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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (185)

Search Parameters:
Keywords = peroxisome-proliferator-associated receptors (PPAR)

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 3771 KiB  
Article
Integrated Transcriptome and Metabolome Analyses Uncover Cholesterol-Responsive Gene Networks
by Ruihao Zhang, Qi Sun, Lixia Huang and Jian Li
Int. J. Mol. Sci. 2025, 26(15), 7108; https://doi.org/10.3390/ijms26157108 - 23 Jul 2025
Viewed by 374
Abstract
Cholesterol stress profoundly modulates cellular processes, but its underlying mechanisms remain incompletely understood. To investigate cholesterol-responsive networks, we performed integrated transcriptome (RNA-seq) and metabolome (LC-MS) analyses on HeLa cells treated with cholesterol for 6 and 24 h. Through transcriptomic analysis of cholesterol-stressed HeLa [...] Read more.
Cholesterol stress profoundly modulates cellular processes, but its underlying mechanisms remain incompletely understood. To investigate cholesterol-responsive networks, we performed integrated transcriptome (RNA-seq) and metabolome (LC-MS) analyses on HeLa cells treated with cholesterol for 6 and 24 h. Through transcriptomic analysis of cholesterol-stressed HeLa cells, we identified stage-specific responses characterized by early-phase stress responses and late-phase immune-metabolic coordination. This revealed 1340 upregulated and 976 downregulated genes after a 6 h cholesterol treatment, including induction and suppression of genes involved in cholesterol efflux and sterol biosynthesis, respectively, transitioning to Nuclear Factor kappa-B (NF-κB) activation and Peroxisome Proliferator-Activated Receptor (PPAR) pathway modulation by 24 h. Co-expression network analysis prioritized functional modules intersecting with differentially expressed genes. We also performed untargeted metabolomics using cells treated with cholesterol for 6 h, which demonstrated extensive remodeling of lipid species. Interestingly, integrated transcriptomic and metabolic analysis uncovered GFPT1-driven Uridine Diphosphate-N-Acetylglucosamine (UDP-GlcNAc) accumulation and increased taurine levels. Validation experiments confirmed GFPT1 upregulation and ANGPTL4 downregulation through RT-qPCR and increased O-GlcNAcylation via Western blot. Importantly, clinical datasets further supported the correlations between GFPT1/ANGPTL4 expression and cholesterol levels in Non-Alcoholic Steatohepatitis (NASH) liver cancer patients. This work establishes a chronological paradigm of cholesterol sensing and identifies GFPT1 and ANGPTL4 as key regulators bridging glycosylation and lipid pathways, providing mechanistic insights into cholesterol-associated metabolic disorders. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

39 pages, 865 KiB  
Review
Current and Emerging Treatments for Metabolic Associated Steatotic Liver Disease and Diabetes: A Narrative Review
by Rachelle Choi, Jatin Vemuri, Alekya Poloju, Rishi Raj, Anurag Mehta, Amon Asgharpour, Mohammad S. Siddiqui and Priyanka Majety
Endocrines 2025, 6(2), 27; https://doi.org/10.3390/endocrines6020027 - 5 Jun 2025
Viewed by 1235
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), previously referred to as Non-Alcoholic Fatty Liver Disease (NAFLD), is a prevalent chronic liver condition strongly linked to Type 2 Diabetes Mellitus (T2DM) and obesity. Globally, MASLD is the most common cause of chronic liver disease. The [...] Read more.
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), previously referred to as Non-Alcoholic Fatty Liver Disease (NAFLD), is a prevalent chronic liver condition strongly linked to Type 2 Diabetes Mellitus (T2DM) and obesity. Globally, MASLD is the most common cause of chronic liver disease. The bidirectional relationship between MASLD and T2DM underscores the pivotal role of insulin resistance in disease progression, which contributes to hepatic steatosis, oxidative stress, and inflammation, forming a vicious cycle. MASLD is also associated with heightened risks of cardiovascular and chronic kidney diseases, necessitating comprehensive treatment approaches. While lifestyle interventions and weight loss remain the cornerstone of management, their sustainability is challenging. This review highlights the evolving pharmacological landscape targeting MASLD and its advanced form, Metabolic Dysfunction-Associated Steatohepatitis (MASH). Currently, Resmetirom is the only FDA-approved drug for MASH. Current and investigational therapies, including insulin-sensitizing agents like peroxisome proliferator-activated receptor (PPAR) agonists, glucose-lowering drugs such as sodium-glucose co-transporter 2 inhibitors (SGLT2i) and glucagon-like peptide-1 receptor agonists (GLP-1 RA), drugs that target intermediary metabolism such as Vitamin E, de novo lipogenesis inhibitors, and emerging agents targeting the gut-liver axis and oxidative stress, are explored. These therapies demonstrate promising effects on hepatic steatosis, inflammation, and fibrosis, providing new avenues to address the multifaceted pathophysiology of MASLD. Full article
(This article belongs to the Special Issue Feature Papers in Endocrines: 2024)
Show Figures

Figure 1

15 pages, 1767 KiB  
Brief Report
β-Hydroxybutyrate Reduces Body Weight by Modulating Fatty Acid Oxidation and Beiging in the Subcutaneous Adipose Tissue of DIO Mice
by Violeta Heras, Virginia Mela, Pallavi Kompella, Elena Rojano, Guillermo Paz-López, Lucia Hurtado-García, Almudena Ortega-Gomez, Maria José García-López, María Luisa García-Martín, Juan A. G. Ranea, Francisco J. Tinahones and Isabel Moreno-Indias
Int. J. Mol. Sci. 2025, 26(11), 5064; https://doi.org/10.3390/ijms26115064 - 24 May 2025
Viewed by 756
Abstract
β-hydroxybutyrate (BHB) serves as an alternative cellular fuel during states of low glucose availability, such as fasting or carbohydrate restriction, when the body shifts to using fats and ketone bodies for energy. While BHB has shown potential metabolic benefits, its mechanisms of action [...] Read more.
β-hydroxybutyrate (BHB) serves as an alternative cellular fuel during states of low glucose availability, such as fasting or carbohydrate restriction, when the body shifts to using fats and ketone bodies for energy. While BHB has shown potential metabolic benefits, its mechanisms of action in the context of obesity are not fully understood. In this study, we examined the effects of BHB supplementation on subcutaneous adipose tissue (SAT) metabolism in a diet-induced obesity (DIO) mouse model. Adult male mice were first fed a high-fat diet for six weeks, followed by a standard diet with or without BHB supplementation for an additional six weeks. BHB supplementation led to significant body weight loss independent of food intake. This weight reduction was associated with decreased adipocyte differentiation, reflected by reduced peroxisome proliferator-activated receptor gamma (PPARγ) protein levels and lower uncoupling protein 1 (UCP1) expression, indicating altered SAT function. Transcriptomic analysis of SAT revealed upregulation of genes involved in fatty acid activation and transport (e.g., Slc27a2, Plin5, Acot4, Acsm3, Rik). Functional enrichment highlighted the activation of the PPAR signaling pathway and enrichment of peroxisomal components in the BHB group. Together, these results suggest that BHB promotes lipid remodeling in SAT, enhancing fatty acid metabolism while suppressing thermogenic pathways, and thus may represent a novel mechanism contributing to adiposity reduction and metabolic improvement. Full article
Show Figures

Graphical abstract

19 pages, 1570 KiB  
Article
Hexaraphane Affects the Activation of Hepatic PPARα Signaling: Impact on Plasma Triglyceride Levels and Hepatic Senescence with Aging
by Manami Higa, Kazuma Naito, Takenari Sato, Ayame Tomii, Yuuka Hitsuda, Miyu Tahara, Katsunori Ishii, Yu Ichisaka, Hikaru Sugiyama, Rin Kobayashi, Fuzuki Sakamoto, Kazuhisa Watanabe, Keisuke Yoshikiyo and Hidehisa Shimizu
Nutrients 2025, 17(11), 1768; https://doi.org/10.3390/nu17111768 - 23 May 2025
Viewed by 522
Abstract
Background/Objectives: Hexaraphane, also known as 6-methylsulfinylhexyl isothiocyanate, derived from wasabi (Eutrema japonicum), increases heme oxygenase-1 (HO-1) and aldehyde dehydrogenase 2 (ALDH2) mRNA expression by activating nuclear factor erythroid 2-related factor 2 (Nrf2) in both HepG2 cells and the mouse liver. [...] Read more.
Background/Objectives: Hexaraphane, also known as 6-methylsulfinylhexyl isothiocyanate, derived from wasabi (Eutrema japonicum), increases heme oxygenase-1 (HO-1) and aldehyde dehydrogenase 2 (ALDH2) mRNA expression by activating nuclear factor erythroid 2-related factor 2 (Nrf2) in both HepG2 cells and the mouse liver. Given the presence of a peroxisome proliferator-activated receptor (PPAR) response element (PPRE) in the HO-1 and ALDH2 promoters, the present study aimed to determine the effects of hexaraphane on PPARα-associated genes, age-related weight gain, plasma triglyceride levels, and hepatic senescence. Methods: HepG2 cells were treated with hexaraphane to evaluate PPARα target gene expression and PPRE transcriptional activity. Male C57BL/6J young control, aged control, and aged mice administered with hexaraphane for 16 weeks were assessed for food and water intake, body and tissue weights, plasma parameters, and hepatic PPARα-related gene expression. Results: Hexaraphane increased HO-1 mRNA expression levels in HepG2 cells, which was inhibited by GW6471, a PPARα antagonist. It elevated PPRE transcriptional activity and increased carnitine palmitoyltransferase 1A (CPT1A) mRNA expression levels, indicating PPARα activation. In aged mice, hexaraphane intake reduced body weight gain by decreasing the adipose tissue weight. Increased CPT1A expression levels and a tendency toward increased acyl-CoA oxidase 1 (ACOX1) expression levels in the liver of aged mice administered hexaraphane were associated with reduced plasma triglyceride levels and body weight gain. Increased hepatic Sirt1 expression levels in aged mice administered hexaraphane was associated with lower plasma triglyceride levels. Increased hepatic PPARα mRNA expression levels in aged mice administered hexaraphane suggest a positive feedback loop between PPARα and Sirt1. The expression levels of hepatic p21 mRNA, a senescence marker regulated by Sirt1, were upregulated in aged mice but suppressed by hexaraphane intake. Conclusions: Hexaraphane may prevent age-related body weight gain, elevated plasma triglyceride levels, and hepatic senescence by activating PPARα, potentially contributing to longevity. Full article
(This article belongs to the Special Issue Association Between Lipid Metabolism and Obesity)
Show Figures

Figure 1

19 pages, 788 KiB  
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 3474
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, 3827 KiB  
Article
Luteolin Relieves Metabolic Dysfunction-Associated Fatty Liver Disease Caused by a High-Fat Diet in Rats Through Modulating the AdipoR1/AMPK/PPARγ Signaling Pathway
by Pongsakorn Taweesap, Prapassorn Potue, Juthamas Khamseekaew, Metee Iampanichakul, Banyaphon Jan-O, Poungrat Pakdeechote and Putcharawipa Maneesai
Int. J. Mol. Sci. 2025, 26(8), 3804; https://doi.org/10.3390/ijms26083804 - 17 Apr 2025
Cited by 2 | Viewed by 903
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a significant global public health issue. Luteolin possesses several beneficial biological properties, including antioxidation and anti-inflammation. This study investigated luteolin’s effect and potential mechanisms on MAFLD in high-fat diet (HFD)-fed rats. Rats were administered an HFD [...] Read more.
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a significant global public health issue. Luteolin possesses several beneficial biological properties, including antioxidation and anti-inflammation. This study investigated luteolin’s effect and potential mechanisms on MAFLD in high-fat diet (HFD)-fed rats. Rats were administered an HFD supplemented with fructose for 12 weeks to induce MAFLD. After that, the HFD-fed rats were given either luteolin (50 or 100 mg/kg/day) or metformin (100 mg/kg/day) for 4 weeks. Luteolin improved metabolic parameters induced by the HFD, since it decreased body weight, blood pressure, fasting blood glucose, serum insulin, free fatty acids, cholesterol, and triglyceride levels (p < 0.05). Luteolin reduced hepatic injury and inflammatory markers in HFD-fed rats (p < 0.05). Additionally, HFD-fed rats treated with luteolin showed reduced malondialdehyde and raised catalase activity in plasma (p < 0.05). Luteolin attenuated hepatic steatosis compared to the untreated rats (p < 0.05). Luteolin also increased plasma adiponectin levels accompanied by upregulation of adiponectin receptor 1 (AdipoR1), AMP-activated protein kinase (AMPK), and peroxisome proliferator-activated receptor γ (PPAR-γ) protein expression in liver (p < 0.05). These findings revealed that luteolin ameliorated HFD-induced MAFLD in rats, possibly by reducing metabolic alterations and oxidative stress and restoring AdipoR1, AMPK, and PPARγ protein expression in HFD-fed rats. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

14 pages, 2032 KiB  
Article
Vaccinium oldhamii Fruit Inhibits Lipid Accumulation in 3T3-L1 Cells and Diet-Induced Obese Animals
by Young-Hyeon Lee, Mikyoung You and Hyeon-A Kim
Nutrients 2025, 17(8), 1346; https://doi.org/10.3390/nu17081346 - 14 Apr 2025
Viewed by 641
Abstract
Background/Objectives: Obesity is a significant global health concern, and the natural bioactive compounds with anti-obesity effects remain challenging. This study aims to examine the anti-obesity effect and the potential mechanism of Vaccinium oldhamii fruit water extract (VOW). Methods: Lipid accumulation, AMP-activated protein kinase [...] Read more.
Background/Objectives: Obesity is a significant global health concern, and the natural bioactive compounds with anti-obesity effects remain challenging. This study aims to examine the anti-obesity effect and the potential mechanism of Vaccinium oldhamii fruit water extract (VOW). Methods: Lipid accumulation, AMP-activated protein kinase (AMPK) activity, and Wnt/β-catenin signaling were evaluated in 3T3-L1 cells. In high-fat and high-sucrose diet (HFHSD)-induced obese mice, body weight, food intake, fat weight, serum lipid profiles, and adipogenic transcription factors were assessed. The most effective VOW fraction was selected by Oil Red O (ORO) staining and its mechanism was studied in 3T3-L1 cells. Results: VOW treatment significantly inhibited cellular lipid accumulation and suppressed phosphorylation of AMPK and its downstream protein, acetyl-CoA carboxylase (ACC). VOW also decreased adipogenic-associated protein expressions such as the peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAAT/enhancer-binding proteins α (C/EBP α), sterol regulatory element binding protein-1c (SREBP-1c), and fatty acid synthase (FAS). The enhanced effect of VOW was abolished by the knockdown of AMPK with siRNA. The inhibitory effect of VOW on differentiation depended on the treatment period, even though VOW treatment downregulated the C/EBP β expression at the early phase of differentiation. VOW dramatically reduced activation of AMPK, thereby downregulating adipogenic-associated proteins. Furthermore, the butanol fraction (BtOH) of VOW showed the most powerful effect of VOW dose-dependently reduced lipid accumulation by suppressing the phosphorylation of AMPK. Consistent with inhibited lipid accumulation in vitro, VOW reduced body weight and white adipose tissue weight in the HFHSD-induced obese animal model. Conclusions: Overall, our study suggested that the anti-adipogenesis effect of VOW and its BtOH fraction involved the activation of AMPK. Full article
(This article belongs to the Special Issue Obesity and Related Diseases: The Role of Nutrition)
Show Figures

Figure 1

42 pages, 1204 KiB  
Review
Metabolic-Dysfunction-Associated Steatotic Liver Disease: Molecular Mechanisms, Clinical Implications, and Emerging Therapeutic Strategies
by Jeysson E. Mejía-Guzmán, Ramón A. Belmont-Hernández, Norberto C. Chávez-Tapia, Misael Uribe and Natalia Nuño-Lámbarri
Int. J. Mol. Sci. 2025, 26(7), 2959; https://doi.org/10.3390/ijms26072959 - 25 Mar 2025
Cited by 5 | Viewed by 2966
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is a highly prevalent metabolic disorder characterized by hepatic steatosis in conjunction with at least one cardiometabolic risk factor, such as obesity, type 2 diabetes, hypertension, or dyslipidemia. As global [...] Read more.
Metabolic-dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is a highly prevalent metabolic disorder characterized by hepatic steatosis in conjunction with at least one cardiometabolic risk factor, such as obesity, type 2 diabetes, hypertension, or dyslipidemia. As global rates of obesity and metabolic syndrome continue to rise, MASLD is becoming a major public health concern, with projections indicating a substantial increase in prevalence over the coming decades. The disease spectrum ranges from simple steatosis to metabolic-dysfunction-associated steatohepatitis (MASH), fibrosis, cirrhosis, and hepatocellular carcinoma, contributing to significant morbidity and mortality worldwide. This review delves into the molecular mechanisms driving MASLD pathogenesis, including dysregulation of lipid metabolism, chronic inflammation, oxidative stress, mitochondrial dysfunction, and gut microbiota alterations. Recent advances in research have highlighted the role of genetic and epigenetic factors in disease progression, as well as novel therapeutic targets such as peroxisome proliferator-activated receptors (PPARs), fibroblast growth factors, and thyroid hormone receptor beta agonists. Given the multifaceted nature of MASLD, a multidisciplinary approach integrating early diagnosis, molecular insights, lifestyle interventions, and personalized therapies is critical. This review underscores the urgent need for continued research into innovative treatment strategies and precision medicine approaches to halt MASLD progression and improve patient outcomes. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

18 pages, 43536 KiB  
Article
Role of Adiponectin in Regulating Cytokines and Its Contribution to the Occurrence and Progression of Clinical Mastitis in Holstein Cows
by Junjun Zhang, Na Chen, Zhen Yang, Yumeng Gao, Bohao Zhang, Jianfu Li, Bin Zhou, Zhixiong Tang, Weitao Dong, Xingxu Zhao, Yong Zhang and Quanwei Zhang
Int. J. Mol. Sci. 2025, 26(7), 2898; https://doi.org/10.3390/ijms26072898 - 22 Mar 2025
Viewed by 561
Abstract
Cytokines are crucial in various physiological and pathological processes, especially in inflammatory diseases in mammals. However, the comprehensive identification of cytokines and their potential regulatory functions in the mammary glands of Holstein cows suffering from clinical mastitis (CM) remains only partially understood. This [...] Read more.
Cytokines are crucial in various physiological and pathological processes, especially in inflammatory diseases in mammals. However, the comprehensive identification of cytokines and their potential regulatory functions in the mammary glands of Holstein cows suffering from clinical mastitis (CM) remains only partially understood. This study aimed to systematically identify biological processes (BPs) and differentially expressed proteins (DEPs) associated with cytokines and to explore their functions through the analysis of previously obtained data from data-independent acquisition (DIA) proteomics. We confirmed that the dynamic balance between pro- and anti-inflammatory factors is closely associated with dairy mastitis. A total of 4 BPs, comprising 75 upregulated and 16 downregulated DEPs, were identified, particularly in relation to adiponectin (ADIPOQ), which strongly interacts with the other DEPs and participates in peroxisome proliferator-activated receptor (PPAR) and adipocytokine signaling pathways. Immunohistochemical and immunofluorescence staining revealed that ADIPOQ was predominantly localized in the cytoplasm of mammary epithelial cells. Moreover, the expression levels of ADIPOQ mRNA and protein in the mammary glands of the CM group were notably reduced compared to those in the healthy group. A potential mechanism of action of ADIPOQ was suggested, with findings indicating that a decrease in ADIPOQ expression could potentially worsen inflammation in CM. These results offer novel insights into cytokines and the regulatory mechanisms of ADIPOQ in Holstein cows with CM which may benefit the prevention and treatment of dairy mastitis. Full article
(This article belongs to the Topic Advances in Adiponectin)
Show Figures

Figure 1

22 pages, 2381 KiB  
Review
Therapeutic Potential and Challenges of Pioglitazone in Cancer Treatment
by Maria Vasileiou, Sotirios Charalampos Diamantoudis, Christina Tsianava and Nam P. Nguyen
Appl. Sci. 2025, 15(4), 1925; https://doi.org/10.3390/app15041925 - 13 Feb 2025
Cited by 2 | Viewed by 2232
Abstract
Pioglitazone (ACTOS) is a thiazolidinedione for peroxisome proliferator-activated receptor γ (PPAR-γ) that has been well established for the second or third line treatment of type 2 diabetes mellitus. Beyond the effects on glucose metabolism, pioglitazone displays positive effects on lipid metabolism, blood pressure, [...] Read more.
Pioglitazone (ACTOS) is a thiazolidinedione for peroxisome proliferator-activated receptor γ (PPAR-γ) that has been well established for the second or third line treatment of type 2 diabetes mellitus. Beyond the effects on glucose metabolism, pioglitazone displays positive effects on lipid metabolism, blood pressure, endothelial function, bone density, and apoptosis of cancer cells. In fact, according to in vitro experiments and preclinical studies, PPAR-γ ligand is currently considered a potential target for both chemoprevention and cancer therapy. PPAR-γ ligands are known to inhibit cancer cell proliferation and metastasis through terminal differentiation and underexpression of inflammatory mediators. Despite its anticancer properties, pioglitazone was withdrawn by the national medicine agencies of France and Germany, due to reports of increased incidence of bladder cancer. These reports were associated with European populations undergoing higher doses and longer durations of treatment. In this review, we discuss the pharmacokinetics, therapeutic potential, and limitations regarding the clinical use of pioglitazone, with a focus on cancer treatment. Full article
Show Figures

Figure 1

33 pages, 3111 KiB  
Review
Targeting Ferroptosis in Parkinson’s: Repurposing Diabetes Drugs as a Promising Treatment
by Carmen Duță, Corina Muscurel, Carmen Beatrice Dogaru and Irina Stoian
Int. J. Mol. Sci. 2025, 26(4), 1516; https://doi.org/10.3390/ijms26041516 - 11 Feb 2025
Cited by 1 | Viewed by 1576
Abstract
This review explores the promising potential of repurposing type 2 diabetes (T2D) medications for the treatment of Parkinson’s disease (PD), highlighting the shared pathophysiological mechanisms between these two age-related conditions, such as oxidative stress, mitochondrial dysfunction, and ferroptosis. The overlap suggests that existing [...] Read more.
This review explores the promising potential of repurposing type 2 diabetes (T2D) medications for the treatment of Parkinson’s disease (PD), highlighting the shared pathophysiological mechanisms between these two age-related conditions, such as oxidative stress, mitochondrial dysfunction, and ferroptosis. The overlap suggests that existing diabetes drugs could target the common pathways involved in both conditions. Specifically, the review discusses how T2D medications, including metformin (Met), peroxisome-proliferator-activated receptor gamma (PPAR-γ) agonists, sodium-glucose cotransporter-2 (SGLT2) inhibitors, incretins, and dipeptidyl-peptidase 4 (DPP-4) inhibitors, can improve mitochondrial function, reduce neuroinflammation and oxidative stress, and potentially inhibit ferroptosis. The connection between ferroptosis and existing treatments, including diabetes medication, are only beginning to be explored. The limited data can be attributed also to the complexity of mechanisms involved in ferroptosis and Parkinson’s disease and to the fact that the specific role of ferroptosis in Parkinson’s disease pathogenesis has not been a primary focus until recent. Despite the promising preclinical evidence, clinical findings are mixed, underscoring the need for further research to elucidate these drugs’ roles in neurodegeneration. Repurposing existing diabetes medications that have well-established safety profiles for Parkinson’s disease treatment could significantly reduce the time and cost associated with drug development and could offer a more comprehensive approach to managing Parkinson’s disease compared to treatments targeting a single mechanism. Full article
(This article belongs to the Special Issue Challenges and Innovation in Neurodegenerative Diseases, 2nd Edition)
Show Figures

Figure 1

17 pages, 1870 KiB  
Review
The General Principle of the Warburg Effect as a Possible Approach for Cancer Immunotherapy: The Regulatory Effect of Plant Extracts Could Change the Game
by Donika Ivanova, Severina Semkova, Boncho Grigorov, Milena Tzanova, Ana Georgieva, Dancho Danchev, Biliana Nikolova and Zvezdelina Yaneva
Molecules 2025, 30(2), 393; https://doi.org/10.3390/molecules30020393 - 18 Jan 2025
Viewed by 1511
Abstract
The interpretation of the biochemistry of immune metabolism could be considered an attractive scientific field of biomedicine research. In this review, the role of glycolysis in macrophage polarization is discussed together with mitochondrial metabolism in cancer cells. In the first part, the focus [...] Read more.
The interpretation of the biochemistry of immune metabolism could be considered an attractive scientific field of biomedicine research. In this review, the role of glycolysis in macrophage polarization is discussed together with mitochondrial metabolism in cancer cells. In the first part, the focus is on the Warburg effect and redox metabolism during macrophage polarization, cancer development, and management of the immune response by the cancer cells. The second part addresses the possibility of impacts on the Warburg effect through targeting peroxisome proliferator-activated receptors (PPARs). This could be an activator of native immune responses. Because of the reported serious adverse effects of using synthetic ligands for PPARs in combination with chemotherapeutics, searches for less toxic and more active PPAR inhibitors, as well as blocking undesirable cellular PPAR-dependent processes, are in progress. On the other hand, recent research in modern immunotherapy has focused on the search for gentle immune-modulating natural compounds with harmless synergistic chemotherapeutic efficacy that can be used as an adjuvant. It is a well-known fact that the plant kingdom is a source of important therapeutic agents with multifaceted effectiveness. One of these is the known association with PPAR activities. In this regard, the secondary metabolites extracted from plants could change the game. Full article
(This article belongs to the Special Issue Bioactive Molecules in Medicinal Plants)
Show Figures

Figure 1

13 pages, 1680 KiB  
Article
Identification of Genetic Markers of APOM and CYP7A1 Genes Affecting Milk Production Traits in Chinese Holstein
by Yanan Liu, Zijiao Guo, Junqing Ni, Chendong Yang, Bo Han, Yabin Ma, Jianming Li, Guie Jiang, Weijie Zheng and Dongxiao Sun
Agriculture 2025, 15(1), 33; https://doi.org/10.3390/agriculture15010033 - 26 Dec 2024
Viewed by 667
Abstract
Our previous study identified the apolipoprotein M (APOM) and cytochrome P450 family 7 subfamily A polypeptide 1 (CYP7A1) genes as candidates for milk traits in dairy cattle, which were significantly up-regulated in liver tissue of Holstein cows between the [...] Read more.
Our previous study identified the apolipoprotein M (APOM) and cytochrome P450 family 7 subfamily A polypeptide 1 (CYP7A1) genes as candidates for milk traits in dairy cattle, which were significantly up-regulated in liver tissue of Holstein cows between the dry and lactation periods. The two genes play critical roles in the peroxisome proliferator-activated receptor (PPAR) pathway. In this study, we further confirmed whether the APOM and CYP7A1 genes had significant genetic impacts on milk production traits in a Chinese Holstein population. By dual-direction sequencing of the polymerase chain reaction (PCR) products of the complete coding sequences and 2000 bp of the 5′ and 3′ flanking regions on pooled DNA sample, seven and three single nucleotide polymorphisms (SNPs) were identified in APOM and CYP7A1, respectively. With SAS 9.2, phenotype-genotype association analysis revealed such SNPs were significantly associated with at least one of the milk production traits, including 305-day milk yield, milk fat yield, milk fat percentage, milk protein yield, and milk protein percentage in the first and second lactations (p = <0.01~0.04). With Haploview 4.2, we further found that six SNPs in APOM and thee SNPs in CYP7A1 formed one haplotype, respectively. The haplotypes were significantly associated with at least one of milk production traits as well (p = <0.01~0.02). Of note, we found the SNPs in the 5′ regulatory region, rs209293266 and rs110721287 in APOM and rs42765359 in CYP7A1, significantly impacted the gene transcriptional activity after mutation (p < 0.01) through changing the transcription factor binding sites by using luciferase assay experiments. Additionally, with RNAfold Web Server, rs110098953 and rs378530166 changed the mRNA secondary structures of APOM and CYP7A1 genes, respectively. In summary, our research is the first to demonstrate that APOM and CYP7A1 genes have significantly genetic effects on milk yield and composition traits, and the identified SNPs may serve as available genetic markers for genomic selection program in dairy cattle. Full article
(This article belongs to the Section Farm Animal Production)
Show Figures

Figure 1

13 pages, 4334 KiB  
Article
Effects of Metabolites Derived from Guava (Psidium guajava L.) Leaf Extract Fermented by Limosilactobacillus fermentum on Hepatic Energy Metabolism via SIRT1-PGC1α Signaling in Diabetic Mice
by Sohyun Jeon, Heaji Lee, Sun-Yeou Kim, Choong-Hwan Lee and Yunsook Lim
Nutrients 2025, 17(1), 7; https://doi.org/10.3390/nu17010007 - 24 Dec 2024
Cited by 1 | Viewed by 1940
Abstract
Background/Objectives: Type 2 diabetes mellitus (T2DM) is considered a serious risk to public health since its prevalence is rapidly increasing worldwide despite numerous therapeutics. Insulin resistance in T2DM contributes to chronic inflammation and other metabolic abnormalities that generate fat accumulation in the liver, [...] Read more.
Background/Objectives: Type 2 diabetes mellitus (T2DM) is considered a serious risk to public health since its prevalence is rapidly increasing worldwide despite numerous therapeutics. Insulin resistance in T2DM contributes to chronic inflammation and other metabolic abnormalities that generate fat accumulation in the liver, eventually leading to the progression of metabolic dysfunction-associated fatty liver disease (MAFLD). Recently, the possibility that microbial-derived metabolites may alleviate MAFLD through enterohepatic circulation has emerged, but the underlying mechanism remains unclear. In this research, we utilized metabolites obtained from the fermentation of guava leaf extract, which is well-known for its antidiabetic activity, to investigate their effects and mechanisms on MAFLD. Methods: Diabetes was induced by a high-fat diet and streptozotocin injection (80 mg/kg body weight) twice in mice. Subsequently, mice whose fasting blood glucose levels were measured higher than 300 mg/dL were administered with metabolites of Limosilactobacillus fermentum (LF) (50 mg/kg/day) or guava leaf extract fermented by L. fermentum (GFL) (50 mg/kg/day) by gavage for 15 weeks. Results: GFL supplementation mitigated hyperglycemia and hepatic insulin resistance. Moreover, GFL regulated abnormal hepatic histological changes and lipid profiles in diabetic mice. Furthermore, GFL enhanced energy metabolism by activating the sirtuin1 (SIRT1)/proliferator-activated receptor γ coactivator 1α (PGC1α)/peroxisome proliferator-activated receptor (PPAR)-α pathway in diabetic mice. Meanwhile, GFL supplementation suppressed hepatic inflammation in diabetic mice. Conclusions: Taken together, the current study elucidated that GFL could be a potential therapeutic to ameliorate hyperglycemia and hepatic steatosis by improving SIRT1/PGC-1α/ PPAR-α-related energy metabolism in T2DM. Full article
(This article belongs to the Section Nutrition and Diabetes)
Show Figures

Figure 1

14 pages, 1083 KiB  
Review
Palmitoylethanolamide in Postmenopausal Metabolic Syndrome: Current Evidence and Clinical Perspectives
by Alessandro Medoro, Sergio Davinelli, Federica Fogacci, Stefania Alfieri, Domenico Tiso, Arrigo F. G. Cicero and Giovanni Scapagnini
Nutrients 2024, 16(24), 4313; https://doi.org/10.3390/nu16244313 - 13 Dec 2024
Cited by 2 | Viewed by 3376
Abstract
Menopause leads to a decline in estrogen levels, resulting in significant metabolic alterations that increase the risk of developing metabolic syndrome—a cluster of conditions including central obesity, insulin resistance, dyslipidemia, and hypertension. Traditional interventions such as hormone replacement therapy carry potential adverse effects, [...] Read more.
Menopause leads to a decline in estrogen levels, resulting in significant metabolic alterations that increase the risk of developing metabolic syndrome—a cluster of conditions including central obesity, insulin resistance, dyslipidemia, and hypertension. Traditional interventions such as hormone replacement therapy carry potential adverse effects, and lifestyle modifications alone may not suffice for all women. This review explores the potential role of palmitoylethanolamide (PEA), an endogenous fatty acid amide, in managing metabolic syndrome during the postmenopausal period. PEA primarily acts by activating peroxisome proliferator-activated receptor-alpha (PPAR-α), influencing lipid metabolism, energy homeostasis, and inflammation. Evidence indicates that PEA may promote the browning of white adipocytes, enhancing energy expenditure and reducing adiposity. It also improves lipid profiles by boosting fatty acid oxidation and decreasing lipid synthesis, potentially lowering low-density lipoprotein cholesterol and triglyceride levels while increasing high-density lipoprotein cholesterol. Additionally, the anti-inflammatory properties of PEA enhance insulin sensitivity by reducing pro-inflammatory cytokines that interfere with insulin signaling. PEA may aid in weight management by influencing appetite regulation and improving leptin sensitivity. Furthermore, its neuroprotective effects may address the mood disturbances and cognitive decline associated with menopause. Given these multifaceted biological activities and a favorable safety profile, PEA may represent a promising non-pharmacological supplement for managing metabolic syndrome in postmenopausal women. However, further large-scale clinical studies are necessary to establish its efficacy, optimal dosing, and long-term safety. If validated, PEA could become an integral part of strategies to improve metabolic and neuropsychological health outcomes in this population. Full article
Show Figures

Figure 1

Back to TopTop