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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,281)

Search Parameters:
Keywords = NLRP3 inflammasomes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 2340 KB  
Article
Biological Activities Underlying the Cardiovascular Benefits of Olive Oil Polyphenols: Focus on Antioxidant, Anti-Inflammatory, and Anti-Atherogenic Effects
by Kaoutar Boumezough, Mehdi Alami, Tamas Fulop, Nada Zoubdane, Ikram Salih, Mhamed Ramchoun, Abdelouahed Khalil and Hicham Berrougui
Int. J. Mol. Sci. 2025, 26(22), 11165; https://doi.org/10.3390/ijms262211165 - 19 Nov 2025
Viewed by 113
Abstract
Extra virgin olive oil (EVOO) polyphenols are recognized for their beneficial effects on human health, yet how their concentration shapes biological outcomes remains insufficiently explored. While a daily intake of 25 mL EVOO is generally regarded as beneficial for cardiovascular protection, the high-phenolic [...] Read more.
Extra virgin olive oil (EVOO) polyphenols are recognized for their beneficial effects on human health, yet how their concentration shapes biological outcomes remains insufficiently explored. While a daily intake of 25 mL EVOO is generally regarded as beneficial for cardiovascular protection, the high-phenolic EVOO examined in this study contains markedly higher levels of polyphenols than most EVOOs reported previously. This suggests that oils richer in polyphenols may exert distinct biological effects. To investigate this, we compared extracts from a standard EVOO and a naturally high-phenolic EVOO, along with their key phenolic compounds, hydroxytyrosol (HT) and tyrosol (Tyr). Antioxidant effects were assessed by quantifying intracellular reactive oxygen species (ROS) and lipid peroxidation. Anti-inflammatory activity was evaluated in THP-1-derived macrophages stimulated with LPS by analyzing inflammatory surface markers’ expression, cytokines’ production, and the NLRP3-inflammasome pathway. Atheroprotective potential was investigated by measuring cholesterol efflux in J774 macrophages. Both EVOO polyphenols extracts and (HT and Tyr) significantly reduced ROS and lipid peroxidation. High phenolic EVOO extract (EVOOPE+) displayed superior antioxidant activity at lower concentrations, while standard EVOO phenolic extract (EVOOPE) showed more consistent effects across doses. Both extracts favored an anti-inflammatory macrophage phenotype, as indicated by increased CD163 and IL-10 expression and reduced CD86, IFN-α, and NLRP3. Moreover, all treatments enhanced cholesterol efflux in a dose-dependent manner, with EVOOPE+ and HT producing the strongest effects. Collectively, these results highlight the capacity of EVOO polyphenols to modulate, through key bioactivity mechanisms, cardioprotective effects and emphasize the importance of polyphenols concentration in their biological efficacy. Full article
(This article belongs to the Special Issue Anti-Inflammatory and Anti-Oxidant Effects of Extracts from Plants)
Show Figures

Figure 1

19 pages, 2439 KB  
Review
The Inflammatory Cell Death in Diabetic Kidney Disease: Integrating Multifactorial Mechanisms into Novel Therapeutics
by Bin Fang, Wei Huang, Sijia Du, Yu Hao, Fangfang He and Chun Zhang
Int. J. Mol. Sci. 2025, 26(22), 11033; https://doi.org/10.3390/ijms262211033 - 14 Nov 2025
Viewed by 398
Abstract
In addition to apoptosis, inflammatory cell death modalities—including pyroptosis, necroptosis, ferroptosis, NETosis, and the integrated paradigm of PANoptosis—are now established as critical drivers of diabetic kidney disease (DKD) pathogenesis. This review summarizes how key inflammatory cell death molecular mediators—such as the NLRP3 inflammasome, [...] Read more.
In addition to apoptosis, inflammatory cell death modalities—including pyroptosis, necroptosis, ferroptosis, NETosis, and the integrated paradigm of PANoptosis—are now established as critical drivers of diabetic kidney disease (DKD) pathogenesis. This review summarizes how key inflammatory cell death molecular mediators—such as the NLRP3 inflammasome, the RIPK1/RIPK3/MLKL axis, executioner caspases, and gasdermin-D (GSDMD)—orchestrate the death of renal cells (podocytes, tubular cells, mesangial cells, endothelium), thereby propagating inflammation and fibrosis. Preclinical studies have demonstrated the efficacy of agents targeting these pathways, highlighting their therapeutic potential. Key challenges include achieving cell type-specific targeting, overcoming redundancy among cell death pathways, and improving the translational applicability of current models. Emerging solutions include the development of precise biomarkers, kidney-targeted delivery systems, and combination therapies that concurrently target multiple cell death axes. This review synthesizes evidence establishing inflammatory cell death as a cornerstone of DKD pathology and provides a conceptual framework to guide future research and therapeutic innovation. Full article
Show Figures

Figure 1

46 pages, 2243 KB  
Review
Inflammasomes as Potential Therapeutic Targets to Prevent Chronic Active Viral Myocarditis—Translating Basic Science into Clinical Practice
by Natalia Przytuła, Jakub Podolec, Tadeusz Przewłocki, Piotr Podolec and Anna Kabłak-Ziembicka
Int. J. Mol. Sci. 2025, 26(22), 11003; https://doi.org/10.3390/ijms262211003 - 13 Nov 2025
Viewed by 205
Abstract
Despite substantial progress in medical care, acute myocarditis remains a life-threatening disorder with a sudden onset, often unexpectedly complicating a simple and common upper respiratory tract infection. In most cases, myocarditis is triggered by viral infections (over 80%), with an estimated incidence of [...] Read more.
Despite substantial progress in medical care, acute myocarditis remains a life-threatening disorder with a sudden onset, often unexpectedly complicating a simple and common upper respiratory tract infection. In most cases, myocarditis is triggered by viral infections (over 80%), with an estimated incidence of 10–106 per 100,000 annually. The clinical course may worsen in cases of mixed etiology, where a primary viral infection is complicated by secondary bacterial pathogens, leading to prolonged inflammation and an increased risk of progression to chronic active myocarditis or dilated cardiomyopathy. We present a case report illustrating the clinical problem of acute myocarditis progression into a chronic active form. A central element of host defense is the inflammasome—an intracellular complex that activates pyroptosis and cytokine release (IL-1β, IL-18). While these processes help combat pathogens, their persistent activation may sustain inflammation and trigger heart failure and cardiac fibrosis, eventually leading to dilated cardiomyopathy. In this review, we summarize the current understanding of inflammasome pathways and their dual clinical role in myocarditis: they are essential for controlling acute infection but may become harmful when overactivated, contributing to chronic myocardial injury. Additionally, we discuss both novel and established therapeutic strategies targeting inflammatory and anti-fibrotic mechanisms, including IL-1 receptor blockers (anakinra, canakinumab), NOD-like receptor protein 3 (NLRP3) inhibitors (colchicine, MCC950, dapansutrile, INF200), NF-κB inhibitors, and angiotensin receptor-neprilysin inhibitors (ARNI), as well as microRNAs. Our aim is to emphasize the clinical importance of early identification of patients at risk of transitioning from acute to chronic inflammation, elucidate the role of inflammasomes, and present emerging therapies that may improve outcomes by balancing effective pathogen clearance with limitation of chronic cardiac damage. Full article
(This article belongs to the Special Issue Molecular Research in Myocarditis)
Show Figures

Figure 1

29 pages, 1818 KB  
Review
Fungal Polysaccharides as Modulators of Molecular Pathways in Liver Health
by Rafał Szelenberger and Magdalena Więckowska
Molecules 2025, 30(22), 4384; https://doi.org/10.3390/molecules30224384 - 13 Nov 2025
Viewed by 336
Abstract
Fungal polysaccharides represent a structurally diverse group of bioactive compounds with increasing recognition for their hepatoprotective potential. This review synthesizes current evidence on their roles in the prevention and treatment of liver diseases, including alcohol-related liver disease (ALD), metabolic dysfunction-associated fatty liver disease [...] Read more.
Fungal polysaccharides represent a structurally diverse group of bioactive compounds with increasing recognition for their hepatoprotective potential. This review synthesizes current evidence on their roles in the prevention and treatment of liver diseases, including alcohol-related liver disease (ALD), metabolic dysfunction-associated fatty liver disease (MAFLD), or toxin-induced injury. The analyzed studies demonstrate that polysaccharides isolated from species such as Lentinula edodes, Grifola frondosa, Ganoderma lucidum, Coriolus versicolor, and Cordyceps militaris exert beneficial effects by reducing oxidative stress, attenuating inflammation, and improving metabolic homeostasis. Mechanistically, these effects are mediated through the regulation of multiple signaling pathways, including Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB), Nuclear factor erythroid 2–related factor 2 (Nrf2), and NOD-like receptor protein 3 (NLRP3) inflammasome, as well as modulation of gut microbiota. Fungal polysaccharides were also shown to improve hepatic function by lowering serum biomarkers of liver injury and ameliorating histopathological damage. Presented evidence indicates that fungal polysaccharides possess considerable potential as multifunctional hepatoprotective agents, highlighting the need for further mechanistic insight and clinical validation. Full article
Show Figures

Figure 1

15 pages, 1340 KB  
Review
Neuroinflammation as a Novel Therapeutic Frontier for Sanfilippo Syndrome
by Donato Rigante and Chiara Veredice
Children 2025, 12(11), 1530; https://doi.org/10.3390/children12111530 - 12 Nov 2025
Viewed by 357
Abstract
Glycosaminoglycans (GAGs), also named ‘mucopolysaccharides’, are nodal constituents of the connective tissue matrix which go through synthesis, demolition, and reconstruction within several cellular structures: an abnormal GAG catabolism is the basis of progressive intra-lysosomal accumulation of non-metabolized GAGs, defining all mucopolysaccharidoses (MPS), protean [...] Read more.
Glycosaminoglycans (GAGs), also named ‘mucopolysaccharides’, are nodal constituents of the connective tissue matrix which go through synthesis, demolition, and reconstruction within several cellular structures: an abnormal GAG catabolism is the basis of progressive intra-lysosomal accumulation of non-metabolized GAGs, defining all mucopolysaccharidoses (MPS), protean disorders characterized by physical abnormalities and multi-organ failure depending on the specific site of non-renewable GAGs stored. A severe cognitive decline is typically observed in the Sanfilippo syndrome, which corresponds to MPS type III, a group of four inherited neurodegenerative diseases resulting from the lack of specific enzymes involved in heparan sulfate (HS) metabolism. As a consequence, the storage of partially degraded HS fragments within lysosomes of the central nervous system elicits chain inflammatory reactions involving the NLRP3-inflammasome in microglia and astrocytes, which cease their homeostatic and immune functions and finally compromise neuron survival. This article provides an overview of the neuroinflammatory picture observed in children with MPS type III, postulating a role of HS accumulation to prime innate immunity responses which culminate with pro-inflammatory cytokine release in the brain and highlighting the relevance of interleukin-1 as a main contributor to neuroinflammation. Full article
Show Figures

Figure 1

18 pages, 3169 KB  
Article
Dehydroandrographolide Alleviates Oxidative Stress, Inflammatory Response, and Pyroptosis in DSS-Induced Colitis Mice by Modulating Nrf2 Signaling Pathway
by Meifen Wang, Zhenyu Li, Xinghua Lei, Ziyue Yang, Shuixing Yu and Guangxin Chen
Biomolecules 2025, 15(11), 1580; https://doi.org/10.3390/biom15111580 - 10 Nov 2025
Viewed by 306
Abstract
Dehydroandrographolide (DA), a bioactive diterpenoid from Andrographis paniculata with diverse biological activity, was investigated for its antioxidant and anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages and dextran sulfate sodium (DSS)-induced murine colitis. In vitro, DA inhibited the inflammatory response by modulating extracellular Signal-Regulated [...] Read more.
Dehydroandrographolide (DA), a bioactive diterpenoid from Andrographis paniculata with diverse biological activity, was investigated for its antioxidant and anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages and dextran sulfate sodium (DSS)-induced murine colitis. In vitro, DA inhibited the inflammatory response by modulating extracellular Signal-Regulated Kinase (Erk), c-Jun N-terminal Kinase (Jnk), p38 Mitogen-Activated Protein Kinase (P38), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 activation, and downregulated interleukin-6 (il-6) and interleukin-1β (il-1β) mRNA. It also had antioxidant effects by upregulating Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2), NAD(P)H quinone dehydrogenase 1 (Nqo-1) and heme oxygenase-1 (Ho-1), promoting protein kinase B (Akt) and 5′-adenosine monophosphate-activated protein kinase-α1 (Ampk-α1) phosphorylation. DA decreased cyclooxygenase-2 (Cox-2) and inducible nitric oxide synthase (iNos) levels and alleviated intracellular reactive oxygen species (ROS) accumulation. In vivo, DA alleviated DSS-induced colitis in wild type (WT) mice by improving weight loss, disease activity index, colonic inflammation, and oxidative stress. The beneficial effects were linked to inhibiting Erk, Jnk, and P38 activation and enhancing Nrf2 signaling pathway. DA inhibited NOD-like receptor family pyrin domain-containing 3 (Nlrp3) inflammasome-mediated pryoptosis. However, DA’s protective effects were abolished in DSS-induced nrf2−/− mice, suggesting its efficacy depends on Nrf2 signaling. Overall, DA alleviates oxidative stress, inflammatory responses, and pyroptosis in experimental colitis mice mainly by activating Nrf2 signaling pathway, highlighting its potential as a promising therapeutic option for inflammatory bowel disease. Full article
(This article belongs to the Special Issue The Value of Natural Compounds as Therapeutic Agents: 3rd Edition)
Show Figures

Figure 1

16 pages, 2739 KB  
Article
Protective Effects of Cyanidin-3-O-Glucoside Against Neurotoxin Acrylamide Through Alleviating Mitochondrial Dysfunction
by Liuqing Yang, Lujia Zhang, Li Dong, Yanli Ma, Lei Zhao, Ruoyang Xu, Fang Chen and Yinghua Luo
Foods 2025, 14(22), 3826; https://doi.org/10.3390/foods14223826 - 8 Nov 2025
Viewed by 305
Abstract
Acrylamide (AA), a well-known neurotoxin, shows obvious damage to the nervous system. Cyanidin-3-O-glucoside (C3G), a representative anthocyanin, is identified as a promising neuroprotective agent as its excellent antioxidant capacity. This study evaluated the mitoprotective effects of C3G against AA-mediated neurotoxicity. The results showed [...] Read more.
Acrylamide (AA), a well-known neurotoxin, shows obvious damage to the nervous system. Cyanidin-3-O-glucoside (C3G), a representative anthocyanin, is identified as a promising neuroprotective agent as its excellent antioxidant capacity. This study evaluated the mitoprotective effects of C3G against AA-mediated neurotoxicity. The results showed that pretreatment with C3G (10 μmol/L) significantly lessened the reduction in AA-induced cell survival rate, increasing cell viability by 1.31 times compared to the AA-only group. C3G reduced intracellular ROS and MDA level accumulation by 84.0% and 61.9%, respectively. Furthermore, C3G suppressed the activation of NLRP3 inflammasome and Caspase-3-dependent apoptosis pathways induced by AA. Further mitochondrial analysis revealed that C3G pretreatment enhanced mitochondrial membrane potential recovery by 1.50 times and preserved the mitochondrial ultrastructure, while also restoring the aerobic respiratory capacity. PCR array demonstrated that C3G reversed the AA-induced downregulation of mitochondrial biogenesis genes PGC-1α and TFAM by 2.67-fold and 1.88-fold, respectively, and mitochondrial dynamics genes Mfn2 and Opa1 by 2.76-fold and 3.08-fold. Further in vivo studies confirmed that the blueberry anthocyanin extracts, which are mainly composed of C3G, showed neuroprotective function through maintaining mitochondrial function, alleviating inflammation, and apoptosis. This article provides new insights into the neuroprotective effects of C3G. Full article
(This article belongs to the Section Food Toxicology)
Show Figures

Graphical abstract

19 pages, 4109 KB  
Article
Modulation of AMPK/NLRP3 Signaling Mitigates Radiation-Induced Lung Inflammation by a Synthetic Lipoxin A4 Analogue
by Sun Ho Min, Jae-Ho Shin, Sunjoo Park, Ronglan Cui, Youn Ji Hur, Woo Hyun Jeong, Sang Yeon Kim, Younghwa Na and Jaeho Cho
Int. J. Mol. Sci. 2025, 26(22), 10832; https://doi.org/10.3390/ijms262210832 - 7 Nov 2025
Viewed by 289
Abstract
Radiation-induced lung inflammation (RILI) is a major complication of thoracic radiotherapy, characterized by excessive inflammation and subsequent fibrosis that compromise pulmonary function and treatment outcomes. This study explores the pharmacological properties of a newly synthesized Lipoxin A4 analogue (CYNC-2) to mitigate RILI by [...] Read more.
Radiation-induced lung inflammation (RILI) is a major complication of thoracic radiotherapy, characterized by excessive inflammation and subsequent fibrosis that compromise pulmonary function and treatment outcomes. This study explores the pharmacological properties of a newly synthesized Lipoxin A4 analogue (CYNC-2) to mitigate RILI by modulating the AMP-activated protein kinase (AMPK)/NOD-like receptor family pyrin domain containing 3(NLRP3) inflammasome pathway. A murine RILI model was established in mice by delivering a single high-dose (ablative) X-ray irradiation to the left lung. Mice in the treatment group received CYNC-2 via tail-vein injection three times per week for 2 weeks. The effects of CYNC-2 on RILI were evaluated histological, immunohistochemical analysis of lung tissues, cytokine profiling, lung function testing using a FlexiVent system, and micro-computed tomography (micro-CT) imaging of lung damage. In parallel, two human lung cell lines—L132 (normal bronchial epithelial cells) and A549 (lung carcinoma cells)—were irradiated with 6 Gy X-rays and treated with CYNC-2 to assess cell viability and changes in AMPK/NLRP3 pathway markers via qPCR and immunofluorescence. Lung tissue sample from patients who underwent thoracic radiotherapy were also examined to validate key findings. CYNC-2 activated AMPK and inhibited mTOR signaling, which suppressed NLRP3 inflammasome activation and led to reduced secretion of pro-inflammatory cytokines (IL-1β, IL-6, and TGF-β1). In vitro, CYNC-2 mitigated radiation-induced inflammatory responses and preserved cellular viability. Overall, CYNC-2 effectively dampened acute pulmonary in the RILI model. These findings suggest that targeting the AMPK/NLRP3 inflammasome pathway via a stable LXA4 analogue such as CYNC-2 is a promising therapeutic strategy to improve clinical outcomes for patients receiving thoracic radiation therapy. Full article
Show Figures

Figure 1

22 pages, 757 KB  
Review
Beyond Lipids and Platelets: A Review of Anti-Inflammatory Strategies in Secondary Prevention of Acute Coronary Syndromes
by Girish Pathangey, Mohamed N. Allam, Mahmoud H. Abdelnabi, Dan Sorajja, Floyd David Fortuin, Kwan S. Lee and Mayurkumar D. Bhakta
J. Clin. Med. 2025, 14(22), 7910; https://doi.org/10.3390/jcm14227910 - 7 Nov 2025
Viewed by 350
Abstract
Despite advances in lipid-lowering and antithrombotic therapy, patients with acute coronary syndromes remain at elevated risk for recurrent events due to persistent atherosclerotic inflammation. This review evaluates inflammation as a therapeutic target in secondary prevention and discusses established, investigational, and emerging strategies. Colchicine, [...] Read more.
Despite advances in lipid-lowering and antithrombotic therapy, patients with acute coronary syndromes remain at elevated risk for recurrent events due to persistent atherosclerotic inflammation. This review evaluates inflammation as a therapeutic target in secondary prevention and discusses established, investigational, and emerging strategies. Colchicine, now FDA-approved for cardiovascular risk reduction, lowered major adverse cardiovascular events in COLCOT and LoDoCo2. Canakinumab (IL-1β inhibition) reduced recurrent events in proportion to IL-6 and hsCRP suppression, while ziltivekimab (IL-6 inhibition) achieved profound biomarker reductions but remains investigational. Early-phase studies of anakinra (IL-1 receptor antagonist) and dapansutrile (oral NLRP3 inhibitor) showed anti-inflammatory effects in early trials, whereas varespladib and darapladib illustrated the challenges of targeting lipid-associated pathways. Beyond direct immunomodulators, GLP-1 receptor agonists and SGLT2 inhibitors provide both cardioprotective and anti-inflammatory benefits, reinforcing their expanding role post-ACS. Additional emerging avenues include triptolidiol, dasatinib, and BTK or JAK/STAT inhibitors, while novel approaches, such as nanozyme delivery systems and CRISPR-based editing, extend the therapeutic horizon. This review highlights the potential of inflammation-targeted therapies to advance secondary prevention in ACS by integrating current evidence and perspectives on future therapeutic developments. Full article
Show Figures

Graphical abstract

27 pages, 2009 KB  
Review
Intracellular Calcium Dysregulation: The Hidden Culprit in the Diabetes–Gout Nexus
by Hongbin Shi, Yisi Shan, Kewei Qian, Ruofei Zhao and Hong Li
Biomedicines 2025, 13(11), 2694; https://doi.org/10.3390/biomedicines13112694 - 2 Nov 2025
Viewed by 509
Abstract
Type 2 diabetes and gout are both common metabolic disorders that frequently occur together. Research indicates that disturbances in intracellular calcium balance may be a key molecular factor linking the development of these two diseases. Calcium signaling disturbances promote the synergistic progression of [...] Read more.
Type 2 diabetes and gout are both common metabolic disorders that frequently occur together. Research indicates that disturbances in intracellular calcium balance may be a key molecular factor linking the development of these two diseases. Calcium signaling disturbances promote the synergistic progression of both diseases through multiple pathways: In pancreatic β-cells, endoplasmic reticulum (ER) calcium imbalance triggers ER stress, mitochondrial dysfunction, and apoptosis, autophagy, and pyroptosis, leading to impaired insulin secretion. Concurrently, calcium overload exacerbates insulin resistance by disrupting insulin signal transduction in peripheral tissues, while hyperinsulinemia further inhibits uric acid excretion through activation of the renal URAT1 transporter, creating a vicious cycle. Additionally, calcium homeostasis dysregulation activates the NLRP3 inflammasome and promotes the release of pro-inflammatory cytokines, aggravating chronic low-grade inflammation, which further deteriorates β-cell function and peripheral metabolic disorders, collectively driving the pathological link between type 2 diabetes and gout. Although calcium channel modulators show potential in improving β-cell function and reducing inflammation, their clinical application faces challenges such as tissue-specific effects and a lack of high-quality clinical trials. We propose that intracellular calcium dysregulation serves as a central pathological amplifier in the diabetes–gout nexus. Future research on targeted calcium signaling interventions, guided by this integrative concept, may help overcome the therapeutic challenges in managing type 2 diabetes complicated by gout. Full article
(This article belongs to the Section Endocrinology and Metabolism Research)
Show Figures

Figure 1

19 pages, 4347 KB  
Article
Lactiplantibacillus plantarum WLPL04 from Human Breast Milk Attenuates Hyperuricemia via Coordinated Purine Salvage Pathway, Renal Transporter Regulation, and Gut Microbiota Remodeling
by Min Wei, Yingsheng Hu, Zhihong Zhang, Liang Qiu, Xueying Tao and Hua Wei
Nutrients 2025, 17(21), 3447; https://doi.org/10.3390/nu17213447 - 31 Oct 2025
Viewed by 354
Abstract
Background: Hyperuricemia (HUA), a metabolic disorder characterized by high serum uric acid (UA) level, presents a growing global health challenge. Method: In this study, a stable murine model of HUA was established by orally administering adenine (100 mg/kg) and potassium oxonate (600 mg/kg) [...] Read more.
Background: Hyperuricemia (HUA), a metabolic disorder characterized by high serum uric acid (UA) level, presents a growing global health challenge. Method: In this study, a stable murine model of HUA was established by orally administering adenine (100 mg/kg) and potassium oxonate (600 mg/kg) in C57BL/6J mice, resulting in significant elevation of serum UA and xanthine oxidase (XOD) activity, as well as renal pathological alterations. Given the anti-hyperuricemia potential of Lactiplantibacillus plantarum WLPL04, a strain from a human breast milk was evaluated. Conclusions: Oral administration of L. plantarum WLPL04 significantly reduced serum UA level and XOD activity in a dose-dependent manner. Moreover, L. plantarum WLPL04 treatment enhanced UA excretion by upregulating ABCG2 and downregulating URAT1 and GLUT9 expression. It ameliorated renal injury and suppressed inflammation via downregulation of the NLRP3 inflammasome pathway. 16S rRNA gene sequencing revealed that L. plantarum WLPL04 restored gut microbial diversity and promoted the enrichment of beneficial genera such as Bacteroides, which was negatively correlated with UA in serum, creatinine, and inflammatory cytokines. Moreover, transcript analysis revealed upregulation of purine salvage genes (hpt and xpt), suggesting enhanced salvage pathway recycling of purine bases and reduced urate production. Those findings suggest that L. plantarum WLPL04 exerted multi-targeted anti-hyperuricemia effects through coordinated regulation of host purine metabolism, urate transport, inflammation, and gut microbiota composition, providing a promising probiotic candidate for HUA management. Full article
Show Figures

Figure 1

45 pages, 6000 KB  
Review
Protein–Ligand Interactions in Cardiometabolic Drug Targets: Focus on Weight Loss and Cardioprotection
by Errikos Petsas, Despoina P. Kiouri, Nikitas Georgiou, Gerasimos Siasos, Thomas Mavromoustakos and Christos T. Chasapis
Molecules 2025, 30(21), 4240; https://doi.org/10.3390/molecules30214240 - 30 Oct 2025
Viewed by 1033
Abstract
Cardiometabolic diseases (CVDs) are the leading cause of premature mortality and disability worldwide, arising from of cardiovascular and metabolic dysregulation. This review focuses on six critical therapeutic targets established in cardiometabolic regulation: GLP-1R, GIPR, FGFR1/β-Klotho, PCSK9, NF-κB, and the NLRP3 inflammasome. Drawing on [...] Read more.
Cardiometabolic diseases (CVDs) are the leading cause of premature mortality and disability worldwide, arising from of cardiovascular and metabolic dysregulation. This review focuses on six critical therapeutic targets established in cardiometabolic regulation: GLP-1R, GIPR, FGFR1/β-Klotho, PCSK9, NF-κB, and the NLRP3 inflammasome. Drawing on curated structural datasets, we analyze the mechanisms of action and map key binding domain features that govern ligand efficacy and specificity. Dual GLP-1R/GIPR agonists, such as tirzepatide, demonstrate superior outcomes in glycemic control and weight reduction. Concurrently, inhibiting PCSK9, NF-κB, and NLRP3 helps to lower cholesterol and reduce harmful inflammation, offering cardioprotection. Structural analysis across these targets reveals complementary motifs (aromatic, hydrophobic, and polar residues). These insights guide the rational design of next-generation multi-target ligands (molecules capable of modulating two or more biological targets involved in related disease pathways, producing integrated therapeutic effects). Such integrated agents are promising for providing combined cardiovascular and metabolic benefits, thus reducing the risks associated with complex therapeutic drug combinations. Full article
(This article belongs to the Section Chemical Biology)
Show Figures

Graphical abstract

22 pages, 2404 KB  
Article
Oxidative Stress and NLRP3 Inflammasome as Markers of Cardiac Injury Following Cardiopulmonary Bypass: Potential Implications for Patients with Preoperative Heart Failure with Reduced Ejection Fraction
by Rodrigo L. Castillo, Rodrigo A. Carrasco, Alejandro Gonzaléz-Candia, Esteban G. Figueroa, Adolfo A. Paz, Alejandro A. Candia, Sawa Kostin, Nikolaos Pagonas, Pamela V. Arias, Emilio A. Herrera, Robert A. Pérez and Sebastián Iturra
Antioxidants 2025, 14(11), 1311; https://doi.org/10.3390/antiox14111311 - 30 Oct 2025
Viewed by 607
Abstract
Cardiopulmonary bypass (CPB) can lead to cardiac damage due to oxidative stress (OS) and inflammation in heart failure (HF). We tested the hypothesis that preoperative HF patients with reduced ejection fraction (HFrEF) subjected to CBP have higher levels of OS and NLRP3 (NOD-, [...] Read more.
Cardiopulmonary bypass (CPB) can lead to cardiac damage due to oxidative stress (OS) and inflammation in heart failure (HF). We tested the hypothesis that preoperative HF patients with reduced ejection fraction (HFrEF) subjected to CBP have higher levels of OS and NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) in heart and plasma and in those that develop postoperative AF (pAF) as a clinical outcome. HF was categorized for preoperative left ventricular EF: preserved (HFpEF > 50%, n = 27) and reduced EF (HFrEF ≤ 40%, n = 25). Samples of atrial tissue, pericardial fluid, and plasma were collected at surgery to assess NLRP3 expression; 3-nitrotyrosine (3-NT), thiobarbituric acid reaction (TBARS), and nuclear factor erythroid 2-related factor 2 (Nrf2) in atrial tissue; NLRP3, IL-1β, and IL-18 expression in pericardial fluid; and antioxidant capacity, 8-isoprostanes, and malondialdehyde (MDA) in plasma. Reactive oxygen species, 3-NT, and NLRP3 in atrial tissue were determined by immunohistochemistry in a subset of pAF patients. Plasma and atrial tissue 3-NT and MDA were higher in HFrEF compared with HFpEF. Lipid peroxidation products were higher in both plasma and atrial tissue in pAF (n = 29), compared to sinus rhythm (SR) (n = 23). In HFrEF patients, the values of tissue ROS, 3-NT, and NLRP3 were higher than in HFpEF patients. In addition, the expression levels of NLRP3, IL-1β, and IL-18 were higher in atrial tissue and pericardial fluid in HFrEF. Patients with preoperative HFrEF showed higher OS in plasma and the expression of NLRP3, ROS, and 3-NT in atrial tissue biopsies and pericardial fluid. This finding suggests a potential pharmacologic therapy for pAF and clinical complications due to CPB. Full article
(This article belongs to the Special Issue Oxidative Stress in Cardiovascular Diseases (CVDs))
Show Figures

Graphical abstract

20 pages, 3935 KB  
Article
In Silico Identification of the NLRP3 Inhibitors from Traditional Chinese Medicine
by Shunjiang Jia, Huanling Lai, Xinyu Chen, Jiajie Lu, Wei Ding, Dongxiao Cui, Peng Zhao, Qiao Zhang, Yuwei Wang and Chunsong Cheng
Int. J. Mol. Sci. 2025, 26(21), 10569; https://doi.org/10.3390/ijms262110569 - 30 Oct 2025
Viewed by 525
Abstract
NOD-like receptor protein 3 (NLRP3) inflammasome is a key mediator of inflammation and a promising therapeutic target. However, the discovery of novel and effective inhibitors of NLRP3 remains limited. A combined docking-based virtual screening (DBVS) and shape-based screening approach was applied to eight [...] Read more.
NOD-like receptor protein 3 (NLRP3) inflammasome is a key mediator of inflammation and a promising therapeutic target. However, the discovery of novel and effective inhibitors of NLRP3 remains limited. A combined docking-based virtual screening (DBVS) and shape-based screening approach was applied to eight traditional Chinese medicine (TCM) databases to identify potential NLRP3 inhibitors. Structural similarity analysis, ADMET prediction, and molecular dynamics (MD) simulations were performed to evaluate structural novelty, pharmacokinetic properties, and binding stability. A total of 25 potential NLRP3 inhibitors were identified, each exhibiting docking scores higher than those of the reference inhibitor XE3. Structural similarity analysis revealed that the screened compounds exhibited low similarity to previously reported NLRP3 inhibitors, demonstrating their structural novelty. ADMET evaluation indicated that compounds C2, C3, and C4 exhibited favorable physicochemical and pharmacokinetic properties. Molecular dynamics (MD) simulations demonstrated that the complexes of compounds C2, C3, and C4 with NLRP3 remained stable throughout the simulations, exhibiting limited backbone fluctuations and compact conformations, as indicated by Rg values of approximately 6 Å. Solvent-accessible surface area (SASA) and polar surface area (PSA) analyses suggested that compounds C3 and C4 were tightly solvated and maintained favorable membrane permeability. Notably, binding free energy calculations revealed that all three compounds exhibited stronger binding than XE3, with compound C3 showing the most favorable energy (–48.81 ± 3.89 kcal/mol), indicating a highly stable and energetically preferred interaction with NLRP3. This study identified promising TCM-derived compounds as potential NLRP3 inhibitors, offering new directions for anti-inflammatory drug development. Full article
Show Figures

Figure 1

18 pages, 2944 KB  
Article
Epicatechin-Loaded Nanocapsules: Development, Physicochemical Characterization, and NLRP3 Inflammasome-Targeting Anti-Inflammatory Activity
by Carolina Bordin Davidson, Éricles Forrati Machado, Amanda Kolinski Machado, Diulie Valente de Souza, Lauren Pappis, Giovana Kolinski Cossettin Bonazza, Djenifer Letícia Ulrich Bick, Taíse Regina Schuster Montagner, André Gündel, Ivana Zanella da Silva, Aline Ferreira Ourique and Alencar Kolinski Machado
Biology 2025, 14(11), 1520; https://doi.org/10.3390/biology14111520 - 30 Oct 2025
Viewed by 464
Abstract
Epicatechin is a flavonoid of the catechin subclass, found in fruits and medicinal plants such as açaí and green tea, widely studied for its anti-inflammatory properties. However, flavonoids often present chemical instability, low aqueous solubility, and poor bioavailability, limiting their therapeutic potential. This [...] Read more.
Epicatechin is a flavonoid of the catechin subclass, found in fruits and medicinal plants such as açaí and green tea, widely studied for its anti-inflammatory properties. However, flavonoids often present chemical instability, low aqueous solubility, and poor bioavailability, limiting their therapeutic potential. This study aimed to incorporate epicatechin into nanocapsules to improve its applicability and evaluate whether the formulation maintains its anti-inflammatory effects via modulation of the NLRP3 inflammasome. Nanocapsules containing 0.25 mg/mL of epicatechin (NC-ECs) were prepared with Eudragit L-100 using interfacial deposition of a preformed polymer. The formulations were characterized for particle size, polydispersity index, zeta potential, and pH, as well as thermal stability over 45 days. Encapsulation efficiency and drug content were determined by high-performance liquid chromatography (HPLC), and morphology analyzed by atomic force microscopy (AFM). Cytocompatibility was assessed in VERO cells, and anti-inflammatory activity was investigated in THP-1-derived macrophages stimulated with LPS + nigericin. The NC-ECs displayed suitable physicochemical properties, high encapsulation efficiency (96%), and full drug loading. The formulation also showed good cytocompatibility and preserved anti-inflammatory activity through NLRP3 inflammasome modulation at low concentrations. These findings indicate NC-ECs as a promising nanotechnological strategy for treating inflammatory diseases involving NLRP3, highlighting its potential contribution to nanomedicine. Full article
(This article belongs to the Special Issue Biology and Function of Inflammasomes)
Show Figures

Figure 1

Back to TopTop