Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
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 (1,109)

Search Parameters:
Keywords = inhibitors of TNF

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 2915 KB  
Article
Neuroprotective Effects of Calpain Inhibition in Parkinson’s Disease: Insights from Cellular and Murine Models
by Vandana Zaman, Amy Gathings, Kelsey P. Drasites, Donald C. Shields, Narendra L. Banik and Azizul Haque
Cells 2025, 14(17), 1310; https://doi.org/10.3390/cells14171310 - 24 Aug 2025
Abstract
Parkinson’s disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra, and key pathways such as neuroinflammation, oxidative stress, and autophagy are believed to significantly contribute to the mechanisms of neurodegeneration. Calpain activation plays a critical role in [...] Read more.
Parkinson’s disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra, and key pathways such as neuroinflammation, oxidative stress, and autophagy are believed to significantly contribute to the mechanisms of neurodegeneration. Calpain activation plays a critical role in neuroinflammation and neurodegeneration, as demonstrated by its impact on microglial activation, reactive oxygen species (ROS) production, and neuronal survival. In this study, we investigated the effects of calpain inhibition using calpeptin (CP) and calpain-2-specific inhibitors in cellular and murine models of neuroinflammation and PD. In BV2 microglial cells, LPS-induced production of pro-inflammatory cytokines (TNF-α, IL-6) and chemokines (MCP-1, IP-10) were significantly reduced by CP treatment with a concomitant decrease in ROS generation. Similarly, in VSC-4.1 motoneuron cells, calpain inhibition attenuated IFN-γ-induced ROS production and improved cell viability, demonstrating its neuroprotective effects. Moreover, in a murine MPTP model of PD, calpain inhibition reduced astrogliosis, ROCK2 expression, and levels of inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-7, and IL12p70) and chemokines (MCP-1 and IP-10) in the dorsal striatum and plasma. The specific role of calpain-2 in immune modulation was further highlighted in human microglia, SV-40 cells. With respect to immune modulation in these cells, siRNA-mediated knockdown of calpain-2, but not calpain-1, significantly reduced antigen presentation to CD4+ T cells. Thus, calpain-2 is likely involved in regulating antigen presentation and activation of inflammatory CD4+ T cells. These findings underscore the therapeutic potential of calpain-2 inhibition in mitigating neuroinflammation and neurodegeneration, particularly in PD, by targeting microglial activation, ROS production, and neuronal survival pathways. Full article
(This article belongs to the Special Issue Role of Calpains in Health and Diseases)
Show Figures

Figure 1

33 pages, 20134 KB  
Article
Paclitaxel and Cephalomannine Synergistically Induce PANoptosis in Triple-Negative Breast Cancer Through Oxygen-Regulated Cell Death Pathways
by Xinyu Gao, Kuilin Chen, Shuhui Jia, Jiapeng Li, Huan Zhang, Yuwei Wang and Weidong Xie
Antioxidants 2025, 14(9), 1037; https://doi.org/10.3390/antiox14091037 - 22 Aug 2025
Viewed by 172
Abstract
Triple-negative breast cancer (TNBC) urgently requires new therapeutic strategies due to the limited efficacy of conventional treatments. Recently, PANoptosis, an integrated form of apoptosis, necroptosis, and pyroptosis, has emerged as a promising target in cancer therapy, though effective agents remain scarce. Paclitaxel, a [...] Read more.
Triple-negative breast cancer (TNBC) urgently requires new therapeutic strategies due to the limited efficacy of conventional treatments. Recently, PANoptosis, an integrated form of apoptosis, necroptosis, and pyroptosis, has emerged as a promising target in cancer therapy, though effective agents remain scarce. Paclitaxel, a Taxus-derived natural product, is often combined with other drugs to enhance efficacy, yet optimal combinations are limited. This study investigates the synergistic antitumor effects of paclitaxel and cephalomannine in TNBC, focusing on oxygen-regulated cell death pathways. Network pharmacology and molecular docking revealed that the combination targets multiple cell death- and inflammation-related proteins, including BCL2L1, MAPK14, SYK, TNF, and ADAM17, suggesting multi-target synergy. In vitro, the combination significantly inhibited MDA-MB-231 cell viability, proliferation, and migration, while inducing apoptosis and necrosis. Mechanistically, co-treatment markedly increased intracellular ROS levels and γ-H2AX expression, indicating oxidative stress and DNA damage, both of which were reversible by ROS inhibition. Further analysis demonstrated that the treatment activated the p38 and p53 pathways, regulated the Bax/Bcl-2 ratio, and initiated mitochondrial apoptosis. It also promoted RIPK1/RIPK3/MLKL phosphorylation and MLKL membrane translocation, triggering necroptosis, as well as upregulated NLRP3, cleaved Caspase-1, and GSDMD, inducing pyroptosis. The use of specific inhibitors partially reversed these effects, confirming the involvement of ROS-mediated PANoptosis. Similar antitumor effects were also observed in BT-549 cells, indicating the broad applicability of this combination in TNBC. MCF-10A cells exhibited mild but acceptable cytotoxicity, reflecting manageable side effects typical of chemotherapeutic agents. In vivo experiments further validated the combination’s antitumor efficacy and safety. In summary, paclitaxel and cephalomannine synergistically induce PANoptosis in TNBC through oxygen-regulated cell death pathways, offering a novel therapeutic strategy based on oxidative stress modulation by natural compounds. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

14 pages, 1022 KB  
Review
Effects of Cytokines (or Activating Factors) on Arterial Endothelial Cells
by Leon M. T. Dicks
Int. J. Mol. Sci. 2025, 26(17), 8142; https://doi.org/10.3390/ijms26178142 - 22 Aug 2025
Viewed by 86
Abstract
The internal mammary arteries (IMAs) and coronary arteries share many common characteristics. The inner layer (tunica intima, or intima) of both arteries is lined with a smooth, longitudinally orientated monolayer of endothelial cells (ECs), connective tissue, and an internal elastic lamina that separates [...] Read more.
The internal mammary arteries (IMAs) and coronary arteries share many common characteristics. The inner layer (tunica intima, or intima) of both arteries is lined with a smooth, longitudinally orientated monolayer of endothelial cells (ECs), connective tissue, and an internal elastic lamina that separates the tunica intima from the tunica media (middle layer). The intima of IMAs is lined with an additional protective layer, the neointima, containing vascular smooth muscle cells (VSMCs). The neointima, located between the intima and internal elastic lamina, protects IMAs from damage by assisting in the remodeling of VSMCs. Coarse longitudinal folds in the internal elastic lamina of IMAs partially prevent the infiltration of VSMCs into damaged IMAs, and intimal thickening is thus less likely to occur. Inflamed IMAs resist the migration of monocytes across the endothelial layer and prevent the formation of lipid-rich macrophages (foam cells) within the subintimal or medial layers of arteries. IMAs are thus less likely to form plaques and develop atherosclerosis (AS). Higher levels of prostacyclin (PGI2) in IMAs prevent blood clotting. The anti-thrombotic agents, and production of tumor necrosis factor α (TNF-α), interferon-γ (INF-γ), and visfatin render IMAs more resistant to inflammation. An increase in the production of nitric oxide (NO) by ECs of IMAs may be due to small ubiquitin-like modifier (SUMO) proteins that alter the nuclear factor kappa B (NF-κB) and TLR pathways. The production of reactive oxygen species (ROS) in IMAs is suppressed due to the inhibition of NADPH oxidase (NOX) by a pigment epithelium-derived factor (PEDF), which is a serine protease inhibitor (SERPIN). In this review, a comparison is drawn between the anatomy of IMAs and coronary arteries, with an emphasis on how ECs of IMAs react to immunological changes, rendering them more suited for coronary artery bypass grafts (CABGs). This narrative review covers the most recent findings published in PubMed and Crossref databases. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

16 pages, 2782 KB  
Article
Photochemically Assisted Synthesis of Thienobenzotriazole-Based Dual Cholinesterase Inhibitors
by Antonija Jelčić, Stanislava Talić, Ilijana Odak, Paula Pongrac, Dora Štefok and Irena Škorić
Molecules 2025, 30(16), 3439; https://doi.org/10.3390/molecules30163439 - 20 Aug 2025
Viewed by 247
Abstract
Background: It has been shown previously that thienobenzo-1,2,3-triazoles exhibit very good selective inhibition toward butyrylcholinesterase (BChE), while the same derivatives converted into salts also display inhibitory activity against acetylcholinesterase (AChE), enzymes relevant to Alzheimer’s disease therapy. They show even better BChE inhibition potential [...] Read more.
Background: It has been shown previously that thienobenzo-1,2,3-triazoles exhibit very good selective inhibition toward butyrylcholinesterase (BChE), while the same derivatives converted into salts also display inhibitory activity against acetylcholinesterase (AChE), enzymes relevant to Alzheimer’s disease therapy. They show even better BChE inhibition potential than neutral analogs. Methods: This study presents the synthesis and biological evaluation of a novel series of charged thienobenzo-1,2,3-triazolinium salts (117) as inhibitors of AChE and BChE. The basic skeleton of the targeted compounds was synthesized via a photochemical method and subsequently converted into corresponding bromide salts. Their structures were confirmed using NMR and HRMS analyses. Results: In vitro testing showed that all synthesized compounds exhibit moderate to strong BChE inhibition and, to a lesser extent, AChE inhibition. Compounds 8 and 11 emerged as the most potent AChE inhibitors (IC50 ~ 2.6–3.2 µM), while compounds 1, 2, and 8 demonstrated excellent and selective BChE inhibition (IC50 ~ 0.3–0.4 µM), outperforming the reference drug galantamine. Anti-inflammatory evaluation revealed limited activity, with compound 17 slightly reducing LPS-induced TNF-α production at the highest tested concentration. Conclusions: These findings highlight the role of the electric charge and substituent type in modulating biological activity and confirm the therapeutic potential of these molecules as dual cholinesterase inhibitors for further development in neurodegenerative disease treatment. Full article
(This article belongs to the Special Issue Synthesis of Bioactive Compounds, 3rd Edition)
Show Figures

Figure 1

22 pages, 1986 KB  
Article
Predictive Microbial Markers Distinguish Responders and Non-Responders to Adalimumab: A Step Toward Precision Medicine in Ulcerative Colitis
by Shaghayegh Baradaran Ghavami, Arfa Moshiri, Carola Bonaretti, Maryam Farmani, Margherita Squillario, Eddi Di Marco, Shabnam Shahrokh, Hedieh Balaii, Maria Valeria Corrias, Mirco Ponzoni, Amir Sadeghi and Roberto Biassoni
Microorganisms 2025, 13(8), 1941; https://doi.org/10.3390/microorganisms13081941 - 20 Aug 2025
Viewed by 170
Abstract
Ulcerative colitis (UC) is a chronic, relapsing inflammatory disease of the colon, often associated with gut microbial dysbiosis. Although anti-TNF-α agents, such as Adalimumab (Cinnora®), are used to treat moderate-to-severe UC, the treatment response is highly variable. Identifying early microbial biomarkers [...] Read more.
Ulcerative colitis (UC) is a chronic, relapsing inflammatory disease of the colon, often associated with gut microbial dysbiosis. Although anti-TNF-α agents, such as Adalimumab (Cinnora®), are used to treat moderate-to-severe UC, the treatment response is highly variable. Identifying early microbial biomarkers of response could help support personalized therapeutic strategies and prevent unnecessary exposure to ineffective treatments. However, the long-term effects of anti-TNF therapy on both stool and mucosal microbiota remain poorly understood. This prospective longitudinal study included 23 corticosteroid-refractory or -dependent UC patients who started Adalimumab after endoscopy-confirmed flare-ups. Stool samples and inflamed colonic biopsies were collected at baseline, and 3 and 6 months. Microbiota profiling was performed using 16S rRNA sequencing. Microbial changes were analyzed over time and compared between responders (Mayo score 0–1) and non-responders (Mayo score ≥ 2). Sixty percent of patients achieved clinical remission. In responders, stool microbiota showed increased Bacteroidetes and decreased Proteobacteria abundances, along with an enrichment of beneficial taxa including Faecalibacterium prausnitzii, Bifidobacterium, and Akkermansia muciniphila. Mucosal microbiota exhibited persistent dysbiosis, characterized by an increase in Proteobacteria and a reduced Firmicutes/Proteobacteria ratio. Notably, responders showed distinct compartment-specific microbial changes, with a decrease in Gammaproteobacteria in stool and an increase in Corynebacterium in tissue. Adalimumab induces divergent microbial changes in stool and mucosa. While stool microbiota trends toward eubiosis in responders, persistent mucosal dysbiosis may reflect asymptomatic inflammation. These findings underscore the importance of niche-specific microbiome profiling in UC and support its integration into personalized treatment monitoring. Full article
(This article belongs to the Section Microbiomes)
Show Figures

Graphical abstract

21 pages, 4617 KB  
Article
Apelin-13-Mediated Upregulation of METTL3 Ameliorates Alzheimer’s Disease via Inhibiting Neuroinflammation Through m6A-Dependent Regulation of lncRNA BDNF-AS
by Li Han, Siwen Wei, Rong Wang, Yiran Liu, Yi Zhong, Juan Fu, Huaiqing Luo and Meihua Bao
Biomolecules 2025, 15(8), 1188; https://doi.org/10.3390/biom15081188 - 18 Aug 2025
Viewed by 299
Abstract
Apelin-13, a neuropeptide, has been recognized for its neuroprotective properties. Our previous study found apelin-13 improves cognitive function in Alzheimer’s disease (AD) rats by inhibiting neuroinflammation through upregulation of BDNF/TrkB signaling pathway. However, the precise mechanism by which apelin-13 modulates BDNF remains unclear. [...] Read more.
Apelin-13, a neuropeptide, has been recognized for its neuroprotective properties. Our previous study found apelin-13 improves cognitive function in Alzheimer’s disease (AD) rats by inhibiting neuroinflammation through upregulation of BDNF/TrkB signaling pathway. However, the precise mechanism by which apelin-13 modulates BDNF remains unclear. Thus, this study aimed to unravel the specific regulatory mechanism by which apelin-13 regulates BDNF. Bilaterally intracerebroventricular injection with Aβ25–35 was used to establish an in vivo model of AD. For the generation of METTL3 KO rats, the Crispr/Cas9 method was applied. PC12 cells were treated with Aβ25–35 to establish an in vitro model of AD. The cognitive function of the rats was evaluated with the Morris water maze and the novel object recognition test. Hippocampal damage and neuron loss were detected through H&E and immunofluorescent staining. METTL3, BDNF, TrkB, and p-TrkB were examined by Western blotting. Inflammation-related cytokines, IBA1, GFAP, IL-1β, and TNF-α were detected by Western blotting, immunofluorescent staining, ELISA, and qRT-PCR. m6A modification level was evaluated through MeRIP. A flow cytometer was applied to evaluate cell apoptosis. Cell proliferation was examined using MTT. m6A methylation inhibitor DAA reverses the improvement effect of apelin-13 on cognitive function, hippocampal nerve damage, neuron loss, and neuroinflammation in Aβ25–35-treated rats. Further results showed that apelin-13 upregulated METTL3, BDNF-AS m6A methylation, inhibited BDNF-AS expression, and subsequently upregulated BDNF/TrkB signaling pathway and reduced neuroinflammation in in vivo and in vitro AD models in a dose-dependent manner. Knockdown of METTL3 abolished apelin-13’s improvement effect in AD rats. Apelin-13-mediated upregulation of METTL3 enhances neuroinflammation inhibition and BDNF/TrkB signaling pathway via m6A-dependent downregulation of lncRNA BDNF-AS, thus ameliorating AD. Our study offers novel insights into the pathogenesis of AD and identifies potential drug targets for its treatment. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

14 pages, 659 KB  
Article
Anti-Inflammatory and Immunomodulatory Effects of 2-(3-Acetyl-5-(4-Chlorophenyl)-2-Methyl-1H-Pyrrol-1-yl)-3-Phenylpropanoic Acid
by Hristina Zlatanova-Tenisheva and Stanislava Vladimirova
Biomedicines 2025, 13(8), 2003; https://doi.org/10.3390/biomedicines13082003 - 18 Aug 2025
Viewed by 328
Abstract
Background: The pursuit of novel anti-inflammatory agents with enhanced efficacy and safety is crucial. Pyrrole-containing compounds, integral to many NSAIDs, exhibit promising anti-inflammatory properties. Compound 3f (2-(3-acetyl-5-(4-chlorophenyl)-2-methyl-1H-pyrrol-1-yl)-3-phenylpropanoic acid), a pyrrole derivative structurally inspired by the COX-2 selective inhibitor celecoxib, was evaluated [...] Read more.
Background: The pursuit of novel anti-inflammatory agents with enhanced efficacy and safety is crucial. Pyrrole-containing compounds, integral to many NSAIDs, exhibit promising anti-inflammatory properties. Compound 3f (2-(3-acetyl-5-(4-chlorophenyl)-2-methyl-1H-pyrrol-1-yl)-3-phenylpropanoic acid), a pyrrole derivative structurally inspired by the COX-2 selective inhibitor celecoxib, was evaluated for its anti-inflammatory and immunomodulatory effects. Methods: Anti-inflammatory activity was assessed in a carrageenan-induced paw edema model in Wistar rats. Compound 3f was administered intraperitoneally at 10, 20, and 40 mg/kg, either as a single dose or daily for 14 days. Diclofenac (25 mg/kg) served as the reference. Edema volume was measured by plethysmometry. Systemic inflammation was induced by lipopolysaccharide (LPS), and serum levels of the pro-inflammatory cytokine TNF-α and anti-inflammatory cytokines IL-10 and TGF-β1 were quantified by ELISA following single and repeated administration of compound 3f. Results: Single-dose administration of compound 3f at 20 mg/kg significantly reduced paw edema at 2 h (p = 0.001). After 14 days, all tested doses significantly inhibited paw edema at all time points (p < 0.001). In the LPS-induced systemic inflammation model, repeated treatment with 40 mg/kg of compound 3f significantly decreased serum TNF-α (p = 0.032). TGF-β1 levels increased significantly after both single and repeated doses (p = 0.002 and p = 0.045, respectively), while IL-10 levels remained unaffected. Conclusions: Compound 3f exhibits potent anti-inflammatory activity, particularly after repeated dosing, reflected by reduced local edema and systemic TNF-α suppression. The marked elevation of TGF-β1 indicates a potential immunomodulatory mechanism, selectively modulating cytokine profiles without altering IL-10. These findings support compound 3f as a promising candidate for targeted anti-inflammatory therapy involving cytokine regulation. Full article
(This article belongs to the Special Issue The Role of Cytokines in Health and Disease: 3rd Edition)
Show Figures

Figure 1

27 pages, 2779 KB  
Article
Cinnamic Acid: A Shield Against High-Fat-Diet-Induced Liver Injury—Exploring Nrf2’s Protective Mechanisms
by Asmahan Taher Alahdal, Laila Naif Al-Harbi, Ghedeir M. Alshammari, Ali Saleh and Mohammed Abdo Yahya
Int. J. Mol. Sci. 2025, 26(16), 7940; https://doi.org/10.3390/ijms26167940 - 17 Aug 2025
Viewed by 372
Abstract
This study investigated the hepatoprotective effects of cinnamic acid (CA) against liver injury and fat accumulation induced by a high-fat diet (HFD), focusing on the role of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Male Wistar rats were divided into six [...] Read more.
This study investigated the hepatoprotective effects of cinnamic acid (CA) against liver injury and fat accumulation induced by a high-fat diet (HFD), focusing on the role of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Male Wistar rats were divided into six groups: a control group receiving carboxymethylcellulose; a CA control group (40 mg/kg); an HFD group; two HFD groups treated with CA (20 mg/kg or 40 mg/kg); and a HFD group co-treated with CA (40 mg/kg) and brusatol (2 mg/kg, i.p.), a selective Nrf2 inhibitor. CA was administered orally, and brusatol intraperitoneally, both twice per week for twelve weeks. CA had no effect on serum glucose or insulin but improved serum and hepatic profiles in HFD rats. It also attenuated liver vacuolization and normalized serum levels of ALT, AST, and γ-GT. CA also reduced hepatic apoptosis by increasing Bcl2 and reducing Bax and caspase-3 levels. CA mitigated oxidative stress by reducing MDA and enhancing SOD and GSH levels. It suppressed inflammatory mediators, including TNF-α, IL-6, and NF-κB. CA also downregulated SREBP1, FAS, ACC-1, and Keap1 while increasing mRNA and nuclear translocation of Nrf2. All these effects were dose-dependent. Similar molecular effects of CA were also seen in control rats while CA protection in HFD rats was abolished with brusatol indicating Nrf2-dependency. Such findings highlight CA as a promising nutraceutical candidate for preventing HFD-induced liver injury. Further studies are warranted to explore its clinical applicability in metabolic liver diseases. Full article
(This article belongs to the Special Issue Liver Diseases: From Pathophysiology to Novel Therapeutic Approaches)
Show Figures

Figure 1

13 pages, 1294 KB  
Review
VEXAS Syndrome: Genetics, Gender Differences, Clinical Insights, Diagnostic Pitfalls, and Emerging Therapies
by Salvatore Corrao, Marta Moschetti, Salvatore Scibetta, Luigi Calvo, Annarita Giardina, Ignazio Cangemi, Carmela Zizzo, Paolo Colomba and Giovanni Duro
Int. J. Mol. Sci. 2025, 26(16), 7931; https://doi.org/10.3390/ijms26167931 - 17 Aug 2025
Viewed by 309
Abstract
VEXAS syndrome (Vacuoles, E1-enzyme, X-linked, Autoinflammation, and Somatic) is a recently identified late-onset autoinflammatory disorder characterized by a unique interplay between hematological and inflammatory manifestations. It results from somatic mutations in the UBA1 gene, located on the short arm of the X chromosome. [...] Read more.
VEXAS syndrome (Vacuoles, E1-enzyme, X-linked, Autoinflammation, and Somatic) is a recently identified late-onset autoinflammatory disorder characterized by a unique interplay between hematological and inflammatory manifestations. It results from somatic mutations in the UBA1 gene, located on the short arm of the X chromosome. Initially, females were considered mere carriers, with the syndrome primarily affecting males over 50. However, recent evidence indicates that heterozygous females can exhibit symptoms as severe as those seen in hemizygous males. The disease manifests as systemic inflammation, macrocytic anemia, thrombocytopenia, chondritis, neutrophilic dermatoses, and steroid-dependent inflammatory symptoms. Due to its overlap with autoimmune and hematologic disorders such as relapsing polychondritis, Still’s disease, and myelodysplastic syndromes, misdiagnosis is common. At the molecular level, VEXAS syndrome is driven by impaired ubiquitination pathways, resulting in dysregulated immune responses and clonal hematopoiesis. A key diagnostic marker is the presence of cytoplasmic vacuoles in myeloid and erythroid precursors, though definitive diagnosis requires genetic testing for UBA1 mutations. Traditional immunosuppressants and TNF inhibitors are generally ineffective, while JAK inhibitors and IL-6 blockade provide partial symptom control. Azacitidine and decitabine have shown promise in reducing disease burden, but hematopoietic stem cell transplantation (HSCT) remains the only curative treatment, albeit with significant risks. This review provides a comprehensive analysis of VEXAS syndrome, examining its clinical features, differential diagnoses, diagnostic challenges, and treatment approaches, including both pharmacological and non-pharmacological strategies. By enhancing clinical awareness and optimizing therapeutic interventions, this article aims to bridge emerging genetic insights with practical patient management, ultimately improving outcomes for those affected by this complex and often life-threatening disease. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

20 pages, 14247 KB  
Article
Comparison of Primary Human Osteoblast-like Cells and hFOB 1.19 Cells: Contrasting Effects of Proinflammatory Cytokines
by Juliana Franziska Bousch, Christoph Beyersdorf, Katharina Schultz, Matthis Schnitker, Christoph Viktor Suschek and Uwe Maus
Cells 2025, 14(16), 1264; https://doi.org/10.3390/cells14161264 - 15 Aug 2025
Viewed by 413
Abstract
Proinflammatory cytokines such as IL-1β, IL-6, and TNF-α are key mediators of inflammatory bone loss and are commonly described as inhibitors of osteoblast function. However, their effects on osteogenesis remain controversial, likely due to the differences in the cell models and experimental settings [...] Read more.
Proinflammatory cytokines such as IL-1β, IL-6, and TNF-α are key mediators of inflammatory bone loss and are commonly described as inhibitors of osteoblast function. However, their effects on osteogenesis remain controversial, likely due to the differences in the cell models and experimental settings in in vitro studies. We recently showed that these cytokines significantly enhanced the mineralization of primary human osteoblast-like cells (OBs). Here, we provide the first analysis of cytokine effects on the osteogenesis of the widely used human osteoblastic cell line hFOB 1.19 and compare them to primary OBs. Unexpectedly, all three cytokines significantly inhibited mineralization in hFOB 1.19 cells without affecting the proliferation. IL-1β and TNF-α also suppressed ALP activity, whereas IL-6 acted ALP-independent but increased the osteogenic marker expression despite the reduced mineralization, indicating a possible uncoupled differentiation and mineralization. Morphological and transcriptional analyses indicated that hFOB 1.19 cells represent an earlier osteogenic differentiation stage, while primary OBs show phenotypic heterogeneity and donor-dependent expression profiles. These data demonstrate that proinflammatory cytokines can have severely different effects on the osteogenesis of different cell models, supported by the highly contradictory findings reported in the literature. Nevertheless, elucidating the mechanisms underlying the inhibition of osteogenesis in hFOB 1.19 cells may provide important insights into the cell model and differentiation-stage-specific cytokine effects. Full article
Show Figures

Figure 1

27 pages, 5167 KB  
Article
Development of Glycyrrhizic Acid Nanoparticles for Modulating Gastric Ulcer Healing: A Comparative In Vivo Study Targeting Oxidative Stress and Inflammatory Pathways
by Mody Albalawi and Sahar Khateeb
Antioxidants 2025, 14(8), 990; https://doi.org/10.3390/antiox14080990 - 12 Aug 2025
Viewed by 421
Abstract
Gastric ulcer (GU) is a common gastrointestinal disorder that impacts quality of life. Currently, several drugs are available for GU treatment, including proton pump inhibitors like omeprazole (OMP); however, their use is limited by numerous potential adverse effects. Glycyrrhizic acid (GLY), a natural [...] Read more.
Gastric ulcer (GU) is a common gastrointestinal disorder that impacts quality of life. Currently, several drugs are available for GU treatment, including proton pump inhibitors like omeprazole (OMP); however, their use is limited by numerous potential adverse effects. Glycyrrhizic acid (GLY), a natural anti-inflammatory agent, exhibits promising gastroprotective properties; however, its use is likewise limited by numerous potential adverse effects. This study aimed to synthesize GLY nanoparticles (GLY-NPs) to enhance their therapeutic potential and to comparatively evaluate their efficacy against OMP in an ethanol-induced GU in male Wistar rats. GLY-NPs were synthesized via a hydrothermal method and characterized using TEM, XRD, FTIR, and zeta potential analyses. In vivo, GLY-NPs significantly attenuated gastric mucosal damage compared to OMP, as evidenced by macroscopic and histopathological analyses. Biochemical assays revealed that GLY-NPs markedly improved antioxidant defenses by elevating SOD, catalase, and glutathione peroxidase activities while reducing MDA levels, surpassing the effects of OMP. Furthermore, GLY-NPs modulated inflammatory responses by downregulating p38 MAPK, NF-κB, and TNF-α expression, concomitant with upregulation of the anti-inflammatory cytokine IL-10. Mechanistic insights indicated that GLY-NPs favorably regulated key signaling pathways implicated in gastric mucosal protection, including suppression of the JAK2/STAT3 and TGF-β1/Smad3 pathways, alongside activation of the SIRT1/FOXO1/PGC-1α axis. In conclusion, these findings indicate that GLY-NPs offer higher gastroprotective effects relative to traditional OMP therapy through comprehensive modulation of oxidative stress, inflammation, and molecular signaling pathways. This study highlights GLY-NPs as a potent nanotherapeutic candidate for the effective management of GU. Full article
Show Figures

Figure 1

14 pages, 9863 KB  
Article
Examination of Behavioral and Neuropsychological Characteristics of Hungarian Patients with Juvenile Idiopathic Arthritis: A Cross-Sectional Analysis
by Diána Garan, Lilla Lengvári, Andrea Ponyi, Márton Szabados, Gyurgyinka Gergev, Imre Bozi, Wouter Wijker and Tamás Constantin
Children 2025, 12(8), 1057; https://doi.org/10.3390/children12081057 - 12 Aug 2025
Viewed by 267
Abstract
Background: Children with juvenile idiopathic arthritis (JIA) may experience chronic pain, contributing to psychological distress. Objective: The objective was to assess neuropsychological functions and behavioral skills in patients with JIA and identify demographic and disease-related factors influencing these outcomes. Methods: This cross-sectional study [...] Read more.
Background: Children with juvenile idiopathic arthritis (JIA) may experience chronic pain, contributing to psychological distress. Objective: The objective was to assess neuropsychological functions and behavioral skills in patients with JIA and identify demographic and disease-related factors influencing these outcomes. Methods: This cross-sectional study evaluated 112 patients at the Division of Pediatric Rheumatology, Semmelweis University (2015–2016). Participants completed psychological assessments using the Child Behavior Checklist and Woodcock–Johnson III Tests. Examined variables included demographic (age and sex), clinical (age at diagnosis and disease activity), and treatment-related factors (therapy type and duration). Treatment groups comprised (a) combination therapy with TNF inhibitor and methotrexate (MTX) (n = 60), (b) MTX monotherapy (n = 34), and (c) TNF inhibitor monotherapy (n = 18). Results: Neuropsychological variables showed no clinically significant differences between treatment groups. These skills were unaffected by age, sex, therapy duration, or disease activity. Pathological behavioral scores were significantly higher (p < 0.05) in younger patients (<7 years), with females showing greater susceptibility to anxiety and depression (p < 0.05). Conclusions: No clinically significant psychological impairments were observed in our cohort. Further research is warranted to clarify the significance of abnormal behavioral scores. Psychological care provision remains vital for improving the quality of life in JIA patients. Full article
Show Figures

Figure 1

14 pages, 1050 KB  
Article
Harringtonine Attenuates Extracellular Matrix Degradation, Skin Barrier Dysfunction, and Inflammation in an In Vitro Skin Aging Model
by Sullim Lee and Sanghyun Lee
Curr. Issues Mol. Biol. 2025, 47(8), 642; https://doi.org/10.3390/cimb47080642 - 10 Aug 2025
Viewed by 350
Abstract
With the growing interest in natural strategies for preventing skin aging, plant-derived compounds are being actively investigated for their potential protective effects against skin inflammation and extracellular matrix (ECM) degradation. In this study, we explored the anti-aging and anti-inflammatory effects of harringtonine, an [...] Read more.
With the growing interest in natural strategies for preventing skin aging, plant-derived compounds are being actively investigated for their potential protective effects against skin inflammation and extracellular matrix (ECM) degradation. In this study, we explored the anti-aging and anti-inflammatory effects of harringtonine, an alkaloid isolated from Cephalotaxus harringtonia, in normal human epidermal keratinocytes (NHEKs) under inflammatory stress induced by tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ). Harringtonine significantly suppressed the expression of matrix metalloproteinases (MMP)-1, MMP-2, and MMP-9 and restored the expression of collagen synthesis-related genes [collagen type I alpha 1 chain (COL1A1), collagen type I alpha 2 chain (COL1A2), and collagen type IV alpha 1 chain COL4A1)], indicating its protective role in ECM degradation. Additionally, harringtonine improved the expression of skin barrier-related genes, such as serine peptidase inhibitor kazal type 5 (SPINK5), loricrin (LOR), quaporin-3 (AQP3), filaggrin (FLG), and keratin 1 (KRT1) although it had no significant effect on involucrin (IVL). Harringtonine also markedly reduced the production of pro-inflammatory cytokines [interleukin (IL)-1β, IL-6, and IL-8] and inflammatory mediators, including prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), and nitric oxide (NO). Our findings suggest that harringtonine may serve as a promising natural compound for mitigating skin aging and inflammation through multi-targeted modulation of ECM remodeling, skin barrier function, and inflammatory response. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

15 pages, 2691 KB  
Review
SGLT2 Inhibitors: Multifaceted Therapeutic Agents in Cardiometabolic and Renal Diseases
by Ana Checa-Ros, Owahabanun-Joshua Okojie and Luis D’Marco
Metabolites 2025, 15(8), 536; https://doi.org/10.3390/metabo15080536 - 7 Aug 2025
Viewed by 384
Abstract
Background: Sodium–glucose cotransporter-2 inhibitors (SGLT2is), initially developed as antihyperglycemic agents, have emerged as multifunctional therapeutics with profound cardiorenal and metabolic benefits. Their unique insulin-independent mechanism, targeting renal glucose reabsorption, distinguishes them from conventional antidiabetic drugs. Mechanisms and Clinical Evidence: SGLT2is induce [...] Read more.
Background: Sodium–glucose cotransporter-2 inhibitors (SGLT2is), initially developed as antihyperglycemic agents, have emerged as multifunctional therapeutics with profound cardiorenal and metabolic benefits. Their unique insulin-independent mechanism, targeting renal glucose reabsorption, distinguishes them from conventional antidiabetic drugs. Mechanisms and Clinical Evidence: SGLT2is induce glycosuria, reduce hyperglycemia, and promote weight loss through increased caloric excretion. Beyond glycemic control, they modulate tubuloglomerular feedback, attenuate glomerular hyperfiltration, and exert systemic effects via natriuresis, ketone utilization, and anti-inflammatory pathways. Landmark trials (DAPA-HF, EMPEROR-Reduced, CREDENCE, DAPA-CKD) demonstrate robust reductions in heart failure (HF) hospitalizations, cardiovascular mortality, and chronic kidney disease (CKD) progression, irrespective of diabetes status. Adipose Tissue and Metabolic Effects: SGLT2is mitigate obesity-associated adiposopathy by shifting macrophage polarization (M1 to M2), reducing proinflammatory cytokines (TNF-α, IL-6), and enhancing adipose tissue browning (UCP1 upregulation) and mitochondrial biogenesis (via PGC-1α/PPARα). Modest weight loss (~2–4 kg) occurs, though compensatory hyperphagia may limit long-term effects. Emerging Applications: Potential roles in non-alcoholic fatty liver disease (NAFLD), polycystic ovary syndrome (PCOS), and neurodegenerative disorders are under investigation, driven by pleiotropic effects on metabolism and inflammation. Conclusions: SGLT2is represent a paradigm shift in managing T2DM, HF, and CKD, with expanding implications for metabolic syndrome. Future research should address interindividual variability, combination therapies, and non-glycemic indications to optimize their therapeutic potential. Full article
(This article belongs to the Special Issue Metabolic Modulators in Cardiovascular Disease Management)
Show Figures

Figure 1

18 pages, 1256 KB  
Article
Longitudinal Assessment of Body Composition and Inflammatory Status in Rheumatoid Arthritis During TNF Inhibitor Treatment: A Pilot Study
by Natalia Mena-Vázquez, Aimara García-Studer, Fernando Ortiz-Márquez, Sara Manrique-Arija, Arkaitz Mucientes, Jose Manuel Lisbona-Montañez, Paula Borregón-Garrido, Patricia Ruiz-Limón, Rocío Redondo-Rodriguez, Laura Cano-García and Antonio Fernández-Nebro
Int. J. Mol. Sci. 2025, 26(15), 7635; https://doi.org/10.3390/ijms26157635 - 7 Aug 2025
Viewed by 393
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease frequently associated with alterations in body composition, including reduced lean mass and increased fat mass. These alterations are thought to be driven by persistent systemic inflammation, which may be influenced by inflammatory activity and by [...] Read more.
Rheumatoid arthritis (RA) is a chronic inflammatory disease frequently associated with alterations in body composition, including reduced lean mass and increased fat mass. These alterations are thought to be driven by persistent systemic inflammation, which may be influenced by inflammatory activity and by therapeutic interventions. Objectives: This pilot study aimed to provide preliminary data on changes in body composition and inflammatory activity in biologic-naive patients with active RA during the initial 6 months of TNF inhibitor treatment, and to compare baseline body composition with healthy controls. We conducted a single-center, observational, 24-week pilot study of 70 biologic-naive RA patients with moderate-to-severe disease activity and 70 matched healthy controls. Lean mass, fat mass, and lean mass index (LMI) were measured using dual-energy X-ray absorptiometry at baseline for both groups, and after 6 months only in the RA group. Clinical, laboratory, adipokines, and cytokine parameters were also recorded. At baseline, RA patients had lower lean mass and LMI than controls. Over 6 months, RA patients showed significant clinical and laboratory improvement, with a corresponding increase in lean mass and LMI. No statistically significant change was observed in fat mass. The increase in lean mass was paralleled by a reduction in inflammatory markers. The LMI was inversely associated with female sex (β = −0.562) and C-reactive protein (β = −0.432) and directly associated with body mass index (β = 0.570). Similar associations were observed for total lean mass and change in lean mass, as well as for DAS28 (β = −0.333). This pilot study provides preliminary evidence that TNF inhibitor therapy may be associated with increased lean mass and decreased inflammation in RA patients. Owing to the absence of a comparator RA group not receiving TNF inhibitors, these findings should be interpreted as hypothesis-generating. Full article
Show Figures

Figure 1

Back to TopTop