error_outline You can access the new MDPI.com website here. Explore and share your feedback with us.
 
 
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,310)

Search Parameters:
Keywords = NLRP6 inflammasome

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 3656 KB  
Article
Free Fatty Acids and Endotoxins Synergically Induce Pyroptosis in Bovine Hepatocytes
by Dan Li, Yuan Tian, Lei Tian, Hang Yu, Le Zhang, Song Wang, Changsheng Lei, Pin Long, Tao Peng, Lei Liu and Yingfang Zhou
Metabolites 2026, 16(1), 53; https://doi.org/10.3390/metabo16010053 - 8 Jan 2026
Abstract
Background/Objectives: Elevated circulating non-esterified fatty acids (NEFAs) are closely associated with hepatic inflammatory injury in dairy cattle, simultaneously with the entry of lipopolysaccharide (LPS) into the liver. This study aimed to investigate the synergistic effects of NEFAs and LPS on pyroptosis in [...] Read more.
Background/Objectives: Elevated circulating non-esterified fatty acids (NEFAs) are closely associated with hepatic inflammatory injury in dairy cattle, simultaneously with the entry of lipopolysaccharide (LPS) into the liver. This study aimed to investigate the synergistic effects of NEFAs and LPS on pyroptosis in bovine hepatocytes. Methods: Primary bovine hepatocytes were allocated into control, NEFA, NEFA + LPS, NEFA + LPS + Caspase-1 inhibitor, and NEFA + LPS + NLRP3 inhibitor groups. Levels and activation of pyroptosis-related markers (NLRP3, ASC, Caspase-1, GSDMD, IL-18 and IL-1β) were measured. Results: NEFAs alone upregulated these markers in a dose-dependent manner. Compared to NEFAs alone, NEFA + LPS co-treatment significantly enhanced levels of the markers, increased IL-1β secretion, and promoted NLRP3/Caspase-1 co-localization and Caspase-1activity. Notably, these effects of NEFA + LPS were attenuated by the NLRP3 or Caspase-1 inhibitors. Similar results were obtained when repeating the experiments in carcinoma HepG2 cells. Also, a random liver section from the subclinical ketotic cows displayed a higher fluorescence intensity of NLRP3 and Caspase-1 and stronger co-localization than that from a healthy cow. Conclusions: NEFAs and LPS synergistically contribute to pyroptosis in bovine hepatocytes by enhancing NLRP3 inflammasome assembly and subsequent Caspase-1 activation, providing a potential target for mitigating hepatic injury. Full article
(This article belongs to the Special Issue Metabolic Research in Dairy Cattle Health)
Show Figures

Graphical abstract

15 pages, 1696 KB  
Article
Luteolin Inhibits Bovine Viral Diarrhea Virus Replication by Disrupting Viral Internalization and Replication and Interfering with the NF-κB/STAT3-NLRP3 Inflammasome Pathway
by Dongjie Cai, Qing Liu, Zifan Shen, Bin Tian, Jiabin Gao, Yulin Lin, Lanjing Ma, Ya Wang and Xiaoping Ma
Vet. Sci. 2026, 13(1), 57; https://doi.org/10.3390/vetsci13010057 - 7 Jan 2026
Abstract
Bovine viral diarrhea virus (BVDV) causes severe mucosal inflammation in cattle, and effective treatment options remain limited. Dysregulated activation of the NLRP3 inflammasome, driven by NF-κB and STAT3 signaling, may exacerbate disease pathogenesis, highlighting this axis as a potential therapeutic target. Although traditional [...] Read more.
Bovine viral diarrhea virus (BVDV) causes severe mucosal inflammation in cattle, and effective treatment options remain limited. Dysregulated activation of the NLRP3 inflammasome, driven by NF-κB and STAT3 signaling, may exacerbate disease pathogenesis, highlighting this axis as a potential therapeutic target. Although traditional Chinese medicine has shown promise in antiviral and anti-inflammatory applications, it remains unclear whether it can inhibit BVDV replication via the NF-κB/STAT3-NLRP3 pathway. The present study aimed to clarify the inhibitory effect of luteolin on bovine viral diarrhea virus (BVDV) replication, and to elucidate its underlying mechanisms from two perspectives: interference with viral internalization and replication processes, as well as regulation of the NF-κB/STAT3-NLRP3 inflammasome pathway. Collectively, this work intended to provide experimental evidence and theoretical support for the development of luteolin as a natural anti-BVDV agent. To this end, BVDV-infected MDBK cells were treated with gradient concentrations of luteolin, followed by quantification of viral load using qRT-PCR and Western blot assays. Meanwhile, the activation status of the NF-κB/STAT3-NLRP3 signaling pathway was evaluated via immunofluorescence staining and luciferase reporter gene assays. Our results demonstrate that luteolin exhibits potent dual antiviral activity against cytopathic BVDV-1m in MDBK (Madin-Darby Bovine Kidney) cells, effectively suppressing both viral replication and inflammatory responses. At non-cytotoxic concentrations, luteolin specifically inhibited the internalization and replication stages of the viral lifecycle, accompanied by reduced NS5B polymerase activity. Importantly, luteolin disrupted the NF-κB/STAT3-NLRP3 axis by suppressing phosphorylation of p65 (Ser536) and STAT3 (Ser727), downregulating NLRP3 and pro-caspase-1 expression, and inhibiting caspase-1 cleavage (p20) as well as maturation of IL-1β and IL-18. Consequently, it attenuated the overexpression of TNF-α and IL-8. To our knowledge, this is the first report of a single compound simultaneously targeting multiple stages of the BVDV lifecycle and counteracting NLRP3-mediated immunopathology, offering a strategic basis for developing flavonoid-based therapies against Flavivirus infections. Full article
Show Figures

Figure 1

27 pages, 673 KB  
Review
Mechanistic and Therapeutic Insights into Nrf2-Mediated Redox Regulation in Periodontitis
by Satoshi Wada, Hiroyuki Nakano, Yasuhisa Sawai, Yota Yamauchi, Miho Hasumoto, Eiji Mitate and Noboru Demura
Antioxidants 2026, 15(1), 72; https://doi.org/10.3390/antiox15010072 - 6 Jan 2026
Viewed by 22
Abstract
Periodontitis is a chronic non-communicable inflammatory disease in which oxidative stress plays an important role in tissue destruction and alveolar bone loss. Excessive production of reactive oxygen species disrupts redox homeostasis, activates inflammatory signaling pathways, and promotes regulated cell death processes such as [...] Read more.
Periodontitis is a chronic non-communicable inflammatory disease in which oxidative stress plays an important role in tissue destruction and alveolar bone loss. Excessive production of reactive oxygen species disrupts redox homeostasis, activates inflammatory signaling pathways, and promotes regulated cell death processes such as pyroptosis and ferroptosis. The Nrf2/Keap1 pathway is a key regulator of antioxidant defense and cellular adaptation to redox imbalance. Impaired Nrf2 signaling has been associated with enhanced oxidative injury, NF-κB and NLRP3 inflammasome activation, osteoclast-driven bone resorption, and reduced regenerative capacity in periodontal tissues. Experimental studies suggest that Nrf2 activation can restore the redox balance and attenuate inflammation and bone destructive responses in a periodontal model. Moreover, therapeutic approaches involving phytochemicals, microbial-derived metabolites, and redox-responsive biomaterials have been reported to influence Nrf2-related signaling in experimental settings. However, the majority of the available evidence is derived from in vitro or animal studies, and the relevance of these findings to clinical periodontitis remains to be established. This review summarizes the current advances linking oxidative stress, redox signaling, cell death pathways, and bone remodeling with Nrf2 dysfunction in periodontitis and outlines the key mechanistic insights while highlighting the existing knowledge gaps. Full article
(This article belongs to the Special Issue Oxidative Stress and NRF2 in Health and Disease—2nd Edition)
Show Figures

Figure 1

31 pages, 1393 KB  
Review
Mitophagy–NLRP3 Inflammasome Crosstalk in Parkinson’s Disease: Pathogenic Mechanisms and Emerging Therapeutic Strategies
by Sahabuddin Ahmed, Tulasi Pasam and Farzana Afreen
Int. J. Mol. Sci. 2026, 27(1), 486; https://doi.org/10.3390/ijms27010486 - 3 Jan 2026
Viewed by 232
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra and pathological α-synuclein aggregation. Growing evidence identifies chronic neuroinflammation—particularly NLRP3 inflammasome activation in microglia—as a central driver for PD onset and progression. Misfolded α-synuclein, [...] Read more.
Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra and pathological α-synuclein aggregation. Growing evidence identifies chronic neuroinflammation—particularly NLRP3 inflammasome activation in microglia—as a central driver for PD onset and progression. Misfolded α-synuclein, mitochondrial dysfunction, and environmental toxins act as endogenous danger signals that prime and activate NLRP3 inflammasome, leading to caspase-1–mediated maturation of IL-1β and IL-18 and subsequent pyroptotic cell death. Impaired mitophagy, due to defects in PINK1/Parkin pathways or receptor-mediated mechanisms, permits accumulation of dysfunctional mitochondria and release DAMPs, thereby amplifying NLRP3 activity. Studies demonstrate that promoting mitophagy or directly inhibiting NLRP3 attenuates neuroinflammation and protects dopaminergic neurons in PD models. Autophagy-inducing compounds, along with NLRP3 inhibitors, demonstrate neuroprotective potential, though their clinical translation remains limited due to poor blood–brain barrier penetration, off-target effects, and insufficient clinical data. Additionally, the context-dependent nature of mitophagy underscores the need for precise therapeutic modulation. This review summarizes current understanding of inflammasome–mitophagy crosstalk in PD, highlights major pharmacological strategies under investigation, and outlines its limitations. Future progress requires development of specific modulators, targeted delivery systems, and robust biomarkers of mitochondrial dynamics and inflammasome activity for slowing PD progression. Full article
Show Figures

Figure 1

26 pages, 1051 KB  
Review
High-Altitude Hypoxia Injury: Systemic Mechanisms and Intervention Strategies on Immune and Inflammatory Responses
by Jingman Zhang, Shujie Guo, Beiebei Dou, Yang Liu, Xiaonan Wang, Yingze Jiao, Qianwen Li, Yan Li and Han Chen
Antioxidants 2026, 15(1), 36; https://doi.org/10.3390/antiox15010036 - 26 Dec 2025
Viewed by 793
Abstract
High-altitude exposure poses significant health challenges to mountaineers, military personnel, travelers, and indigenous residents. Altitude-related illnesses encompass acute conditions such as acute mountain sickness (AMS), high-altitude pulmonary edema (HAPE), and high-altitude cerebral edema (HACE), and chronic manifestations like chronic mountain sickness (CMS). Hypobaric [...] Read more.
High-altitude exposure poses significant health challenges to mountaineers, military personnel, travelers, and indigenous residents. Altitude-related illnesses encompass acute conditions such as acute mountain sickness (AMS), high-altitude pulmonary edema (HAPE), and high-altitude cerebral edema (HACE), and chronic manifestations like chronic mountain sickness (CMS). Hypobaric hypoxia induces oxidative stress and inflammatory cascades, causing alterations in multiple organ systems through co-related amplification mechanisms. Therefore, this review aims to systematically discuss the injury mechanisms and comprehensive intervention strategies involved in high-altitude diseases. In summary, these pathologies involve key damage pathways: oxidative stress activates inflammatory pathways through NF-κB and NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasomes; energy depletion impairs calcium homeostasis, leading to cellular calcium overload; mitochondrial dysfunction amplifies injury through mitochondrial permeability transition pore (mPTP) opening and apoptotic factor release. These mechanisms could be converged in organ-specific patterns—blood–brain barrier disruption in HACE, stress failure in HAPE, and right heart dysfunction in chronic exposure. Promising strategies include multi-level therapeutic approaches targeting oxygenation (supplemental oxygen, acetazolamide), specific pathway modulation (antioxidants, calcium channel blockers, HIF-1α regulators), and damage repair (glucocorticoids). Notably, functional foods show significant therapeutic potential: dietary nitrates (beetroot) enhance oxygen delivery, tea polyphenols and anthocyanins (black goji berry) provide antioxidant effects, and traditional herbal bioactives (astragaloside, ginsenosides) offer multi-targeted organ protection. Full article
(This article belongs to the Special Issue Redox Regulation of Immune and Inflammatory Responses)
Show Figures

Figure 1

21 pages, 3788 KB  
Article
Dose-Sparing Efficacy of d-Limonene with Low-Dose Allopurinol in a Dual Model of Hyperuricemia and Gouty Arthritis in Rats
by Krishnaraju Venkatesan, Pooja Muralidharan, Durgaramani Sivadasan, Manimekalai Pichaivel, Yahya I. Asiri, Khalid A. Asseri, Nizar Sirag, Hassabelrasoul Elfadil, Mahmoud Elodemi, Kousalya Prabahar, Premalatha Paulsamy and Kumarappan Chidambaram
Nutrients 2026, 18(1), 72; https://doi.org/10.3390/nu18010072 - 25 Dec 2025
Viewed by 240
Abstract
Background: d-Limonene (LIM) is a food-derived monoterpenoid phytocompound predominantly found in citrus peels, endowed with potent antioxidant and anti-inflammatory properties, and has been reported to inhibit xanthine oxidase (XO) activity in vitro. This study investigated the dose-sparing efficacy of this dietary [...] Read more.
Background: d-Limonene (LIM) is a food-derived monoterpenoid phytocompound predominantly found in citrus peels, endowed with potent antioxidant and anti-inflammatory properties, and has been reported to inhibit xanthine oxidase (XO) activity in vitro. This study investigated the dose-sparing efficacy of this dietary bioactive compound in combination with low-dose allopurinol (ALP) using a dual rat model combining potassium oxonate (PO)-induced hyperuricemia and monosodium urate (MSU)-triggered gouty arthritis, thereby capturing both metabolic and inflammatory dimensions of gout. Methods: Female Wistar rats were PO-primed and MSU-challenged, then treated with LIM (50 mg/kg), ALP (5 or 10 mg/kg), or LIM + ALP. Outcomes included paw thickness, dysfunction and inflammation indices, serum uric acid, urea, creatinine, AST/ALT, cytokines (IL-1β, TNF-α, IL-6), oxidative stress markers (MDA, SOD, catalase, GSH), and NLRP3 immunoreactivity, supported by radiographic and histopathological analyses. Data were analyzed by one-way ANOVA with Tukey’s post hoc test. Results: LIM improved clinical and biochemical outcomes versus monotherapies. However, LIM + low-dose ALP exhibited the greatest overall efficacy. On Day 30, paw thickness was significantly lower with LIM + ALP than with LIM alone (3.25 ± 0.31 vs. 3.98 ± 0.72 mm; p < 0.001). Serum uric acid and hepatic transaminases declined most with the combination (p < 0.0001 vs. LIM), accompanied by improved renal indices (p < 0.001). Pro-inflammatory cytokines were markedly reduced, NLRP3 immunostaining was minimal, and oxidative balance shifted toward homeostasis (↓ MDA; ↑ SOD, catalase, GSH). Radiographic and histological evaluations corroborated attenuation of joint inflammation and tissue damage. Conclusions: In the PO + MSU gout model, co-administration of the food-derived compound LIM with low-dose ALP achieved additive, dose-sparing benefits across metabolic, inflammatory, and histological endpoints. While in vivo XO activity was not directly assessed, the findings are consistent with XO-pathway modulation, NLRP3–IL-1β suppression, and redox restoration. These results highlight the potential of dietary bioactives such as d-Limonene to complement standard urate-lowering therapy, warranting further pharmacokinetic and safety validation. Full article
(This article belongs to the Special Issue Food-Derived Bioactive Compounds and Their Health Benefits)
Show Figures

Figure 1

28 pages, 12906 KB  
Article
Integrative Multi-Omics Elucidates the Therapeutic Effect of Coix Seed Oil on Rheumatoid Arthritis via the Gut-Butyrate-Joint Axis and NLRP3 Inflammasome Suppression
by Fanxin Ouyang, Xiaoyu Zhang, Rui Miao, Hongxi Kong, Wenxin Zhang, Zhidan Wang, Xu Han, Shuang Ren, Jie Zhang and Fanyan Meng
Pharmaceuticals 2026, 19(1), 48; https://doi.org/10.3390/ph19010048 - 25 Dec 2025
Viewed by 265
Abstract
Background: Rheumatoid arthritis (RA) is a chronic and debilitating autoimmune disease with a complex etiology, creating a significant unmet clinical need for safer and more effective therapeutics. Coix seed oil (CSO), a traditional Chinese medicine with a long history of use against RA, [...] Read more.
Background: Rheumatoid arthritis (RA) is a chronic and debilitating autoimmune disease with a complex etiology, creating a significant unmet clinical need for safer and more effective therapeutics. Coix seed oil (CSO), a traditional Chinese medicine with a long history of use against RA, represents a promising candidate; however, its precise mechanisms of action remain largely unexplored. Objectives: This study aimed to elucidate the mechanistic basis for the anti-arthritic effects of CSO, with a specific focus on its role in modulating the gut-joint axis. Methods: A collagen-induced arthritis (CIA) rat model was employed. The therapeutic efficacy of CSO was evaluated through detailed assessments of arthritic symptoms, joint histopathology, and Micro-CT analysis. To unravel the mechanism, an integrative multi-omics approach was applied, combining untargeted fecal metabolomics with targeted serum metabolomics, which pinpointed butyric acid as a key differential metabolite. This was integrated with 16S rRNA sequencing to profile gut microbiota remodeling. The causal role of butyrate was further verified by exogenous sodium butyrate supplementation in CIA mice. Finally, network pharmacology predictions of potential effector proteins were experimentally validated in vivo using immunofluorescence and qPCR. Results: CSO treatment significantly alleviated joint swelling and bone damage in CIA rats after the treatment of 7 days, especially on day 35. CSO primarily restored gut dysbiosis in the CIA model by upregulating butyrate levels, increasing four butyrate-producing probiotics at the genus level, and reducing two pathogenic bacteria. Further exogenous butyrate supplementation validated its ability to improve RA phenotypes. Network pharmacology analysis speculated that there were 142 common targets between CSO and RA, among which NLRP3 was its potential effector protein. In vivo studies verified the suppression of NLRP3 inflammasome activation and reduced expression of subsequent inflammatory mediators by CSO. Conclusions: Coix Seed Oil alleviates RA by orchestrating a dual-mechanism action, it remodels the gut microbiota to enhance the production of the microbiotic metabolite butyrate, while also inhibiting the NLRP3 inflammasome pathway. These findings collectively elucidate that CSO mediates its anti-arthritic effects through a novel “gut-butyrate-joint” axis, underscoring its potential as a promising dietary supplement or therapeutic agent derived from medicine-food homology for the management of RA. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

18 pages, 1596 KB  
Review
Interplay Among Gut Microbiota-Derived TMAO, Autonomic Nervous System Dysfunction, and Heart Failure Progression
by Laura Calvillo, Emilio Vanoli, Fulvio Ferrara and Eugenio Caradonna
Int. J. Mol. Sci. 2026, 27(1), 203; https://doi.org/10.3390/ijms27010203 - 24 Dec 2025
Viewed by 275
Abstract
The gut microbiota is crucial for metabolic homeostasis and cardiovascular health. Dysbiosis triggers a gut–brain–heart axis dysfunction: vagal signaling promotes neuroinflammation and cerebral damage, which in turn impairs cardiac function. This bidirectional cycle is further exacerbated by reduced cerebral perfusion. Trimethylamine-N-oxide (TMAO), a [...] Read more.
The gut microbiota is crucial for metabolic homeostasis and cardiovascular health. Dysbiosis triggers a gut–brain–heart axis dysfunction: vagal signaling promotes neuroinflammation and cerebral damage, which in turn impairs cardiac function. This bidirectional cycle is further exacerbated by reduced cerebral perfusion. Trimethylamine-N-oxide (TMAO), a metabolite of dietary choline and L-carnitine, acts as a primary mediator in this network. Elevated TMAO levels—resulting from bacterial conversion and hepatic oxidation—are linked to atherosclerosis and heart failure. Mechanistically, TMAO activates the NLRP3 inflammasome, inhibits the SIRT3-SOD2 pathway, and promotes platelet hyperreactivity. Furthermore, it modulates the autonomic nervous system, enhancing sympathetic activity and cardiac arrhythmias. Clinical evidence suggests TMAO is a potent predictor of mortality in HF. While current HF therapies focus on end-organ response (beta-blockers) or humoral pathways (ACE inhibitors), directly targeting the microbiota and TMAO offers a novel therapeutic frontier. Integrating TMAO assessment into risk models and utilizing advanced in vitro gut–brain models will be essential for developing personalized, groundbreaking cardiovascular interventions. Within this framework, the main aim of the present review is to describe how cardiac autonomic control can be directly modulated by the microbiota and its byproducts like TMAO. This latter is a leading target candidate for novel HF prevention and therapy interventions. Full article
Show Figures

Figure 1

22 pages, 1501 KB  
Review
The Convergent Immunopathogenesis of Cigarette Smoke Exposure: From Oxidative Stress to Epigenetic Reprogramming in Chronic Disease
by Aysen Kutan Fenercioglu, Hafize Uzun and Durisehvar Ozer Unal
Int. J. Mol. Sci. 2026, 27(1), 187; https://doi.org/10.3390/ijms27010187 - 24 Dec 2025
Viewed by 253
Abstract
Cigarette smoking is the leading preventable cause of chronic diseases (e.g., COPD, cardiovascular disease, cancer), largely driven by persistent immune-inflammatory mechanisms. This review synthesizes the molecular and cellular cascades linking cigarette smoke (CS) exposure to chronic pathology. CS constituents, particularly ROS/RNS, induce rapid [...] Read more.
Cigarette smoking is the leading preventable cause of chronic diseases (e.g., COPD, cardiovascular disease, cancer), largely driven by persistent immune-inflammatory mechanisms. This review synthesizes the molecular and cellular cascades linking cigarette smoke (CS) exposure to chronic pathology. CS constituents, particularly ROS/RNS, induce rapid oxidative stress that overwhelms antioxidant defenses and generates damage-associated molecular patterns (DAMPs). These DAMPs activate pattern recognition receptors (PRRs) and the NLRP3 inflammasome, initiating NF-κB signaling and the release of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). CS exposure causes profound innate immune dysregulation, including airway epithelial barrier disruption, hyperactivated neutrophils, and dysfunctional alveolar macrophages (AMs) that release destructive proteases (e.g., MMP-12) and acquire foam-cell–like characteristics. Furthermore, CS drives adaptive immunity toward a Th1/Th17-dominant phenotype while suppressing regulatory T-cell (Treg) function, thereby promoting autoimmunity and chronic tissue injury. Critically, CS induces epigenetic reprogramming (e.g., DNA methylation, miRNA dysregulation), locking immune cells into a persistent pro-inflammatory state. This convergence of oxidative stress, innate and adaptive immune dysregulation, and epigenetic alterations underlies the systemic low-grade inflammation that fuels smoking-related chronic diseases, highlighting key targets for novel therapeutic interventions. Full article
Show Figures

Graphical abstract

18 pages, 2449 KB  
Article
NLRP3 Inflammasome as Potential Predictor of Non-Responsiveness to Immunosuppressive Treatment in Lupus Nephritis
by Camila Barbosa Lyra de Oliveira, Werbson Lima Guaraná, Gisele Vajgel, Braziliano Miguel da Silva Júnior, Camilla Albertina Dantas Lima, Stanley de Almeida Araújo, Fabrício Oliveira Souto, Denise Maria do Nascimento Costa, Lucila Maria Valente and Paula Sandrin-Garcia
Int. J. Mol. Sci. 2026, 27(1), 43; https://doi.org/10.3390/ijms27010043 - 19 Dec 2025
Viewed by 300
Abstract
Lupus nephritis (LN) can affect up to 60% of patients with systemic lupus erythematosus (SLE). The NLRP3 inflammasome has been implicated in the pathogenesis of LN. This study aimed to evaluate the role of the NLRP3 inflammasome as a predictor of response to [...] Read more.
Lupus nephritis (LN) can affect up to 60% of patients with systemic lupus erythematosus (SLE). The NLRP3 inflammasome has been implicated in the pathogenesis of LN. This study aimed to evaluate the role of the NLRP3 inflammasome as a predictor of response to immunosuppressive treatment in patients with active LN. A prospective cohort study was conducted with 20 adult patients with active LN, classes III, IV, and V, from January 2021 to September 2023. Patients were followed up at biopsy (T0) and 6 months (T6) and 12 months (T12) after treatment and classified according to the primary efficacy renal response (PERR) at 12 months. Gene expression of NLRP3, CARD8, CASP1, IL1B, and IL18 was evaluated by RT-qPCR in PBMCs. Immunohistochemistry (IHC) for NLRP3 was performed on kidney tissue. The concentration of cytokine IL-1β was measured using the BD™ Cytometric Bead Array (CBA). The mean age was 31.9 ± 8.3 years, with 19 females and 1 male. After 12 months, 65% of patients achieved PERR. The IHC intensity in inflammatory cells was higher in patients with no PERR (p = 0.0426). In the no-PERR group, the gene expression of IL1B showed a significant increase at T6 (FC = 2.22: p = 0.0037) and T12 (FC = 2.91; p = 0.0001) compared with T0. Relative expression of IL1B was higher in no-PERR patients at T12 compared to the PERR group (p = 0.0477). The no-PERR group also had higher serum IL-1β levels compared to the PERR group at 12 months (2.9 ± 0.5 vs. 2.5 ± 0.7, p = 0.0164). In conclusion, our study evidenced an increase in IL1B expression and IL-1β levels over the 12 months of treatment in no-PERR patients, suggesting a potential biomarker of disease activity. Furthermore, a strong NLRP3 IHC staining score was associated with a higher likelihood of no PERR, highlighting the potential of the NLRP3 inflammasome as a predictor of worse clinical outcomes. Full article
Show Figures

Figure 1

14 pages, 990 KB  
Article
Temporal Expression of NLRP3 Inflammasome Components in Patients with Acute Coronary Syndrome
by Paraskevi Papanikolaou, Andreas Aggelopoulos, Alexios S. Antonopoulos, Panagiotis Theofilis, Maria Gazouli, Konstantinos Tsioufis and Dimitris Tousoulis
Life 2026, 16(1), 1; https://doi.org/10.3390/life16010001 - 19 Dec 2025
Viewed by 510
Abstract
Background: Inflammation is a central driver of atherothrombosis, yet the temporal behavior of key inflammasome mediators following acute coronary syndrome (ACS) is not well characterized. The NLRP3 inflammasome, a major regulator of interleukin (IL)-1β activation, has been implicated in plaque destabilization and recurrent [...] Read more.
Background: Inflammation is a central driver of atherothrombosis, yet the temporal behavior of key inflammasome mediators following acute coronary syndrome (ACS) is not well characterized. The NLRP3 inflammasome, a major regulator of interleukin (IL)-1β activation, has been implicated in plaque destabilization and recurrent cardiovascular risk. This study aims to investigate the temporal expression of NLRP3 inflammasome components in peripheral blood mononuclear cells (PBMCs) of patients with ACS. Methods: In this prospective observational study, PBMCs were collected from 73 patients with ACS during the early in-hospital phase and at 8–12 weeks follow-up. Gene expression of NLRP3, caspase-1, and IL-1β was quantified by qRT-PCR, and fold-change was calculated using the 2−ΔΔCT method. Associations with clinical and biochemical variables were evaluated using multivariable linear regression. Results: Expression of all measured inflammasome-related genes increased significantly at follow-up compared with baseline: caspase-1 (≈2-fold, p = 0.003), NLRP3 (>10-fold, p < 0.001), and IL-1β (≈4-fold, p < 0.001). Subgroup analyses showed that the post-ACS upregulation of NLRP3, caspase-1, and IL-1β was consistent across STEMI and NSTEMI presentations and was not significantly modified by diabetes status. Caspase-1 fold-change correlated positively with IL-1β, LDL-cholesterol, peak troponin I, and high sensitivity C reactive protein, whereas NLRP3 showed minimal correlations with clinical variables. In multivariable analysis, caspase-1 upregulation was independently associated with STEMI presentation and low-density lipoprotein-cholesterol, and IL-1β with type 2 diabetes. Conclusions: Patients with ACS exhibit significant and persistent upregulation of NLRP3 inflammasome components weeks after the acute event, indicating sustained immune cell priming during recovery. These findings highlight a potential molecular substrate for residual inflammatory risk and support further exploration of inflammasome-targeted therapies in the post-ACS period. Full article
Show Figures

Figure 1

26 pages, 1377 KB  
Review
The Role of Inflammasomes in Chronic Oral Inflammatory Disease and Oral Cancer: A Narrative Review
by Banan Al-Natour, Issam Rasheed and Ikhlas A. El Elkarim
Dent. J. 2025, 13(12), 609; https://doi.org/10.3390/dj13120609 - 18 Dec 2025
Viewed by 467
Abstract
Background: Chronic inflammation is a hallmark of many oral and systemic diseases and has long been recognised as a risk factor for cancer development. Central to inflammatory responses are inflammasomes—multiprotein complexes that, upon activation, trigger caspase-1–mediated release of the pro-inflammatory cytokines interleukin-1β (IL-1β) [...] Read more.
Background: Chronic inflammation is a hallmark of many oral and systemic diseases and has long been recognised as a risk factor for cancer development. Central to inflammatory responses are inflammasomes—multiprotein complexes that, upon activation, trigger caspase-1–mediated release of the pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18). Their emerging contribution to chronic oral inflammatory conditions has generated interest in understanding whether persistent inflammasome activity may also influence pathways involved in oral carcinogenesis. This review summarises current evidence on the role of inflammasomes in oral inflammatory diseases and explores their potential involvement in the transition from chronic inflammation to malignant transformation. Methods: A narrative review of the literature was conducted by searching major scientific databases for studies investigating inflammasome activation in oral tissues, inflammatory oral diseases, and mechanisms linking chronic inflammation to oral cancer. Eligible articles included experimental studies, animal models, observational clinical research, and review papers that provided mechanistic or associative insights. Due to heterogeneity in study designs, a qualitative synthesis was performed. Results: Available evidence indicates that inflammasomes, particularly NLRP3 and AIM2, contribute to the pathophysiology of pulpitis, periodontitis, and several systemic conditions that affect oral health. Preclinical and observational findings also suggest potential involvement of inflammasome-related pathways in early tumorigenic processes, although these associations require further clarification. Preliminary biomarker-based studies demonstrate that inflammasome components measurable in saliva, pulpal blood, or gingival crevicular fluid may offer minimally invasive indicators of inflammatory burden and oral health status. Conclusions: Inflammasomes appear to play a meaningful role in oral inflammatory diseases, and growing evidence links their persistent activation to mechanisms relevant to oral carcinogenesis. However, current findings are largely associative and derived primarily from experimental and early clinical research. Additional work is needed to define precisely how inflammasomes contribute to the progression from chronic oral inflammation toward malignant change and to evaluate whether targeting inflammasome pathways offers viable therapeutic or diagnostic potential. Full article
(This article belongs to the Special Issue Feature Review Papers in Dentistry: 2nd Edition)
Show Figures

Graphical abstract

22 pages, 10520 KB  
Article
Lycopene Protects Deoxynivalenol-Induced Intestinal Barrier Dysfunction and NLRP3 Inflammasome Activation by Targeting the ERK Pathway
by Zihui Cai, Zhi Lu, Youshuang Wang, Wenxi Song, Xu Yang and Cong Zhang
Antioxidants 2025, 14(12), 1513; https://doi.org/10.3390/antiox14121513 - 17 Dec 2025
Viewed by 382
Abstract
In agricultural production, Deoxynivalenol (DON) generally exists and contaminates wheat, corn, and other grains, causing intestinal damage and immunotoxicity. Lycopene (LYC), an antioxidant, anti-inflammatory carotenoid, is mainly found in red fruits such as tomatoes and has been investigated for its great medicinal advantages. [...] Read more.
In agricultural production, Deoxynivalenol (DON) generally exists and contaminates wheat, corn, and other grains, causing intestinal damage and immunotoxicity. Lycopene (LYC), an antioxidant, anti-inflammatory carotenoid, is mainly found in red fruits such as tomatoes and has been investigated for its great medicinal advantages. This study aimed to investigate the protective effect of LYC against DON-induced enterotoxicity. Our findings demonstrated that incubation of IPEC-J2 cells with 0.5 μM DON for 24 h caused intestinal barrier impairment and oxidative stress induction, which subsequently led to increased secretion of pro-inflammatory factors (TNF-α, IL-1β, IL-18, and IL-6) and decreased secretion of the counterregulatory factor (IL-10). Furthermore, DON ultimately induced NLRP3 inflammasome activation through the stimulation of the MAPK/NF-κB pathway. It is worth mentioning that the above changes were reversed after adding 30 μg/mL of LYC to DON-exposed IPEC-J2 cells. In addition, further experiments confirmed that ERK activator (4-Methylbenzylidene camphor, 4-MBC) eliminated the positive effect of LYC on alleviating enterotoxicity induced by DON in IPEC-J2 cells. In addition, further experiments confirmed that 4-MBC eliminated the positive effect of LYC on alleviating enterotoxicity induced by DON. In general, our study certified that ERK is a therapeutic target for LYC protecting DON-induced intestinal barrier dysfunction and NLRP3 inflammasome activation. Full article
Show Figures

Figure 1

17 pages, 2645 KB  
Article
Interplay Between NLRP3 Activation by DENV-2 and Autophagy and Its Impact on Lipid Metabolism in HMEC-1 Cells
by Giovani Visoso-Carvajal, Julio García-Cordero, Yandy Ybalmea-Gómez, Margarita Diaz-Flores, Moisés León-Juárez, Rosaura Hernández-Rivas, Porfirio Nava, Nicolás Villegas-Sepúlveda and Leticia Cedillo-Barrón
Pathogens 2025, 14(12), 1292; https://doi.org/10.3390/pathogens14121292 - 16 Dec 2025
Viewed by 362
Abstract
Dengue Virus (DENV) induces assembly of the NOD-like receptor (NLR) family pyrin domain containing-3 (NLRP3) inflammasome and autophagy, which are closely interconnected processes playing crucial roles in lipid metabolism and DENV replication. However, the autophagy–NLRP3 activation interplay during DENV infection in human endothelial [...] Read more.
Dengue Virus (DENV) induces assembly of the NOD-like receptor (NLR) family pyrin domain containing-3 (NLRP3) inflammasome and autophagy, which are closely interconnected processes playing crucial roles in lipid metabolism and DENV replication. However, the autophagy–NLRP3 activation interplay during DENV infection in human endothelial cells remains incompletely understood. We aimed to elucidate effects of NLRP3 activation on autophagy during DENV-2 infection. We investigated how autophagy-related molecules are altered by NLRP3 inhibition and how this regulation affects lipid metabolism, through the master lipid transcription factors SREBP-1 and 2, which increase the expression of their target lipid-synthesizing genes such as fatty acid synthase (FAS) in a model of microvascular endothelial cells (HMEC-1). We demonstrated a dynamic interplay between inflammasome activity and autophagy in DENV-infected HMEC-1 cells: autophagy increases early during infection and decreases as inflammasome activity increases. NLRP3 inflammasome inhibition affects viral replication. Glyburide (an inflammasome inhibitor) treatment partially inhibited DENV-induced NLRP3 inflammasome activation. Non-structural viral protein expression (NS3 and NS5) and infectious viral-particle formation were significantly reduced. NLRP3 inhibition also downregulated SREBP-1 and SREBP-2 activation. These findings provide new insights into the modulation of the interconnected NLRP3 inflammasome, autophagy, and lipid metabolism pathways, presenting a promising therapeutic strategy for severe clinical forms of dengue. Full article
Show Figures

Graphical abstract

37 pages, 3637 KB  
Article
Lemon Juice-Assisted Green Extraction of Strawberry Enhances Neuroprotective Phytochemicals: Insights into Alzheimer’s-Related Pathways
by Youssef Mohamed Sharaf, Jilan A. Nazeam, Karema Abu-Elfotuh, Ayah M. H. Gowifel, Ahmed M. Atwa, Ehsan Khedre Mohamed, Ahmed M. E. Hamdan, Reema Almotairi, Amira M. Hamdan, Samir M. Osman and Hala M. El Hefnawy
Pharmaceuticals 2025, 18(12), 1892; https://doi.org/10.3390/ph18121892 - 15 Dec 2025
Viewed by 946
Abstract
Background/Objective: Alzheimer’s disease (AD) is a neurodegenerative condition characterized by oxidative stress, neuroinflammation, amyloidogenesis, and tau-related pathology. This study investigated the macronutrient and phytochemical composition of strawberry (S), lemon (L), and lemon juice-assisted strawberry (S/L) extracts and evaluated their neuroprotective efficacy relative [...] Read more.
Background/Objective: Alzheimer’s disease (AD) is a neurodegenerative condition characterized by oxidative stress, neuroinflammation, amyloidogenesis, and tau-related pathology. This study investigated the macronutrient and phytochemical composition of strawberry (S), lemon (L), and lemon juice-assisted strawberry (S/L) extracts and evaluated their neuroprotective efficacy relative to selenium (Se) in an aluminum chloride (AlCl3)-induced rat model of AD. Methods: Macronutrients and phenolics were quantified in S, L, and S/L, and the extracts were profiled using high-performance liquid chromatography and electrospray ionization tandem mass-spectrometry. Male Sprague–Dawley rats received AlCl3 with or without S, L, S/L, or Se, and their cognitive performance was assessed using the Morris water maze, Y-maze, and conditioned avoidance tests. Markers of oxidative status, inflammation, cholinergic function, apoptotic signaling, and Wnt3/β-catenin pathway activity were quantified in the brain tissue, and cortico-hippocampal morphology was examined. Results: The S/L extract showed the highest carbohydrate, protein, and lipid content. The total phenolic content was highest in S/L (60.46 mg gallic acid equivalents/g), followed by L (55.08) and S (44.75), with S/L also being the richest in gallic, ellagic, and chlorogenic acids. S/L attenuated AlCl3-induced cognitive deficits, restored antioxidant status, suppressed neuroinflammation, improved cholinergic indices, modulated apoptotic signaling, and downregulated amyloidogenic and NLRP3 inflammasome markers, consistent with histological evidence of neuronal preservation. Conclusions: Lemon juice-assisted extraction enhanced the macronutrient and phenolic richness and multitarget neuroprotection of strawberries. S/L co-extracts represent promising functional food–derived adjuvants for AD management and support integrative compositional–mechanistic profiling to optimize natural product–based interventions. Full article
Show Figures

Graphical abstract

Back to TopTop