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

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

Search Results (753)

Search Parameters:
Keywords = proliferator-activated receptor γ

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 594 KiB  
Review
PEDF and Its Role in Metabolic Disease, Angiogenesis, Cardiovascular Disease, and Diabetes
by Crispin R. Dass
Biomedicines 2025, 13(7), 1780; https://doi.org/10.3390/biomedicines13071780 - 21 Jul 2025
Viewed by 447
Abstract
This review highlights recent findings on the potent anti-angiogenic serpin protein, pigment epithelium-derived factor (PEDF) as it relates to metabolic disease, diabetes, angiogenesis and cardiovascular disease (CVD), listing a majority of all the publicly available studies reported to date. PEDF is involved in [...] Read more.
This review highlights recent findings on the potent anti-angiogenic serpin protein, pigment epithelium-derived factor (PEDF) as it relates to metabolic disease, diabetes, angiogenesis and cardiovascular disease (CVD), listing a majority of all the publicly available studies reported to date. PEDF is involved in various physiological roles in the body, and when awry, it triggers various disease states clinically. Biomarkers such as insulin, AMP-activated protein kinase alpha (AMPK-α), and peroxisome proliferator-activated receptor gamma (PPAR-γ) are involved in PEDF effects on metabolism. Wnt, insulin receptor substate (IRS), Akt, extracellular signal-regulated kinase (ERK), and mitogen-activated protein kinase (MAPK) are implicated in diabetes effects displayed by PEDF. For CVD, oxidised LDL, Wnt/β-catenin, and reactive oxygen species (ROS) are players intertwined with PEDF activity. The review also presents an outlook on where efforts could be devoted to bring this serpin closer to clinical trials for these diseases and others in general. Full article
Show Figures

Figure 1

27 pages, 2385 KiB  
Review
Butyrate Produced by Gut Microbiota Regulates Atherosclerosis: A Narrative Review of the Latest Findings
by Leon M. T. Dicks
Int. J. Mol. Sci. 2025, 26(14), 6744; https://doi.org/10.3390/ijms26146744 - 14 Jul 2025
Viewed by 641
Abstract
Atherosclerosis (AS), a progressive inflammatory disease of coronary arteries, the aorta, and the internal carotid artery, is considered one of the main contributors to cardiovascular disorders. Blood flow is restricted by accumulating lipid-rich macrophages (foam cells), calcium, fibrin, and cellular debris into plaques [...] Read more.
Atherosclerosis (AS), a progressive inflammatory disease of coronary arteries, the aorta, and the internal carotid artery, is considered one of the main contributors to cardiovascular disorders. Blood flow is restricted by accumulating lipid-rich macrophages (foam cells), calcium, fibrin, and cellular debris into plaques on the intima of arterial walls. Butyrate maintains gut barrier integrity and modulates immune responses. Butyrate regulates G-protein-coupled receptor (GPCR) signaling and activates nuclear factor kappa-B (NF-κB), activator protein-1 (AP-1), and interferon regulatory factors (IFRs) involved in the production of proinflammatory cytokines. Depending on the inflammatory stimuli, butyrate may also inactivate NF-κB, resulting in the suppression of proinflammatory cytokines and the stimulation of anti-inflammatory cytokines. Butyrate modulates mitogen-activated protein kinase (MAPK) to promote or suppress macrophage inflammation, muscle cell growth, apoptosis, and the uptake of oxidized low-density lipoprotein (ox-LDL) in macrophages. Activation of the peroxisome proliferator-activated receptor γ (PPARγ) pathway plays a role in lipid metabolism, inflammation, and cell differentiation. Butyrate inhibits interferon γ (IFN-γ) signaling and suppresses NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) involved in inflammation and scar tissue formation. The dual role of butyrate in AS is discussed by addressing the interactions between butyrate, intestinal epithelial cells (IECs), endothelial cells (ECs) of the main arteries, and immune cells. Signals generated from these interactions may be applied in the diagnosis and intervention of AS. Reporters to detect early AS is suggested. This narrative review covers the most recent findings published in PubMed and Crossref databases. Full article
Show Figures

Figure 1

13 pages, 2934 KiB  
Article
Mechanotransductive Activation of PPAR-γ by Low-Intensity Pulsed Ultrasound Induces Contractile Phenotype in Corpus Spongiosum Smooth Muscle Cells
by Huan Yu, Jianying Li, Zihan Xu, Zhiwei Peng, Min Wu, Yiqing Lv, Fang Chen, Mingming Yu and Yichen Huang
Biomedicines 2025, 13(7), 1701; https://doi.org/10.3390/biomedicines13071701 - 11 Jul 2025
Viewed by 347
Abstract
Background: Previously, we found that the pathological changes in the corpus spongiosum (CS) in hypospadias were mainly localized within smooth muscle tissue, presenting as a transformation from the contraction phenotype to synthesis. The role of low-intensity pulsed ultrasound (LIPUS) in regulating smooth muscle [...] Read more.
Background: Previously, we found that the pathological changes in the corpus spongiosum (CS) in hypospadias were mainly localized within smooth muscle tissue, presenting as a transformation from the contraction phenotype to synthesis. The role of low-intensity pulsed ultrasound (LIPUS) in regulating smooth muscle cells (SMCs) and angiogenesis has been confirmed. Objectives: To demonstrate the feasibility of regulating the phenotypic transformation of corpus spongiosum smooth muscle cells (CSSMCs) in hypospadias using LIPUS and to explore the potential mechanisms. Materials and Methods: The CSSMCs were extracted from CS in patients with proximal hypospadias. In vitro experiments were conducted to explore the appropriate LIPUS irradiation intensity and duration which could promote the phenotypic transformation of CSSMCs. A total of 71 patients with severe hypospadias were randomly divided into a control group and a LIPUS group to verify the in vivo transition effect of LIPUS. Consequently, the potential mechanisms by which LIPUS regulates the phenotypic transformation of CSSMCs were explored in vitro. Results: In vitro experiments showed that LIPUS with an intensity of 100 mW/cm2 and a duration of 10 min could significantly increase the expression of contraction markers in CSSMCs and decrease the expression of synthesis markers. Moreover, LIPUS stimulation could alter the phenotype of CSSMCs in patients with proximal hypospadias. RNA sequencing results revealed that peroxisome proliferator-activated receptor gamma (PPAR-γ) significantly increased after LIPUS stimulation. Overexpression of PPAR-γ significantly increased the expression of contraction markers in CSSMCs, and the knockdown of PPAR-γ blocked this effect. Conclusions: LIPUS can regulate the transition of CSSMCs from a synthetic to a contractile phenotype in hypospadias. The PPAR-γ-mediated signaling pathway is a possible mechanism involved in this process. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

16 pages, 823 KiB  
Review
GABAergic Influences on Medulloblastoma
by Viviane Aline Buffon, Jurandir M. Ribas Filho, Osvaldo Malafaia, Isadora D. Tassinari, Rafael Roesler and Gustavo R. Isolan
Brain Sci. 2025, 15(7), 746; https://doi.org/10.3390/brainsci15070746 - 11 Jul 2025
Viewed by 403
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children and typically arises in the cerebellum, likely due to disruptions in neuronal precursor development. The primary inhibitory neurotransmitter in the central nervous system (CNS), γ-aminobutyric acid (GABA), exerts its effects through GABA [...] Read more.
Medulloblastoma (MB) is the most common malignant brain tumor in children and typically arises in the cerebellum, likely due to disruptions in neuronal precursor development. The primary inhibitory neurotransmitter in the central nervous system (CNS), γ-aminobutyric acid (GABA), exerts its effects through GABAA, GABAB, and GABAC receptors. GABA receptor activity regulates the development and function of cerebellar neurons, including glutamatergic cerebellar granule cells (CGCs). Beyond the nervous system, GABA is also a common metabolite in non-neuronal cell types. An increasing body of evidence indicates that GABA can influence cell proliferation, differentiation, and migration in several types of adult solid tumors, including brain cancers. GABA and GABAA receptor agonists can impair the viability and survival of MB cells, primarily acting on GABAA receptors containing the α5 subunit. A marked expression of the gene encoding the α5 subunit is found across all MB tumor molecular subgroups, particularly Group 3 MB, which has a poor prognosis. Importantly, high levels of the γ-aminobutyric acid type A receptor subunit α5 (GABRA5) gene are associated with shorter patient overall survival in Group 3 and Group 4 MB. In contrast, high γ-aminobutyric acid type A receptor subunit β1 (GABRB1) gene expression is related to longer survival in all MB subgroups. The GABAergic system may, therefore, regulate MB cell function and tumor progression and influence patient prognosis, and is worthy of further investigation as a biomarker and therapeutic target in MB. Full article
(This article belongs to the Special Issue Editorial Board Collection Series: Advances in Neuro-Oncology)
Show Figures

Figure 1

13 pages, 830 KiB  
Article
Nutritional and Metabolic Consequences of Camelina Seed Oil Compared to Flaxseed Oil in a Rat Diet
by Reshma Susan Babu and Adam Jurgoński
Molecules 2025, 30(13), 2738; https://doi.org/10.3390/molecules30132738 - 25 Jun 2025
Viewed by 532
Abstract
Camelina seeds are rich in α-linolenic acid (ALA), but also contain small amounts of erucic acid, which is considered toxic to laboratory rats. This experiment compares the dietary inclusion of camelina oil to that of flaxseed oil, a well-known source of ALA, and [...] Read more.
Camelina seeds are rich in α-linolenic acid (ALA), but also contain small amounts of erucic acid, which is considered toxic to laboratory rats. This experiment compares the dietary inclusion of camelina oil to that of flaxseed oil, a well-known source of ALA, and evaluates their effects on the nutritional and metabolic status of growing rats. The oils were chemically analyzed and incorporated into a semi-purified diet for 4 weeks. The experiment was divided into 3 groups: PO (control-fed palm oil with a trace of ALA), FO (comparative-fed flaxseed oil), and CO (experimental-fed camelina seed oil). Both CO and FO showed a higher percentage of lean body mass, greater lean mass gain, and a lower fat percentage compared to PO. Similar to the body composition, the blood lipid profile also improved in CO and FO, with higher HDL cholesterol and lower triglyceride levels, which was associated with upregulation of the peroxisome proliferator-activated receptor γ gene. However, in FO and CO, higher plasma liver enzyme activity and malondialdehyde concentrations were observed in the heart and liver. The results suggest that camelina oil has a similarly beneficial impact on the metabolic processes of the growing body as flaxseed oil, while also indicating a potential for increased organ-specific lipid peroxidation and hepatic burden when consumed in excess. Full article
(This article belongs to the Special Issue Food Bioactive Components in Functional Foods and Nutraceuticals)
Show Figures

Figure 1

16 pages, 2264 KiB  
Article
Ethanolic Extract of Ganoderma mexicanum Pat. Mycelium: A Source of Bioactive Compounds with Antiproliferative Activity and Potential PPAR-γ Natural Ligands
by Lucia T. Angulo-Sanchez, Max Vidal-Gutiérrez, Heriberto Torres-Moreno, Martín Esqueda, Aldo Gutiérrez, Georgina Vargas, Juan Luis Monribot-Villanueva, José A. Guerrero-Analco, César Muñoz-Bacasehua and Ramón Enrique Robles-Zepeda
Pharmaceuticals 2025, 18(6), 909; https://doi.org/10.3390/ph18060909 - 18 Jun 2025
Viewed by 701
Abstract
Background/Objective: Ganoderma spp. have long been studied for their bioactive pharmacological properties, and their biomass and extracts have been obtained from various sources. This study adopts a novel approach: enriching a liquid culture of Ganoderma mexicanum with a vineyard pruning waste extract [...] Read more.
Background/Objective: Ganoderma spp. have long been studied for their bioactive pharmacological properties, and their biomass and extracts have been obtained from various sources. This study adopts a novel approach: enriching a liquid culture of Ganoderma mexicanum with a vineyard pruning waste extract to identify bioactive compounds with antiproliferative activity through enriched chromatographic fractions. Methods: The ethanolic extract from a mycelial culture was separated following a partitioning process, and the hexane fraction was subsequently separated in a chromatographic column. The fractions were evaluated for their antiproliferative properties against cancer cell lines. The interactions of the molecules identified with peroxisome proliferator-activated receptor gamma (PPAR-γ) were analyzed via molecular docking. Results: Three chromatographic fractions (FH11–FH13) exhibited antiproliferative activity which was significantly more effective against non-small lung cancer cells (A549). The cells treated with the crude extract and fractions presented a balloon-like morphology. A chemical analysis of the active fractions allowed us to identify four compounds: one fatty acid (9-Hydroxy-10E,12Z-octadecadienoic acid) and three triterpenes (ganoderic acids DM, TQ, and X). These compounds showed interactions with the PPAR-γ receptor through molecular docking. Conclusions: Ganoderma mexicanum is a promising source of compounds with antiproliferative activity that could serve as natural ligands for PPAR-γ and has possible applications in lung cancer therapy. Full article
Show Figures

Graphical abstract

13 pages, 665 KiB  
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
Viewed by 581
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

21 pages, 2278 KiB  
Review
Orphan Nuclear Receptors TR2 and TR4 in Erythropoiesis: From Mechanisms to Therapies
by Yunlong Liu, Helian Yang, Mengtian Ren, Qing Yu, Qingyang Xu and Xiuping Fu
Biomolecules 2025, 15(6), 798; https://doi.org/10.3390/biom15060798 - 31 May 2025
Viewed by 668
Abstract
Testicular orphan receptors TR2 and TR4 serve as central regulators of erythropoiesis, orchestrating the entire continuum of erythroid progenitor cell proliferation, differentiation, and maturation. As core components of the direct repeat erythroid determinant (DRED) complex, they activate erythroid-specific transcriptional programs to dynamically control [...] Read more.
Testicular orphan receptors TR2 and TR4 serve as central regulators of erythropoiesis, orchestrating the entire continuum of erythroid progenitor cell proliferation, differentiation, and maturation. As core components of the direct repeat erythroid determinant (DRED) complex, they activate erythroid-specific transcriptional programs to dynamically control the spatiotemporal expression of globin genes. These nuclear receptors not only engage in functional interactions with key erythroid transcription factors GATA1 and KLF1 to coregulate erythroid differentiation and maturation but also recruit epigenetic modifier complexes such as DNMT1 and LSD1 to modulate chromatin states dynamically. Research has established that dysfunctions in TR2/TR4 are implicated in β-thalassemia and sickle cell disease (SCD): β-thalassemia is associated with the defective silencing of γ-globin genes, while in SCD, TR2/TR4 antagonizes BCL11A to reactivate fetal hemoglobin (HbF) expression. This review systematically dissects the molecular regulatory networks of TR2/TR4 in erythroid cells, interprets their dual regulatory properties across different stages of erythroid differentiation, and explores the therapeutic potential of targeting TR2/TR4 for treating erythroid-related disorders such as β-thalassemia and SCD, thereby providing novel directions for hematological disorder therapy. Full article
(This article belongs to the Section Biomacromolecules: Proteins, Nucleic Acids and Carbohydrates)
Show Figures

Figure 1

15 pages, 2682 KiB  
Article
Effects of Diethylstilbestrol on the Structure and Function of the Spleen in Male Golden Hamsters
by Jian Li, Ruiping Xu, Qingwei Wang, Xue Bai, Yanhua Su, Yaoxing Chen and Jing Cao
Toxics 2025, 13(5), 397; https://doi.org/10.3390/toxics13050397 - 15 May 2025
Viewed by 576
Abstract
With industrial development, endocrine-disrupting chemicals have continued to accumulate in the environment, attracting growing attention due to their potential effects on biological health. The reproductive toxicity of diethylstilbestrol (DES), a synthetic estrogen widely present in the environment, is widely documented; however, studies on [...] Read more.
With industrial development, endocrine-disrupting chemicals have continued to accumulate in the environment, attracting growing attention due to their potential effects on biological health. The reproductive toxicity of diethylstilbestrol (DES), a synthetic estrogen widely present in the environment, is widely documented; however, studies on its effects on the immune system remain limited. In this study, adult male golden hamsters were subcutaneously administered varying doses of DES (0, 0.01, 0.1, and 1.0 mg/kg) for seven consecutive days to assess its immunomodulatory impact on peripheral blood and the spleen. We found that the DES treatment significantly reduced spleen index, white pulp area, and splenic lymphocyte proliferation while increasing caspase-3-positive apoptotic cells and inducible nitric oxide synthase expression. In peripheral blood, DES induced a dose-dependent suppression of lymphocyte proliferation, with lipopolysaccharide- and concanavalin A-stimulated proliferation reduced by 47.68–71.76% and 44.23–72.7%, respectively. Concurrently, DES significantly downregulated the pro-inflammatory cytokines IL-2 and IFN-γ (p < 0.01) while upregulating the anti-inflammatory cytokines IL-4 and IL-10 (p < 0.01). Furthermore, DES treatment impaired antioxidant defenses, decreasing the activity of superoxide dismutase, glutathione peroxidase, and catalase while elevating malondialdehyde levels. Notably, DES led to the upregulation of G protein-coupled estrogen receptor and estrogen receptor α at both transcriptional and protein levels, whereas estrogen receptor β mRNA expression increased despite a decline in protein levels. This study provides critical experimental evidence elucidating the immunoregulatory effects of endocrine-disrupting environmental estrogens. Full article
Show Figures

Figure 1

23 pages, 7191 KiB  
Article
Interleukin-15Rα-Sushi-Fc Fusion Protein Co-Hitchhikes Interleukin-15 and Pheophorbide A for Cancer Photoimmunotherapy
by Zhe Li, Jiaojiao Xu, Hongzheng Lin, Sheng Yu, Jingwen Sun, Chen Zhang, Sihang Zhang, Tingting Li, Afeng Yang and Wei Lu
Pharmaceutics 2025, 17(5), 615; https://doi.org/10.3390/pharmaceutics17050615 - 5 May 2025
Viewed by 611
Abstract
Background: Interleukin-15 (IL-15) stimulates the proliferation of natural killer cells or T cells, which, in combination with photodynamic therapy (PDT), has emerged as an effective strategy for cancer photoimmunotherapy. Instead of direct cytokine receptor activation, IL-15 necessitates first binding to the IL-15 [...] Read more.
Background: Interleukin-15 (IL-15) stimulates the proliferation of natural killer cells or T cells, which, in combination with photodynamic therapy (PDT), has emerged as an effective strategy for cancer photoimmunotherapy. Instead of direct cytokine receptor activation, IL-15 necessitates first binding to the IL-15 receptor α chain subunit (IL-15Rα), followed by trans-presentation to the IL-15 receptor β/γ chain subunit on the effector cells for pharmacologic activation. Therefore, the delivery of IL-15 remains a major challenge owing to its short half-life, its lack of targeting activity, and the limited availability of IL-15Rα. Methods: A co-hitchhiking delivery approach using recombinant IL-15 (rIL-15) and a photosensitizer, pheophorbide A (PhA), is developed for enhanced combinatorial cancer immunotherapy with PDT. A recombinant IL-15Rα-sushi-Fc fusion protein (rILR-Fc) is designed to load rIL-15 through the IL-15Rα sushi domain, which mimics its trans-presentation. Moreover, the Fc moiety of rILR-Fc can load PhA based on its high binding affinity. Results: Through self-assembly, rILR-Fc/PhA/rIL-15 nanoparticles (NPs) are formulated to co-hitchhike PhA and rIL-15, which improves the tumor accumulation of PhA and rIL-15 through receptor-mediated transcytosis. Moreover, the nanoparticles prolong the blood half-life of rIL-15 but do not alter the elimination rate of PhA from the blood. The rILR-Fc/PhA/rIL-15 NPs effectively elicit potent systemic antitumor immunity and long-lasting immune memory against tumor rechallenge in model mice bearing orthotopic colon tumors. Conclusions: The enhanced antitumor therapeutic effect demonstrates that the co-hitchhiking delivery strategy, optimizing the pharmacokinetics of both the photosensitizer and IL-15, provides a promising strategy for combinatorial photodynamic and IL-15 immunotherapy. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

14 pages, 2998 KiB  
Article
In Vitro Immunomodulatory Effects of Equine Adipose Tissue-Derived Mesenchymal Stem Cells Primed with a Cannabidiol-Rich Extract
by Lorena Battistin, Luís Felipe Arantes Moya, Lucas Vinícius de Oliveira Ferreira, Aline Márcia Marques Braz, Márcio de Carvalho, Marjorie de Assis Golim and Rogério Martins Amorim
Int. J. Mol. Sci. 2025, 26(9), 4208; https://doi.org/10.3390/ijms26094208 - 29 Apr 2025
Viewed by 518
Abstract
Cell-based therapy using mesenchymal stem cells (MSCs) shows promise for treating several diseases due to their anti-inflammatory and immunomodulatory properties. To enhance the therapeutic potential of MSCs, in vitro priming strategies have been explored. Cannabidiol (CBD), a non-psychoactive compound derived from cannabis, may [...] Read more.
Cell-based therapy using mesenchymal stem cells (MSCs) shows promise for treating several diseases due to their anti-inflammatory and immunomodulatory properties. To enhance the therapeutic potential of MSCs, in vitro priming strategies have been explored. Cannabidiol (CBD), a non-psychoactive compound derived from cannabis, may influence MSC proliferation, differentiation, and immunomodulatory properties. This study evaluates the immunomodulatory potential of equine adipose tissue-derived MSCs (EqAT-MSCs) primed with a CBD-rich cannabis extract. EqAT-MSCs (P3) were primed with CBD concentrations of 5 µM and 7 µM for 24 h. Morphological analysis, MTT assay, β-galactosidase activity, apoptosis assays, and gene expression of interleukins IL-1β, IL-6, IL-10, interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α) were conducted. Additionally, cannabinoid receptor 1 (CB1) and 2 (CB2) expression were evaluated in naïve EqAT-MSCs (P2–P5). The naïve EqAT-MSCs expressed CB1 and CB2 receptors. Priming with 5 µM significantly increased the expression of IL-10, TNF-α, and IFN-γ, while 7 µM decreased IL-1β and IL-6 expression. No significant changes were observed in other cytokines, MTT, β-galactosidase activity, or apoptosis. These findings demonstrate that naïve EqAT-MSCs express CB1 and CB2 receptors and priming with the extract modulates the expression of pro- and anti-inflammatory cytokines, highlighting its potential immunomodulatory role in EqAT-MSC-based therapies. Full article
(This article belongs to the Special Issue Biomedical Applications of Mesenchymal Stem Cells)
Show Figures

Figure 1

17 pages, 5597 KiB  
Article
Role of T3 in the Regulation of GRP78 on Granulosa Cells in Rat Ovaries
by Yan Liu, Yilin Yao, Yakun Yu, Ying Sun, Mingqi Wu, Rui Chen, Haoyuan Feng, Shuaitian Guo, Yanzhou Yang and Cheng Zhang
Int. J. Mol. Sci. 2025, 26(9), 4196; https://doi.org/10.3390/ijms26094196 - 28 Apr 2025
Viewed by 637
Abstract
Thyroid hormone (TH) plays a vital role in ovarian follicle development, and glucose-regulated protein 78 (GRP78) is involved in these processes, which is regulated by TH. However, the mechanisms are still unclear. To evaluate the possible mechanism of TH on the regulation of [...] Read more.
Thyroid hormone (TH) plays a vital role in ovarian follicle development, and glucose-regulated protein 78 (GRP78) is involved in these processes, which is regulated by TH. However, the mechanisms are still unclear. To evaluate the possible mechanism of TH on the regulation of GRP78 expression, Cleavage Under Targets and Tagmentation (CUT & Tag) sequencing, luciferase assays, and Electrophoretic Mobility Shift Assays (EMSA) were employed to delineate the binding sites of thyroid hormone receptor β (TRβ) on the GRP78 promoter and to confirm the interactions. Additionally, Co-Immunoprecipitation (Co-IP) and Immunofluorescence (IF) assays were used to investigate the interactions between TRβ and the coactivator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) after triiodothyronine (T3) treatment with different concentrations. Our findings identified a thyroid hormone response element (TRE) on the GRP78 promoter and demonstrated that TRβ can activate GRP78 expression by interacting with PGC-1α. In order to simulate the condition of hyperthyroidism, granulosa cells (GCs) extracted from rats were treated by T3 with high concentrations, which decreased the expression of PGC-1α, resulting in decreased expressions of GRP78 and other ferroptosis-related markers such as glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11, xCT), thereby inducing ferroptosis in GCs. Taken together, the present study demonstrates that T3 induces cellular ferroptosis by binding TRE of the GRP78 promoter in ovarian GCs via TRβ. As a switcher, PGC-1α is also involved in these processes. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

13 pages, 2292 KiB  
Article
The Soluble Cytoplasmic Tail of CD45 (ct-CD45) Regulates Dendritic Cell Activation and Function via TLR4 Signaling
by Sara Gil-Cantero, Alexander Puck, Sarojinidevi Künig, Veronica Pinnarò, Petra Waidhofer-Söllner and Johannes Stöckl
Int. J. Mol. Sci. 2025, 26(8), 3888; https://doi.org/10.3390/ijms26083888 - 20 Apr 2025
Viewed by 548
Abstract
The soluble cytoplasmic tail of the prototypic receptor-like protein tyrosine phosphatase (PTP) CD45 (ct-CD45) is cleaved and released into the human plasma by activated phagocytes. Released ct-CD45 was found to inhibit T cell proliferation and cytokine production via engagement of Toll-like receptor 4 [...] Read more.
The soluble cytoplasmic tail of the prototypic receptor-like protein tyrosine phosphatase (PTP) CD45 (ct-CD45) is cleaved and released into the human plasma by activated phagocytes. Released ct-CD45 was found to inhibit T cell proliferation and cytokine production via engagement of Toll-like receptor 4 (TLR4). In this study, we analyzed the impact of the ct-CD45/TLR4 pathway on the function of human monocyte-derived dendritic cells (DCs). We could demonstrate that activation of DCs by ct-CD45 upregulated the expression of certain cell surface markers (e.g., CD71 and CD86) and induced IL-10 production via TLR4. Yet, in contrast to stimulation with LPS, other typical cell surface markers and cytokines were not upregulated or induced in DCs by ct-CD45. The T cell proliferation–stimulatory capacity of DCs was not modulated by ct-CD45 treatment. However, treatment of DCs with ct-CD45 modulated the cytokine profile in co-cultured T cells. While IFN-γ production induced by DCs was strongly inhibited, the release of IL-4 was increased in T cells upon stimulation with ct-CD45-treated DCs. In contrast, ct-CD45-stimulated DCs induced IL-2 and IL-10 production in co-cultured T cells comparable to untreated DCs. In summary, we could demonstrate that ct-CD45 acts as an immunoregulatory factor for DCs via a non-canonical TLR4-dependent activation pathway. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

18 pages, 6741 KiB  
Article
Competitive Ligand-Induced Recruitment of Coactivators to Specific PPARα/δ/γ Ligand-Binding Domains Revealed by Dual-Emission FRET and X-Ray Diffraction of Cocrystals
by Shotaro Kamata, Akihiro Honda, Sayaka Yashiro, Chihiro Kaneko, Yuna Komori, Ayumi Shimamura, Risa Masuda, Takuji Oyama and Isao Ishii
Antioxidants 2025, 14(4), 494; https://doi.org/10.3390/antiox14040494 - 20 Apr 2025
Viewed by 830
Abstract
Peroxisome proliferator-activated receptors (PPARs), composed of the α/δ/γ subtypes, are ligand-activated nuclear receptors/transcription factors that sense endogenous fatty acids or therapeutic drugs to regulate lipid/glucose metabolism and oxidative stress. PPAR forms a multiprotein complex with a retinoid X receptor and corepressor complex in [...] Read more.
Peroxisome proliferator-activated receptors (PPARs), composed of the α/δ/γ subtypes, are ligand-activated nuclear receptors/transcription factors that sense endogenous fatty acids or therapeutic drugs to regulate lipid/glucose metabolism and oxidative stress. PPAR forms a multiprotein complex with a retinoid X receptor and corepressor complex in an unliganded/inactive state, and ligand binding induces the replacement of the corepressor complex with the coactivator complex to initiate the transcription of various genes, including the metabolic and antioxidant ones. We investigated the processes by which the corepressor is replaced with the coactivator or in which two coactivators compete for the PPARα/δ/γ-ligand-binding domains (LBDs) using single- and dual-emission fluorescence resonance energy transfer (FRET) assays. Single-FRET revealed that the respective PPARα/δ/γ-selective agonists (pemafibrate, seladelpar, and pioglitazone) induced the dissociation of the two corepressor peptides, NCoR1 and NCoR2, from the PPARα/δ/γ-LBDs and the recruitment of the two coactivator peptides, CBP and TRAP220. Meanwhile, dual-FRET demonstrated that these processes are simultaneous and that the four coactivator peptides, CBP, TRAP220, PGC1α, and SRC1, were competitively recruited to the PPARα/δ/γ-LBDs with different preferences upon ligand activation. Furthermore, the five newly obtained cocrystal structures using X-ray diffraction, PPARα-LBDs–NCoR2/CBP/TRAP220/PGC1α and PPARγ-LBD–NCoR2, were co-analyzed with those from our previous studies. This illustrates that these coactivators bound to the same PPARα-LBD loci via their consensus LXXLL motifs in the liganded state; that NCoR1/NCoR2 corepressors bound to the same loci via the IXXXL sequences within their consensus LXXXIXXXL motifs in the unliganded state; and that ligand activation induced AF-2 helix 12 formation that interfered with corepressor binding and created a binding space for the coactivator. These PPARα/γ-related biochemical and physicochemical findings highlight the coregulator dynamics on limited PPARα/δ/γ-LBDs loci. Full article
Show Figures

Graphical abstract

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

Back to TopTop