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

Article Types

Countries / Regions

Search Results (206)

Search Parameters:
Keywords = peroxisomal targeting signal

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
38 pages, 1248 KB  
Review
Targeting Inflammation with Natural Products: A Mechanistic Review of Iridoids from Bulgarian Medicinal Plants
by Rositsa Mihaylova, Viktoria Elincheva, Reneta Gevrenova, Dimitrina Zheleva-Dimitrova, Georgi Momekov and Rumyana Simeonova
Molecules 2025, 30(17), 3456; https://doi.org/10.3390/molecules30173456 - 22 Aug 2025
Viewed by 861
Abstract
Chronic, low-grade inflammation is a key contributor to the development of numerous non-communicable diseases (NCDs), including cardiovascular, metabolic, and neurodegenerative disorders. Conventional anti-inflammatory drugs, such as nonsteroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, often present safety concerns with prolonged use, highlighting the need for [...] Read more.
Chronic, low-grade inflammation is a key contributor to the development of numerous non-communicable diseases (NCDs), including cardiovascular, metabolic, and neurodegenerative disorders. Conventional anti-inflammatory drugs, such as nonsteroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, often present safety concerns with prolonged use, highlighting the need for safer, multi-targeted therapeutic options. Iridoids, a class of monoterpenoid compounds abundant in several medicinal plants, have emerged as promising bioactive agents with diverse pharmacological properties. They exert anti-inflammatory and metabolic regulatory effects by modulating key signaling pathways, including nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK), Janus kinase/signal transducer and activator of transcription (JAK/STAT), adenosine monophosphate-activated protein kinase (AMPK), and peroxisome proliferator-activated receptor (PPAR) pathways. This review provides a comprehensive summary of the major iridoid metabolites derived from ten Bulgarian medicinal plant species, along with mechanistic insights from in vitro and in vivo studies. Documented biological activities include anti-inflammatory, antioxidant, immunomodulatory, antifibrotic, organoprotective, antibacterial, antiviral, analgesic, and metabolic effects. By exploring their phytochemical profiles and pharmacodynamics, we underscore the therapeutic potential of iridoid-rich Bulgarian flora in managing inflammation-related and metabolic diseases. These findings support the relevance of iridoids as complementary or alternative agents to conventional therapies and highlight the need for further translational and clinical research. Full article
(This article belongs to the Special Issue Role of Natural Products in Inflammation)
Show Figures

Figure 1

23 pages, 1467 KB  
Article
Effects of Dietary Rhodotorula mucilaginosa on Muscle Composition, Serum Biochemical Indicators, Antioxidant Capacity, Lipid Metabolism, and mTOR Signaling Pathway in Red Claw Crayfish (Cherax quadricanatus)
by Liuqing Meng, Luoqing Li, Ziyi Ma, Wenyan He, Qin Zhang, Tong Tong, Dapeng Wang, Rui Wang, Huizan Yang, Yongqiang Liu and Yin Huang
Biology 2025, 14(8), 1089; https://doi.org/10.3390/biology14081089 - 20 Aug 2025
Viewed by 496
Abstract
This study investigated the effects of dietary supplementation with varying levels (CK: 0.0 g/kg; RL: 0.1 g/kg; RM: 1.0 g/kg; RH: 10.0 g/kg) of Rhodotorula mucilaginosa on muscle composition, serum biochemical indicators, antioxidant capacity, lipid metabolism, and the mTOR signaling pathway in red [...] Read more.
This study investigated the effects of dietary supplementation with varying levels (CK: 0.0 g/kg; RL: 0.1 g/kg; RM: 1.0 g/kg; RH: 10.0 g/kg) of Rhodotorula mucilaginosa on muscle composition, serum biochemical indicators, antioxidant capacity, lipid metabolism, and the mTOR signaling pathway in red claw crayfish (Cherax quadricarinatus). Results showed that, compared to CK, treatment groups had higher muscle crude protein, fat, leucine, histidine, arginine, and essential amino acids (p < 0.05), and lower saturated fatty acids (p < 0.05). Treatment groups also exhibited increased activities of alkaline phosphatase, acid phosphatase, superoxide dismutase, catalase, glutathione S-transferase, lysozyme, albumin, total protein, and antioxidant capacity (p < 0.05), with reduced activities of aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and triglycerides (p < 0.05). In the hepatopancreas, treatment groups showed significant downregulation of AMP-activated protein kinase α, β, and γ, and carnitine palmitoyltransferase 1 genes (p < 0.05). Conversely, genes involved in lipid anabolism (peroxisome proliferator-activated receptor γ, acetyl-CoA carboxylase, fatty acid synthase, sterol regulatory element-binding protein, protein kinase B, and mammalian target of rapamycin 1 and 2) were upregulated (p < 0.05). In conclusion, R. mucilaginosa supplementation affects muscle composition, lipid metabolism, and mTOR signaling. The optimal dose is 1.0 g/kg. Full article
(This article belongs to the Special Issue Nutrition, Environment, and Fish Physiology)
Show Figures

Figure 1

16 pages, 3830 KB  
Article
5,7-Dimethoxyflavone Attenuates Sarcopenic Obesity by Enhancing PGC-1α–Mediated Mitochondrial Function in High-Fat-Diet-Induced Obese Mice
by Changhee Kim, Mi-Bo Kim, Sanggil Lee and Jae-Kwan Hwang
Nutrients 2025, 17(16), 2642; https://doi.org/10.3390/nu17162642 - 14 Aug 2025
Viewed by 741
Abstract
Background/Objectives: Sarcopenic obesity, defined by the coexistence of excessive fat accumulation and progressive muscle loss, is associated with an increased risk of metabolic dysfunction and physical disability. While 5,7-dimethoxyflavone (DMF), a bioactive flavone derived from Kaempferia parviflora, has demonstrated anti-obesity and [...] Read more.
Background/Objectives: Sarcopenic obesity, defined by the coexistence of excessive fat accumulation and progressive muscle loss, is associated with an increased risk of metabolic dysfunction and physical disability. While 5,7-dimethoxyflavone (DMF), a bioactive flavone derived from Kaempferia parviflora, has demonstrated anti-obesity and muscle-preserving properties, its effects on sarcopenic obesity remain unclear. Methods: Four-week-old male C57BL/6J mice were fed a high-fat diet (HFD) for 6 weeks to induce sarcopenic obesity, followed by 8 weeks of continued HFD with the oral administration of DMF. Muscle function was assessed through grip strength and treadmill running tests, while muscle and fat volumes were measured using micro-CT. Mechanistic analyses were performed using gene expression and Western blot analysis. Results: DMF significantly reduced body weight, fat mass, and adipocyte size while enhancing grip strength, endurance, skeletal muscle mass, and the muscle fiber cross-sectional area. In the gastrocnemius muscle, DMF increased the gene expression of peroxisome proliferator-activated receptor gamma coactivator-1α (Ppargc1a) and its isoform Ppargc1a4, thereby promoting mitochondrial biogenesis. It also improved protein turnover by modulating protein synthesis and degradation via the phosphatidylinositol 3-kinase/protein kinase B/mechanistic target of rapamycin signaling pathway. In subcutaneous and brown adipose tissues, DMF increased mitochondrial DNA content and the expression of thermogenic and beige adipocyte-related genes. These findings suggest that DMF alleviates sarcopenic obesity by improving mitochondrial function and regulating energy metabolism in both skeletal muscle and adipose tissues via PGC-1α-mediated pathways. Thus, DMF represents a promising therapeutic candidate for the integrated management of sarcopenic obesity. Full article
Show Figures

Figure 1

17 pages, 4198 KB  
Article
Proteomic Analysis of Protein Ubiquitination Events in Dairy Goats with Fatty Liver
by Yuli Zhu, Zhenhua Liu, Yuming Zhang, Yao Meng, Xunuo Song, Jinyu Li, Yue Zhang, Junkang Zhao, Liyin Du and Qinghua Deng
Animals 2025, 15(14), 2010; https://doi.org/10.3390/ani15142010 - 8 Jul 2025
Viewed by 499
Abstract
Fatty liver is a major metabolic disease in periparturient dairy goats. Protein ubiquitination, a type of dynamic and multifaceted post-translational modification, plays an important role in metabolism by regulating the stability and function of target proteins. However, the hepatic protein ubiquitination profile in [...] Read more.
Fatty liver is a major metabolic disease in periparturient dairy goats. Protein ubiquitination, a type of dynamic and multifaceted post-translational modification, plays an important role in metabolism by regulating the stability and function of target proteins. However, the hepatic protein ubiquitination profile in dairy goats with fatty liver is yet to be elucidated. In this study, we collected liver and blood samples from healthy dairy goats (Con, n = 3) and dairy goats with fatty liver (FL, n = 3). Then, we analyzed the overall ubiquitination of hepatic proteins in dairy goats with fatty liver through quantitative ubiquitin label-free proteomics and bioinformatics. Proteins showing significantly altered levels of ubiquitination were identified via bioinformatics, and related regulatory pathways were screened. The results showed that the blood levels of beta-hydroxybutyric acid and non-esterified fatty acids were significantly upregulated in dairy goats with fatty liver, and a total of 238 ubiquitination sites across 921 proteins were found to be differentially altered in the fatty liver group. Among them, ubiquitination was upregulated at 351 sites across 93 proteins and downregulated at 570 sites across 145 proteins. In addition, GO and KEGG pathway analysis revealed that the differentially ubiquitinated proteins were enriched in pathways regulating lipid metabolism, such as the PPAR signaling pathway, fatty acid degradation, and peroxisome activity. Notably, by observing the overlap among these three sub-networks, we found that proteins with downregulated ubiquitination—such as ACSL1, ACSL5, EHHADH, and ACAA1—were transcriptionally upregulated in dairy goats with fatty liver. This study reveals the key ubiquitinated proteins in dairy goats with fatty liver and provides a more comprehensive understanding of the pathogenesis of fatty liver in dairy goats. Full article
Show Figures

Figure 1

29 pages, 1939 KB  
Review
Peroxisomal Alterations in Prostate Cancer: Metabolic Shifts and Clinical Relevance
by Mohamed A. F. Hussein, Celien Lismont, Hongli Li, Ruizhi Chai, Frank Claessens and Marc Fransen
Cancers 2025, 17(13), 2243; https://doi.org/10.3390/cancers17132243 - 4 Jul 2025
Cited by 1 | Viewed by 1460
Abstract
Cancer is hallmarked by uncontrolled cell proliferation and enhanced cell survival, driven by a complex interplay of factors—including genetic and epigenetic changes—that disrupt metabolic and signaling pathways and impair organelle function. While the roles of mitochondria and the endoplasmic reticulum in cancer are [...] Read more.
Cancer is hallmarked by uncontrolled cell proliferation and enhanced cell survival, driven by a complex interplay of factors—including genetic and epigenetic changes—that disrupt metabolic and signaling pathways and impair organelle function. While the roles of mitochondria and the endoplasmic reticulum in cancer are widely recognized, emerging research is now drawing attention to the involvement of peroxisomes in tumor biology. Peroxisomes are essential for lipid metabolism, including fatty acid α- and β-oxidation, the synthesis of docosahexaenoic acid, bile acids, and ether lipids, as well as maintaining redox balance. Despite their critical functions, the role of peroxisomes in oncogenesis remains inadequately explored. Prostate cancer (PCa), the second most common cancer in men worldwide, exhibits a unique metabolic profile compared to other solid tumors. In contrast to the glycolysis-driven Warburg effect, primary PCa relies primarily on lipogenesis and oxidative phosphorylation. Peroxisomes are intricately involved in the metabolic adaptations of PCa, influencing both disease progression and therapy resistance. Key alterations in peroxisomal activity in PCa include the increased oxidation of branched-chain fatty acids, upregulation of α-methylacyl coenzyme A racemase (a prominent PCa biomarker), and downregulation of 1-alkyl-glycerone-3-phosphate synthase and catalase. This review critically examines the role of peroxisomes in PCa metabolism, progression, and therapeutic response, exploring their potential as biomarkers and targets for therapy. We also consider their relationship with androgen receptor signaling. A deeper understanding of peroxisome biology in PCa could pave the way for new therapies to improve patient outcomes. Full article
(This article belongs to the Special Issue Advancements in Molecular Research of Prostate Cancer)
Show Figures

Graphical abstract

36 pages, 1531 KB  
Review
Orchestration of Gut–Liver-Associated Transcription Factors in MAFLD: From Cross-Organ Interactions to Therapeutic Innovation
by Ao Liu, Mengting Huang, Yuwen Xi, Xiaoling Deng and Keshu Xu
Biomedicines 2025, 13(6), 1422; https://doi.org/10.3390/biomedicines13061422 - 10 Jun 2025
Cited by 2 | Viewed by 1636
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) represents a global health burden, however, therapeutic advancements remain hindered by incomplete insights on mechanisms and suboptimal clinical interventions. This review focused on the transcription factors (TFs) associated with the gut–liver axis, emphasizing their roles as molecular [...] Read more.
Metabolic dysfunction-associated fatty liver disease (MAFLD) represents a global health burden, however, therapeutic advancements remain hindered by incomplete insights on mechanisms and suboptimal clinical interventions. This review focused on the transcription factors (TFs) associated with the gut–liver axis, emphasizing their roles as molecular interpreters of systemic crosstalk in MAFLD. We delineate how TF networks integrate metabolic, immune, and gut microbial signals to manage hepatic steatosis, inflammation, and fibrosis. For instance, metabolic TFs such as peroxisome proliferator-activated receptor α (PPARα) and farnesoid X receptor (FXR) are responsible for regulating lipid oxidation and bile acid homeostasis, while immune-related TFs like signal transducer and activator of transcription 3 (STAT3) modulate inflammatory cascades involving immune cells. Emerging evidence highlights microbiota-responsive TFs, like hypoxia-inducible factor 2α (HIF2α) and aryl hydrocarbon receptor (AHR), linking microbial metabolite signaling to hepatic metabolic reprogramming. Critically, TF-centric therapeutic strategies, including selective TF-agonists, small molecules targeted to degrade TF, and microbiota modulation, hold considerable promise for treating MAFLD. By synthesizing these insights, this review underscores the necessity to dissect TF-mediated interorgan communication and proposes a roadmap for translating mechanism discoveries into precision therapies. Future research should prioritize the use of multi-omics approaches to map TF interactions and validate their clinical relevance to MAFLD. Full article
(This article belongs to the Special Issue New Insights Into Non-Alcoholic Fatty Liver Diseases)
Show Figures

Figure 1

13 pages, 665 KB  
Review
Galectin-12 in the Regulation of Sebocyte Proliferation, Lipid Metabolism, and Immune Responses
by Ching-Han Tsao, Wei-Chen Hsieh, Feng-Jen Lin, Fu-Tong Liu and Ri-Yao Yang
Biomolecules 2025, 15(6), 837; https://doi.org/10.3390/biom15060837 - 8 Jun 2025
Cited by 1 | Viewed by 892
Abstract
Galectin-12, a member of the galectin family of glycan-binding proteins, exhibits restricted tissue distribution, primarily in adipocytes and sebocytes. In sebocytes, it modulates the cell cycle, influences lipid metabolism through interactions with peroxisome proliferator-activated receptor γ (PPARγ), and exerts immunomodulatory functions by activating [...] Read more.
Galectin-12, a member of the galectin family of glycan-binding proteins, exhibits restricted tissue distribution, primarily in adipocytes and sebocytes. In sebocytes, it modulates the cell cycle, influences lipid metabolism through interactions with peroxisome proliferator-activated receptor γ (PPARγ), and exerts immunomodulatory functions by activating immune signaling pathways. Dysregulation of sebocyte homeostasis, lipid metabolism, and immune responses contributes to the pathogenesis of a number of skin diseases, underscoring the therapeutic potential of targeting galectin-12. The review summarizes and discusses current developments in the field to foster future research in this important but underexplored galectin. Full article
(This article belongs to the Special Issue Cell Biology and Biomedical Application of Galectins)
Show Figures

Figure 1

17 pages, 3590 KB  
Article
Amelioration of Acetaminophen-Induced Hepatic Oxidative Stress and Inflammation by RNAi Targeting Cyp2e1 In Vivo
by Wenwen Liu, Liwen Huan, Cai Zhang, Runting Yin, Zhen Ouyang and Yuan Wei
Curr. Issues Mol. Biol. 2025, 47(5), 372; https://doi.org/10.3390/cimb47050372 - 19 May 2025
Cited by 1 | Viewed by 1612
Abstract
The overdose of acetaminophen (APAP) has become the leading cause of acute liver failure in the United States and some Western countries. As a principal member of the cytochrome P450 enzymes (CYPs), CYP2E1 is a vital enzyme in regard to the production of [...] Read more.
The overdose of acetaminophen (APAP) has become the leading cause of acute liver failure in the United States and some Western countries. As a principal member of the cytochrome P450 enzymes (CYPs), CYP2E1 is a vital enzyme in regard to the production of toxic APAP metabolites and in the development of APAP-induced liver injury (AILI). In this study, we investigated the therapeutic effects and mechanisms of lipid nanoparticle (LNP)-based delivery of small interfering RNA targeting Cyp2e1 (si-Cyp2e1 LNPs) on AILI in mice. C57BL/6J male mice were injected with 300 mg/kg APAP to establish an AILI model, and si-Cyp2e1 LNPs were administered via the tail vein. The results showed that the levels of serum alanine aminotransferase and aspartate aminotransferase were lower than those in APAP mice after treatment with si-Cyp2e1 LNPs immediately. Moreover, si-Cyp2e1 LNPs significantly inhibited liver necrosis and oxidative stress in APAP mice. RNA sequencing revealed that si-Cyp2e1 LNPs exerted regulatory effects on pathways and genes related to peroxisome proliferator-activated receptor (PPAR). Consistent with this finding, we also proved that si-Cyp2e1 LNPs markedly regulated the expressions of genes involved in the PPAR signaling pathway (CYP4A, PPARα, FABP 1, and CD36) in APAP mice, as well as inflammatory factors (Il-6, Il-1β, and Tnf-α). These findings suggested that si-Cyp2e1 LNPs may alleviate APAP-induced oxidative stress and inflammation by regulating lipid metabolism via PPAR-related pathways. Full article
(This article belongs to the Special Issue Advances in Molecular Biology Methods in Hepatology Research)
Show Figures

Graphical abstract

16 pages, 3543 KB  
Article
PPARα Genetic Deletion Reveals Global Transcriptional Changes in the Brain and Exacerbates Cerebral Infarction in a Mouse Model of Stroke
by Milton H. Hamblin, Austin C. Boese, Rabi Murad and Jean-Pyo Lee
Int. J. Mol. Sci. 2025, 26(9), 4082; https://doi.org/10.3390/ijms26094082 - 25 Apr 2025
Viewed by 893
Abstract
Ischemic stroke is a leading cause of death and disability worldwide. Currently, there is an unmet clinical need for pharmacological treatments that can improve ischemic stroke outcomes. In this study, we investigated the role of brain peroxisome proliferator-activated receptor alpha (PPARα) in ischemic [...] Read more.
Ischemic stroke is a leading cause of death and disability worldwide. Currently, there is an unmet clinical need for pharmacological treatments that can improve ischemic stroke outcomes. In this study, we investigated the role of brain peroxisome proliferator-activated receptor alpha (PPARα) in ischemic stroke pathophysiology. We used a well-established model of cerebral ischemia in PPARα transgenic mice and conducted the RNA sequencing (RNA-seq) of mouse stroke brains harvested 48 h post-middle cerebral artery occlusion (MCAO). PPARα knockout (KO) increased brain infarct size following stroke, indicating a protective role of PPARα in brain ischemia. Our RNA-seq analysis showed that PPARα KO altered the expression of genes in mouse brains with known roles in ischemic stroke pathophysiology. We also identified many other differentially expressed genes (DEGs) upon the loss of PPARα that correlated with increased infarct size in our stroke model. Gene set enrichment analysis (GSEA) and Gene Ontology (GO) analysis revealed the upregulation of gene signatures for the positive regulation of leukocyte proliferation, apoptotic processes, acute-phase response, and cellular component disassembly in mouse stroke brains with PPARα KO. In addition, pathway analysis of our RNA-seq data revealed that TNFα signaling, IL6/STAT3 signaling, and epithelial–mesenchymal transition (EMT) gene signatures were increased in PPARα KO stroke brains. Our study highlights PPARα as an attractive drug target for ischemic stroke due to its transcriptional regulation of inflammation-, apoptosis-, and EMT-related genes in brain tissue following ischemia. Full article
(This article belongs to the Special Issue Inflammatory Biomarkers in Ischemic Stroke)
Show Figures

Figure 1

19 pages, 788 KB  
Review
Omega-3 Fatty Acids and Exercise in Obesity Management: Independent and Synergistic Benefits in Metabolism and Knowledge Gaps
by Viviana Sandoval, Álvaro Vergara-Nieto, Amanda Bentes, Saulo Silva, Carolina Núñez and Sergio Martínez-Huenchullán
Biology 2025, 14(5), 463; https://doi.org/10.3390/biology14050463 - 24 Apr 2025
Viewed by 5298
Abstract
Obesity is a significant global health issue, profoundly affecting metabolic and cardiovascular health and other related chronic conditions. In Chile, the prevalence of obesity is among the highest within the Organisation for Economic Cooperation and Development (OECD) countries, highlighting a critical public health [...] Read more.
Obesity is a significant global health issue, profoundly affecting metabolic and cardiovascular health and other related chronic conditions. In Chile, the prevalence of obesity is among the highest within the Organisation for Economic Cooperation and Development (OECD) countries, highlighting a critical public health challenge. This narrative review examines current evidence on the independent and potential synergistic roles of omega-3 fatty acids and exercise in managing obesity-related metabolic dysfunction. Omega-3 fatty acids, particularly eicosapentaenoic acid (EPA), have been shown to lower triglyceride levels, enhance lipid metabolism, and modulate inflammation via pathways involving peroxisome proliferator-activated receptors (PPARs) and sterol regulatory element-binding protein-1c (SREBP-1c). Exercise interventions, such as moderate-intensity continuous training (MICT) and high-intensity interval training (HIIT), provide distinct yet complementary metabolic benefits. Specifically, MICT improves body fat distribution and mitochondrial efficiency, whereas HIIT has notable effects on metabolic adaptability and insulin signaling. Additionally, emerging evidence points toward a potential role of the kinin-kallikrein system, particularly kallikrein 7 (KLK7), in obesity-associated insulin resistance. Despite these promising findings, several knowledge gaps persist regarding optimal dosing, intervention timing, population-specific effects, and the exact mechanisms behind the potential synergistic interactions between omega-3 supplementation and structured exercise. This review emphasizes the importance of conducting further research, particularly controlled clinical trials, to clarify these combined interventions’ effectiveness and establish targeted therapeutic strategies tailored to individual metabolic profiles. Full article
(This article belongs to the Section Physiology)
Show Figures

Figure 1

15 pages, 1622 KB  
Article
Rutin Ameliorates BHBA-Induced Inflammation and Lipid Accumulation in Calf Hepatocytes Through NF-κB Signaling Pathway
by Kun Yang, Haixia Zhao, Min Gao, Honglian Hu and Dabiao Li
Curr. Issues Mol. Biol. 2025, 47(4), 274; https://doi.org/10.3390/cimb47040274 - 14 Apr 2025
Cited by 1 | Viewed by 730
Abstract
When subclinical ketosis (SCK) occurs in dairy cows, it leads to an excessive production of β-hydroxybutyrat (BHBA), which disrupts liver lipid metabolism and triggers a series of inflammatory responses. Rutin (RT), a flavonoid extracted from plants, exhibits diverse biological activities. However, its potential [...] Read more.
When subclinical ketosis (SCK) occurs in dairy cows, it leads to an excessive production of β-hydroxybutyrat (BHBA), which disrupts liver lipid metabolism and triggers a series of inflammatory responses. Rutin (RT), a flavonoid extracted from plants, exhibits diverse biological activities. However, its potential to mitigate BHBA-induced liver inflammation and lipid accumulation in dairy cows remains unexplored. In this study, we investigated the effect of RT on the BHBA-induced injury of hepatocytes and the possible mechanism. First, hepatocytes were treated with BHBA (0, 0.3, 0.6, 1.2, 2.4 mM) to assess its effects on inflammation impairment and lipid accumulation. Second, hepatocytes were pretreated with RT (0, 25, 50, 100, 150 μg/mL) to evaluate its protective effects. Third, hepatocytes were divided into five treatment groups: blank control, BHBA treatment, RT + BHBA treatment, NF-κB activator (PDTC) + BHBA treatment, and RT + PDTC + BHBA treatment. This experiment further explored the underlying mechanism of RT in mitigating BHBA-induced hepatocyte injury. The results demonstrated that RT at 100 and 150 μg/mL mitigated the increases in hepatocyte interleukin-1 beta (IL-1β), IL-6, triglyceride (TG), and total cholesterol (TC) contents induced by high concentrations of BHBA (p < 0.05). Compared to the BHBA treatment, 100 μg/mL RT significantly downregulated the relative protein expression of P-NF-κB p65 and the relative mRNA expression of NF-κB p65, tumor necrosis factor-alpha (TNF-α), IL-1β, IL-6, peroxisome proliferator-activated receptor gamma (PPARγ), and microsomal triglyceride transfer protein (MTP), while upregulating the relative mRNA expression of IKBα (p < 0.05). Additionally, these effects were more pronounced with the combined pretreatment of the PDTC and RT. In conclusion, RT inhibits BHBA-triggered hepatocyte inflammation and lipid accumulation by modulating the NF-κB signaling pathway, implying that RT may be a promising target for ameliorating damage in SCK cows. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

12 pages, 2262 KB  
Review
From Skeletal Muscle to Myocardium: Molecular Mechanisms of Exercise-Induced Irisin Regulation of Cardiac Fibrosis
by Zhao Wang, Lin Li, Meng Yang, Biao Li and Siyuan Hu
Int. J. Mol. Sci. 2025, 26(8), 3550; https://doi.org/10.3390/ijms26083550 - 10 Apr 2025
Cited by 2 | Viewed by 1203
Abstract
This study systematically elucidates the regulatory mechanisms and potential therapeutic value of the exercise-induced hormone Irisin in the pathological progression of cardiac fibrosis. Through comprehensive analysis and multidimensional data integration, we constructed a complete regulatory network of Irisin within the cardiovascular system, spanning [...] Read more.
This study systematically elucidates the regulatory mechanisms and potential therapeutic value of the exercise-induced hormone Irisin in the pathological progression of cardiac fibrosis. Through comprehensive analysis and multidimensional data integration, we constructed a complete regulatory network of Irisin within the cardiovascular system, spanning its secretion, signal transduction, and precise regulatory control. Our findings demonstrate that exercise intervention significantly elevates circulating Irisin levels via the skeletal muscle–peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)–fibronectin type III domain-containing protein 5 (FNDC5) signaling axis. Irisin establishes a multidimensional molecular barrier against cardiac fibrosis by targeting Sirtuin 1 (Sirt1) activation, inhibiting the transforming growth factor-beta (TGF-β)/Smad3 signaling pathway, and modulating the transcriptional activity of the mitochondrial biogenesis core factors PGC-1α and nuclear respiratory factor 1 (NRF-1). Moreover, the dual regulatory mechanism of the exercise–skeletal muscle–heart axis not only effectively suppresses the aberrant activation of cardiac fibroblasts but also significantly reduces collagen deposition, oxidative stress, and inflammatory infiltration by restoring mitochondrial dynamics balance. Taken together, this study reveals a novel exercise-mediated cardioprotective mechanism at the molecular interaction network level, thereby providing a theoretical basis for the development of non-pharmacological bio-intervention strategies targeting the Irisin signaling pathway and laying a translational foundation for precise exercise prescriptions in cardiovascular diseases. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

16 pages, 3962 KB  
Article
Ark Shell-Derived Peptides AWLNH (P3) and PHDL (P4) Mitigate Foam Cell Formation by Modulating Cholesterol Metabolism and HO-1/Nrf2-Mediated Oxidative Stress in Atherosclerosis
by Chathuri Kaushalya Marasinghe and Jae-Young Je
Mar. Drugs 2025, 23(3), 111; https://doi.org/10.3390/md23030111 - 5 Mar 2025
Cited by 2 | Viewed by 1310
Abstract
Atherosclerosis, a leading contributor to cardiovascular diseases (CVDs), is characterized by foam cell formation driven by excessive lipid accumulation in macrophages and vascular smooth muscle cells. This study elucidates the anti-atherosclerotic potential of AWLNH (P3) and PHDL (P4) peptides by assessing their effects [...] Read more.
Atherosclerosis, a leading contributor to cardiovascular diseases (CVDs), is characterized by foam cell formation driven by excessive lipid accumulation in macrophages and vascular smooth muscle cells. This study elucidates the anti-atherosclerotic potential of AWLNH (P3) and PHDL (P4) peptides by assessing their effects on foam cell formation, lipid metabolism, and oxidative stress regulation. P3 and P4 effectively suppressed intracellular lipid accumulation in RAW264.7 macrophages and human aortic smooth muscle cells (hASMCs), thereby mitigating foam cell formation. Mechanistically, both peptides modulated cholesterol homeostasis by downregulating cholesterol influx mediators, cluster of differentiation 36 (CD36), and class A1 scavenger receptor (SR-A1), while upregulating cholesterol efflux transporters ATP-binding cassette subfamily A member 1 (ABCA1) and ATP-binding cassette subfamily G member 1 (ABCG1). The activation of peroxisome proliferator-activated receptor-gamma (PPAR-γ) and liver X receptor-alpha (LXR-α) further substantiated their role in promoting cholesterol efflux and restoring lipid homeostasis. Additionally, P3 and P4 peptides exhibited potent antioxidative properties by attenuating reactive oxygen species (ROS) generation through activation of the HO-1/Nrf2 signaling axis. HO-1 silencing via siRNA transfection abolished these effects, confirming HO-1-dependent regulation of oxidative stress and lipid metabolism. Collectively, these findings highlight P3 and P4 peptides as promising therapeutic agents for atherosclerosis by concurrently targeting foam cell formation, cholesterol dysregulation, and oxidative stress, warranting further exploration for potential clinical applications. Full article
(This article belongs to the Special Issue Bioactive Proteins and Peptides from Marine Mollusks)
Show Figures

Figure 1

17 pages, 4608 KB  
Article
Proteomics Profiling Reveals Pharmaceutical Excipient PEG400 Induces Nuclear-Receptor-Activation-Affected Lipid Metabolism and Metabolic Enzyme Expression
by Mei Zhao, Siyuan Cao, Dan Yang, Leyuan Shang, Ye Hang, Pengjiao Wang, Shuo Zhang, Chaoji Li, Min Zhang and Xiuli Gao
Int. J. Mol. Sci. 2025, 26(4), 1732; https://doi.org/10.3390/ijms26041732 - 18 Feb 2025
Cited by 2 | Viewed by 1306
Abstract
PEG400 is widely used as a pharmaceutical excipient in the biomedical field. Increasing evidence suggests that PEG400 is not an inert drug carrier; it can influence the activity of various drug-metabolizing enzymes and transporters, thereby affecting the in vivo process of drugs. It [...] Read more.
PEG400 is widely used as a pharmaceutical excipient in the biomedical field. Increasing evidence suggests that PEG400 is not an inert drug carrier; it can influence the activity of various drug-metabolizing enzymes and transporters, thereby affecting the in vivo process of drugs. It can also alleviate obesity and adipose tissue inflammation induced by a high-fat diet. In this study, we employed proteomics to investigate the impact of PEG400 on hepatic protein expression in rats. We found that over 40 metabolic enzymes were altered, with UDP-glucuronosyltransferase 1a9 (Ugt1a9) showing the most significant upregulation. This observation is consistent with our previous findings. KEGG pathway enrichment analysis revealed that PEG400 influences retinol metabolism, steroid hormone biosynthesis, drug metabolism, bile secretion, fatty acid degradation, peroxisome proliferator-activated receptor (PPAR) signaling pathway, and pentose and glucuronate interconversions. Western blot and molecular docking were used to quantitatively analyze related proteins. The results demonstrated that PEG400 promotes the metabolism of retinol to produce retinoic acid; enhances bile secretion by upregulating bile acid synthesis and transporter proteins; and activates the PPARα signaling pathway to regulate the expression of fat metabolism-related proteins, thereby reducing lipid accumulation. Furthermore, as natural ligands for nuclear receptors, retinoic acid and bile acids may activate nuclear receptors and initiate the regulation of target gene expression. We found upregulation of the nuclear receptors PPARα, retinoid X receptor alpha (RXRα), and pregnane X receptor (PXR). RXRα can form a dimer with PPARα or PXR to regulate the expression of target genes, which may explain the changes in the expression of numerous metabolic enzymes. This study provides a comprehensive understanding of the effects of PEG400 on liver metabolism in rats, reveals its potential biological functions, and offers new insights into the application and development of PEG400. Full article
(This article belongs to the Special Issue The Twist and Turn of Lipids in Human Diseases 2.0)
Show Figures

Figure 1

15 pages, 1079 KB  
Review
Pexophagy and Oxidative Stress: Focus on Peroxisomal Proteins and Reactive Oxygen Species (ROS) Signaling Pathways
by Xiaofan Wei, Laxman Manandhar, Hyunsoo Kim, Arun Chhetri, Jaetaek Hwang, Gyuho Jang, Channy Park and Raekil Park
Antioxidants 2025, 14(2), 126; https://doi.org/10.3390/antiox14020126 - 23 Jan 2025
Cited by 6 | Viewed by 2750
Abstract
Peroxisomes generate reactive oxygen species (ROS) and also play a role in protecting cells from the damaging effects of such radicals. Dysfunctional peroxisomes are recognized by receptors and degraded by a selective type of macroautophagy called pexophagy. Oxidative stress is one of the [...] Read more.
Peroxisomes generate reactive oxygen species (ROS) and also play a role in protecting cells from the damaging effects of such radicals. Dysfunctional peroxisomes are recognized by receptors and degraded by a selective type of macroautophagy called pexophagy. Oxidative stress is one of the signals that activates pexophagy through multiple signaling pathways. Conversely, impaired pexophagy results in the accumulation of damaged peroxisomes, which in turn leads to elevated ROS levels and oxidative stress, resulting as cellular dysfunction and the progression of diseases such as neurodegeneration, cancer, and metabolic disorders. This review explores the molecular mechanisms driving pexophagy and its regulation by oxidative stress with a particular focus on ROS. This highlights the role of peroxisomal proteins and ROS-mediated signaling pathways in regulating pexophagy. In addition, emerging evidence suggests that the dysregulation of pexophagy is closely linked to neurological disorders, underscoring its potential as a therapeutic target. Understanding the intricate crosstalk between pexophagy and oxidative stress provides new insights into the maintenance of cellular homeostasis and offers promising directions for addressing neurological disorders that are tightly associated with pexophagy and oxidative stress. Full article
(This article belongs to the Special Issue Crosstalk between Autophagy and Oxidative Stress)
Show Figures

Figure 1

Back to TopTop