM ST32db. In an in vivo model, C57BL/6 mice were fed a high-fat diet (HFD) for 18 weeks to induce obesity and treated or not with ST32db (1 mg kg−1). ST32db significantly decreased intracellular lipid accumulation in OA-treated HepG2 cells. In these cells, ST32db remarkably decreased mRNA and protein levels of adipogenesis- and lipogenesis-related genes and increased mRNA levels of adipose triglyceride lipase (ATGL), a lipolytic enzyme. In HFD-fed mice, the ST32db treatment significantly decreased the liver weight, serum triglycerides, and fat vacuole and triglyceride accumulation in the liver. Livers from these mice also showed significantly decreased CCAAT/enhancer-binding protein β mRNA and protein levels, increased ATF3 mRNA and protein and ATGL mRNA levels, and increased levels of phosphorylated AMP-activated protein kinase (AMPK) and protein kinase A (PKA). These findings suggest that ST32db may exert protective effects against MASLD through activating hepatic AMPK and PKA pathways. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

18 pages, 6370 KB  
Article
Molecular Mechanisms Underlying Divergent Biochemical Compositions in Longissimus Dorsi of Huainan and Yunan Black Pigs: Insights from Fatty Acids, Amino Acids, and Transcriptomic Profiling
by Yuliang Wen, Chengshui Liao, Ruibiao Wang, Bo Wen, Weiyu Luo, Wei Zhang, Chunling Zhu, Huarun Sun, Longfei Zhang, Xuehan Liu, Jiyuan Shen, Yuping Xiang, Hao Li, Jianhe Hu, Yueyu Bai, Ke Ding and Lei Wang
Agriculture 2025, 15(24), 2532; https://doi.org/10.3390/agriculture15242532 - 6 Dec 2025
Viewed by 342
Abstract
This study investigated meat quality, nutritional characteristics, and transcriptomic regulation in Yunan (YN) black pigs and Huainan (HN) black pigs (n = 6 each). Analysis of fatty acid composition revealed that HN black pigs possessed significantly higher levels of most fatty acids compared [...] Read more.
This study investigated meat quality, nutritional characteristics, and transcriptomic regulation in Yunan (YN) black pigs and Huainan (HN) black pigs (n = 6 each). Analysis of fatty acid composition revealed that HN black pigs possessed significantly higher levels of most fatty acids compared to YN black pigs. Notably, the contents of monounsaturated fatty acid C18:1n9c and polyunsaturated fatty acid C18:2n6c in HN black pigs were 1.94-fold and 2.65-fold higher, respectively, than those in YN black pigs. The α-linolenic acid content was also significantly elevated in HN black pigs, indicating an overall higher fatty acid content. Regarding amino acid differences, HN black pigs exhibited significantly higher levels of aspartic acid, glutamic acid, histidine, as well as superior composition of total amino acids, total umami amino acids, and essential amino acids, which contribute to enhanced flavor characteristics and nutritional balance. Transcriptome analysis identified 526 differentially expressed genes in HN vs. YN. KEGG enrichment analysis showed that these genes were involved in many adipogenesis and lipid metabolism signaling pathways, such as biosynthesis of unsaturated fatty acids, fatty acid elongation, apelin signaling pathway and lysine degradation. By integrating transcriptome and protein–protein interaction (PPI) network analyses, we identified key meat quality-related genes: ELOVL6, PRKAG3, ROCK2, and MYH11. miRNA profiling identified ssc-miR-133b, ssc-miR-206, and miR-205 as key regulators of meat quality. This study provides a valuable theoretical foundation for understanding the molecular mechanisms underlying pork quality and offers insights for its future improvement. Full article
(This article belongs to the Section Farm Animal Production)
Show Figures

Figure 1

12 pages, 1550 KB  
Article
Extracellular Vesicles from Lactobacillus rhamnosus BS-Pro-08, Kefir Grain, Suppress Adipogenesis and Enhance Lipolysis in Adipocytes
by Bi-Oh Park, Ho Woon Lee, Chang-Hyun Song, Miji Yeom, Seoungwoo Shin, Hyesoo Wang, Junbo Sim, Eunae Cho, Deokhoon Park and Eunsun Jung
Int. J. Mol. Sci. 2025, 26(23), 11732; https://doi.org/10.3390/ijms262311732 - 4 Dec 2025
Viewed by 497
Abstract
Extracellular vesicles (EVs) derived from probiotic bacteria have recently emerged as postbiotic mediators that regulate host cellular responses. This study investigated the effects of EVs from Lactobacillus rhamnosus BS-Pro-08, isolated from kefir grains (Lacto EV), on adipocyte differentiation and lipid metabolism. Lacto [...] Read more.
Extracellular vesicles (EVs) derived from probiotic bacteria have recently emerged as postbiotic mediators that regulate host cellular responses. This study investigated the effects of EVs from Lactobacillus rhamnosus BS-Pro-08, isolated from kefir grains (Lacto EV), on adipocyte differentiation and lipid metabolism. Lacto EV treatment markedly suppressed the differentiation of 3T3-L1 preadipocytes into mature adipocytes, as reflected by reduced lipid accumulation and decreased expression of the adipogenic transcription factors peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα). This inhibitory effect was most pronounced at the early stage of adipogenesis. In mature adipocytes, Lacto EV enhanced lipolysis in a dose-dependent manner, accompanied by increased glycerol release and total lipase activity. Interestingly, these lipolytic responses occurred despite reduced protein levels of adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL), suggesting that Lacto EVs may mediate an EV-enhanced lipolysis that is not fully explained by canonical ATGL/HSL signaling. Collectively, these findings demonstrate that Lacto EV modulates both adipogenic and lipolytic processes in vitro, providing insight into the metabolic actions of probiotic-derived vesicles. Full article
(This article belongs to the Special Issue The Interactions Between Nutrients and Adipose Tissue)
Show Figures

Figure 1

29 pages, 1924 KB  
Review
Marine-Algal-Derived Postbiotics Modulating the Gut Microbiota–Adipose Tissue Axis in Obesity: A New Frontier
by Edward Kurnia Setiawan Limijadi, Kevin Christian Tjandra, Happy Kurnia Permatasari, Piko Satria Augusta, Reggie Surya, Dante Saksono Harbuwono and Fahrul Nurkolis
Nutrients 2025, 17(23), 3774; https://doi.org/10.3390/nu17233774 - 30 Nov 2025
Viewed by 1018
Abstract
Background: Obesity is increasingly recognized as a metabolic disorder driven by gut microbiota dysbiosis and chronic low-grade inflammation within adipose tissue. Emerging evidence highlights the gut–adipose tissue axis as a critical mediator of energy balance and metabolic regulation. Marine algae—rich in polysaccharides, polyphenols, [...] Read more.
Background: Obesity is increasingly recognized as a metabolic disorder driven by gut microbiota dysbiosis and chronic low-grade inflammation within adipose tissue. Emerging evidence highlights the gut–adipose tissue axis as a critical mediator of energy balance and metabolic regulation. Marine algae—rich in polysaccharides, polyphenols, and carotenoids—offer bioactive compounds that modulate gut microbial composition and generate beneficial metabolites termed “postbiotics.” Objective: This review aims to comprehensively summarize current advances in understanding how marine-algal-derived postbiotics influence the gut microbiota–adipose tissue axis and contribute to obesity prevention and management. Methods: A structured literature search was conducted across PubMed, Scopus, Web of Science, ScienceDirect, and SpringerLink for studies published between 2015 and October 2025. Eligible studies included in vitro, in vivo, and human trials examining the effects of marine-algal compounds on gut microbiota composition, short-chain fatty acid (SCFA) production, adipose inflammation, and metabolic outcomes. Results: Marine-algal polysaccharides (fucoidan, alginate, laminarin, carrageenan, and ulvan) act as fermentable fibers that enhance SCFA production and enrich beneficial taxa such as Akkermansia, Lactobacillus, and Bacteroides, while reducing endotoxin-producing bacteria. Polyphenols and carotenoids (fucoxanthin, phlorotannins, astaxanthin) directly target adipogenesis, oxidative stress, and adipose browning. Animal studies consistently demonstrate reduced body weight, improved insulin sensitivity, and decreased inflammation following algae supplementation. Human trials—though limited—confirm safety and show microbiota modulation with modest weight loss. Conclusions: Marine-algal-derived postbiotics represent a promising, natural, and sustainable strategy to target the gut microbiota–adipose tissue axis in obesity. They offer multi-targeted mechanisms through microbial and host pathways, supporting their integration into functional food and nutraceutical development. Further clinical research and regulatory standardization are warranted to translate these findings into evidence-based interventions. Full article
(This article belongs to the Special Issue Interaction Between Gut Microbiota and Obesity)
Show Figures

Figure 1

Back to TopTop