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
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
remove_circle_outline

Search Results (3,366)

Search Parameters:
Keywords = anti-IL-1 treatment

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 3029 KB  
Article
Selective STAT3 Allosteric Inhibitors HCB-5300 and HCB-5400 Alleviate Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice
by Wook-Young Baek, Ji-Won Kim, So-Won Park, Nan Kim, Sun-Gyo Lim and Chang-Hee Suh
Int. J. Mol. Sci. 2025, 26(24), 11981; https://doi.org/10.3390/ijms262411981 - 12 Dec 2025
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract, marked by persistent mucosal inflammation and structural damage. We evaluated the efficacy of HCB-5300 and HCB-5400, novel selective STAT3 allosteric inhibitors, in a mouse model of dextran sulfate sodium (DSS)-induced [...] Read more.
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract, marked by persistent mucosal inflammation and structural damage. We evaluated the efficacy of HCB-5300 and HCB-5400, novel selective STAT3 allosteric inhibitors, in a mouse model of dextran sulfate sodium (DSS)-induced ulcerative colitis. Colitis was induced in C57BL/6 mice using 3% DSS in water for 5 d. HCB-5300 (25 mg/kg) or HCB-5400 (12.5 mg/kg) was administered orally during induction. Disease progression was assessed using the disease activity index (DAI), considering body weight, stool consistency, and rectal bleeding. Colon length and histopathological analyses were used to evaluate mucosal integrity and inflammatory damage. Interleukin (IL)-6 levels were quantified using enzyme-linked immunosorbent assay, and kidney pathology was assessed for systemic effects. HCB-5300 and HCB-5400 significantly mitigated DSS-induced colitis, as evidenced by reduced body weight loss, improved DAI scores, preserved colon length, and decreased mucosal damage and inflammation in the treated mice. IL-6 levels were significantly lower in both treatment groups, indicating effective STAT3 inhibition. HCB-5400 exhibited superior efficacy for most parameters. HCB-5300 and HCB-5400 are potent and selective STAT3 allosteric inhibitors with notable anti-inflammatory effects. HCB-5400’s efficacy underscores its potential as a therapeutic candidate for managing inflammatory flares in IBD. Full article
Show Figures

Figure 1

13 pages, 1802 KB  
Article
Lactose-Modified Hyaluronic Acid Molecule Attenuates In Vitro Chondrocyte Inflammation
by Alice Cristina Donato, Elisa Belluzzi, Valentina Masola, Pietro Ruggieri and Paola Brun
Cells 2025, 14(24), 1977; https://doi.org/10.3390/cells14241977 - 12 Dec 2025
Abstract
Background: Osteoarthritis (OA) is a chronic degenerative whole joint disease characterized by cartilage breakdown and inflammation. Galectin-3 (Gal-3), a β-galactoside-binding lectin secreted into the extracellular space, binds to glycosylated components of the extracellular matrix (ECM), modulating cell–matrix interactions and inflammation. This study aims [...] Read more.
Background: Osteoarthritis (OA) is a chronic degenerative whole joint disease characterized by cartilage breakdown and inflammation. Galectin-3 (Gal-3), a β-galactoside-binding lectin secreted into the extracellular space, binds to glycosylated components of the extracellular matrix (ECM), modulating cell–matrix interactions and inflammation. This study aims to evaluate the anti-inflammatory effects of Hylach®, a hyaluronic acid (HA) derivative conjugated with lactose-based residues that bind Gal-3, on in vitro inflamed primary human chondrocytes. Methods: Chondrocyte viability, after both Hylach® and HA treatments at different concentrations was assessed using the MTT assay. Two-dimensional and 3D cell cultures exposed to the conditioned medium (CM) of activated U937 monocytes and subsequently treated with Hylach or HA, were analyzed for the expression of IL-1β, IL-6, TNF-α, and Gal-3 at different time points (4, 10, and 24 h). Results: HA and Hylach® did not affect cell viability at any of the tested concentrations. Both molecules reduced the overexpression of Gal-3 and pro-inflammatory molecules in 2D inflamed cell cultures, at both gene and protein levels. Notably, IL-1β, IL-6 and Gal-3 showed a more pronounced inhibitory effect at 4 h, with Hylach demonstrating a stronger reduction compared to native HA. Moreover, in inflamed 3D chondrocyte cultures, Hylach® but not HA, significantly reduced IL-1β, TNF-α and Gal-3 gene expression. Conclusions: Hylach® exerts an early and more potent anti-inflammatory effect in inflamed 2D and 3D chondrocyte cultures when compared to HA. These findings suggest that targeting Gal-3 through selective HA derivatives may represent a promising strategy for modulating both inflammation and matrix remodelling in OA. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

21 pages, 5226 KB  
Article
Agathisflavone Modulates the Kynurenine Pathway and Glial Inflammatory Responses with Implications for Neuroprotection
by Deivison Silva Argolo, Lucas Matheus Gonçalves Oliveira, Cleonice Creusa dos Santos, Lilian Vanessa da Penha Gonçalves, Erick Correia Loiola, Bruno Solano de Freitas Souza, George E. Barreto, Arthur Morgan Butt, Jorge Mauricio David, Alexsandro Branco, Isabella Mary Alves Reis, Annabel Azevedo-Silva, Silvia Lima Costa and Maria de Fátima Dias Costa
Int. J. Mol. Sci. 2025, 26(24), 11951; https://doi.org/10.3390/ijms262411951 - 11 Dec 2025
Abstract
The cells in the central nervous system (CNS) can adapt to injury and inflammation through structural and functional changes, many of which are mediated by the kynurenine pathway (KP). Studies using glia–neuron co-cultures showed that the biflavonoid agathisflavone (FAB), purified from the leaves [...] Read more.
The cells in the central nervous system (CNS) can adapt to injury and inflammation through structural and functional changes, many of which are mediated by the kynurenine pathway (KP). Studies using glia–neuron co-cultures showed that the biflavonoid agathisflavone (FAB), purified from the leaves of Cenostigma pyramidale Tul., a plant native to the Brazilian caatinga, exerts strong neuroprotective effects. This study evaluated whether agathisflavone (1 µM) modulates these responses in human and murine astrocytes and microglia exposed to inflammatory activation with lipopolysaccharide (LPS, 1 µg/mL), excitotoxic activation of NMDA receptors with quinolinic acid (QUIN, 500 µM), or inhibition of the KP rate-limiting enzyme indoleamine 2,3-dioxygenase 1 (IDO1) with 1-methyl tryptophan (1-MT, 1.5 μM). Co-treatment with FAB increased astrocyte viability relative to LPS, QUIN, or 1-MT alone, by up to 35% (p < 0.05), while reducing GFAP overexpression and other features of reactive astrogliosis. FAB decreased the proportion of Iba-1+ microglia, indicating anti-inflammatory effects. When combined with QUIN or 1-MT, FAB reversed the elevation of iNOS (p < 0.0001) and reduced IL1β upregulation. FAB also modulated KP activity in a cell type-specific manner. In astrocytes, FAB with QUIN or with 1-MT increased IDO activity, whereas in microglia, FAB alone reduced it. In microglia, kynurenine-3-monooxygenase (KMO) expression was significantly increased under FAB+QUIN or FAB+1-MT (p < 0.0001). Finally, astrocyte-conditioned medium from FAB-treated cells increased the viability of neuron-like PC12 cells by up to 40%. Collectively, these findings show that FAB confers cytoprotective and anti-inflammatory actions on glial cells, modulates KP signalling in a context-dependent manner, and supports neuronal survival under neuroinflammatory conditions. Full article
(This article belongs to the Special Issue Recent Advances in Bioactive Compounds in Human Health)
Show Figures

Figure 1

24 pages, 4674 KB  
Article
Combination Therapy with Azithromycin and Clostridium butyricum Retains Anti-Diarrheal Efficacy but Partially Compromises Gut Microbiota Restoration Compared to Probiotics Monotherapy
by Cai-Yun Wang, Meng-Yue Zhang, Hai-Ying Zhang, Yi-Meng Yang, Lu Zhang and Yi-Xuan Zhang
Microorganisms 2025, 13(12), 2812; https://doi.org/10.3390/microorganisms13122812 - 10 Dec 2025
Viewed by 1
Abstract
Most probiotics require separate administration from antibiotics due to sensitivity issues. Clostridium butyricum, however, exhibits intrinsic resistance, making it a promising candidate for combined therapy against diarrhea. In this study, a diarrhea model was established in mice induced by Escherichia coli, [...] Read more.
Most probiotics require separate administration from antibiotics due to sensitivity issues. Clostridium butyricum, however, exhibits intrinsic resistance, making it a promising candidate for combined therapy against diarrhea. In this study, a diarrhea model was established in mice induced by Escherichia coli, followed by treatment with azithromycin (AZM), C. butyricum (RH2), or their combination (COM) to assess therapeutic efficacy. The results demonstrated that mice in RH2 and COM groups achieved full body weight recovery and significant alleviation of diarrhea, accompanied by normalized fecal E. coli loads, preserved tissue integrity, reduced pro-inflammatory cytokines (IL-1β, IL-6, TNF-α), and elevated anti-inflammatory IL-10. In contrast, AZM treatment led to sex-specific disparities in weight recovery and E. coli loads, and both sexes experienced relapse-prone diarrhea. Furthermore, the AZM group displayed shortened colons, sustained inflammatory infiltration, epithelial damage, and elevated IL-1β and male-specific IL-6. Gut microbiota analysis revealed that the COM group retained beneficial genera (e.g., Parabacteroides, Blautia) from the AZM group while uniquely enriching Lachnospiraceae taxa (e.g., NK4A136_group, FCS020_group). Untargeted metabolomics demonstrated the COM group activated GABA/arginine pathways, enhancing anti-inflammatory and barrier functions, whereas azithromycin disrupted butyrate synthesis and steroid metabolism. These findings highlight the advantage of combining C. butyricum with azithromycin for intestinal protection. Full article
(This article belongs to the Section Antimicrobial Agents and Resistance)
Show Figures

Graphical abstract

17 pages, 1852 KB  
Article
Glypican-3-Specific CAR NK Cells Co-Secreting IL-15 and IFN-α Have Increased Anti-Tumor Function Versus Hepatocellular Carcinoma In Vitro
by Rosalia Busà, Gioacchin Iannolo, Bruno Douradinha, Duilio Pagano, Anna Gallina, Giancarlo Cappello, Antonio La Rocca, Salvatore Gruttadauria, Pier Giulio Conaldi and Ester Badami
Int. J. Mol. Sci. 2025, 26(24), 11892; https://doi.org/10.3390/ijms262411892 - 10 Dec 2025
Viewed by 52
Abstract
Chimeric antigen receptor (CAR)-modified natural killer (NK) cells represent a promising immunotherapeutic approach for the treatment of oncological malignancies such as hepatocellular carcinoma (HCC). In this work, we have engineered primary human NK cells, re-directing them so they can specifically recognize Glypican-3 (GPC3), [...] Read more.
Chimeric antigen receptor (CAR)-modified natural killer (NK) cells represent a promising immunotherapeutic approach for the treatment of oncological malignancies such as hepatocellular carcinoma (HCC). In this work, we have engineered primary human NK cells, re-directing them so they can specifically recognize Glypican-3 (GPC3), an immunotherapeutic target for HCC. In previous studies, we have demonstrated that IFN-α significantly enhances NK cells’ anti-tumor and anti-viral cytotoxicity. Fourth-generation self-inactivating lentiviral vectors were used to deliver a transgenic expression of IFN-α or its co-expression with IL-15 (which induces NK cells expansion, survival, and function), aiming to enhance CAR-GPC3 NK cells’ anti-tumor response against HCC. We optimized a protocol for efficient transduction of primary NK cells, demonstrating that CAR expression is maintained at high levels over time. Exposure of HCC ectopically expressing GPC3+ to CAR-GPC3-IL15 and CAR-GPC3-IL15-IFNα NK cells demonstrated significant in vitro cytotoxicity and cytokine production, dependent on GPC3 expression. To prevent undesired side effects of CAR-NK cell immunotherapy, co-delivery with a suicide gene is advised as a safety measure. Thus, a truncated epidermal growth factor receptor (tEGFR) was co-delivered with the anti-GPC3 CAR, which efficiently promoted the suicide of the CAR-NK used in this work. Our study demonstrates the efficacy of re-directed CAR-GPC3 primary NK cells, encouraging further preclinical and clinical translation studies and strengthening the potential of these cells as a novel treatment option for patients with HCC. Full article
(This article belongs to the Special Issue Chimeric Antigen Receptors Against Cancers and Autoimmune Diseases)
Show Figures

Figure 1

9 pages, 407 KB  
Article
Rapamycin Modulates the Polarisation of CD4+ T Cells Towards TH1 Cells in Patients with Active Granulomatosis with Polyangiitis and Microscopic Polyangiitis
by Jang Woo Ha, Taejun Yoon, Oh Chan Kwon, Yong-Beom Park and Sang-Won Lee
J. Clin. Med. 2025, 14(24), 8720; https://doi.org/10.3390/jcm14248720 - 9 Dec 2025
Viewed by 100
Abstract
Objective: This study investigated whether rapamycin could modulate the polarisation of CD4+ T cells towards TH1, TH2, TH17, and Treg cells using peripheral blood mononuclear cell (PBMC) obtained from patients with granulomatosis with polyangiitis and microscopic polyangiitis [...] Read more.
Objective: This study investigated whether rapamycin could modulate the polarisation of CD4+ T cells towards TH1, TH2, TH17, and Treg cells using peripheral blood mononuclear cell (PBMC) obtained from patients with granulomatosis with polyangiitis and microscopic polyangiitis (GPA/MPA). Methods: Twenty patients with GPA/MPA were included in this study. Their stored PBMCs were cultured and stimulated with anti-CD3 and anti-CD28 antibodies for 72 h in the presence or absence of rapamycin (10 nM). The cells were stained for surface markers with anti-CD4-FITC and anti-CD25-APC, followed by intracellular staining using anti-interferon (IFN)-γ-PE, anti-IL-4-PerCP-Cy5, anti-IL17A-APC, and anti-Foxp3-PE. The stained cells were analysed using a flow cytometer. Results: The median age of the 20 GPA/MPA patients (10 men and 10 women) was 65.5 years. Rapamycin treatment significantly modulated the polarisation of CD4+IFN-γ+ T (TH1) cells compared to no treatment among GPA/MPA patients. In addition, the polarisation of CD4+IFN-γ+ T (TH1) cells was also significantly reduced in rapamycin-treated PBMC obtained from active patients compared to untreated PBMC from the same patients; however, these alterations were not observed in inactive patients. Conversely, rapamycin treatment did not affect the polarisation of CD4+IL-4+ T (TH2), CD4+IL-17+ T (TH17), or CD4+FoxP3+CD25+ T (Treg) cells, regardless of GPA/MPA activity. Conclusions: This study was the first pilot study to demonstrate that rapamycin modulates the polarisation of CD4+ T cells towards CD4+IFN-γ+ T cells in active GPA/MPA. Full article
(This article belongs to the Section Immunology & Rheumatology)
Show Figures

Figure 1

17 pages, 2907 KB  
Article
Potential Anti-Inflammatory Effects of Gintonin-Enriched Fraction in TNF-α-Stimulated Keratinocytes
by Rami Lee, Kyung-Jong Won, Ji-Hun Kim, Sun-Hye Choi, Sung-Hee Hwang and Seung-Yeol Nah
Int. J. Mol. Sci. 2025, 26(24), 11864; https://doi.org/10.3390/ijms262411864 - 9 Dec 2025
Viewed by 68
Abstract
Gintonin-enriched fraction (GEF), a bioactive glycolipoprotein derived from Panax ginseng is known for its potential as a natural anti-inflammatory agent. Keratinocytes are closely related to the development and progression of various inflammatory skin conditions. However, the effect of GEF on inflammation-related responses in [...] Read more.
Gintonin-enriched fraction (GEF), a bioactive glycolipoprotein derived from Panax ginseng is known for its potential as a natural anti-inflammatory agent. Keratinocytes are closely related to the development and progression of various inflammatory skin conditions. However, the effect of GEF on inflammation-related responses in keratinocytes remains unclear. This study aimed to investigate whether GEF modulates key inflammatory responses in keratinocytes stimulated by tumor necrosis factor (TNF)-α. The effect of GEF on biological activities in TNF-α-stimulated keratinocytes (HaCaT cells) was evaluated using water-soluble tetrazolium salt, enzyme-linked immunosorbent, immunostaining, and immunoblotting assays. In TNF-α-stimulated HaCaT cells, GEF attenuated reactive oxygen species production, nitric oxide release, and inducible nitric oxide synthase expression. Moreover, GEF reduced the release of interleukin (IL)-6 and RANTES, while increasing the release of IL-10 in TNF-α-exposed HaCaT cells. Additionally, GEF treatment resulted in reduced cyclooxygenase-2 expression and prostaglandin E2 release and inhibited TNF-α-induced translocation of nuclear factor-κB in HaCaT cells. Furthermore, TNF-α and IL-6 levels in ultraviolet B-irradiated HaCaT cells were reduced by GEF treatment. These findings indicated that GEF exerts anti-inflammatory effects on keratinocytes. This study provides a basis for the development of novel therapeutic approaches for the prevention and treatment of inflammatory skin disorders. Full article
(This article belongs to the Special Issue The Role of Natural Products in Treating Human Diseases)
Show Figures

Graphical abstract

19 pages, 2137 KB  
Article
IL-12 as a Potential Prognostic Marker in Penile Cancer: Implications for Immune Dysregulation
by Mateusz Czajkowski, Michał Kunc, Jacek Kieżun, Bartłomiej Emil Kraziński, Marcin Matuszewski, Weronika Łyzińska, Oliver W. Hakenberg and Piotr M. Wierzbicki
Int. J. Mol. Sci. 2025, 26(24), 11829; https://doi.org/10.3390/ijms262411829 - 7 Dec 2025
Viewed by 209
Abstract
Penile cancer (PeCa) is a rare malignancy with few validated tissue biomarkers to guide prognosis and treatment, despite growing evidence for a key role of inflammation in its biology. This retrospective study evaluated whether the immuno-expression of selected pro-inflammatory cytokines is associated with [...] Read more.
Penile cancer (PeCa) is a rare malignancy with few validated tissue biomarkers to guide prognosis and treatment, despite growing evidence for a key role of inflammation in its biology. This retrospective study evaluated whether the immuno-expression of selected pro-inflammatory cytokines is associated with disease progression and cancer-specific survival (CSS) in PeCa. Immunohistochemistry (IHC) analysis of eight cytokines (IL-1A, IL-1B, IL-2, IL-6, IL-12, TGF-β1, TNF-α and IFN-γ) was performed in paired tumour tissues and corresponding negative surgical margins from 94 patients with penile squamous cell carcinoma. Compared with surgical margins, tumour tissues showed a characteristic inflammatory shift, with markedly increased IL-1β and IL-6 and relatively reduced TNF-α, IFN-γ, IL-12 and IL-2. Receiver Operating Characteristic (ROC) analysis indicated that TNF-α, IL-6 and IL-12 had the strongest ability to discriminate tumour from normal tissue and provided data-driven cut-offs for subsequent analyses. Within tumour samples, high IL-1α, IL-12 and TGF-β1 immuno-expression was significantly associated with advanced UICC TNM prognostic stage and lymph node involvement. Importantly, in contrast to the classically anti-tumour role of IL-12 described in many other solid cancers, increased IL-12 immuno-expression in tumour tissues in our cohort was independently associated with poorer CSS in multivariable Cox regression (HR 2.42, 95% CI: 1.08–5.41, p = 0.031), alongside advanced TNM stage (HR 5.03, 95% CI: 2.12–11.95, p = 0.0002). These findings highlight IL-1α, IL-12 and TGF-β1 as promising tissue biomarkers of aggressive PeCa and support a central role for cytokine-driven immune dysregulation in penile cancer. The prognostic value of IL-12 should be considered exploratory and warrants validation in larger, multicentre cohorts. Full article
Show Figures

Figure 1

21 pages, 2706 KB  
Article
Anti-Inflammatory and Immunomodulatory Effects of Aqueous Extracts from Green Leaves and Rhizomes of Posidonia oceanica (L.) Delile on LPS-Stimulated RAW 264.7 Macrophages
by Giulia Abruscato, Daniela Ganci, Federica Bellistrì, Roberto Chiarelli, Manuela Mauro, Aiti Vizzini, Vincenzo Arizza, Mirella Vazzana and Claudio Luparello
Molecules 2025, 30(24), 4685; https://doi.org/10.3390/molecules30244685 - 7 Dec 2025
Viewed by 125
Abstract
The marine angiosperm Posidonia oceanica (Linnaeus) Delile, 1813 is a rich source of phytotherapeutic compounds whose potential applications for human health remain largely uninvestigated. Here, we determined the differential impact of aqueous extracts from P. oceanica’s green leaves (GLE) and rhizomes (RE) [...] Read more.
The marine angiosperm Posidonia oceanica (Linnaeus) Delile, 1813 is a rich source of phytotherapeutic compounds whose potential applications for human health remain largely uninvestigated. Here, we determined the differential impact of aqueous extracts from P. oceanica’s green leaves (GLE) and rhizomes (RE) on the inflammation-related mRNA expressions and protein levels, nitric oxide (NO) release, and endocytic activity in LPS-stimulated RAW 264.7 macrophages. We also examined the influence of the extracts in modulating the activation of components of intracellular signaling pathways. Co-treatments of LPS-stimulated RAW 264.7 cells in the presence of either GLE or RE resulted in a reduction in NO production, associated with a down-regulation of Nos2 expression, reduced levels of COX-2 and TNFα proteins, and a decrease in Nfkb1 expression and NF-κB activation. No effect was exerted on the release of IL-6. Moreover, co-exposures to LPS and the extracts led to an elevation in pJNK and pAKT levels alongside a reduction in pERK. In contrast to GLE, RE specifically lowered IL-1β production, induced a more robust increase in IL-10, positively influenced the endocytic function of RAW 264.7 cells, and drastically up-regulated the phosphorylation of p38. The data obtained indicate that GLE and RE exhibit considerable promise as prospective anti-inflammatory and immunomodulatory agents. Full article
Show Figures

Graphical abstract

18 pages, 3893 KB  
Article
Bcl3 Deficiency Leads to Hyperinflammation in Zebrafish
by Chengjian Fan, Nana Ai, Wei Ge and Vivien Ya-Fan Wang
Cells 2025, 14(24), 1935; https://doi.org/10.3390/cells14241935 - 5 Dec 2025
Viewed by 265
Abstract
B-cell leukemia/lymphoma protein 3 (Bcl3), a member of the IκB family proteins, regulates the transcriptional activities of the NF-κB family of transcription factors. It is known that aberrant Bcl3 activities induce malignancies of both hematologic and non-hematologic origins. Overexpressed, mutated and/or phosphorylated Bcl3 [...] Read more.
B-cell leukemia/lymphoma protein 3 (Bcl3), a member of the IκB family proteins, regulates the transcriptional activities of the NF-κB family of transcription factors. It is known that aberrant Bcl3 activities induce malignancies of both hematologic and non-hematologic origins. Overexpressed, mutated and/or phosphorylated Bcl3 has been implicated in several cancers due to its altered transcriptional activities. However, the physiological function of Bcl3 in immune homeostasis remained elusive to date. In this study, Bcl3 knockout zebrafish were generated to investigate its role in immune regulation. Bcl3 deficient zebrafish exhibited growth retardation and significantly reduced survival. Histological analyses revealed the absence of Hassall bodies in the thymus and hepatocellular nuclear abnormalities, indicating compromised integrity of the immune organs. Zebrafish with Bcl3 deficiency further showed enhanced immune responses and increased susceptibility to both bacterial and viral infections, resulting in significantly elevated levels of pro-inflammatory cytokines il1b, il6, il8, and tnfa. Treatment with the anti-inflammatory drug dexamethasone (Dex) effectively alleviated inflammation, downregulated pro-inflammatory cytokine expressions and improved survival. Collectively, our findings demonstrate Bcl3 as a key regulator of immune activation in vivo, highlighting its role in maintaining immune homeostasis and promoting organismal survival. Full article
Show Figures

Figure 1

24 pages, 15211 KB  
Article
Integrative Network Pharmacology and Multi-Omics Analysis Reveal Key Targets and Mechanisms of Saikosaponin B1 Against Acute Lung Injury
by Yuanfei Niu, Meiting Liu, Shuang Cui, Kaiyang Liu, Mengyuan Yang, Xiaozhen Hu, Changhui Zheng, Lianmei Wang and Junling Cao
Metabolites 2025, 15(12), 782; https://doi.org/10.3390/metabo15120782 - 4 Dec 2025
Viewed by 199
Abstract
Background/Objectives: Acute lung injury (ALI) is a severe condition driven largely by inflammation and has limited therapeutic options. Although saikosaponin B1 (SSB1), a primary bioactive saponin from Bupleurum Radix, has demonstrated anti-inflammatory properties, its efficacy against ALI and its corresponding molecular mechanisms [...] Read more.
Background/Objectives: Acute lung injury (ALI) is a severe condition driven largely by inflammation and has limited therapeutic options. Although saikosaponin B1 (SSB1), a primary bioactive saponin from Bupleurum Radix, has demonstrated anti-inflammatory properties, its efficacy against ALI and its corresponding molecular mechanisms remain largely unexplored. This study employed an integrated approach combining network pharmacology, transcriptomics, and metabolomics to decipher the protective mechanisms of SSB1 against ALI. Methods: Potential targets were identified via network pharmacology, and core targets were validated through molecular docking, dynamics simulations, and independent GEO transcriptomic datasets. Experimental validation was performed in an LPS-induced murine ALI model, combining histopathology, ELISA, and integrated transcriptomic and metabolomic analyses. Results: Integrated analyses identified IL1B, TNF, and IL6 as core targets through which SSB1 exerts its anti-ALI effects. These targets were validated by high-affinity binding in simulations, confirmed in independent GEO transcriptomic datasets, and shown to be normalized by SSB1 treatment in vivo. Mechanistically, SSB1 appears to modulate the NOD-like receptor and cGAS-STING signaling pathways and rectify the key metabolic pathways orchestrated by these targets, including glycerophospholipid, arachidonic acid, and linoleic acid metabolism. Conclusions: This study systematically investigates the therapeutic effects of SSB1 against ALI by identifying its potential targets and underlying pathways. These results provide crucial mechanistic insights and robust experimental support, thereby paving the way for the clinical translation of SSB1. Full article
(This article belongs to the Section Pharmacology and Drug Metabolism)
Show Figures

Figure 1

30 pages, 1934 KB  
Review
Low pH, High Stakes: A Narrative Review Exploring the Acid-Sensing GPR65 Pathway as a Novel Approach in Renal Cell Carcinoma
by Michael Grant, Barbara Cipriani, Alastair Corbin, David Miller, Alan Naylor, Stuart Hughes, Tom McCarthy, Sumeet Ambarkhane, Danish Memon, Michael Millward, Sumanta Pal and Ignacio Melero
Cancers 2025, 17(23), 3883; https://doi.org/10.3390/cancers17233883 - 4 Dec 2025
Viewed by 177
Abstract
Renal cell carcinoma (RCC) is a biologically heterogeneous malignancy accounting for 3% of adult cancers globally. Despite advances in immune checkpoint inhibitors (ICIs) and vascular endothelial growth factor (VEGF)-targeted therapies, durable disease control remains elusive for many patients. Increasing evidence implicates the acidic [...] Read more.
Renal cell carcinoma (RCC) is a biologically heterogeneous malignancy accounting for 3% of adult cancers globally. Despite advances in immune checkpoint inhibitors (ICIs) and vascular endothelial growth factor (VEGF)-targeted therapies, durable disease control remains elusive for many patients. Increasing evidence implicates the acidic tumour microenvironment (TME) as a critical mediator of RCC progression, immune evasion, and therapeutic resistance. Solid tumours, including RCC, exhibit reversed pH gradients, characterised by acidic extracellular (pH 6.2–6.9) and alkaline intracellular conditions. This dysregulation arises from enhanced glycolysis, hypoxia-driven lactate accumulation, and the overexpression of pH-regulating enzymes such as carbonic anhydrase (CA9). Acidic TMEs impair cytotoxic T-cell and NK-cell activity, promote tumour-associated macrophage (TAM) polarisation towards an immunosuppressive phenotype, and upregulate alternative immune checkpoints. These mechanisms collectively undermine ICI efficacy and contribute to primary and secondary treatment resistance. Proton-sensing G-protein-coupled receptors (GPCRs), notably GPR65, have emerged as pivotal mediators linking extracellular acidosis to immune dysfunction. Preclinical studies demonstrate that GPR65 antagonists restore anti-tumour immune activity by reversing acidosis-driven immunosuppression and enhancing antigen processing. In RCC models, selective GPR65 inhibitors have shown the ability to reduce immunosuppressive cytokine IL-10 production, induce immunoproteasome activation, and synergise with anti-PD-1 therapy. The first-in-class GPR65 inhibitor, PTT-4256, is now under evaluation in the Phase I/II RAISIC-1 trial (NCT06634849) in solid tumours, including RCC. Targeting acid-sensing pathways represents a novel and promising therapeutic strategy in RCC, aiming to remodel the TME and overcome ICI resistance. Integrating GPR65 inhibition with existing immunotherapies may define the next era of RCC management, warranting continued translational and clinical investigation. Full article
Show Figures

Figure 1

21 pages, 4041 KB  
Article
Network Pharmacology Analysis and Experimental Study of Yinchen Against Neuroinflammation in Ischemic Stroke
by Minmin Guo, Yijie Ma, Linlin Wang, Ruipeng Ge, You Wang, Gefei Ma, Guanhua Du and Li Li
Pharmaceuticals 2025, 18(12), 1852; https://doi.org/10.3390/ph18121852 - 4 Dec 2025
Viewed by 232
Abstract
Objective: Ischemic stroke (IS) is an acute neurologic injury in which inflammatory responses play a key role. Yinchen, a common medicinal plant used in Traditional Chinese Medicine (TCM), has been proven to possess strong anti-inflammatory effects. However, its efficacy in treating IS remains [...] Read more.
Objective: Ischemic stroke (IS) is an acute neurologic injury in which inflammatory responses play a key role. Yinchen, a common medicinal plant used in Traditional Chinese Medicine (TCM), has been proven to possess strong anti-inflammatory effects. However, its efficacy in treating IS remains unclear. In this study, we aimed to investigate the therapeutic potential of Yinchen for IS and the material basis of this potential. Methods: The main active components in Artemisia scoparia extract (ASE, the extract of Yinchen), were identified by HPLC and MS. The targets of Yinchen and IS were obtained from public databases. Network pharmacology, molecular docking, and experimental investigation were further applied to acquire the core constituents in Yinchen that work against the neuroinflammation that occuring during IS. The neurological outcomes were evaluated in a transient Middle Cerebral Artery Occlusion (tMCAO) rat model. Additionally, the changes in the inflammatory responses in both the ischemic brain and in lipopolysaccharide (LPS)-treated microglial cells were examined using real-time qPCR. Results: Four active compounds of ASE, including isochlorogenic acid C (ICGA-C), isochlorogenic acid B (ICGA-B), isochlorogenic acid A (ICGA-A), and chlorogenic acid (CGA), were identified by HPLC and MS. Network pharmacology predicted that 103 compounds of Yinchen had 198 intersection targets with IS. The top five of these targets were TNF, STAT3, IL1B, AKT1, and SRC. Molecular docking results demonstrated that the abovementioned four compounds detected in ASE showed good interaction with all of the above five core targets. Moreover, both the four compounds and ASE were observed to attenuate NO release and suppress the release of various inflammatory factors (TNF-α, IL-1β, IL-6, and MCP-1) in a dose-dependent manner in LPS-induced BV2 microglial cells. ASE was further found to exert neuroprotective effects against ischemia–reperfusion (I/R) injury and inhibit the production of inflammatory factors in tMCAO rats. Conclusions: Yinchen exerts an anti-neuroinflammatory effect on IS, and its constituents with high scores binding to five core targets contribute to this effect. This supports its potential as an anti-inflammatory agent for the treatment of IS. Full article
(This article belongs to the Special Issue Network Pharmacology of Natural Products, 2nd Edition)
Show Figures

Graphical abstract

12 pages, 922 KB  
Review
The Role of Short-Chain Fatty Acids (SCFAs) in Colic and Anti-Inflammatory Pathways in Horses
by Nathan Schank, Ashley Cottone, Michelle Wulf, Keely Seiter, Brinley Thomas, Lynda M. J. Miller, Stacy L. Anderson, Amal Sahyoun, Ammaar H. Abidi, Modar Kassan and Ashutosh Verma
Animals 2025, 15(23), 3482; https://doi.org/10.3390/ani15233482 - 3 Dec 2025
Viewed by 267
Abstract
Equine colic remains a prevalent and potentially life-threatening condition with multifactorial origins, including dietary imbalances, stress, and microbial dysbiosis. Central to equine gut health is the production of short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate, generated through microbial fermentation of dietary [...] Read more.
Equine colic remains a prevalent and potentially life-threatening condition with multifactorial origins, including dietary imbalances, stress, and microbial dysbiosis. Central to equine gut health is the production of short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate, generated through microbial fermentation of dietary fibers in the hindgut. These metabolites not only serve as vital energy sources but also play crucial roles in maintaining intestinal barrier integrity, modulating motility, and suppressing inflammation. This review explores the role of SCFAs in equine gastrointestinal health, with particular emphasis on their anti-inflammatory effects and potential to prevent or mitigate colic. We examine how SCFAs interact with immune pathways, via G-protein-coupled receptors and regulatory T-cell promotion, to reduce pro-inflammatory cytokines such as TNF-α and IL-6. Evidence suggests that dietary shifts toward high-starch or low-fiber intake can reduce SCFA production, contributing to microbial imbalance, increased gut permeability, and systemic inflammation, all hallmarks of colic pathophysiology. Strategies to enhance SCFA levels, including high-forage diets, targeted prebiotic and probiotic supplementation, and emerging approaches like fecal microbiota transplantation, are discussed. Despite promising findings, significant gaps remain in equine-specific research, highlighting the need for longitudinal and mechanistic studies. Understanding and harnessing the therapeutic potential of SCFAs could pave the way for novel, microbiome-based interventions in colic prevention and treatment. Full article
(This article belongs to the Section Equids)
Show Figures

Figure 1

13 pages, 2191 KB  
Article
Microvesicles from Turmeric Extracts Contain Curcuminoids and Modulate Macrophage Polarization and Migration
by Stefano Tacconi, Audrey Jalabert, Emmanuelle Berger, César Cotte, Elizabeth Errazuriz-Cerda, Valérie Bardot, Anne Leblanc, Lucile Berthomier, Michel Dubourdeaux and Sophie Rome
Pharmaceutics 2025, 17(12), 1555; https://doi.org/10.3390/pharmaceutics17121555 - 3 Dec 2025
Viewed by 323
Abstract
Background/Objectives: Recent studies have revealed that plants produce lipid-derived microvesicles with potent anti-inflammatory properties. In turmeric (Curcuma longa L.), such microvesicles have been identified in rhizome juice and shown to exert beneficial effects in murine models of colitis. In this study, we [...] Read more.
Background/Objectives: Recent studies have revealed that plants produce lipid-derived microvesicles with potent anti-inflammatory properties. In turmeric (Curcuma longa L.), such microvesicles have been identified in rhizome juice and shown to exert beneficial effects in murine models of colitis. In this study, we investigated whether turmeric extracts commonly used in phytotherapy (30% ethanolic or aqueous extracts, and freeze-dried or spray-dried preparations) contain Curcuma-derived microvesicles (CuMVs), and we evaluated the influence of extraction processes on their aggregation and morphology. Methods: All extracts were processed using a standardized protocol involving differential centrifugation, filtration, and ultracentrifugation. CuMVs with sizes from 50 to 200 nm were detected in all pellets, but CuMVs from dehydrated extracts were markedly aggregated compared to those from liquid preparations. Results: The 30% ethanolic extract yielded the most polydisperse CuMVs and was therefore selected for functional immunomodulatory analyses on macrophages. Protein quantification indicated that 600 mL of 30% ethanolic extract contained approximately 60 µg of CuMVs which contained curcumin and its derivatives demethoxycurcumin (DMC), and bisdemethoxycurcumin (BDMC) identified by high-performance thin-layer chromatography (HPTLC). Green fluorescence in the form of small dots close to the nuclei was detected in recipient THP-1 macrophages, indicating the incorporation of CuMVs and therefore the transfer of the naturally fluorescent curcumin. CuMV treatment reduced ROS production, downregulated CD86, and upregulated CD163 expression. Furthermore, CuMVs increased the expression of IL-10 and TGF-β, as well as antibacterial cytokines (IL-1β, IL-6, and TNF-α), and enhanced RAW macrophage migration. Depletion of CuMVs from turmeric extracts markedly reduced their immunomodulatory effects. Conclusions: Collectively, these findings emphasize the importance of preserving CuMVs during the industrial processing of turmeric, as they play a crucial role in curcuminoid delivery and in mediating the immunomodulatory properties of turmeric extracts. Full article
Show Figures

Graphical abstract

Back to TopTop