1
|
Ran X, Li H, Wang Z, Wu F, Deng Z, Zhou Q, Dai C, Peng J, Lu L, Zhou K, Ran P, Zhou Y. Increased plasma interleukin-1β is associated with accelerated lung function decline in non-smokers. Pulmonology 2025; 31:2411811. [PMID: 39883490 DOI: 10.1080/25310429.2024.2411811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 06/26/2024] [Indexed: 01/31/2025] Open
Abstract
Interleukin-1β is one of the major cytokines involved in the initiation and persistence of airway inflammation in chronic obstructive pulmonary disease (COPD). However, the association between plasma interleukin-1β and lung function decline remains unclear. We aimed to explore the association between plasma interleukin-1β and lung function decline. This longitudinal evaluation of data from the Early COPD study analysed the association between the plasma interleukin-1β concentration, lung function decline, and COPD exacerbation. Overall, 1,328 participants were included in the baseline analysis, and 1,135 (85%) completed the 1-year follow-up. Increased plasma interleukin-1β was associated with accelerated lung function decline in non-smokers (forced expiratory volume in 1 s: per unit natural log-transformed increase, adjusted unstandardised β [95% confidence interval] 101.46 [16.73-186.18] mL/year, p=0.019; forced vital capacity: per unit natural log-transformed increase, adjusted unstandardised β [95% confidence interval] 146.20 [93.65-198.75] mL/year, p<0.001), but not in smokers. In non-smokers, participants with an interleukin-1β concentration in the top 30% (>5.02 pg/mL) had more respiratory symptoms, more severe emphysema and air trapping, and higher levels of inflammation-related biomarkers. In this study, a subgroup with increased plasma interleukin-1β was identified among non-smokers, and increased plasma interleukin-1β was associated with lung function accelerated decline.
Collapse
Affiliation(s)
- Xinru Ran
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Haiqing Li
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zihui Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fan Wu
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International BioIsland, Guangzhou, China
| | - Zhishan Deng
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiaorui Zhou
- The First Clinical College of Guangzhou Medical University, Guangzhou, China
| | - Cuiqiong Dai
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jieqi Peng
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lifei Lu
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kunning Zhou
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pixin Ran
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International BioIsland, Guangzhou, China
| | - Yumin Zhou
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine & Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Binte Hanafi Z, Mei Y, Teo HY, Zhu Y, Yong Lionel CC, Chiu JW, Lu J, Liu H. Calpain 2 regulates IL-1α secretion and inhibits tumor development via modulating calpain 1 expression in the tumor microenvironment. Oncoimmunology 2025; 14:2451444. [PMID: 39803956 PMCID: PMC11730618 DOI: 10.1080/2162402x.2025.2451444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Tumor-promoting inflammation significantly impacts cancer progression, and targeting inflammatory cytokines has emerged as a promising therapeutic approach in clinical trials. Interleukin (IL)-1α, a member of the IL-1 cytokine family, plays a crucial role in both inflammation and carcinogenesis. How IL-1α is secreted in the tumor microenvironment has been poorly understood, and we previously showed that calpain 1 cleaves pro-IL-1α for mature IL-1α secretion, which exacerbates hepatocellular carcinoma by recruiting myeloid-derived suppressor cells. In this study, we report that calpain 2 also modulates IL-1α secretion. Notably, a deficiency in calpain 2 resulted in enhanced hepatocellular carcinoma development within an IL-1α-enriched tumor microenvironment. Further investigations revealed that calpain 2 deficiency increased calpain 1 expression, implying a compensatory mechanism between the two calpains. Mechanistically, calpain 2 deficiency led to increased expression of FoxO3, which is a forkhead transcription factor that promotes calpain 1 expression. Collectively, these results suggest that calpain 2 modulates calpain 1 expression, and therefore IL-1α secretion through the induction of FoxO3, offering novel potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Zuhairah Binte Hanafi
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Mei
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chew Chin Yong Lionel
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Wen Chiu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinhua Lu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Kulkarni MM, Popovic B, Nolfi AL, Skillen CD, Brown BN. Distinct impacts of aging on the immune responses to extracellular matrix-based versus synthetic biomaterials. Biomaterials 2025; 320:123204. [PMID: 40056612 DOI: 10.1016/j.biomaterials.2025.123204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 03/10/2025]
Abstract
All implanted materials inevitably trigger an acute inflammatory response. The long-term outcome, however, is dependent on the trajectory of this response. This study investigates the effects of aging on the immune response to two commercially available biomaterials. Extracellular matrix-based urinary bladder matrix (UBM) and synthetic polypropylene mesh (PPM) were implanted in young (4 months) and aged (18 months) C57BL/6J mice. Overall, PPM led to a sustained inflammatory response regardless of the age of the mice. In contrast, UBM induced an initial inflammatory response that matured into a pro-regenerative/remodeling response with time, though aged mice exhibited a delayed resolution of inflammation. The PPM-induced response was predominantly pro-inflammatory with consistently higher M1-like macrophage phenotype, whereas the response to UBM was characterized by an anti-inflammatory M2-like phenotype, especially in young mice. RNA sequencing revealed marked age-related differences in gene transcription. At day 7 post-implantation, the young mice with UBM showed a robust upregulation of both pro- and anti-inflammatory pathways as compared to young mice implanted with PPM, however, by day 14, the gene expression profile transitioned into an anti-inflammatory profile. Intriguingly, in aged mice, the response to UBM was distinct with consistent downregulation of inflammatory genes compared to PPM, while the response to PPM in both young and aged animals was largely consistent. Upstream analysis identified cytokines as key drivers of the host response, with IL-4 and IL-13 in young mice, and TNF-α and IL-1β driving chronic inflammation in aged mice. These findings highlight the importance of host age in biomaterial outcome, and the potential of ECM-based materials to mount a favorable response even in the presence of age-related immune dysregulation.
Collapse
Affiliation(s)
- Mangesh M Kulkarni
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Branimir Popovic
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alexis L Nolfi
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Clint D Skillen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Bryan N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
4
|
Hatamipour M, Saremi H, Kesharwani P, Sahebkar A. Identification of potential therapeutic targets for stroke using data mining, network analysis, enrichment, and docking analysis. Comput Biol Chem 2025; 117:108431. [PMID: 40127530 DOI: 10.1016/j.compbiolchem.2025.108431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/26/2025]
Abstract
Stroke is a leading cause of disability and death worldwide. In this study, we identified potential therapeutic targets for stroke using a data mining, network analysis, enrichment, and docking analysis approach. We first identified 1991 genes associated with stroke from two publicly available databases: GeneCards and DisGeNET. We then constructed a protein-protein interaction (PPI) network using the STRING database and identified 1301 nodes and 5413 edges. We used Metascape to perform GO enrichment analysis and KEGG pathway enrichment analysis. The results of these analyses identified ten hub genes (TNF, IL6, ACTB, AKT1, IL1B, TP53, VEGFA, STAT3, CASP3, and CTNNB1) and five KEGG pathways (cancer, lipid and atherosclerosis, cytokine-cytokine receptor interaction, AGE RAGE signaling pathway in complications, and TNF signaling pathway) that are enriched in stroke genes. We then performed molecular docking analysis to screen potential drug candidates for these targets. The results of this analysis identified several promising drug candidates that could be used to develop new therapeutic strategies for stroke.
Collapse
Affiliation(s)
- Mahdi Hatamipour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Saremi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Shawky A, Saber S, Abd El-Kader EM, El-Kashef HA. Verapamil inhibits TXNIP-dependent NLRP3 Inflammasome activation in an ulcerative colitis rat model: A new evolving role of the calcium channel blocker. Int Immunopharmacol 2025; 158:114751. [PMID: 40359884 DOI: 10.1016/j.intimp.2025.114751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Ulcerative colitis (UC) is a long-term inflammatory bowel disease (IBD) associated with significant morbidity. It is marked by inflammation and damage to the colon's mucosal lining. Studies have shown that NLRP3 inflammasome activation, apoptosis, and impaired autophagy are critical in its pathogenesis. Verapamil, a calcium channel blocker, has been found to inhibit NLRP3 inflammasome activation in various preclinical models. However, the potential influence of verapamil on the TXNIP in UC remains unexplored. This study investigates the effects of verapamil on an UC rat model induced chemically by acetic acid. Verapamil effectively inhibited the TXNIP-NLRP3-caspase-1 axis, reducing inflammasome activation and the release of IL-1β and IL-18. Additionally, verapamil suppressed NFκB, the priming step of NLRP3 activation. The drug enhanced autophagic activity, as indicated by increased expression of LC3-II and Beclin-1, along with reduced LC3-I and mTOR expression. Moreover, it demonstrated anti-apoptotic effects mediated by regulating Bax and cleaved caspase-3. These molecular changes contributed to mucosal healing and improved microscopic and macroscopic outcomes in the colitis model. Furthermore, verapamil improved the colon weight-to-length ratio and disease activity scores and mitigated oxidative stress. As verapamil has been safely used in clinics to treat hypertension, our findings suggest it may be a safe therapeutic option for ameliorating inflammation and apoptosis and activating autophagy in UC pathology. Since hypertension demonstrates a strong association with UC, the use of verapamil merits particular attention in hypertensive patients fighting against IBD.
Collapse
Affiliation(s)
- Ahmed Shawky
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| | - Eman M Abd El-Kader
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| | - Hassan A El-Kashef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
6
|
Li W, Li Y, Qiu Y, Huang R, Niu J, Chen J, Liu Y, Chen L. Kurarinone and Nor-kurarinone inhibit NLRP3 inflammasome activation and regulate macrophage polarization against ulcerative colitis. Int Immunopharmacol 2025; 157:114758. [PMID: 40318276 DOI: 10.1016/j.intimp.2025.114758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 04/01/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Activation of NOD-like receptor protein 3 (NLRP3) can lead to the production of inflammatory factors and perturbation of macrophage polarization, leading to an intestinal immune imbalance that promotes the progression of ulcerative colitis (UC). In this study, we investigated the therapeutic effect of Kurarinone and Nor-kurarinone on UC and their regulatory mechanisms relating to NLRP3 inflammasome activation and macrophage polarization. UC mice were induced using dextran sulfate sodium (DSS) and treated with Kurarinone and Nor-kurarinone. Results showed that Kurarinone and Nor-kurarinone could alleviate weight loss, decrease the disease activity index (DAI) score, shorten colon length, inhibit formation of the NLRP3 inflammasome in the colon and regulate macrophage polarization in UC mice. The THP-1 cells were used as an in vitro model of the NLRP3 inflammasome, conducted by treatment with lipopolysaccharide (LPS) and ATP/Nigericin. Kurarinone and Nor-kurarinone can inhibit the NLRP3 inflammasome formation response by disrupting the NLRP3/ASC interaction to inhibit NLRP3 assembly and then regulating the polarization of macrophages. In conclusion, Kurarinone and Nor-kurarinone inhibited NLRP3 inflammasome assembly to counteract activation of the NLRP3 inflammasome. This inhibition led to a reduction in M1 polarization of intestinal macrophages in UC mice to keep the balance of M1/ M2 macrophages. Our study suggests that Kurarinone and Nor-kurarinone may be novel therapeutic modalities for UC.
Collapse
Affiliation(s)
- Wanyu Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yadi Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yongyi Qiu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ruiting Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jing Niu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiawen Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Liu
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Lei Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
7
|
Altun S, Özdemir S, Arslan H, Kiliçlioğlu M, Yaprak E, Bolat İ, Aydın Ş. Impact of long-term deltamethrin exposure on Alzheimer's-related neurodegeneration in rats. Exp Neurol 2025; 388:115223. [PMID: 40120660 DOI: 10.1016/j.expneurol.2025.115223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/27/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
In recent years, epidemiological studies have emerged indicating a potential association between chronic exposure to pesticides and the development of chronic neurodegenerative nervous system diseases such as Alzheimer's disease. In this study, we aimed to investigate the potential role of long-term nonfatal exposure to Deltamethrin in spreading this disease. To this end, a range of aspects of brain damage were discussed in rats administered deltamethrin in oral doses of 0.65 mg/kg b.w. and 1.3 mg/kg b.w. for 30 days. The activation of beta-amyloid, the primary component of plaques characteristic of Alzheimer's disease, and the NG2, a type 1 transmembrane protein, was assessed by immunohistochemistry and western blot methods in rat brain. In addition, the expression level of the APP, GFAP, NfL, TNF-alpha, CXCL9, CCL5, and IL-1 alpha genes in deltamethrin-exposed brain tissue was measured using qRT-PCR. In addition, levels of pTau181 and Abeta42 were measured with ELISA. A strong positive immunohistochemical reaction for beta-amyloid was detected in the deltamethrin-exposed brain tissues. A decrease in NG2 immunofluorescence positivity was found in the application groups compared to the control group. It was demonstrated that deltamethrin exposure significantly up-regulated the expressions of APP, GFAP, NfL, TNF-alpha, CXCL9, CCL5, and IL-1 alpha genes, also significantly higher the levels of pTau181 and Abeta42 (pg/ml) in rat brain tissues. This study provides scientific evidence that exposure to chronic doses of deltamethrin may play a positive role in the development of diseases such as Alzheimer's. Future studies should investigate similar projects and expand knowledge on the topic.
Collapse
Affiliation(s)
- Serdar Altun
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey.
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Harun Arslan
- Department of Basic Sciences, Faculty of Fisheries, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Metin Kiliçlioğlu
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Esra Yaprak
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Yakutiye, 25240 Erzurum, Turkey
| | - İsmail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| | - Şeyma Aydın
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240 Erzurum, Turkey
| |
Collapse
|
8
|
Seaton-Terry A, Hunter Z, Lewis M, Fisher S, Bray E, Townsend B, Gabure S, Daniel L, Whalen M. Toll-Like Receptors in Pentachlorophenol- and Dibutyltin-Induced Production of Pro-Inflammatory Cytokines, Interleukin (IL)-1β, and IL-6, by Human Immune Cells. J Appl Toxicol 2025; 45:976-993. [PMID: 39914831 PMCID: PMC12064381 DOI: 10.1002/jat.4762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 05/11/2025]
Abstract
Pentachlorophenol (PCP) and dibutyltin dichloride (DBT) contaminate the environment due to their diverse applications. PCP has been found from 0.26 to 5 μM in the serum of exposed individuals and at an average of 0.15 μM in the unexposed. DBT has been detected in human blood at levels up to 0.3 μM. Exposure to these contaminants is linked to pathological conditions including cancer. Interleukin-1 beta (IL-1β) and IL-6 are pro-inflammatory cytokines that when produced inappropriately can cause chronic inflammation, which is linked to pathologies including autoimmune diseases and cancer. PCP and DBT have been shown to increase the production of IL-1β and IL-6 by immune cells in a MAP kinase (MAPK) dependent process. Toll-like receptors (TLRs) activate the signaling pathways linked to MAPK that lead to production of these cytokines. This study demonstrates that PCP-induced production of IL-1β and IL-6 is dependent on TLR4 and TLR8, and independent of TLR1/2, TLR2, and TLR3. Additionally, DBT-induced IL-6 production depends on TLR1/2, whereas IL-1β production does not. Blocking the TLR-linked adapter protein, MyD88, lead to a loss of both PCP and DBT stimulation of IL-1β and IL-6. These findings indicate that both PCP and DBT interact with selected TLRs as part of their mechanisms of elevating the levels of critical pro-inflammatory cytokines, which may contribute to chronic inflammation and its related pathologies.
Collapse
Affiliation(s)
| | - Zinia Hunter
- Department of Biology, Tennessee State University, Nashville, Tennessee, USA
| | - Meaghan Lewis
- Department of Biology, Tennessee State University, Nashville, Tennessee, USA
| | - Sophia Fisher
- Department of Biology, Tennessee State University, Nashville, Tennessee, USA
| | - Ellie Bray
- Department of Biology, Tennessee State University, Nashville, Tennessee, USA
| | - Brian Townsend
- Department of Biology, Tennessee State University, Nashville, Tennessee, USA
| | - Saleban Gabure
- Deapartment of Chemistry, Tennessee State University, Nashville, Tennessee, USA
| | - Latoya Daniel
- Department of Biology, Tennessee State University, Nashville, Tennessee, USA
| | - Margaret Whalen
- Deapartment of Chemistry, Tennessee State University, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Wang J, Ji L, Gao Y, Sun J, Zhou X, Ding Y, Zhou Z, Guo X, Liu C, Wang Y, Zhang Q, Lv Z, Ma D. Inhibition of CDK5 signaling mediated inflammation in macrophages promotes cutaneous wound healing. Sci Rep 2025; 15:18509. [PMID: 40425656 PMCID: PMC12116921 DOI: 10.1038/s41598-025-02488-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Cyclin-dependent kinase 5 (CDK5) plays a critical role in the inflammatory response. Macrophages are pivotal orchestrators of inflammation, fibrosis, and wound repair. However, the effectiveness of CDK5 in macrophages on cutaneous wound healing remains inadequately characterized. We determined the role of CDK5 signaling pathway in macrophages in mouse cutaneous wound healing through the established macrophage-specific deletion of CDK5 (myeCDK5-/-) mice and the pharmacological CDK5 inhibitor Roscovitine. Phosphorylated proteomics, western blotting, Masson staining, and dualimmunofluorescence staining were performed to investigate the potential mechanisms underlying CDK5-mediated inflammatory regulation in macrophages in wound healing. CDK5 expression and phosphorylation were both elevated significantly in cutaneous wound healing process in mice. Moreover, an accelerated wound healing in myeCDK5-/- mice was exhibited with the reduced pro-inflammatory mediators (IL-1β and iNOS) and the elevated anti-inflammatory markers (IL-10 and CD163) expression significantly. CDK5 deficiency in macrophages enhanced tissue remodeling, evidenced by increased collagen deposition and capillary density (CD31+ cells). Consistently, Roscovitine-treated mice also showed accelerated wound healing, accompanied by decreased pro-inflammatory factors and increased anti-inflammatory markers at the wound site. Mechanistically, the decreased phosphorylation of SIRT1 at the Ser14 and Ser47 sites, as a substrate of CDK5, was confirmed in myeCDK5-/- mice. These data are the first to indicate that CDK5 signaling-dependent regulation of SIRT1 phosphorylation in macrophage-mediated inflammation is required for the wound healing process, warranting consideration of the CDK5-SIRT1 pathway as a therapeutic target for cutaneous wound healing.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- The Fourth Department of Bone Injury, The First Affiliated Hospital of Hebei University of Traditional Chinese Medicine, Shijiazhuang, 050011, China
| | - Lin Ji
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- The Fourth Department of Bone Injury, The First Affiliated Hospital of Hebei University of Traditional Chinese Medicine, Shijiazhuang, 050011, China
| | - Yingbo Gao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Jingyu Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Xiaobin Zhou
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yujia Ding
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Zihan Zhou
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Xiaofan Guo
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Chao Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yujie Wang
- The Fourth Department of Bone Injury, The First Affiliated Hospital of Hebei University of Traditional Chinese Medicine, Shijiazhuang, 050011, China
| | - Qingfu Zhang
- Burn and Wound Repair Center, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Zhenmu Lv
- Burn and Wound Repair Center, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Fourth Department of Bone Injury, The First Affiliated Hospital of Hebei University of Traditional Chinese Medicine, Shijiazhuang, 050011, China
| | - Dong Ma
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, and Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
10
|
Karim SU, Nazneen F, Denyoh PMD, Bai DS, Romero DG, Bai F. Heterozygous interferon signaling deficient mice as animal models for Chikungunya virus infection in the heart. Sci Rep 2025; 15:18022. [PMID: 40410198 PMCID: PMC12102211 DOI: 10.1038/s41598-025-02191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
Although chikungunya virus (CHIKV)-caused cardiovascular diseases are frequently reported in clinics, the underlying mechanisms are poorly understood, which is primarily due to a lack of animal models. In this study, we report that CHIKV infection in homozygous interferon α/β receptor-deficient (ifnar1-/-) and interferon α/β/γ receptor-deficient (ifnag-/-) mice resulted in high viral loads in the hearts as early as day (D) 1 post-infection (p.i.) but with 100% mortality within three days p.i. In contrast, the heterozygous ifnar1+/-and ifnag+/- mice survived CHIKV infection and bore higher viral burdens in the heart tissues than the wild-type (WT) controls. Immunohistochemistry and flow cytometry revealed that more leukocytes, particularly neutrophils, infiltrated the heart of ifnag+/- and ifnar1+/- mice than WT mice. In addition, the Hematoxylin and Eosin staining analysis showed that CHIKV infection caused vasculitis in the left ventricles on D5 p.i. in both heterozygous groups and the vacuole formation and pyknosis in ifnar1+/- mice. Moreover, CHIKV infection may also lead to cardiac fibrosis, as indicated by the upregulation of the expression of the Connective Tissue Growth Factor gene in the hearts of ifnar1+/- mice. In summary, our data suggest that the heterozygous ifnar1+/- and ifnag+/- mice are invaluable for studying pathogenesis and testing therapeutic interventions for CHIKV-caused cardiac diseases.
Collapse
Affiliation(s)
- Shazeed-Ul Karim
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA
| | - Farzana Nazneen
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA
| | - Prince M D Denyoh
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA
| | - David S Bai
- Oak Grove High School, Hattiesburg, MS, 39402, USA
| | - Damian G Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Mississippi Center of Excellence in Perinatal Research, Jackson, MS, 39216, USA
- Women's Health Research Center, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Fengwei Bai
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA.
| |
Collapse
|
11
|
Lee SY, Lee GH, Maeng J, Kim SY, Yun HY, Jeong GS, Jeong HG. Anti-Inflammasome Effect of Impressic Acid on Diesel Exhaust Particulate Matter-Induced NLRP1 Inflammasome via the Keap1/p62/Nrf2-Signaling Pathway in Keratinocytes. Antioxidants (Basel) 2025; 14:610. [PMID: 40427491 PMCID: PMC12109400 DOI: 10.3390/antiox14050610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Diesel exhaust particulate (DEP) is widely recognized to weaken lung function and skin diseases. When the skin, which defends against external factors, is exposed to PM2.5, various chronic inflammatory diseases occur. When keratinocytes recognize harmful signals, they synthesize the NOD-like receptor protein 1 (NLRP1) inflammasome. DEP enhances NF-κB signaling and NLRP1 inflammasome expression through the interaction of TXNIP with NLRP1 in keratinocytes. Although many studies have reported the anti-inflammatory and antioxidant characteristics of Impressic acid (IPA), the umbrella consequences of IPA for PM2.5-influenced inflammasomes and the associated mechanisms remain unknown. Therefore, this study aimed to examine the protective function of IPA against inflammation in human keratinocytes. IPA attenuated the NLRP1 expression, caspase-1, IL-1β actuation, and NF-κB and IκB phosphorylation induction by DEP. IPA upregulated the Nrf2, HO-1, and NQO1 expression through CaMKKβ, AMPK, and GSK3β phosphorylation. Also, IPA led to the elevation of p62 and the degradation of the Keap1 protein. ML385 reversed the suppressive effect of IPA on the NLRP1 inflammasome, which was enhanced by DEP, and NAC counteracted the effect of ML385. These findings indicate that IPA can suppress inflammation induced by PM2.5 by expressing antioxidant enzymes through the Keap1/p62/Nrf2-signaling pathway in human keratinocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hye Gwang Jeong
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea; (S.Y.L.); (G.H.L.); (J.M.); (S.Y.K.); (H.-Y.Y.); (G.-S.J.)
| |
Collapse
|
12
|
Wu X, Zhang Y, Yi F, Geng Z, Guo M, Ling X, Li J, Li L. Anti-inflammatory and barrier repair mechanisms of active components in Daemonorops draco Bl. for UVB-induced skin damage. Sci Rep 2025; 15:17124. [PMID: 40382359 DOI: 10.1038/s41598-025-01289-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Daemonorops draco Bl. extract and its active ingredients can remove blood stasis and promote muscle and wound healing and are widely used in skin health and other fields. Modern pharmacological studies have demonstrated that this extract exerts excellent anti-inflammatory effects beneficial for skin barrier repair. However, the mechanism of action and monomeric components of D. draco remain unclear. Seven active monomers (XJ-1 ~ XJ-7) were extracted and purified from D. draco. The successful construction of the HaCaT inflammation model was achieved through the detection of IL-1β and TNF-α expressions in UVB-irradiated HaCaT cells. Based on this cellular model, (2 S)-5-methoxy-6-methylflavan-7-ol (XJ-2) was determined to be the best-screened monomer. The effects of XJ-2 on the production of reactive oxygen species (ROS) and Ca2+ in HaCaT cells were investigated using fluorescent probes and flow cytometry, respectively. The impact of XJ-2 on the expression of crucial proteins within the NF-κB pathway was examined via immunofluorescence and western blotting. The expression levels of downstream inflammatory factors, namely IL-1β and TNF-α, were detected through PCR. The effects of XJ-2 on the expression of skin barrier-related factors filaggrin (FLG), aquaporin 3 (AQP-3), and claudin1 (CLDN1) were investigated using PCR, immunofluorescence, and western blotting. Based on these findings, we comprehensively examined the mechanisms underlying the anti-inflammatory and barrier repair effects of XJ-2. XJ-2 primarily protected the internal structure and function of the cells by inhibiting the mass production of ROS and Ca2+ inflow. XJ-2 exerts anti-inflammatory effects by regulating the key proteins of the NF-κB/IKKα pathway and reducing the expression of inflammatory factors. XJ-2 repairs skin barrier damage by regulating multiple factors. Compound XJ-2 from D. draco exerts excellent anti-inflammatory and barrier repair effects, possesses great potential for the treatment of skin diseases, and can be used as a dermatological drug to repair skin barrier damage.
Collapse
Affiliation(s)
- Xingyi Wu
- Beijing Key Lab of Plant Resource Research and Development, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing, 100048, China
| | - Ying Zhang
- Beijing Key Lab of Plant Resource Research and Development, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing, 100048, China
| | - Fan Yi
- Beijing Key Lab of Plant Resource Research and Development, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing, 100048, China
| | - Zaijun Geng
- Beijing Key Lab of Plant Resource Research and Development, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing, 100048, China
| | - Miaomiao Guo
- Beijing Key Lab of Plant Resource Research and Development, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing, 100048, China
| | - Xiao Ling
- Beijing Lan Divine Technology Co. LTD, Culture Building, No. A59, Zhongguancun Street, Haidian District, Beijing, 100872, China
| | - Jun Li
- School of Chinese Materia Medica, Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, North Third Ring East Road, Chaoyang District, Beijing, 100029, China.
| | - Li Li
- Beijing Key Lab of Plant Resource Research and Development, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing, 100048, China.
| |
Collapse
|
13
|
Phillips KM, Lavere PF, Hanania NA, Adrish M. The Emerging Biomarkers in Chronic Obstructive Pulmonary Disease: A Narrative Review. Diagnostics (Basel) 2025; 15:1245. [PMID: 40428238 PMCID: PMC12110743 DOI: 10.3390/diagnostics15101245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
The burden of chronic obstructive pulmonary disease (COPD) is increasing, especially for women in low-to-middle income countries. Biomarkers provide ever-increasing diagnostic precision for COPD and show promise for primary, secondary, and tertiary disease prevention. This review describes emerging applications for biomarkers in COPD, especially as they align with the Global Initiative for Chronic Obstructive Lung Disease (GOLD) emphasis on prevention, early diagnosis, and response to therapy. These biomarkers include blood eosinophils; IgE; C-reactive protein; fibrinogen; procalcitonin; interleukins 6, 8, and 33; tumor necrosis factor alpha; and soluble receptor for advanced glycated products (sRAGE). They have been used in various ways to identify COPD endotypes, predict exacerbations, predict mortality, and monitor the response to therapy. The fraction of exhaled nitric oxide (FENO) is increasingly studied in eosinophilic COPD endotypes and can be a diagnostic and predictive non-invasive biomarker. Imaging biomarkers, especially the quantitative computerized tomography (QCT) assessment of airway remolding, functional small airway disease, air trapping, lung function, and volume surrogates, all serve as non-invasive biomarkers for screening, early detection, and disease progression. Biomarkers facilitate all the phases of COPD care from detecting early airflow obstruction to predicting exacerbation and mortality. Biomarkers will be increasingly used as precise diagnostic tools to improve the COPD outcomes. The aim of this narrative review is to summarize the recent investigations in COPD biomarkers and their clinical applications.
Collapse
Affiliation(s)
| | | | | | - Muhammad Adrish
- Section of Pulmonary, Critical Care, and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (K.M.P.); (P.F.L.); (N.A.H.)
| |
Collapse
|
14
|
Mezni G, Issa H, Dahdah M, Poulin A, Daïch A, Alamri A, Rouabhia M, Semlali A. New Curcumin Analogue (PAC) Inhibits Candida albicans Virulence, Restricts Its Adhesion Potential, and Relieves Oral Epithelial Cell Inflammation and Defense Mechanisms. Antibiotics (Basel) 2025; 14:495. [PMID: 40426561 PMCID: PMC12108166 DOI: 10.3390/antibiotics14050495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/21/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Objectives: The oral cavity hosts one of the most complex microbial communities in the body. A disruption of the balance favors the growth of pathogenic species, contributing to oral diseases. The rise in microbial resistance has limited the effectiveness of conventional treatments, shifting the interest to natural product-based alternatives. Given its superior bioavailability and bioactivity in other models, this study investigates the antifungal potential of a novel curcumin derivative, PAC (3,5-bis(4-hydroxy-3-methoxybenzylidene)-N-methyl-4-piperidone), and studies its impact on host-pathogen dynamics and host defense mechanisms. Methods:Candida albicans was used as the model organism. Viability, growth kinetics, and colony formation were evaluated using optical density, agar culture, and MTT assay. Biofilm formation was assessed through electron microscopy and total sugar quantification. The morphological transition from hyphae to the less virulent blastospore was monitored using an optical microscope. The gene expression of adhesion factors and host defense markers was analyzed using RT-PCR. Results: PAC impairs C. albicans viability and reduces virulence by compromising biofilm formation and ensuring phenotypic transition to a blastospore form. Also, PAC controls C. albicans growth via necrosis/ROS pathways. As a result, PAC appears to repress host-pathogen interaction by downregulating SAPs, EAP1, and HWP1 adhesion genes, thus relieving the need to activate gingival epithelial cell defense mechanisms. This is highlighted by recording baseline levels of IL-6, IL-8, and IL-1β cytokines and antimicrobial β-defensin peptides in the presence of less virulent candida forms. Conclusions: PAC effectively reduces C. albicans virulence by limiting biofilm formation and adhesion while minimizing inflammatory responses. These findings support its potential as a promising therapeutic agent for infectious disease control.
Collapse
Affiliation(s)
- Ghazoua Mezni
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada; (G.M.); (H.I.); (M.D.); (A.P.); (M.R.)
| | - Hawraa Issa
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada; (G.M.); (H.I.); (M.D.); (A.P.); (M.R.)
| | - Manal Dahdah
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada; (G.M.); (H.I.); (M.D.); (A.P.); (M.R.)
| | - Anaïs Poulin
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada; (G.M.); (H.I.); (M.D.); (A.P.); (M.R.)
| | - Adam Daïch
- Normandie Univ., UNILEHAVRE, INC3M FR 3038 CNRS, URCOM, 76600 Le Havre, France. UR 3221, UFR ST, BP: 1123, 25 rue Philipe Lebon, 76063 Le Havre Cedex, France;
| | - Abdulaziz Alamri
- Biochemistry Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Mahmoud Rouabhia
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada; (G.M.); (H.I.); (M.D.); (A.P.); (M.R.)
| | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada; (G.M.); (H.I.); (M.D.); (A.P.); (M.R.)
| |
Collapse
|
15
|
Roy-Biswas S, Hibma M. The Epithelial Immune Response to Human Papillomavirus Infection. Pathogens 2025; 14:464. [PMID: 40430784 PMCID: PMC12114228 DOI: 10.3390/pathogens14050464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
The skin is a complex organ, containing an intricate network of immune cells that are crucial for host barrier function and defence against pathogens. Human papillomavirus (HPV) exclusively infects the skin, and its lifecycle is intimately associated with epithelial cell division and differentiation. There are over 450 HPV types, 12 of which are classified as carcinogenic. The primary focus of this review is the epithelial immune response to HPV infection of the cervix during the initial stages of infection, productive infection, and disease progression. During the early stages of infection, cells are HPV-positive; however, there are no attributable histological changes to the epithelium. The HPV-infected cells have the capacity for innate sensing and signalling through toll-like receptors in response to viral nucleic acids. However, HPV has evolved multiple mechanisms to evade the innate response. During productive infection, all viral antigens are expressed and there are visible histological changes to the epithelium, including koilocytosis. Disease regression is associated with Tbet positive cells in the infected epithelium and the presence of CD4 and CD8 T cells in the lamina propria. Disease progression is associated with the overexpression of the E6 and E7 oncoproteins after integration of viral genomes into the host chromosomal DNA. Histologically, the epithelium is less differentiated, and changes to cells include a higher nuclear-to-cytoplasmic ratio and an increased mitotic index. Immune changes associated with disease progression include increased numbers of cells expressing suppressor molecules, such as FoxP3, Blimp-1, and HMGB1, and myeloid cell infiltrates with an M2-like phenotype. This review highlights the gaps in the understanding of the immune response in HPV-positive cervical neoplasia, and in regression and progression of disease. This knowledge is critical for the development of effective immunotherapies that reliably cause HPV-positive cervical neoplasia to regress.
Collapse
Affiliation(s)
| | - Merilyn Hibma
- Department of Pathology, Dunedin School of Medicine, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand;
| |
Collapse
|
16
|
Xia LY, Yu NR, Huang SL, Qu H, Qin L, Zhao QS, Leng Y. Dehydrotrametenolic acid methyl ester, a triterpenoid of Poria cocos, alleviates non-alcoholic steatohepatitis by suppressing NLRP3 inflammasome activation via targeting Caspase-1 in mice. Acta Pharmacol Sin 2025:10.1038/s41401-025-01569-9. [PMID: 40329004 DOI: 10.1038/s41401-025-01569-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025]
Abstract
Non-alcoholic steatohepatitis (NASH) has emerged as a prevalent chronic liver disease with a huge unmet clinical need. A few studies have reported the beneficial effects of Poria cocos Wolf (P. cocos) extract on NASH mice, but the active components were still unknown. In this study we investigated the therapeutic effects of dehydrotrametenolic acid methyl ester (ZQS5029-1), a lanosterol-7,9(11)-diene triterpenes in P. cocos, in a high-fat diet plus CCl4 induced murine NASH model and a GAN diet induced ob/ob murine NASH model. The NASH mice were treated with ZQS5029-1 (75 mg·kg-1·d-1, i.g.) for 6 and 8 weeks, respectively. We showed that ZQS5029-1 treatment markedly relieved liver injury, inflammation and fibrosis in both the murine NASH models. We found that ZQS5029-1 treatment significantly suppressed hepatic NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation in both the NASH murine models, and blocked lipopolysaccharides (LPS)+adenosine 5'-triphosphate (ATP)/Nigericin-induced NLRP3 inflammasome activation in bone marrow-derived macrophages (BMDMs) and Kupffer cells in vitro. We demonstrated that ZQS5029-1 directly bound to the H236 residue of mouse Caspase-1, thereby inhibiting NLRP3 inflammasome activation. The effects of ZQS5029-1 on macrophage-hepatocyte/HSC crosstalk were analyzed using the supernatants from macrophages preconditioned with LPS + ATP introduced into hepatocytes and hepatic stellate cells (HSCs). We found that the conditioned medium from the BMDMs induced injury and death, as well as lipid accumulation in hepatocytes, and activation of HSCs; these effects were blocked by conditioned medium from BMDMs treated with ZQS5029-1. Moreover, the protective effects of ZQS5029-1 on hepatocytes and HSCs were eliminated by H236A-mutation of Caspase-1. We conclude that ZQS5029-1 is a promising lead compound for the treatment of NASH by inhibiting NLRP3 inflammasome activation through targeting Caspase-1 and regulating the macrophage-hepatocyte/HSC crosstalk.
Collapse
Affiliation(s)
- Ling-Yan Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nai-Rong Yu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Su-Ling Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hui Qu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li Qin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qin-Shi Zhao
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Ying Leng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
17
|
Wu J, Qiu W, Li G, Guo H, Dai S, Li G. Effects of glutamine supplementation on the growth performance, antioxidant capacity, immunity and intestinal morphology of cold-stressed prestarter broiler chicks. Vet Res Commun 2025; 49:183. [PMID: 40310539 DOI: 10.1007/s11259-025-10756-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Cold stress has important effects on the growth and production of broiler chickens. Glutamine (Gln) is a conditionally essential amino acid that plays an important role in promoting intestinal development and enhancing immune function. The aim of this study was to investigate the effects of Gln supplementation on the growth performance and health of cold-stressed prestarter broiler chicks. METHODS AND RESULTS A total of 375 1-day-old male SZ901 broilers were randomly divided into five groups (CON, CS, GLN1, GLN2, GLN3). Birds in the CON and CS groups were provided with normal drinking water, while the GLN1, GLN2 and GLN3 groups were provided with water supplemented with 0.4%, 0.8% and 1.2% Gln, respectively. At d7, birds in groups CS, GLN1, GLN2, and GLN3 were stressed at 12 ± 1 ℃ for 12 h. The results showed that cold stress significantly decreased the growth performance, serum antioxidant capacity and antibody concentrations, small intestine villus structure, and increased the gene expression of intestinal inflammatory factors of broiler chicks compared with the CON group (P < 0.05). Compared with the CS group, Gln supplementation exhibited increased growth performance, serum antioxidant capacity and antibody concentrations, gene expression levels of intestinal tight junction protein, villus height and villus height to crypt depth ratio (V/C) of small intestine, and decreased mRNA expression level of intestinal inflammatory factors (P < 0.05). CONCLUSIONS Gln supplementation ameliorated the impact of cold stress to a large extent as it promoted the development of the intestine and immune system and enhanced the antioxidant enzyme system in cold-stressed prestarter chicks.
Collapse
Affiliation(s)
- Juanjuan Wu
- College of Animal Science and Technology, Jiangxi Agriculture University, Nanchang City, 330045, Jiangxi Province, China
| | - Wenxin Qiu
- College of Animal Science and Technology, Jiangxi Agriculture University, Nanchang City, 330045, Jiangxi Province, China
| | - Guiyao Li
- College of Animal Science and Technology, Jiangxi Agriculture University, Nanchang City, 330045, Jiangxi Province, China
| | - Haoneng Guo
- College of Animal Science and Technology, Jiangxi Agriculture University, Nanchang City, 330045, Jiangxi Province, China
| | - Sifa Dai
- Department of Pharmaceutical and Life Sciences, Jiujiang University, Jiujiang City, 332005, Jiangxi Province, China
| | - Guanhong Li
- College of Animal Science and Technology, Jiangxi Agriculture University, Nanchang City, 330045, Jiangxi Province, China.
| |
Collapse
|
18
|
Alarcón‐Sánchez MA, Rodríguez‐Montaño R, Mosaddad SA, Heboyan A. Levels of IL-1β, MMP-8, and MMP-9 in the Saliva of Subjects With Periodontitis: A Systematic Review and Meta-Analysis. J Clin Lab Anal 2025; 39:e70040. [PMID: 40289477 PMCID: PMC12089797 DOI: 10.1002/jcla.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/23/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Proinflammatory cytokines and enzymes responsible for tissue destruction are important in the development of periodontitis. This study compared salivary concentrations of interleukin-1 beta (IL-1β), matrix metalloproteinases (MMP-8), and (MMP-9) in individuals with and without periodontitis to evaluate their diagnostic utility as potential biomarkers. MATERIALS AND METHODS A comprehensive search was performed across PubMed, Scopus, ScienceDirect, and Google Scholar, supplemented by manual searches in relevant journals up to January 2024. Eligibility criteria focused on human studies with defined diagnostic criteria for periodontitis and saliva samples analyzed for IL-1β, MMP-8, and MMP-9. Data were extracted to compare salivary levels of these markers between periodontitis patients and healthy controls. The Joanna Briggs Institute tool was used to evaluate the risk of bias and quality of the included studies. Statistical analysis employed a random effects model to calculate standardized mean differences and assess heterogeneity and publication bias. RESULTS The search yielded 122 articles, with 27 meeting the inclusion criteria. Fifteen percent of these studies presented a moderate risk of bias, while the remaining 85% exhibited a low risk of bias. The meta-analyses indicated significantly higher levels of IL-1β, MMP-8, and MMP-9 in the saliva of subjects with periodontitis compared to healthy individuals: IL-1β: Standardized Mean Difference (SMD) = 163.29 (95% CI = 104.64-221.95), p < 0.001; MMP-8: SMD = 282.22 (95% CI = 209.68-354.77), p < 0.001; MMP-9: SMD = 311.85 (95% CI = 179.64-444.05), p < 0.001. CONCLUSION Elevated salivary levels of IL-1β, MMP-8, and MMP-9 are linked to periodontitis.
Collapse
Affiliation(s)
- Mario Alberto Alarcón‐Sánchez
- Doctor of Science in Molecular Biology in Medicine Program, University Center of Health SciencesUniversity of Guadalajara (CUCS‐UdeG)GuadalajaraJaliscoMexico
- Institute of Research in Dentistry, Department of Integral Dental Clinics, University Center of Health SciencesUniversity of Guadalajara (CUCS‐UdeG)GuadalajaraJaliscoMexico
| | - Ruth Rodríguez‐Montaño
- Institute of Research in Dentistry, Department of Integral Dental Clinics, University Center of Health SciencesUniversity of Guadalajara (CUCS‐UdeG)GuadalajaraJaliscoMexico
- Department of Health and Illness as an Individual and Collective Process, University Center of TlajomulcoUniversity of Guadalajara (CUTLAJO‐UdeG)Tlajomulco de ZuñigaJaliscoMexico
| | - Seyed Ali Mosaddad
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Department of Conservative Dentistry and Bucofacial Prosthesis, Faculty of OdontologyComplutense University of MadridMadridSpain
- Department of Prosthodontics, School of DentistryShiraz University of Medical SciencesShirazIran
| | - Artak Heboyan
- Department of Prosthodontics, Faculty of StomatologyYerevan State Medical University after Mkhitar HeratsiYerevanArmenia
| |
Collapse
|
19
|
Zuo Z, Wang Y, Fang Y, Zhao M, Wang Z, Yang Z, Jia B, Sun Y. A novel regulator of NLRP3 inflammasome: Peptides. Peptides 2025; 187:171381. [PMID: 40064242 DOI: 10.1016/j.peptides.2025.171381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
The NLRP3 inflammasome plays a crucial role as a critical regulator of the immune response and has been implicated in the pathogenesis of numerous diseases. Peptides, known for their remarkable potency, selectivity, and low toxicity, have been extensively employed in disease treatment. Recent research has unveiled the potential of peptides in modulating the activity of the NLRP3 inflammasome. This review begins by examining the structure of the NLRP3 inflammasome, encompassing NLRP3, ASC, and Caspase-1, along with the three activation pathways: canonical, non-canonical, and alternative. Subsequently, we provide a comprehensive summary of peptide modulators targeting the NLRP3 inflammasome and elucidate their underlying mechanisms. The efficacy of these modulators has been validated through in vitro and in vivo experiments on NLRP3 inflammasome regulation. Furthermore, we conduct sequence alignment of the identified peptides and investigate their binding sites on the NLRP3 protein. This work is a foundational exploration for advancing peptides as potential therapeutic agents for NLRP3-related diseases.
Collapse
Affiliation(s)
- Zhuo Zuo
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yaxing Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yanwei Fang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Mengya Zhao
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhe Wang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhouqi Yang
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Bin Jia
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yulong Sun
- School of Life Sciences, Key Laboratory for Space Biosciences & Biotechnology, Institute of Special Environmental Biophysics, Research Center of Special Environmental Biomechanics and Medical Engineering, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
20
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025; 19:15189-15219. [PMID: 40249331 PMCID: PMC12045021 DOI: 10.1021/acsnano.5c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
21
|
Li JY, Chen HK, Huang YH, Zhi YP, Li YE, Lin KY, Chen C, Guo YS. Identification of mitophagy-related genes in patients with acute myocardial infarction. Hereditas 2025; 162:70. [PMID: 40287718 PMCID: PMC12034215 DOI: 10.1186/s41065-025-00424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Mitophagy is involved in acute myocardial infarction (AMI) process. However, the role of mitophagy-related genes (MRGs) in the AMI process is not well illustrated. We identified MRGs involved in AMI by bioinformatics analysis. The external datasets were employed for the validation of the MRGs, alongside the execution of cellular and animal experiments. Forty-five MRGs were detected, and machine learning identified the top four hub genes, namely ALDH2, ACSL1, IL1B, and GABARAPL1. Additionally, an external validation set was used to screen for three diagnostic markers (ACSL1, IL1B, and GABARAPL1) among these hub genes. Immune infiltration analysis revealed changes in the immune microenvironment among patients with AMI. Finally, the significant upregulation of ACSL1, IL1B, and GABARAPL1 in both cellular and animal models was confirmed. The occurrence of mitophagy was observed in the cell model through transmission electron microscopy (TEM). Our study demonstrated that ACSL1, IL1B, and GABARAPL1 possess potential biomarkers for AMI.
Collapse
Affiliation(s)
- Ju-Ying Li
- The First People's Hospital of Yibin, Yibin, Sichuan Province, 644000, China
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Hong-Kui Chen
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Yi-Hao Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Yu-Peng Zhi
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Yue-E Li
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Kai-Yang Lin
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| | - Chun Chen
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350000, China.
| | - Yan-Song Guo
- Shenli Clincal Medical College of Fujian Medical University, Fuzhou, Fujian Province, 350000, China.
| |
Collapse
|
22
|
Luan H, Song Y, Hu H, Zhang W, Zhang H, Su T, Wang J, Ye G, Yin Z, Zhao X, Zhou X, Li L, Zou Y, Zhang Y, Song X. Resveratrol exerts antiviral activity against pseudorabies virus through regulation of the OPN-ERK/JNK-IL-1β signaling axis. J Proteomics 2025; 317:105444. [PMID: 40274095 DOI: 10.1016/j.jprot.2025.105444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025]
Abstract
Pseudorabies virus (PRV) can infect most mammals and has caused significant economic losses in global pig production. The emergence of new mutants significantly reduces the protective effect of vaccination, indicating an urgent need for the development of specific therapeutic agents against PRV infection. In this study, we analyzed the changes in the cellular proteome after PRV infection in resveratrol-treated PK-15 cells using TMT quantitative proteomics combined with LC-MS/MS. The results identified the differential proteins osteopontin (iOPN) and interleukin-1 receptor accessory protein (IL-1RAP), which have significant biological implications. The regulation of OPN-IL-1β signaling by PRV infection was further studied through the OPN-ERK/JNK-IL-1β signaling axis. The transcriptional levels of OPN, C-JUN, IL-1RAP, and IL-1β, along with the protein levels of ERK, JNK, C-Jun, and their phosphorylated forms at 8, 12, and 16 h post-infection, were determined. The results showed that PRV infection inhibited the activation of this signaling axis, which was upregulated by resveratrol treatment. Down-regulation of OPN by siRNA increased PRV proliferation and inhibited the activation of the signaling axis, which was antagonized by resveratrol treatment. In PRV-infected mice, resveratrol treatment produced the same changes observed in vitro. The present study demonstrated that resveratrol can promote innate immune responses by regulating the OPN-ERK/JNK-IL-1β signaling axis, thereby activating host antiviral defenses against PRV infection. SIGNIFICANCE: Resveratrol targets the OPN-ERK/JNK-IL-1β axis to enhance innate immunity, offering a novel antiviral strategy against PRV infection. This study identifies OPN as a key regulator of host defense, linking ERK/JNK signaling to IL-1β-mediated antiviral responses. In vivo validation demonstrates resveratrol's therapeutic potential, reducing PRV replication and mortality in mice via immune pathway activation.
Collapse
Affiliation(s)
- Hongliang Luan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Qilu Animal Health Products Co., Ltd, Jinan 250100, China
| | - Yizhen Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongqiao Hu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wenrui Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Tianli Su
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Juan Wang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinhong Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xun Zhou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yingying Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China. @sicau.edu.cn
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
23
|
Larcombe AN, Chivers EK, Landwehr KR, Berry LJ, de Jong E, Huxley RR, Musk A, Franklin PJ, Mullins BJ. Partial amelioration of a chronic cigarette-smoke-induced phenotype in mice by switching to electronic cigarettes. Arch Toxicol 2025:10.1007/s00204-025-04055-7. [PMID: 40249508 DOI: 10.1007/s00204-025-04055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Electronic cigarettes ("e-cigarettes") are often marketed as smoking cessation tools and are used by smokers to reduce/quit cigarette smoking. The objective of this study was to assess the health effects of switching to e-cigarettes after long-term smoking in a mouse model and compare these effects with continued smoking, or quitting entirely. Adult BALB/c mice were whole-body exposed to mainstream cigarette smoke (2 h/day, 5 days/week) for 12 weeks prior to switching to flavoured e-cigarette aerosol (50:50 propylene glycol and glycerine) containing 18 mg/mL nicotine (2 h/day and 5 days/week), continuing cigarette smoking (2 h/day and 5 days/week), or quitting entirely for an additional 2 weeks. We then assessed a range of respiratory health outcomes including lung function and structure, pulmonary inflammation and changes in gene expression in the lung. Switching to e-cigarettes led to improvements in some aspects of respiratory health in mice compared with continued smoking, such as reduced neutrophilic inflammation in the lung. However, total cellular lung inflammation was still elevated and lung function was still impaired, in terms of airway responsiveness to methacholine, for e-cigarette use compared with quitting. Larger effects were typically seen in female mice compared to male. This study shows that switching to e-cigarettes after long-term cigarette smoking leads to improvements in some aspects of respiratory health, such as neutrophilic inflammation and the volume dependence of lung function compared with continued smoking. However, switching to e-cigarettes was not as effective as quitting smoking entirely.
Collapse
Affiliation(s)
- Alexander N Larcombe
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia.
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia.
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| | - Emily K Chivers
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
| | - Katherine R Landwehr
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia
| | - Luke J Berry
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
| | - Emma de Jong
- Centre for Health Research, The Kids Research Institute Australia, The University of Western Australia, Perth, WA, Australia
| | - Rachel R Huxley
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
- Faculty of Health, Deakin University, Geelong, VIC, Australia
| | - Arthur Musk
- School of Population and Global Health, University of Western Australia, Perth, WA, Australia
| | - Peter J Franklin
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, 15 Hospital Avenue, Nedlands, Perth, WA, 6009, Australia
- School of Population and Global Health, University of Western Australia, Perth, WA, Australia
| | - Benjamin J Mullins
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA, Australia
| |
Collapse
|
24
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liping Yang
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
25
|
Guerrero GG, Madrid-Marina V, Martínez-Romero A, Torres-Poveda K, Favela-Hernández JM. Host-Pathogen Interaction Interface: Promising Candidate Targets for Vaccine-Induced Protective and Memory Immune Responses. Vaccines (Basel) 2025; 13:418. [PMID: 40333316 PMCID: PMC12031405 DOI: 10.3390/vaccines13040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccine formulations are a successful strategy against pathogen transmission because vaccine candidates induce effective and long-lasting memory immune responses (B and CD4+ T cells) at systemic and mucosal sites. Extracellular vesicles of lipoproteins, bioactive compounds from plants and invertebrates (sponges) encapsulated in liposomes, and glycoproteins can target these sites. The vaccine candidates developed can mimic microbial pathogens in a way that successfully links the innate and adaptive immune responses. In addition, vaccines plus adjuvants promote and maintain an inflammatory response. In this review, we aimed to identify the host-pathogen interface as a rich source of candidate targets for vaccine-induced protective and long-lasting memory immune responses.
Collapse
Affiliation(s)
- Gloria G. Guerrero
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas, Zacatecas 98600, Zac., Mexico
| | - Vicente Madrid-Marina
- Centro de Investigación en Enfermedades infecciosas (CISEI), Instituto Nacional de Salud Pública (INSP), Cuernavaca 62100, Mor., Mexico
| | - Aurora Martínez-Romero
- Facultad de Química, Universidad Juárez del Estado de Durango, Gómez Palacio 34100, Dgo., Mexico
| | - Kirvis Torres-Poveda
- Centro de Investigación en Enfermedades infecciosas (CISEI), Instituto Nacional de Salud Pública (INSP), Cuernavaca 62100, Mor., Mexico
- Secretaria de Ciencia, Humanidades y Tecnologías (SECIHTI), Instituto Nacional de Salud Pública, Cuernavaca 62100, Mor., Mexico
| | - Juan Manuel Favela-Hernández
- Facultad de Química, Universidad Juárez del Estado de Durango, Gómez Palacio 34100, Dgo., Mexico
- Instituto Multidisciplinario de Ciencias “Avicena”, Torreón 27250, Coah., Mexico
| |
Collapse
|
26
|
Khattab MSA, Haiqiang L, Shaoxun T, Qiongxian Y, Yong L, Zhiliang T, Qi L. Immunological response enhancement in cows with subclinical mastitis fed diet supplemented with Macleaya cordata. Front Vet Sci 2025; 12:1503487. [PMID: 40303387 PMCID: PMC12037520 DOI: 10.3389/fvets.2025.1503487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction The present study explored the immune response, milk production and health status of mastitis-infected lactating cows fed diets supplemented with Macleaya cordata extract. Methods Twenty-four Holstein and Jersey cows were equally assigned to two experimental groups: the first group was fed a control diet (control), and the second experimental group was fed a control diet plus Macleaya extract at 8 g/head/d (Macleaya). The experiment was conducted for 60 days. The daily milk yield was recorded, and the milk samples were analyzed for total solids, fat, protein, and lactose contents. Results Blood samples were analyzed for different blood constituents, biochemical parameters, antioxidant capacity and immune indices. Compared with the control, the addition of Macleaya improved immune indices (p < 0.05). No significant differences (p > 0.05) were recorded between the two groups for different rumen liquor parameters, antioxidant capacities, milk yields or compositions. However, supplementing the diet with Macleaya significantly decreased SCC, SAA, and endotoxin. Conclusion This study suggested that supplementing the diets of lactating cows with Macleaya extract potentially improved their immune competence without adversely impacting their productive performance.
Collapse
Affiliation(s)
- Mostafa S. A. Khattab
- State Key Laboratory of Forage Breeding-by-Design and Utilization, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- Dairy Department, National Research Centre, Giza, Egypt
| | - Li Haiqiang
- State Key Laboratory of Forage Breeding-by-Design and Utilization, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Animal Science, Guizhou University, Guiyang, China
| | - Tang Shaoxun
- State Key Laboratory of Forage Breeding-by-Design and Utilization, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yan Qiongxian
- State Key Laboratory of Forage Breeding-by-Design and Utilization, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Liu Yong
- State Key Laboratory of Forage Breeding-by-Design and Utilization, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Tan Zhiliang
- State Key Laboratory of Forage Breeding-by-Design and Utilization, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Lu Qi
- College of Animal Science, Guizhou University, Guiyang, China
| |
Collapse
|
27
|
Borim PA, Gatto M, Mota GAF, Meirelles ALB, dos Santos ACC, Pagan LU, Ojopi EPB, Rodrigues EA, Souza LM, Damatto FC, Oliveira LRDS, Zornoff LAM, Okoshi K, Okoshi MP. Nlrc4 Inflammasome Expression After Acute Myocardial Infarction in Rats. Int J Mol Sci 2025; 26:3697. [PMID: 40332346 PMCID: PMC12028149 DOI: 10.3390/ijms26083697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Acute myocardial necrosis activates the immune response and inflammatory processes. Although the initial response is helpful in restoring tissue injury, dysregulated and exacerbated inflammation contributes to the progression of cardiac remodeling. Inflammasomes play important roles in post-infarction inflammation. NALP1/NLRP1, NLRP 3, and NLRC4 are the best-known inflammasomes. NLRP3, which has received the most study in cardiovascular disease, has been linked to increased IL-1β (IL1B) production and caspase-1 activity, as well as impaired cardiac function. The role of NLRP1 and NLRC4 inflammasomes after acute myocardial infarction (MI) is poorly understood. We evaluated the expression of myocardial inflammasomes and inflammatory markers 72 h after MI in rats. Male Wistar rats were divided into Sham (n = 15) and MI (n = 16) groups. MI was induced by ligating the left anterior descending coronary artery. Infarct size was assessed by histology. Myocardial protein and gene expression was analyzed by Western blot and RT-qPCR, respectively. IL-1β (Il1b) concentrations in serum and heart macerate supernatant were evaluated by ELISA. Statistical analysis was performed using Student's t test. Rats with an MI size less than 30% of the total left ventricle (LV) area were excluded; infarct size was 46 ± 11% of the total LV area in MI. The interstitial collagen fraction was higher in MI. Nlrc4, caspase-1 (Casp1), and IL-1β (Il1b) protein expressions were higher in MI. Nlrp3, Nlrp1, ASC (Pycard), pro-caspase-1, and pro-IL-1β (Il1b) expressions did not differ between groups. Expression of the Nlrp3 and ASC (Pycard) genes, as well as myocardial and serum IL-1β (Il1b) concentrations, was higher in MI. Acute post-myocardial infarction inflammation is characterized by increased protein expression of Nlrc4, caspase-1, and interleukin-1β; increased gene expression of Nlrp3 and ASC (Pycard); and elevated serum and myocardial concentrations of interleukin-1β in combination with an increased myocardial collagen interstitial fraction.
Collapse
Affiliation(s)
- Patricia Aparecida Borim
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Mariana Gatto
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Gustavo Augusto Ferreira Mota
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Ana Luiza Barioni Meirelles
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Anna Clara Consorti dos Santos
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Luana Urbano Pagan
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Elida Paula Benquique Ojopi
- Clinic Hospital, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
| | - Eder Anderson Rodrigues
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Lidiane Moreira Souza
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Felipe Cesar Damatto
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Leiliane Rodrigues dos Santos Oliveira
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Leonardo Antonio Mamede Zornoff
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Katashi Okoshi
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Marina Politi Okoshi
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| |
Collapse
|
28
|
Jiang W, Pang X, Ha P, Li C, Chang GX, Zhang Y, Bossong LA, Ting K, Soo C, Zheng Z. Fibromodulin selectively accelerates myofibroblast apoptosis in cutaneous wounds by enhancing interleukin 1β signaling. Nat Commun 2025; 16:3499. [PMID: 40221432 PMCID: PMC11993684 DOI: 10.1038/s41467-025-58906-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Activated myofibroblasts deposit extracellular matrix material to facilitate rapid wound closure that can heal scarlessly during fetal development. However, adult myofibroblasts exhibit a relatively long life and persistent function, resulting in scarring. Thus, understanding how fetal and adult tissue regeneration differs may serve to identify factors that promote more optimal wound healing in adults with little or less scarring. We previously found that matricellular proteoglycan fibromodulin is one such factor promoting more optimal repair, but the underlying molecular and cellular mechanisms for these effects have not been fully elucidated. Here, we find that fibromodulin induces myofibroblast apoptosis after wound closure to reduce scarring in small and large animal models. Mechanistically, fibromodulin accelerates and prolongs the formation of the interleukin 1β-interleukin 1 receptor type 1-interleukin 1 receptor accessory protein ternary complex to increase the apoptosis of myofibroblasts and keloid- and hypertrophic scar-derived cells. As the persistence of myofibroblasts during tissue regeneration is a key cause of fibrosis in most organs, fibromodulin represents a promising, broad-spectrum anti-fibrotic therapeutic.
Collapse
Affiliation(s)
- Wenlu Jiang
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral, Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Pin Ha
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Grace Xinlian Chang
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuxin Zhang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral, Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Lawrence A Bossong
- Department of Neuroscience, Princeton University, Princeton, NJ, 08540, USA
| | - Kang Ting
- American Dental Association Forsyth Institute, Cambridge, MA, 02142, USA.
- School of Dentistry, National Yang-Ming Chiao Tung University, Taipei, 30010, Taiwan.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Zhong Zheng
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
29
|
Lee JJ, Yang L, Kotzin JJ, Ahimovic D, Bale MJ, Nigrovic PA, Josefowicz SZ, Mathis D, Benoist C. Early transcriptional effects of inflammatory cytokines reveal highly redundant cytokine networks. J Exp Med 2025; 222:e20241207. [PMID: 39873673 PMCID: PMC11865922 DOI: 10.1084/jem.20241207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/25/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
Inflammatory cytokines are fundamental mediators of the organismal response to injury, infection, or other harmful stimuli. To elucidate the early and mostly direct transcriptional signatures of inflammatory cytokines, we profiled all immunologic cell types by RNAseq after systemic exposure to IL1β, IL6, and TNFα. Our results revealed a significant overlap in the responses, with broad divergence between myeloid and lymphoid cells, but with very few cell-type-specific responses. Pathway and motif analysis identified several main controllers (NF-κB, IRF8, and PU.1), but the largest portion of the response appears to be mediated by MYC, which was also implicated in the response to γc cytokines. Indeed, inflammatory and γc cytokines elicited surprisingly similar responses (∼50% overlap in NK cells). Significant overlap with interferon-induced responses was observed, paradoxically in lymphoid but not myeloid cell types. These results point to a highly redundant cytokine network, with intertwined effects between disparate cytokines and cell types.
Collapse
Affiliation(s)
- Juliana J. Lee
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Liang Yang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Jonathan J. Kotzin
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dughan Ahimovic
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Michael J. Bale
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Peter A. Nigrovic
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven Z. Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
30
|
Yeshna, Singh M, Monika, Kumar A, Garg V, Jhawat V. Pathophysiology and emerging therapeutic strategies for cervical spondylosis: The role of pro-inflammatory mediators, kinase inhibitors, and Organogel based drug delivery systems. Int Immunopharmacol 2025; 151:114350. [PMID: 40010157 DOI: 10.1016/j.intimp.2025.114350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Cervical spondylosis is a prevalent ailment characterized by chronic wear and degenerative changes affecting the cervical spine, leading to various clinical syndromes such as axial neck pain, cervical myelopathy, and cervical radiculopathy. The pathophysiology of the development of cervical alterations is multifaceted, with alterations in the normal physiology and pathogenesis of intervertebral disc degeneration. The involvement of pro-inflammatory mediators, such as interleukin-1, tumor necrosis factor-α, interleukin-4, interleukin-6, and interleukin-10, in the pathological processes associated with intervertebral disc degeneration offers potential therapeutic targets. The review also introduces kinase inhibitors as potential treatments for cervical spondylosis. Protein kinase inhibitors, including mitogen-activated protein kinase (MAPK), Janus kinase (JAK), and spleen tyrosine kinase (SYK), are explored for their anti-inflammatory properties. The article discusses their potential in modulating inflammatory signaling cascades and presents them as attractive candidates for treating immune-mediated disorders. Inhibitors of Nuclear Factor-κB, p38 MAPK, Jun-N terminal kinase (JNK), and Extracellular signal-regulated kinase (ERK) have shown efficacy in suppressing inflammatory responses, offering potential avenues for intervention in this prevalent condition. Organogels are semi-solid materials formed by trapping an organic solvent within a three-dimensional cross-linked network. They hold considerable potential in drug delivery, especially in enhancing drug solubility, facilitating controlled release, and improving skin penetration. These properties of organogels can help treat or alleviate the symptoms of cervical spondylosis.
Collapse
Affiliation(s)
- Yeshna
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika Singh
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Ashok Kumar
- Faculty of Pharmacy, Kalinga University, Naya Raipur, Chhattisgarh, India
| | - Vandana Garg
- Department of Pharmaceutical Science, MD University, Rohtak, India
| | - Vikas Jhawat
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India.
| |
Collapse
|
31
|
Davoodi Karsalari P, Asna Ashari K, Rezaei N. NLRP3 inflammasome: significance and potential therapeutic targets to advance solid organ transplantation. Expert Opin Ther Targets 2025; 29:281-301. [PMID: 40317257 DOI: 10.1080/14728222.2025.2500425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome, integral to innate immunity, has become a pivotal figure in the inflammatory cascade. AREAS COVERED This article provides an overview of the NLRP3 inflammasome, reviewing its complicated structure, as well as the diverse signals that trigger its assembly. Furthermore, we explored the intricate relationship between the NLRP3 inflammasome and acute and chronic rejection in solid organ transplantation. Solid organ transplantation stands as a crucial medical intervention, yet its efficacy is challenged by immune-mediated complications, including acute rejection, ischemia-reperfusion injury, and chronic allograft rejection. We also investigated the encouraging potential of immunosuppressive therapies targeting NLRP3 signaling to alleviate inflammatory responses linked to transplantation. EXPERT OPINION In recent years, the NLRP3 inflammasome has garnered considerable attention owing to its critical functions spanning diverse fields. This study highlights the critical function of the NLRP3 inflammasome and presents insights, offering fresh perspectives on how its modulation might help to improve the outcomes among patients who undergo solid organ transplantations.
Collapse
Affiliation(s)
- Pershia Davoodi Karsalari
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Kosar Asna Ashari
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Pediatrics, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Ji G, Feng X, Hu C, Zhang J, Sheng H, Na R, Li F, Wang Y, Ma Y, Cai B, Ma Y. HADHA promotes apoptosis and inflammatory response in bovine endometrial epithelial cells by regulating transcription and metabolism. Int J Biol Macromol 2025; 304:140980. [PMID: 39952496 DOI: 10.1016/j.ijbiomac.2025.140980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Endometritis in dairy cows significantly impacts their reproductive performance. However, its underlying mechanisms remain unclear. Hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit-alpha (HADHA) is known to regulate the occurrence of various diseases, but its role in bovine endometritis is poorly understood. In the present study, an in vitro bovine endometrial epithelial cells (BEECs) inflammation model was constructed to explore the effects of HADHA on inflammation, proliferation, and apoptosis. Functional analyses based on HADHA interference and overexpression revealed that it positively regulated the expression of IL-6, IL-8, and IL-1β in lipopolysaccharide (LPS)-induced BEECs, enhancing reactive oxygen species (ROS) production and promoting inflammation. Concurrently, HADHA decreased the expression of PCNA, CDK2, and CDK4, inhibited mitotic transition of BEECs from S to G2 phase, and negatively regulated BEEC proliferation. It also increased BAX and Caspase-3 expression while decreasing BCL2 expression, hence promoting BEECs apoptosis. Transcriptomic and metabolomic analyses indicated that HADHA modulated inflammation in BEECs by affecting pathways such as the TGF-beta signaling pathway, fatty acid metabolism, and p53 signaling. These findings provide novel insights into HADHA's role in bovine endometritis and reveal future research directions on its regulatory mechanisms.
Collapse
Affiliation(s)
- Guoshang Ji
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Xue Feng
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Chunli Hu
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Junxing Zhang
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Hui Sheng
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381,China
| | - Rina Na
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Fen Li
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Yachun Wang
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, National Engineering Laboratory of Animal Breeding, State Key Laboratory of Farm Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yanfen Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Bei Cai
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Yun Ma
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
33
|
Alqudah A, Qnais E, Gammoh O, Bseiso Y, Wedyan M, Alqudah M, Aljabali AAA, Tambuwala M. Exploring Scopoletin's Therapeutic Efficacy in DSS-Induced Ulcerative Colitis: Insights into Inflammatory Pathways, Immune Modulation, and Microbial Dynamics. Inflammation 2025; 48:575-589. [PMID: 38918333 PMCID: PMC12053357 DOI: 10.1007/s10753-024-02048-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 06/27/2024]
Abstract
This study aimed to investigate the therapeutic potential of scopoletin in ulcerative colitis, with a primary focus on its impact on crucial inflammatory pathways and immune responses. A male mouse model of DSS-induced colitis was employed with six distinct groups: a control group, a group subjected to DSS only, three groups treated with varying scopoletin doses, and the final group treated with dexamethasone. The investigation included an assessment of the effects of scopoletin on colitis symptoms, including alterations in body weight, Disease Activity Index (DAI), and histopathological changes in colonic tissue. Furthermore, this study scrutinized the influence of scopoletin on cytokine production, PPARγ and NF-κB expression, NLRP3 inflammasome, and the composition of intestinal bacteria. Scopoletin treatment yielded noteworthy improvements in DSS-induced colitis in mice, as evidenced by reduced weight loss and colonic shortening (p < 0.05, < 0.01, respectively). It effectively diminished TNF-α, IL-1β, and IL-12 cytokine levels (p < 0.01, p < 0.05), attenuated NLRP3 inflammasome activation and the associated cytokine release (p < 0.05, p < 0.01), and modulated the immune response by elevating PPARγ expression while suppressing NF-κB pathway activation (p < 0.05, p < 0.01). Additionally, scopoletin induced alterations in the gut microbiota composition, augmenting beneficial Lactobacillus and Bifidobacteria while reducing E. coli (p < 0.05). It also enhanced tight junction proteins, signifying an improvement in the intestinal barrier integrity (p < 0.05, < 0.01). Scopoletin is a promising therapeutic agent for managing ulcerative colitis, showing benefits that extend beyond mere anti-inflammatory actions to encompass regulatory effects on gut microbiota and restoration of intestinal integrity.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Yousra Bseiso
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammad Alqudah
- Physiology Department, School of Medicine and Biomedical Sciences, Arabian Gulf University, Manama, Bahrain
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Murtaza Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
| |
Collapse
|
34
|
Singh V, Ubaid S, Kashif M, Singh T, Singh G, Pahwa R, Singh A. Role of inflammasomes in cancer immunity: mechanisms and therapeutic potential. J Exp Clin Cancer Res 2025; 44:109. [PMID: 40155968 PMCID: PMC11954315 DOI: 10.1186/s13046-025-03366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025] Open
Abstract
Inflammasomes are multi-protein complexes that detect pathogenic and damage-associated molecular patterns, activating caspase-1, pyroptosis, and the maturation of pro-inflammatory cytokines such as IL-1β and IL-18Within the tumor microenvironment, inflammasomes like NLRP3 play critical roles in cancer initiation, promotion, and progression. Their activation influences the crosstalk between innate and adaptive immunity by modulating immune cell recruitment, cytokine secretion, and T-cell differentiation. While inflammasomes can contribute to tumor growth and metastasis through chronic inflammation, their components also present novel therapeutic targets. Several inhibitors targeting inflammasome components- such as sensor proteins (e.g., NLRP3, AIM2), adaptor proteins (e.g., ASC), caspase-1, and downstream cytokines- are being explored to modulate inflammasome activity. These therapeutic strategies aim to modulate inflammasome activity to enhance anti-tumor immune responses and improve clinical outcomes. Understanding the role of inflammasomes in cancer immunity is crucial for developing interventions that effectively bridge innate and adaptive immune responses for better therapeutic outcomes.
Collapse
Affiliation(s)
- Vivek Singh
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Saba Ubaid
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Mohammad Kashif
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tanvi Singh
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Gaurav Singh
- Department of Biochemistry, King George'S Medical University (KGMU), U.P, Lucknow, 226003, India
| | - Roma Pahwa
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anand Singh
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
35
|
Rodríguez-Fernández MA, Tristán-Flores FE, Casique-Aguirre D, Negrete-Rodríguez MDLLX, Cervantes-Montelongo JA, Conde-Barajas E, Acosta-García G, Silva-Martínez GA. Virtual Screening and Molecular Dynamics of Cytokine-Drug Complexes for Atherosclerosis Therapy. Int J Mol Sci 2025; 26:2931. [PMID: 40243563 PMCID: PMC11988346 DOI: 10.3390/ijms26072931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease remains the leading global cause of mortality, largely driven by atherosclerosis, a chronic inflammatory condition characterized by lipid accumulation and immune-cell infiltration in arterial walls. Macrophages play a central role by forming foam cells and secreting pro-atherogenic cytokines, such as TNF-α, IFN-γ, and IL-1β, which destabilize atherosclerotic plaques, expanding the lipid core and increasing the risk of thrombosis and ischemia. Despite the significant health burden of subclinical atherosclerosis, few targeted therapies exist. Current treatments, including monoclonal antibodies, are limited by high costs and immunosuppressive side effects, underscoring the urgent need for alternative therapeutic strategies. In this study, we employed in silico drug repositioning to identify multitarget inhibitors against TNF-α, IFN-γ, and IL-1β, leveraging a virtual screening of 2750 FDA-approved drugs followed by molecular dynamics simulations to assess the stability of selected cytokine-ligand complexes. This computational approach provides structural insights into potential inhibitors. Additionally, we highlight nutraceutical options, such as fatty acids (oleic, linoleic and eicosapentaenoic acid), which exhibited strong and stable interactions with key cytokine targets. Our study suggests that these bioactive compounds could serve as effective new therapeutic approaches for atherosclerosis.
Collapse
Affiliation(s)
- María Angélica Rodríguez-Fernández
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
| | - Fabiola Estefanía Tristán-Flores
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ciencias Básicas, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico
| | - Diana Casique-Aguirre
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Delegación Puebla, Puebla 06600, Mexico;
- Secretaría de Ciencia, Humanidades, Tecnología e Innovación (SECIHTI), Ciudad de México 03940, Mexico
| | - María de la Luz Xochilt Negrete-Rodríguez
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| | - Juan Antonio Cervantes-Montelongo
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
- Escuela de Medicina, Universidad de Celaya, Celaya 38080, Guanajuato, Mexico
| | - Eloy Conde-Barajas
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| | - Gerardo Acosta-García
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| | - Guillermo Antonio Silva-Martínez
- Posgrado de Ingeniería Bioquímica, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico; (M.A.R.-F.); (F.E.T.-F.); (M.d.l.L.X.N.-R.); (E.C.-B.); (G.A.-G.)
- Secretaría de Ciencia, Humanidades, Tecnología e Innovación (SECIHTI), Ciudad de México 03940, Mexico
- Departamento de Ingeniería Bioquímica y Ambiental, Tecnológico Nacional de México/IT de Celaya, Celaya 38010, Guanajuato, Mexico;
| |
Collapse
|
36
|
Zakaria N, Menze ET, Elsherbiny DA, Tadros MG, George MY. Lycopene mitigates paclitaxel-induced cognitive impairment in mice; Insights into Nrf2/HO-1, NF-κB/NLRP3, and GRP-78/ATF-6 axes. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111262. [PMID: 39848561 DOI: 10.1016/j.pnpbp.2025.111262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Chemotherapy-induced cognitive impairment, referred to as "chemobrain", is widely acknowledged as a significant adverse effect of cancer therapy. Paclitaxel, a chemotherapeutic drug, has been reported to cause cognitive impairment clinically and in animal models. However, the precise mechanisms are not fully understood. The current study explored the potential neuroprotective effect of lycopene in paclitaxel-induced cognitive impairment in mice and its potential underlying mechanisms. Mice were randomly allocated into six groups: control, paclitaxel-treated (10 mg/kg), lycopene-treated (5, 10, and 20 mg/kg) + paclitaxel, and lycopene alone-treated (20 mg/kg) groups. The effect of lycopene treatment on behavioral function and histological examination was assessed. Lycopene (20 mg/kg) was selected for additional investigation into the underlying mechanisms. Lycopene treatment counteracted paclitaxel-induced oxidative stress by reducing lipid peroxidation and enhancing catalase levels. Additionally, lycopene-treated mice demonstrated a significant elevation in nuclear factor erythroid 2-related factor 2 with no significant effect on hemeoxygenase-1. Moreover, paclitaxel administration elevated endoplasmic reticulum stress markers; glucose-regulated protein78, activating Transcription Factor 6, C/EBP homologous protein, and apoptosis marker annexin V which were significantly reduced by lycopene treatment. Furthermore, lycopene mitigated paclitaxel-induced neuroinflammation through the reduction of the levels of the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome axis markers; nuclear factor-κB, NLRP3, caspase-1, interleukin-1β, and interleukin-18. Our study findings may provide new evidence that lycopene mitigates paclitaxel-induced cognitive impairment in mice by reversing oxidative stress, endoplasmic reticulum stress, and inflammatory mechanisms.
Collapse
Affiliation(s)
- Nora Zakaria
- Armed Forces Medical Complex- Kobry El-Qobba, Ministry of Defense, Kobry El-Qobba, Cairo 11766, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| |
Collapse
|
37
|
Matveyenka M, Sholukh M, Kurouski D. Cytotoxicity of Amyloid β1-42 Fibrils to Brain Immune Cells. ACS Chem Neurosci 2025; 16:1144-1149. [PMID: 40056144 PMCID: PMC11926782 DOI: 10.1021/acschemneuro.4c00835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/01/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive pathology that is linked to abrupt aggregation of amyloid β1-42 (Aβ1-42) peptide in the central nervous system. Aβ1-42 aggregation yields amyloid oligomers and fibrils, toxic protein aggregates that cause progressive neuronal degeneration in the frontal lobe of the brain. Although neurons remain the focus of AD for decades, a growing body of evidence suggests that the degeneration of immune cells in the brain can be the major cause of AD. However, the extent to which Aβ1-42 aggregates are toxic to the major classes of immune cells in the brain remains unclear. In the current study, we examine the cytotoxic effects of Aβ1-42 fibrils on macrophages, dendritic cells, and microglia. These cells play vitally important roles in development and homeostasis of the central nervous system. We found that Aβ1-42 fibrils caused calcium release and enhanced levels of reactive oxygen species in macrophages, dendritic cells, and microglia as well as neurons. We also investigated the extent to which the lysozymes of these immune cells could alter the aggregation properties of Aβ1-42. Our results showed that lysosomes extracted from macrophages, dendritic cells, and microglia drastically accelerated Aβ1-42 aggregation as well as altered cytotoxicity of these protein aggregates. These results indicate that impairment of immune cells in the brain can be a critically important aspect of neurodegenerative processes that are taking place upon the onset of AD.
Collapse
Affiliation(s)
- Mikhail Matveyenka
- Department
of Biochemistry and Biophysics, Texas A&M
University, College
Station, Texas 77843, United States
| | - Mikhail Sholukh
- Department
of Biology, Belarussian State University, Minsk 220030, Belarus
| | - Dmitry Kurouski
- Department
of Biochemistry and Biophysics, Texas A&M
University, College
Station, Texas 77843, United States
| |
Collapse
|
38
|
Beesetti S. Ubiquitin Ligases in Control: Regulating NLRP3 Inflammasome Activation. FRONT BIOSCI-LANDMRK 2025; 30:25970. [PMID: 40152367 DOI: 10.31083/fbl25970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 03/29/2025]
Abstract
Ubiquitin ligases play pivotal roles in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, a critical process in innate immunity and inflammatory responses. This review explores the intricate mechanisms by which various E3 ubiquitin ligases exert both positive and negative influences on NLRP3 inflammasome activity through diverse post-translational modifications. Negative regulation of NLRP3 inflammasome assembly is mediated by several E3 ligases, including F-box and leucine-rich repeat protein 2 (FBXL2), tripartite motif-containing protein 31 (TRIM31), and Casitas B-lineage lymphoma b (Cbl-b), which induce K48-linked ubiquitination of NLRP3, targeting it for proteasomal degradation. Membrane-associated RING-CH 7 (MARCH7) similarly promotes K48-linked ubiquitination leading to autophagic degradation, while RING finger protein (RNF125) induces K63-linked ubiquitination to modulate NLRP3 function. Ariadne homolog 2 (ARIH2) targets the nucleotide-binding domain (NBD) domain of NLRP3, inhibiting its activation, and tripartite motif-containing protein (TRIM65) employs dual K48 and K63-linked ubiquitination to suppress inflammasome assembly. Conversely, Pellino2 exemplifies a positive regulator, promoting NLRP3 inflammasome activation through K63-linked ubiquitination. Additionally, ubiquitin ligases influence other components critical for inflammasome function. TNF receptor-associated factor 3 (TRAF3) mediates K63 polyubiquitination of apoptosis-associated speck-like protein containing a CARD (ASC), facilitating its degradation, while E3 ligases regulate caspase-1 activation and DEAH-box helicase 33 (DHX33)-NLRP3 complex formation through specific ubiquitination events. Beyond direct inflammasome regulation, ubiquitin ligases impact broader innate immune signaling pathways, modulating pattern-recognition receptor responses and dendritic cell maturation. Furthermore, they intricately control NOD1/NOD2 signaling through K63-linked polyubiquitination of receptor-interacting protein 2 (RIP2), crucial for nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) activation. Furthermore, we explore how various pathogens, including bacteria, viruses, and parasites, have evolved sophisticated strategies to hijack the host ubiquitination machinery, manipulating NLRP3 inflammasome activation to evade immune responses. This comprehensive analysis provides insights into the molecular mechanisms underlying inflammasome regulation and their implications for inflammatory diseases, offering potential avenues for therapeutic interventions targeting the NLRP3 inflammasome. In conclusion, ubiquitin ligases emerge as key regulators of NLRP3 inflammasome activation, exhibiting a complex array of functions that finely tune immune responses. Understanding these regulatory mechanisms not only sheds light on fundamental aspects of inflammation but also offers potential therapeutic avenues for inflammatory disorders and infectious diseases.
Collapse
Affiliation(s)
- Swarna Beesetti
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
39
|
Fahey DL, Patel N, Watford WT. TPL2 kinase activity is required for Il1b transcription during LPS priming but dispensable for NLRP3 inflammasome activation. Front Immunol 2025; 16:1496613. [PMID: 40170849 PMCID: PMC11958189 DOI: 10.3389/fimmu.2025.1496613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
The NLRP3 inflammasome complex is an important mechanism for regulating the release of pro-inflammatory cytokines, IL-1β and IL-18, in response to harmful pathogens. Overproduction of pro-inflammatory cytokines has been linked to cryopyrin-associated periodic syndrome, arthritis, and other inflammatory conditions. It has been previously shown that tumor progression locus 2, a serine-threonine kinase, promotes IL-1β synthesis in response to LPS stimulation; however, whether TPL2 kinase activity is required during inflammasome priming to promote Il1b mRNA transcription and/or during inflammasome activation for IL-1β secretion remained unknown. In addition, whether elevated type I interferons, a consequence of either Tpl2 genetic ablation or inhibition of TPL2 kinase activity, decreases IL-1β expression or inflammasome function has not been explored. Using LPS-stimulated primary murine bone marrow-derived macrophages, we determined that TPL2 kinase activity is required for transcription of Il1b, but not Nlrp3, Il18, caspase-1 (Casp1), or gasdermin-D (Gsdmd) during inflammasome priming. Both Casp1 and Gsdmd mRNA synthesis decreased in the absence of type I interferon signaling, evidence of crosstalk between type I interferons and the inflammasome. Our results demonstrate that TPL2 kinase activity is differentially required for the expression of inflammasome precursor cytokines and components but is dispensable for inflammasome activation. These data provide the foundation for the further exploration of TPL2 kinase inhibitor as a potential therapeutic in inflammatory diseases.
Collapse
Affiliation(s)
- Denise L. Fahey
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Niki Patel
- College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Wendy T. Watford
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
40
|
Feng A, Gonzalez MV, Kalaycioglu M, Yin X, Mumau M, Shyamsundar S, Bustamante MS, Chang SE, Dhingra S, Dodig-Crnkovic T, Schwenk JM, Garg T, Yoshizaki K, van Rhee F, Fajgenbaum DC, Utz PJ. Common connective tissue disorder and anti-cytokine autoantibodies are enriched in idiopathic multicentric castleman disease patients. Front Immunol 2025; 16:1528465. [PMID: 40181993 PMCID: PMC11966032 DOI: 10.3389/fimmu.2025.1528465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Idiopathic Multicentric Castleman Disease (iMCD) is a polyclonal lymphoproliferative disorder involving cytokine storms that can lead to organ failure and death. The cause of iMCD is unknown, but some clinical evidence suggests an autoimmune etiology. For example, connective tissue disorders (CTDs) and iMCD share many clinical features, and autoantibodies have been anecdotally reported in individual iMCD patients. This study investigates whether common autoantibodies are shared across iMCD patients. Methods We assembled custom bead-based protein arrays consisting of 52 autoantigens traditionally associated with CTDs and 38 full-length cytokines and screened serum samples from 101 iMCD patients for IgG autoantibodies. We also screened samples with a 1,103-plex array of recombinant human protein fragments to identify additional autoantibody targets. Finally, we performed receptor blocking assays on select samples with anti-cytokine autoantibodies (ACAs) identified by array. Results We found that an increased proportion of iMCD patients (47%) tested positive for at least one CTD-associated autoantibody compared to healthy controls (HC) (17%). Commonly detected CTD-associated autoantibodies were associated with myositis and overlap syndromes as well as systemic lupus erythematosus (SLE) and Sjögren's Syndrome (SS). ACAs were also detected in a greater proportion of iMCD patients (38%) compared to HC (10%), while the protein fragment array identified a variety of other autoantibody targets. One iMCD sample tested positive for receptor blocking against interferon-ω (IFNω). Discussion IgG autoantibodies binding autoantigens associated with common CTDs and cytokines are elevated in iMCD patients compared to HC, suggesting that autoimmunity may be involved in iMCD pathogenesis.
Collapse
Affiliation(s)
- Allan Feng
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael V. Gonzalez
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Muge Kalaycioglu
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Xihui Yin
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Melanie Mumau
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Saishravan Shyamsundar
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Mateo Sarmiento Bustamante
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah E. Chang
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaurya Dhingra
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Tea Dodig-Crnkovic
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jochen M. Schwenk
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tarun Garg
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Kazuyuki Yoshizaki
- Department of Biomolecular Science and Regulation, The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Frits van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - David C. Fajgenbaum
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Paul J. Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
41
|
Boonhok R, Senghoi W, Sangkanu S, Lim CL, Pudla M, Pereira MDL, Wilairatana P, Mahboob T, Rahman MA, Utaisincharoen P, Hiransai P, Nissapatorn V. Acanthamoeba castellanii-Mediated Reduction of Interleukin-1β Secretion and Its Association With Macrophage Autophagy. SCIENTIFICA 2025; 2025:3430892. [PMID: 40109888 PMCID: PMC11922611 DOI: 10.1155/sci5/3430892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Noncanonical autophagy including unconventional protein secretion has been extensively studied. Our work focused on a leaderless IL-1β protein secretion from human macrophage in response to Acanthamoeba castellanii components, Acanthamoeba culture supernatant (CS) and cell lysate (CL), as well as its association with macrophage autophagy. Phorbol 12-myristate 13-acetate (PMA)-induced THP-1 macrophages were treated with Acanthamoeba components of pathogenic (ATCC50739) and nonpathogenic (ATCC30010) strains in vitro. The data showed that Acanthamoeba treatment resulted in low IL-1β secretion from macrophages. In addition, Acanthamoeba CL of both strains was able to upregulate autophagy-related (Atg) protein 8, an autophagy marker, whereas Acanthamoeba CS downregulated Atg8 expression. We further manipulated autophagy and found that autophagy induction by starvation diminished IL-1β secretion while autophagy inhibition by 3-methyladenine (3MA) increased IL-1β secretion. Interestingly, in the presence of Acanthamoeba components either under starvation or 3MA treatment, IL-1β secretion was significantly reduced. Transcriptional expression of other ATG genes, i.e., ATG6, ATG7, and ATG5, were investigated and showed that their mRNA expression was maintained at the basal level under A. castellanii CS or CL treatment. Inflammasome-related genes, NLRP3 and CASPASE1, were upregulated following A. castellanii 50739 CS treatment but not in A. castellanii 50739 CL-treated condition. However, both conditions were able to increase IL-1β mRNA expression. TEM micrographs revealed that 3MA treatment induced the formation of large vacuoles and accumulation of autophagosome at the edge of THP-1 macrophages. However, the number and size of their structures were declined in the presence of A. castellanii 50739 CS with 3MA. Furthermore, immunofluorescence staining demonstrated the association between Atg8/LC3 and IL-1β expression, where downregulation of Atg8 by A. castellanii 50739 CS led to the upregulation of IL-1β. Altogether, the data indicate that Acanthamoeba can manipulate macrophage autophagy, thereby controlling low IL-1β secretion. The expression of autophagy- and inflammasome-related genes also indicates multiple mechanisms in IL-1β secretion in response to Acanthamoeba components. However, further characterization of Atg proteins and investigations into other intracellular pathways or defense mechanisms are needed to fully understand the unconventional secretion of IL-1β in macrophages. This knowledge could eventually lead to the development of innovative therapeutic strategies against Acanthamoeba infection by modulating autophagy or macrophage responses.
Collapse
Affiliation(s)
- Rachasak Boonhok
- Department of Medical Technology, School of Allied Health Sciences, and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Wilaiwan Senghoi
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence Research for Melioidosis and Microorganisms (CERMM), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Suthinee Sangkanu
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 90112, Thailand
| | - Chooi Ling Lim
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Matsayapan Pudla
- Department of Oral Microbiology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials and Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tooba Mahboob
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Pongsak Utaisincharoen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Poonsit Hiransai
- Department of Medical Technology, School of Allied Health Sciences, and Center of Excellence in Marijuana, Hemp, and Kratom, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team) and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
42
|
Abe K, Yokota S, Matsumoto S, Ujiie H, Kikuchi E, Satoh K, Ishisaki A, Chosa N. Proinflammatory cytokine-induced matrix metalloproteinase-9 expression in temporomandibular joint osteoarthritis is regulated by multiple intracellular mitogen-activated protein kinase pathways. J Oral Biosci 2025; 67:100609. [PMID: 39755166 DOI: 10.1016/j.job.2024.100609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
OBJECTIVES Temporomandibular joint (TMJ) osteoarthritis (OA) is an inflammatory disease that involves periarthritis of the TMJ and destruction of cartilage tissue in the mandibular condyle. However, the role of proinflammatory cytokines in the expression levels of matrix metalloproteinase (MMP) remains inconclusive. Thus, in this study, we aimed to investigate the effect of proinflammatory cytokines on the expression of MMPs. METHODS FLS1 cells (mouse TMJ-derived synovial cell line) were treated with tumor necrosis factor alpha (TNF-α) or interleukin (IL)-1β in the presence or absence of mitogen-activated protein kinase (MAPK) inhibitors. The mRNA expression levels of MMP-2 and MMP-9 were examined by reverse transcription-quantitative polymerase chain reaction. Additionally, the phosphorylation status of extracellular signal-regulated kinase (ERK)1/2 and p38 MAPK in the FLS1 cells treated with TNF-α or IL-1β was evaluated by performing western blotting analysis. RESULTS TNF-α and IL-1β significantly increased the expression of MMP-9 in the FLS1 cells; however, MMP-2 expression remained unaffected. Mitogen-activated protein kinase kinase (MEK) and p38 MAPK inhibitors significantly suppressed cytokine-induced MMP-9 upregulation. Conversely, Jun amino-terminal kinase (JNK) inhibitors further increased MMP-9 expression in the cells treated with TNF-α or IL-1β. Moreover, TNF-α and IL-1β enhanced ERK1/2 and p38 MAPK phosphorylation in the FLS1 cells. CONCLUSIONS TNF-α and IL-1β induced MMP-9 expression in the FLS1 cells via the MEK/ERK and p38 MAPK pathways and suppressed it via the JNK pathway. Thus, proinflammatory cytokines control MMP-9 expression in TMJ-OA by regulating multiple MAPK pathways.
Collapse
Affiliation(s)
- Karen Abe
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan; Division of Orthodontics, Department of Developmental Oral Health Science, Iwate Medical University School of Dentistry, Morioka, Iwate, 020-8505, Japan
| | - Seiji Yokota
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan
| | - Shikino Matsumoto
- Division of Orthodontics, Department of Developmental Oral Health Science, Iwate Medical University School of Dentistry, Morioka, Iwate, 020-8505, Japan
| | - Hayato Ujiie
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan
| | - Emiko Kikuchi
- Division of Orthodontics, Department of Developmental Oral Health Science, Iwate Medical University School of Dentistry, Morioka, Iwate, 020-8505, Japan
| | - Kazuro Satoh
- Division of Orthodontics, Department of Developmental Oral Health Science, Iwate Medical University School of Dentistry, Morioka, Iwate, 020-8505, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan
| | - Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate, 028-3694, Japan.
| |
Collapse
|
43
|
Koga T, Sato S, Furukawa K, Yamamoto H, Kawakami A. Investigating the impact of tocilizumab on serum cytokines concentrations in Japanese FMF patients: a sub-analysis of the NUH01FMF study. Immunol Med 2025; 48:70-77. [PMID: 39440870 DOI: 10.1080/25785826.2024.2418164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024] Open
Abstract
Familial Mediterranean Fever (FMF) is the most common hereditary autoinflammatory disease, characterized by recurrent fever, arthritis, rash, and serositis, and is caused by mutations in the MEFV gene coding for the pyrin protein. The primary treatment goal is to prevent acute attacks and minimize subclinical inflammation to avoid secondary amyloidosis with colchicine as the first-line treatment. However, 10-20% of patients are colchicine-resistant or intolerant. While the therapeutic potential of IL-6 inhibitors such as tocilizumab (TCZ) has been suggested, the detailed serum cytokine profiles after TCZ treatment in patients with FMF remain largely unexplored. This study focused on a sub-analysis of a clinical trial evaluating TCZ in patients with colchicine-resistant FMF (crFMF). We analyzed the serum cytokine profiles at 0, 2, 4, 8, 12, 16, 20, and 24 weeks in the TCZ and placebo groups. Our findings revealed a decrease in serum C-X-C motif chemokine ligand 1 and vascular endothelial growth factor levels in the TCZ group at week 4 compared to baseline, which persisted until week 24, indicating the potential of TCZ to manage crFMF by modulating specific inflammatory cytokines. Further research is required to confirm these findings and optimize the treatment strategies for FMF.
Collapse
Affiliation(s)
- Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Kaori Furukawa
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Hiroshi Yamamoto
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
44
|
Kodagoda YK, Hanchapola HACR, Rodrigo DCG, Lim C, Liyanage DS, Omeka WKM, Ganepola GANP, Dilshan MAH, Kim J, Lee JH, Jeong T, Wan Q, Kim G, Lee J. Expression profiling and functional role of cyclooxygenase-2 in the immune and inflammatory responses of red-spotted grouper (Epinephelus akaara). FISH & SHELLFISH IMMUNOLOGY 2025; 158:110158. [PMID: 39890039 DOI: 10.1016/j.fsi.2025.110158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/08/2025] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Cyclooxygenase-2 (Cox-2) is a well-studied enzyme and a significant medicinal target associated with various inflammatory disorders. However, its role in pathogen-induced inflammatory responses in fish remains poorly understood. This study characterized the structural and functional properties of a Cox-2 homolog from red-spotted grouper (Epinephelus akaara) (EaCox-2). The three-dimensional structure of EaCox-2 revealed a homodimer with two functional domains: a catalytic domain with two active sites and a membrane-binding domain. EaCox-2 transcripts were ubiquitously expressed in all tested tissues of E. akaara, with the highest expression in the gills, followed by the spleen. Immune stimulation with polyinosinic:polycytidylic acid (poly I:C), lipopolysaccharides (LPS), and nervous necrosis virus (NNV) led to significant upregulation in EaCox-2 transcripts 12 and 24 h post-injection in both gill and spleen tissues. EaCox-2 overexpression in murine macrophages triggered a pro-inflammatory response characterized by M1 macrophage polarization, upregulation of pro-inflammatory mediators such as TNF-α, IL-1β, and IL-6, and iNOS enzyme, enhanced production of reactive nitric oxide (NO), and mitochondrial depolarization. These findings highlight the crucial role of EaCox-2 in regulating immune and inflammatory responses in E. akaara, providing valuable insights into the molecular mechanisms underlying teleost immunity.
Collapse
Affiliation(s)
- Yasara Kavindi Kodagoda
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - H A C R Hanchapola
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - D C G Rodrigo
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Chaehyun Lim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - D S Liyanage
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - W K M Omeka
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - G A N P Ganepola
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - M A H Dilshan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jeongeun Kim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Ji Hun Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Taehyug Jeong
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Qiang Wan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Gaeun Kim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
45
|
Johnson DE, Cui Z. Triggering Pyroptosis in Cancer. Biomolecules 2025; 15:348. [PMID: 40149884 PMCID: PMC11940180 DOI: 10.3390/biom15030348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Pyroptosis is an inflammatory programmed cell death recently identified as a crucial cellular process in various diseases, including cancers. Unlike other forms of cell death, canonical pyroptosis involves the specific cleavage of gasdermin by caspase-1, resulting in cell membrane damage and the release of the pro-inflammatory cytokines IL-1β and IL-18. Initially observed in innate immune cells responding to external pathogens or internal death signals, pyroptotic cell death has now been observed in numerous cell types. Recent studies have extensively explored different ways to trigger pyroptotic cell death in solid tumors, presenting a promising avenue for cancer treatment. This review outlines the mechanisms of both canonical and noncanonical pyroptosis pertinent to cancer and primarily focuses on various biomolecules that can induce pyroptosis in malignancies. This strategy aims not only to eliminate cancer cells but also to promote an improved tumor immune microenvironment. Furthermore, emerging research indicates that targeting pyroptotic pathways may improve the effectiveness of existing cancer treatments, making them more potent against resistant tumor types, offering new hope for overcoming treatment resistance in aggressive malignancies.
Collapse
Affiliation(s)
- Daniel E. Johnson
- Department of Otolaryngology—Head and Neck Surgery, University of California at San Francisco, San Francisco, CA 94143, USA;
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Zhibin Cui
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| |
Collapse
|
46
|
Nashtahosseini Z, Eslami M, Paraandavaji E, Haraj A, Dowlat BF, Hosseinzadeh E, Oksenych V, Naderian R. Cytokine Signaling in Diabetic Neuropathy: A Key Player in Peripheral Nerve Damage. Biomedicines 2025; 13:589. [PMID: 40149566 PMCID: PMC11940495 DOI: 10.3390/biomedicines13030589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a debilitating complication of diabetes mellitus, characterized by progressive nerve damage driven by chronic hyperglycemia and systemic inflammation. The pathophysiology of DPN is significantly influenced by pro-inflammatory cytokines, such as IL-1β, IL-6, and TNF-α. These cytokines promote oxidative stress, vascular dysfunction, and neuronal degeneration by activating important signaling pathways including NF-κB and MAPK. While IL-6 promotes a pro-inflammatory microenvironment, increasing neuronal damage and neuropathic pain, TNF-α and IL-1β worsen Schwann cell failure by compromising axonal support and causing demyelination. Immune cell infiltration and TLR activation increase the inflammatory cascade in DPN, resulting in a persistent neuroinflammatory state that sustains peripheral nerve injury. The main characteristics of DPN are axonal degeneration, decreased neurotrophic support, and Schwann cell dysfunction, which weaken nerve transmission and increase susceptibility to damage. Advanced glycation end-products, TNF-α, and CXCL10 are examples of biomarkers that may be used for early diagnosis and disease progression monitoring. Additionally, crucial molecular targets have been found using proteomic and transcriptome techniques, enabling precision medicine for the treatment of DPN. This review emphasizes the importance of cytokine signaling in the pathogenesis of DPN and how cytokine-targeted treatments might reduce inflammation, restore nerve function, and improve clinical outcomes for diabetic patients.
Collapse
Affiliation(s)
| | - Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
| | - Elham Paraandavaji
- Clinical Research Development Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran 13399-73111, Iran
| | - Alireza Haraj
- Student Research Committee, Faculty of Medicine, Iran University of Medical Sciences, Tehran 14496-1453, Iran
| | - Bahram Fadaee Dowlat
- Faculty of Medicine, Iran University of Medical Sciences, Tehran 14496-1453, Iran
| | - Ehsan Hosseinzadeh
- Department of Surgery, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | | | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
47
|
M Bader S, Scherer L, Schaefer J, Cooney JP, Mackiewicz L, Dayton M, Georgy SR, Davidson KC, Allison CC, Herold MJ, Strasser A, Pellegrini M, Doerflinger M. IL-1β drives SARS-CoV-2-induced disease independently of the inflammasome and pyroptosis signalling. Cell Death Differ 2025:10.1038/s41418-025-01459-x. [PMID: 40016339 DOI: 10.1038/s41418-025-01459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 03/01/2025] Open
Abstract
Excessive inflammation and cytokine release are hallmarks of severe COVID-19. Certain programmed cell death processes can drive inflammation, however, their role in the pathogenesis of severe COVID-19 is unclear. Pyroptosis is a pro-inflammatory form of regulated cell death initiated by inflammasomes and executed by the pore-forming protein gasdermin D (GSDMD). Using an established mouse adapted SARS-CoV-2 virus and a panel of gene-targeted mice we found that deletion of the inflammasome (NLRP1/3 and the adaptor ASC) and pore forming proteins involved in pyroptosis (GSDMA/C/D/E) only marginally reduced IL-1β levels and did not impact disease outcome or viral loads. Furthermore, we found that SARS-CoV-2 infection did not trigger GSDMD activation in mouse lungs. Finally, we did not observe any difference between WT animals and mice with compound deficiencies in the pro-inflammatory initiator caspases (C1/11/12-/-). This indicates that the classical canonical and non-canonical pro-inflammatory caspases known to process and activate pro-IL-1β, pro-IL-18 and GSDMD do not substantially contribute to SARS-CoV-2 pathogenesis. However, the loss of IL-1β, but not the absence of IL-18, ameliorated disease and enhanced survival in SARS-CoV-2 infected animals compared to wildtype mice. Collectively, these findings demonstrate that IL-1β is an important factor contributing to severe SARS-CoV-2 disease, but its release was largely independent of inflammasome and pyroptotic pathways.
Collapse
Affiliation(s)
- Stefanie M Bader
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Lena Scherer
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Jan Schaefer
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - James P Cooney
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Liana Mackiewicz
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Merle Dayton
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Smitha Rose Georgy
- Anatomic Pathology-Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Werribee, VIC, 3030, Australia
| | - Kathryn C Davidson
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Cody C Allison
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Marc Pellegrini
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
- Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia.
| | - Marcel Doerflinger
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
48
|
Zhang Y, Liu J, Li M, Dong Y, Li Z, Yi D, Wu T, Wang L, Zhao D, Hou Y. Zinc Oxide Administration Relieves the Diarrhea of ETEC K88-Infected Piglets by Reducing Ileal Apoptosis and Maintaining Gut Microbial Balance. Vet Sci 2025; 12:115. [PMID: 40005874 PMCID: PMC11861302 DOI: 10.3390/vetsci12020115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
The impact of ZnO as a feed additive on growth-performance and intestinal function of Enterotoxigenic Escherichia coli (ETEC) K88-infected piglets remains unclear. Fecal scores of piglets in ETEC group were significantly increased compared to control group. ETEC K88 significantly damages the small intestine, including a reduction in villus height in the jejunum, duodenum, and ileum, and a decrease in total superoxide dismutase activity in the jejunum and catalase activity in the ileum and jejunum. Compared to control group, ETEC K88 infection significantly elevated the mRNA level of gene IL-1β and the level of ileal epithelial cell apoptosis. ZnO administration significantly alleviated these negative effects and improved the antioxidative capability of the ileum. Moreover, ZnO supplementation alleviated the imbalance of gut microbiota by restoring the reduced amount of Enterococcus and Lactobacillus in the jejunum, Clostridium in the ileum, and Lactobacillus in the cecum, as well as the increased amount of total eubacteria in the ileum and Enterococcus in the cecum induced by the ETEC K88 infection. In conclusion, ZnO administration can reduce the diarrhea of piglets infected with ETEC K88 by reducing the structural damage of the intestine, attenuating intestinal oxidative stress and epithelial cell apoptosis, and modulating the gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yongqing Hou
- Engineering Research Center of Feed Protein Resources on Agricultural By-Products, Ministry of Education, Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430024, China
| |
Collapse
|
49
|
Wu Q, Zeng Y, Geng K, Guo M, Teng FY, Yan PJ, Lei Y, Long Y, Jiang ZZ, Law BYK, Xu Y. The role of IL-1 family cytokines in diabetic cardiomyopathy. Metabolism 2025; 163:156083. [PMID: 39603339 DOI: 10.1016/j.metabol.2024.156083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Diabetic cardiomyopathy (DCM) is the primary cause of heart failure in patients with diabetes and is characterised by contractile dysfunction and left ventricular hypertrophy. The complex pathological and physiological mechanisms underlying DCM have contributed to a limited number of available treatment options. A substantial body of evidence has established that DCM is a low-grade inflammatory cardiovascular disorder, with the interleukin-1 (IL-1) family of cytokines playing crucial roles in initiating inflammatory responses and shaping innate and adaptive immunity. In this review, we aim to provide an overview of the underlying mechanisms of the IL-1 family and their relevance in DCM of various aetiologies. Furthermore, we highlighted potential therapeutic targets within the IL-1 family for the management of DCM.
Collapse
Affiliation(s)
- Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Pathology, and Luzhou Key Laboratory of Precision Pathology Diagnosis for Serious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kang Geng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Plastic and burns surgery, National Key Clinical Construction Specialty, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Man Guo
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fang-Yuan Teng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pi-Jun Yan
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yi Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yang Long
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China.
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
50
|
Drake LY, Roos BB, Wicher SA, Khalfaoui L, Nesbitt LL, Fang YH, Pabelick CM, Prakash YS. Aging, brain-derived neurotrophic factor, and allergen-induced pulmonary responses in mice. Am J Physiol Lung Cell Mol Physiol 2025; 328:L290-L300. [PMID: 39437757 DOI: 10.1152/ajplung.00145.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Asthma in the elderly is being recognized as more severe, resistant to standard therapies, and having greater morbidity. Therefore, it becomes important to understand the impact of aging-associated airway structure and functional changes toward pathogenesis of asthma in the elderly. Here, airway smooth muscle plays important roles in airway hyperreactivity and structural remodeling. The role of smooth muscle in asthma can be modulated by growth factors [including neurotrophins such as brain-derived neurotrophic factor (BDNF)] and proinflammatory senescence factors. In this study, we investigated aging effects on airway hyperreactivity, structural remodeling, inflammation, and senescence in a mouse model of allergic asthma. C57BL/6J wild-type mice or smooth muscle-specific BDNF knockout mice at 4, 18, and 24 mo of age were intranasally exposed to mixed allergens (MAs, ovalbumin, Aspergillus, Alternaria, and house dust mite) over 4 wk. Assessing lung function by flexiVent, we found that compared with 4-mo-old mice, 18- and 24-mo-old C57BL/6J mice showed decreased airway resistance and increased airway compliance after PBS or MA treatment. Deletion of smooth muscle BDNF blunted airway hyperreactivity in aged mice. Lung histology analysis revealed that aging increased bronchial airway thickness and decreased lung inflammation. Multiplex assays showed that aging largely reduced allergen-induced lung expression of proinflammatory chemokines and cytokines. By immunohistochemistry staining, we found that aging increased bronchial airway expression of senescence markers, including p21, phospho-p53, and phospho-γH2A.X. Our data suggest that aging-associated increase of airway senescence in the context of allergen exposure may contribute to asthma pathology in the elderly.NEW & NOTEWORTHY The pathogenesis of asthma in elderly is not well understood. Using a mouse model of asthma, we show that aging results in decreased lung function and less responsiveness to allergen exposure, impacted by locally produced brain-derived neurotrophic factor. Aging also decreases allergen-induced inflammation but increases airway remodeling and senescence. Our results suggest that senescence pathways may contribute to asthma pathogenesis in elderly.
Collapse
Affiliation(s)
- Li Y Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Benjamin B Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Sarah A Wicher
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Latifa Khalfaoui
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Lisa L Nesbitt
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Yun Hua Fang
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|