1
|
Hu C, Yuan F, Wu Y, Xiao S, Xu Y, Peng X, He L. Disruption of the caspase-1/IL-1β axis alleviates myocardial Ischemia/Reperfusion injury via improvement of mitochondrial homeostasis and reduction of Pyroptosis. Clin Exp Hypertens 2025; 47:2506619. [PMID: 40373207 DOI: 10.1080/10641963.2025.2506619] [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: 03/06/2025] [Revised: 04/28/2025] [Accepted: 05/09/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Pyroptosis is a novel kind of programmed cell death and Caspase-1 plays key roles in driving pyroptosis. The current study aims to elucidate the molecular mechanism affecting cardiomyocyte pyroptosis in myocardial ischemia/reperfusion (I/R) injury, both in vivo and in vitro. METHODS A murine model of myocardial I/R injury was established and then treated with lentivirus-mediated shRNA targeting Caspase-1 to evaluate the effect of Caspase-1 on myocardial I/R injury. Further, Caspase-1 was silenced in the cardiomyocytes following hypoxia-reoxygenation (H/R) to detect the function of Caspase-1 in mitochondrial homeostasis and cardiomyocyte pyroptosis. RESULTS Knockdown of Caspase-1 inhibited the secretion of interleukin-1 beta (IL-1β), improved cardiac dysfunction and decreased pyroptosis in vivo. The cardio-protective effect was verified in the H/R-induced cardiomyocyte model. Recombinant IL-1β protein reversed the inhibitory effect of Caspase-1 knockdown on pyroptosis. CONCLUSION Overall, activating the Caspase-1/IL-1β axis by myocardial I/R injury causes mitochondrial homeostasis imbalance, pyroptosis, and the consequent cardiomyocyte injury.
Collapse
Affiliation(s)
- ChenKai Hu
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - FengXia Yuan
- Department of Pharmacy, the Second Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, China
| | - YingXing Wu
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shan Xiao
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yuan Xu
- Medical Big Data Research Center, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiang Peng
- Information Department, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Lei He
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
2
|
Naidovski N, Chong SKT, Liu F, Riordan SM, Wehrhahn MC, Yuwono C, Zhang L. Human macrophage response to the emerging enteric pathogen Aeromonas veronii: Inflammation, apoptosis, and downregulation of histones. Virulence 2025; 16:2440554. [PMID: 39663607 PMCID: PMC11702953 DOI: 10.1080/21505594.2024.2440554] [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: 08/05/2024] [Revised: 11/11/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
This study investigated the pathogenic mechanisms of Aeromonas veronii in macrophages. THP-1 derived macrophages were used as a human macrophage model and were treated with A. veronii strain AS1 isolated from intestinal biopsies of an IBD patient, or Escherichia coli strain K-12. RNA was extracted and subjected to RNA sequencing and comparative transcriptomic analyses. Protein levels of IL-8, IL-1β, IL-18, and TNFα were measured using ELISA, and apoptosis was assessed using caspase 3/7 assays. Both A. veronii AS1 and E. coli K-12 significantly upregulated the expression of many genes involving inflammation. At the protein level, A. veronii AS1 induced significantly higher levels of IL-8, TNFα, mature IL-18 and IL-1β than E. coli K-12, and led to greater elevation of caspase 3/7 activities. Both A. veronii AS1 and E. coli K-12 upregulated the expression of CASP5, but not other caspase genes. A. veronii AS1 significantly downregulated the expression of 20 genes encoding histone proteins that E. coli K-12 did not. The more profound pathogenic effects of A. veronii in inducing inflammation and apoptosis in macrophages than E. coli K-12 are consistent with its role as a human enteric pathogen. The upregulated expression of CASP5 and increased release of IL-1β and IL-18 support the role of CASP5 in activation of non-canonical inflammasome. The downregulation of histone genes by A. veronii suggests a unique impact on host cell gene expression, which may represent a novel virulence strategy. These findings advance the understanding of pathogenic mechanisms of the emerging human enteric pathogen A. veronii.
Collapse
Affiliation(s)
- Nicholas Naidovski
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Sarah K. T. Chong
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Fang Liu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Stephen M. Riordan
- Gastrointestinal and Liver Unit, Prince of Wales Hospital, University of New South Wales, Sydney, Australia
| | - Michael C. Wehrhahn
- Douglass Hanly Moir Pathology, a Sonic Healthcare Practice, Macquarie Park, NSW, Australia
| | - Christopher Yuwono
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
3
|
Fior Ribeiro G, Priest EL, Heaney H, Richardson JP, Childers DS. Mannan is a context-dependent shield that modifies virulence in Nakaseomyces glabratus. Virulence 2025; 16:2491650. [PMID: 40233931 PMCID: PMC12001547 DOI: 10.1080/21505594.2025.2491650] [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: 07/25/2024] [Revised: 02/17/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
Fungal-host interaction outcomes are influenced by how the host recognizes fungal cell wall components. Mannan is a major cell wall carbohydrate and can be a glycoshield that blocks the inner cell wall β-1,3-glucan from activating pro-inflammatory immune responses. Disturbing this glycoshield in Candida albicans results in enhanced antifungal host responses and reduced fungal virulence. However, deletions affecting mannan synthesis can lead to systemic hypervirulence for Nakaseomyces glabratus (formerly Candida glabrata) suggesting that proper mannan architecture dampens virulence for this organism. N. glabratus is the second leading cause of invasive and superficial candidiasis, but little is known about how the cell wall affects N. glabratus pathogenesis. In order to better understand the importance of these species-specific cell wall adaptations in infection, we set out to investigate how the mannan polymerase II complex gene, MNN10, contributes to N. glabratus cell wall architecture, immune recognition, and virulence in reference strains BG2 and CBS138. mnn10Δ cells had thinner inner and outer cell wall layers and elevated mannan, chitin, and β-1,3-glucan exposure compared to wild-type cells. Consistent with these observations, mnn10Δ cells activated the β-1,3-glucan receptor in oral epithelial cells (OECs), EphA2, and caused less OEC damage than wild-type. mnn10Δ replication was also restricted in macrophages compared to wild-type controls. Yet, during systemic infection in Galleria mellonella larvae, mnn10Δ cells induced rapid larval melanization and BG2 mnn10Δ cells killed larvae significantly faster than wild-type. Thus, our data suggest that mannan plays context-dependent roles in N. glabratus pathogenesis, acting as a glycoshield in superficial disease models and modulating virulence during systemic infection.
Collapse
Affiliation(s)
- Gabriela Fior Ribeiro
- Institute of Medical Sciences, Aberdeen Fungal Group, University of Aberdeen, Aberdeen, UK
| | - Emily L. Priest
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
| | - Helen Heaney
- Institute of Medical Sciences, Aberdeen Fungal Group, University of Aberdeen, Aberdeen, UK
| | - Jonathan P. Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
| | - Delma S. Childers
- Institute of Medical Sciences, Aberdeen Fungal Group, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
4
|
de Brito Monteiro L, Archambault AS, Starchuk LF, Alcazar A, Oh JH, Dubland JA, Rakić B, Patterson AE, Verchere CB, Klein Geltink RI. Inhibition of xanthine oxidoreductase with febuxostat, but not allopurinol, prevents inflammasome assembly and IL-1β release. Life Sci Alliance 2025; 8:e202403191. [PMID: 40399065 PMCID: PMC12095928 DOI: 10.26508/lsa.202403191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025] Open
Abstract
Xanthine oxidoreductase (XOR) inhibitors are used to treat gout, inhibiting uric acid production, which causes clinical symptoms. Commonly used XOR inhibitors are the small molecule febuxostat (Fbx) and the purine analogue allopurinol (Allo). Recent studies show that XOR inhibitors can reduce mature interleukin (IL)-1β production by activated macrophages. This effect is not due to reduced uric acid crystal formation, which can induce NLRP3 inflammasome activation, but an independent effect. Fbx and Allo have been used interchangeably in in vitro studies to highlight the role of XOR in pro-inflammatory macrophage function. Here, we analysed the effects of Fbx and Allo on pro-inflammatory macrophage signatures. Both XOR inhibitors maintain pro-inflammatory macrophage metabolic and phenotypic hallmarks. However, only Fbx reduces the activity of caspase-1 and the release of IL-1β by preventing inflammasome assembly in macrophages isolated from both mice and humans. Our study identified an Fbx-specific reduction in IL-1β production, which could be used clinically to reduce the deleterious effects of macrophage-derived IL-1β.
Collapse
Affiliation(s)
- Lauar de Brito Monteiro
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Surgery, Vancouver, Canada
| | - Anne-Sophie Archambault
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, Canada
| | - Lucas F Starchuk
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, Canada
| | - Armando Alcazar
- University of British Columbia, Department of Biochemistry and Molecular Biology, Vancouver, Canada
| | - Ju Hee Oh
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, Canada
| | - Joshua A Dubland
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, Canada
- University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Bojana Rakić
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, Canada
| | - Annette E Patterson
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, Canada
| | - C Bruce Verchere
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Surgery, Vancouver, Canada
- University of British Columbia, Centre for Molecular Medicine and Therapeutics, Vancouver, Canada
| | - Ramon I Klein Geltink
- BC Children's Hospital Research Institute, Vancouver, Canada
- University of British Columbia, Department of Pathology and Laboratory Medicine, Vancouver, Canada
- University of British Columbia, Edwin S.H. Leong Centre for Healthy Aging, Vancouver, Canada
- BC Cancer Research Centre, Department of Basic and Translational Research, Vancouver, Canada
| |
Collapse
|
5
|
Chao P, Zhang X, Zhang L, Han Z, Jie R, Duan P, Cao M, Yang A. Electroacupuncture as a promising therapeutic strategy for doxorubicin-induced heart failure: Insights into the PI3K/AKT/mTOR/ULK1 and AMPK /mTOR /ULK1 pathways. Colloids Surf B Biointerfaces 2025; 251:114590. [PMID: 40024111 DOI: 10.1016/j.colsurfb.2025.114590] [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: 02/07/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Electroacupuncture (EA), a traditional Chinese medicine therapy, exhibits cardioprotective and therapeutic effects against cardiac injury. However, the precise mechanisms underlying these benefits remain unclear. PURPOSE The aim of this study is to examine the impact of EA on Doxorubicin-Induced heart failure and elucidate the mechanisms involved. METHODS C57BL/6 mice were randomly assigned to six experimental groups, including a control group, a DCM group, a DCM group receiving non-acupoint EA (NEA), and a DCM group receiving acupoint EA (EA). The cardiac function, levels of inflammatory factors, and markers of apoptosis were assessed both in vivo and in vitro. The presence of AMPK/mTOR/ULK1(Ser317) and PI3K/AKT/mTOR/ULK1(Ser757) was confirmed. RESULTS EA stimulation significantly improved cardiac function, as evidenced by increased left ventricular ejection fraction (LVEF), E/A ratio, and fractional shortening (FS%) compared to the DCM group (p < 0.05). After EA stimulation, the phosphorylation levels of PI3K/AKT increase, leading to elevated expression of mTOR/ULK1(Ser757), which ultimately inhibited the expression of apoptosis-related proteins and inflammatory factors. Simultaneously, EA stimulation could inhibit the phosphorylation levels of AMPK, reducing the expression of mTOR/ULK1(Ser317), and thereby also inhibiting the expression of apoptosis-related proteins and inflammatory factors. CONCLUSIONS This study showed that EA stimulation can counteract myocardial damage caused by apoptosis and inflammation, thereby significantly improving cardiac function and prognosis in HF mice. The mechanism may be that EA stimulation activates the PI3K/AKT/mTOR/ULK1(ser757) pathway and inhibits the AMPK/ULK1(ser317) pathway. EA stimulation exerts the same effect by regulating these two pathways in different directions, ultimately reducing myocardial cell apoptosis and cardiac inflammation.
Collapse
Affiliation(s)
- Peng Chao
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xueqin Zhang
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Lei Zhang
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Zhengyang Han
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Runda Jie
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Pingxiu Duan
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Min Cao
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Aiping Yang
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
6
|
Spunde K, Korotkaja K, Sominskaya I, Zajakina A. Genetic adjuvants: A paradigm shift in vaccine development and immune modulation. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102536. [PMID: 40336572 PMCID: PMC12056970 DOI: 10.1016/j.omtn.2025.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
The COVID-19 pandemic underscored the urgency of developing effective vaccines to combat infectious diseases, especially in vulnerable populations such as the elderly and immunocompromised. While recombinant protein vaccines offer safety, their poor immunogenicity highlights the need for advanced vaccination platforms. New genetic/nucleic acid vaccine formulations like plasmid DNA and mRNA showed efficiency and safety in preclinical and clinical studies; however, they demand innovative adjuvants because their mechanism of action differs from traditional protein vaccines. Genetic adjuvants-encoded by nucleic acids within DNA, RNA, or viral vectors-emerge as a promising solution by targeting and modulating specific immune pathways, including antigen presentation, T cell activation, and memory formation. These innovative adjuvants enhance vaccine efficacy by fine-tuning innate and adaptive immune responses, overcoming immune senescence, and addressing the challenges of CD8+ T cell activation in immunocompromised populations. This review explores the potential of genetically encoded adjuvants, including cytokines, chemokines, and other immune modulators. By comparing these adjuvants to traditional formulations, we highlight their capacity to address the limitations of modern vaccines while discussing their integration with emerging technologies like RNA-based vaccines. As genetic adjuvants advance toward clinical application, understanding their mechanisms and optimizing their delivery is pivotal to unlocking next-generation immunization strategies.
Collapse
Affiliation(s)
- Karina Spunde
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Ksenija Korotkaja
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Irina Sominskaya
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| | - Anna Zajakina
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k. 1, LV-1067 Riga, Latvia
| |
Collapse
|
7
|
Lippincott MJ, Tomkinson J, Bunten D, Mohammadi M, Kastl J, Knop J, Schwandner R, Huang J, Ongo G, Robichaud N, Dagher M, Mansilla-Soto A, Saravia-Estrada C, Tsuboi M, Basualto-Alarcón C, Way GP. A morphology and secretome map of pyroptosis. Mol Biol Cell 2025; 36:ar63. [PMID: 40202832 DOI: 10.1091/mbc.e25-03-0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Pyroptosis represents one type of programmed cell death. It is a form of inflammatory cell death that is canonically defined by caspase-1 cleavage and Gasdermin-mediated membrane pore formation. Caspase-1 initiates the inflammatory response (through IL-1β processing), and the N-terminal cleaved fragment of Gasdermin D polymerizes at the cell periphery forming pores to secrete proinflammatory markers. Cell morphology also changes in pyroptosis, with nuclear condensation and membrane rupture. However, recent research challenges canon, revealing a more complex secretome and morphological response in pyroptosis, including overlapping molecular characterization with other forms of cell death, such as apoptosis. Here, we take a multimodal, systems biology approach to characterize pyroptosis. We treated human peripheral blood mononuclear cells (PBMCs) with 36 different combinations of stimuli to induce pyroptosis or apoptosis. We applied both secretome profiling (nELISA) and high-content fluorescence microscopy (Cell Painting). To differentiate apoptotic, pyroptotic, and control cells, we used canonical secretome markers and modified our Cell Painting assay to mark the N-terminus of Gasdermin D. We trained hundreds of machine learning (ML) models to reveal intricate morphology signatures of pyroptosis that implicate changes across many different organelles and predict levels of many proinflammatory markers. Overall, our analysis provides a detailed map of pyroptosis which includes overlapping and distinct connections with apoptosis revealed through a mechanistic link between cell morphology and cell secretome.
Collapse
Affiliation(s)
- Michael J Lippincott
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Jenna Tomkinson
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Dave Bunten
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| | | | | | | | | | | | - Grant Ongo
- Nomic Bio, Montreal, Québec, Canada H2T 1C1
| | | | | | | | | | - Masafumi Tsuboi
- Department of Chemistry and Biotechnology, University of Tokyo, Tokyo, Japan 113-0033
| | - Carla Basualto-Alarcón
- Health Sciences Department, University of Aysén, Coyhaique, Chile
- Anatomy and Legal Medicine Department, University of Chile, Santiago, Chile
| | - Gregory P Way
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
8
|
Zhang S, Qian Y, Li N, Zhu Q, Zhang S, Wen P, Xiao Y, Yan C, Lin Z, Zhong J, Ma J, Wu X, Zhuang G, Zhang K. Specific MSI2 deletion maintains intestinal barrier integrity by down-regulating ILC3s-derived IL-17 a in mice with colitis. Int Immunopharmacol 2025; 156:114717. [PMID: 40279942 DOI: 10.1016/j.intimp.2025.114717] [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: 02/12/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory bowel disease with an unknown cause. Previous studies have shown that Group 3 innate lymphoid cells (ILC3s) are crucial for maintaining intestinal mucosal immune homeostasis by producing key cytokines such as IL-22 and IL-17 A. While the RNA-binding protein Musashi-2 (MSI2) is recognized as essential for promoting intestinal epithelial regeneration post-injury, its impact on immune regulation remains unclear. Therefore, we aim to investigate the protective mechanisms associated with ILC3s-specific MSI2 deletion in a mouse model of ulcerative colitis. METHODS Dextran sulfate sodium (DSS) was used to induce a mouse colitis model. Colitis severity was evaluated through weight loss, diarrhea, fecal traits, colon length, and pathological scoring. Transcriptome sequencing was utilized to identify differentially expressed genes in colon tissues. Flow cytometry was employed to measure the quantity and functionality of ILC3s. Western blot was conducted to analyze protein expression, while real-time polymerase chain reaction and enzyme-linked immunosorbent assay were employed to quantify inflammatory factors. Additionally, immunofluorescence, AB-PAS staining, and immunohistochemistry were employed to evaluate the integrity of the intestinal barrier. RESULTS Following DSS treatment, colon damage was milder in Msi2∆Rorc mice than in Msi2fl/fl mice. Transcriptomic analysis revealed the down-regulation of cytokines and pro-inflammatory factors in the colon tissue of Msi2∆Rorc mice. Flow cytometry showed that specific deletion of MSI2 reduced the infiltration of ILC3s in the intestinal lamina propria of Msi2∆Rorc mice and decreased IL-17 A production. The reduction of IL-17 A-mediated immune responses lessened inflammatory damage to the intestinal barrier, thereby reducing colitis severity. CONCLUSIONS Specific deletion of MSI2 alleviates DSS-induced colitis in mice by reducing ILC3s infiltration and IL-17 A secretion in the lamina propria of the colon. This decrease in inflammatory mediators and cell infiltration dampens the inflammatory response in the intestinal mucosa, helping to maintain the integrity of the intestinal barrier in mice with colitis. These findings enhance our understanding of UC pathogenesis and offer novel avenues for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Shuaishuai Zhang
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Yunyun Qian
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Nengneng Li
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Qiang Zhu
- Department of General Surgery, First General Hospital of Fuzhou, Fujian Medical University, 350005 Fuzhou, Fujian, China
| | - Shiying Zhang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Peizhen Wen
- Department of General Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, 200003 Shanghai, China
| | - Yi Xiao
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Changxiu Yan
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Zeyang Lin
- Department of Pathology, Zhongshan Hospital, Xiamen University, 361001 Xiamen, Fujian, China
| | - Jianfa Zhong
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Jingmiao Ma
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Xia Wu
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Guohong Zhuang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China.
| | - Kun Zhang
- Department of General Surgery, First General Hospital of Fuzhou, Fujian Medical University, 350005 Fuzhou, Fujian, China.
| |
Collapse
|
9
|
Arnesdotter E, Stoffels CBA, Alker W, Gutleb AC, Serchi T. Per- and polyfluoroalkyl substances (PFAS): immunotoxicity at the primary sites of exposure. Crit Rev Toxicol 2025:1-21. [PMID: 40400477 DOI: 10.1080/10408444.2025.2501420] [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: 01/09/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/23/2025]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are persistent synthetic chemicals widely used in industrial and consumer products, leading to environmental contamination and human exposure. This review focuses on perfluoroalkyl acids, a subset of PFAS, which are primarily encountered through diet, including drinking water, and other pathways such as dust ingestion, and dermal contact. Impaired vaccine antibody response has been identified as the most critical effect for risk assessment by the European Food Safety Authority. Furthermore, human epidemiological studies have linked exposure to certain PFAS to various immune-related outcomes, such as asthma, allergies, and inflammatory bowel disease. This review examines potential immunomodulatory effects of perfluoroalkyl acids at the primary sites of exposure: lungs, intestines, and skin, using human epidemiological data as the basis for investigating these impacts. While animal studies are referenced for context, this paper highlights the need for further human-based research to address key questions about PFAS and their immunological impacts. The state of in vitro toxicity testing related to these effects is thoroughly reviewed and critical issues pertaining to this topic are discussed.
Collapse
Affiliation(s)
- Emma Arnesdotter
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology, Esch-sur-Alzette, Luxembourg
| | - Charlotte B A Stoffels
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology, Esch-sur-Alzette, Luxembourg
| | - Wiebke Alker
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Arno C Gutleb
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology, Esch-sur-Alzette, Luxembourg
| | - Tommaso Serchi
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
10
|
Yang B, Rutkowski N, Ruta A, Gray-Gaillard E, Maestas DR, Kelly SH, Krishnan K, Wu X, Wu S, Chen A, Mejías JC, Hooks JST, Vanderzee I, Mensah P, Celik N, Eric M, Abraham P, Tam A, Housseau F, Pardoll DM, Sears CL, Elisseeff JH. Murine gut microbiota dysbiosis via enteric infection modulates the foreign body response to a distal biomaterial implant. Proc Natl Acad Sci U S A 2025; 122:e2422169122. [PMID: 40354538 DOI: 10.1073/pnas.2422169122] [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: 11/26/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
The gut microbiota influences systemic immunity and the function of distal tissues, including the brain, liver, skin, lung, and muscle. However, the role of the gut microbiota in the foreign body response and fibrosis is largely unexplored. To investigate this connection, we perturbed the homeostasis of the murine gut microbiota via infection with the pathogenic bacterial species enterotoxigenic Bacteroides fragilis (ETBF) and implanted particulate material (mean particle size <600 μm) of the synthetic polymer polycaprolactone (PCL) into a distal muscle injury. ETBF infection in mice led to increased neutrophil and γδ T cell infiltration into the PCL implant site. ETBF infection alone promoted systemic inflammation, increased levels of neutrophils in lymphoid tissues, and altered skeletal muscle gene expression. At the PCL implant site, we found significant changes in the transcriptome of sorted stromal cells between infected and control mice, including differences related to ECM components such as proteoglycans and glycosaminoglycans. However, we did not observe ETBF-induced differences in fibrosis levels. These results demonstrate the ability of the gut microbiota to mediate long-distance effects such as immune and stromal responses to a distal biomaterial implant.
Collapse
Affiliation(s)
- Brenda Yang
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Natalie Rutkowski
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Anna Ruta
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Elise Gray-Gaillard
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - David R Maestas
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Sean H Kelly
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Kavita Krishnan
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Xinqun Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Shaoguang Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Allen Chen
- Department of Biomedical Engineering, Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218
| | - Joscelyn C Mejías
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Joshua S T Hooks
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Isabel Vanderzee
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Patricia Mensah
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Nazmiye Celik
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Marie Eric
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Peter Abraham
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Ada Tam
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Franck Housseau
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Drew M Pardoll
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Cynthia L Sears
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287
| | - Jennifer H Elisseeff
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| |
Collapse
|
11
|
Kaur J, Sharma V, Khan H, Singh S, Singh TG. Intersecting molecular pathways in Synucleinopathies and Amyloidogenesis: Exploring shared mechanisms and therapeutic potential. Brain Res 2025; 1855:149568. [PMID: 40090446 DOI: 10.1016/j.brainres.2025.149568] [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: 01/16/2025] [Revised: 02/21/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
Synucleinopathies and amyloidogenic disorders are the two most prevalent neurodegenerative conditions, characterized by progressive loss of neurons and aggregation of proteins in the central nervous system. Emerging evidence suggests that despite their distinct pathological hallmarks: α-synuclein in Parkinson's disease (PD) and amyloid-β in Alzheimer's disease (AD), both disorders share common molecular pathways, including oxidative stress, neuroinflammation, misfolding/aggregation of proteins and mitochondrial dysfunction. This review explores the molecular intersections between synucleinopathies and amyloidogenesis. Furthermore, this review highlights how these pathways drive neuronal loss and suggest that targeting them could provide broad therapeutic benefits. By elucidating the shared mechanisms between PD and AD, the multi-targeted therapies could address the underlying molecular disruptions common to both disorders, offering new avenues for effective disease-modifying treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jashanpreet Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
12
|
Jia L, Meng X, Ma Y, Luo J, Luo X, Yang H, Zhou J. Effect of cypermethrin on thyroid follicular epithelial cell injury via the ROS-NF-κB-NLRP3 pathway. Toxicol Lett 2025; 409:121-129. [PMID: 40381747 DOI: 10.1016/j.toxlet.2025.05.008] [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: 11/05/2024] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Cypermethrin (CYP, IUPAC name: [cyano-(3-phenoxyphenyl)methyl] 3-(2,2-dichloroethenyl)-2,2-dimethylcyclopropane-1-carboxylate), a type II pyrethroid insecticide, is suggested to have potential adverse effects on human endocrine, immune, and neurotoxic systems. OBJECTIVES In this study, we aimed to investigate whether CYP causes oxidative stress and pyroptosis in thyroid follicle epithelial cells through the activation of the ROS-NFκB-NLRP3 pathway, thereby causing inflammatory responses. METHODS Nthy-ori 3-1 cells were used as an in vitro model and exposed to CYP at different doses (0, 100, 200, 400, 800, and 1600 μmol/L) for 24 h. RESULTS CYP treatment enhanced oxidative damage in Nthy-ori 3-1 cells, characterized by cellular crumpling, indistinct borders, increased cellular gaps, appearance of intercellular grease droplets, and increased pyroptosis. Subsequent treatment with the caspase-1 inhibitor VX-765 enhanced cell viability in the VX-765 +CYP group, improving cell morphology, reducing pyroptotic vesicles, suppressing oxidative stress, and downregulating Interleukin-18(IL-18) (an inflammatory factor) level. CONCLUSION The results demonstrate that CYP induces pyroptosis in Nthy-ori 3-1 cells through activation of the reactive oxygen species (ROS)-NF-κB-NLRP3 pathway.
Collapse
Affiliation(s)
- Leina Jia
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| | - Xiaoning Meng
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| | - Yu Ma
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| | - Jinyu Luo
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| | - Xiaoqi Luo
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| | - Huifang Yang
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| | - Jian Zhou
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| |
Collapse
|
13
|
Wijerathne SVT, Pandit R, Ezeuko CC, Matthews QL. Comparative Examination of Feline Coronavirus and Canine Coronavirus Effects on Extracellular Vesicles Acquired from A-72 Canine Fibrosarcoma Cell Line. Vet Sci 2025; 12:477. [PMID: 40431570 DOI: 10.3390/vetsci12050477] [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: 04/01/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction: Coronavirus (CoV) is an extremely contagious, enveloped positive-single-stranded RNA virus, which has become a global pandemic that causes several illnesses in humans and animals. Hence, it is necessary to investigate viral-induced reactions across diverse hosts. Herein, we propose utilizing naturally secreted extracellular vesicles (EVs), mainly focusing on exosomes to examine virus-host responses following CoV infection. Exosomes are small membrane-bound vesicles originating from the endosomal pathway, which play a pivotal role in intracellular communication and physiological and pathological processes. We suggested that CoV could impact EV formation, content, and diverse immune responses in vitro. Methods: In this study, we infected A-72, which is a canine fibroblast cell line, with a feline coronavirus (FCoV) and canine coronavirus (CCoV) independently in an exosome-free media at 0.001 multiplicity of infection (MOI), with incubation periods of 48 and 72 h. The cell viability was significantly downregulated with increased incubation time following FCoV and CCoV infection, which was identified by performing the 3-(4,5-dimethylthiazo-1-2yl)-2,5-diphenyltetrazolium bromide (MTT) assay. After the infection, EVs were isolated through ultracentrifugation, and the subsequent analysis involved quantifying and characterizing the purified EVs using various techniques. Results: NanoSight particle tracking analysis (NTA) verified that EV dimensions fell between 100 and 200 nm at both incubation periods. At both periods, total protein and RNA levels were significantly upregulated in A-72-derived EVs following FCoV and CCoV infections. However, total DNA levels were gradually upregulated with increased incubation time. Dot blot analysis indicated that the expression levels of ACE2, IL-1β, Flotillin-1, CD63, caspase-8, and Hsp90 were modified in A-72-derived EVs following both CoV infections. Conclusions: Our results indicated that FCoV and CCoV infections could modulate the EV production and content, which could play a role in the development of viral diseases. Investigating diverse animal CoV will provide in-depth insight into host exosome biology during CoV infection. Hence, our findings contribute to the comprehension and characterization of EVs in virus-host interactions during CoV infection.
Collapse
Affiliation(s)
| | - Rachana Pandit
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA
| | - Chioma C Ezeuko
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA
| | - Qiana L Matthews
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA
- Department of Biological Sciences, College of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| |
Collapse
|
14
|
El-Khawaga OY, ElSaid AM, Mustafa W, El-Daw AN, Saad M. Interleukins 10 and 1β gene polymorphisms in ischemic stroke risk in the Egyptian population. Sci Rep 2025; 15:16792. [PMID: 40369205 PMCID: PMC12078794 DOI: 10.1038/s41598-025-98531-w] [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: 12/04/2024] [Accepted: 04/14/2025] [Indexed: 05/16/2025] Open
Abstract
Stroke remains the leading cause for lasting disability and death globally. Two frequent proinflammatory cytokine variants in the IL-10 and IL-1β genes, rs16944 T/C and rs1800896 G/A, may be major candidate gene loci impacting ischemic stroke vulnerability. The present case-control research aims to establish a link between both of these polymorphisms and ischemic stroke risk in the Egyptian population. The study demonstrates that the TC genotype, over dominant (TT + CC vs. TC) and dominant models (TC + CC vs. TT), exhibited greater prevalence among stroke groups as compared to the healthy group. Regarding IL-10, GA genotype, A allele, over dominant (GG + AA vs. GA), and dominant (GG vs. GA + AA) genotyping models in patients, they show highly significant differences from controls that increased the risk of stroke. Stroke patients with higher cholesterol, LDL, SBP, DBP, and lower HDL levels were more likely to develop stroke. The study found no significant link between genetic polymorphisms and smoking, gender, diabetes, or hypertension in stroke patients, except for the IL-1β heterozygous TC and dominant model, which was associated with troubles in chewing and swallowing and consciousness issues in the patients. Only troubles in chewing and swallowing manifestations were associated with the IL-10 variant. The rs16944 and rs1800896 polymorphisms differ substantially between groups, providing a more definite outcome for the Egyptian population and IS. The results validate the utilization of the rs16944 T/C and rs1800896 G/A variations in stroke prediction for Egyptians.
Collapse
Affiliation(s)
- Omali Y El-Khawaga
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt.
| | - Afaf M ElSaid
- Genetic Unit, Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Wessam Mustafa
- Neurology Department, Mansoura University Hospital, Mansoura, 35516, Egypt
| | - A N El-Daw
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
- Genetic Unit, Department of Pediatrics, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
- Neurology Department, Mansoura University Hospital, Mansoura, 35516, Egypt
| | - Mariam Saad
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
15
|
Liu Y, Jiang Y, Song C, Zuo T, Zhang J, Zhao L. The role of miR-146a/IRAK1/JNK1 pathway in mediating the effects of Yiqi Congming decoction on dry eye: A mechanistic study in rat models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119698. [PMID: 40188896 DOI: 10.1016/j.jep.2025.119698] [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: 02/08/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yiqi Congming Decoction (YQCM) is a traditional Chinese medicine formula widely used as a complementary and alternative therapy for dry eye and other ophthalmic disorders. OBJECTIVE This study aims to investigate the potential effects of YQCM on dry eye and to identify the active components responsible for its therapeutic efficacy using a rat model. MATERIALS AND METHODS Using Ultra-High Performance Liquid Chromatography/Quadrupole Time-of-Flight Mass Spectrometry (HPLC-QTOF/MS), the chemical constituents of YQCM were identified. In vivo experiments demonstrated the protective effects of YQCM on the corneal barrier in a rat model of dry eye and determined the optimal therapeutic dose. YQCM and agomiR-146a were administered either individually or in combination to rat over 14 days, followed by evaluations of Schirmer I test (SⅠT) results, tear film breakup time (BUT), fluorescein staining (FL) levels, corneal epithelial cell inflammation, and apoptosis. Furthermore, the expression of proteins in the miR-146a/IRAK1/JNK1 pathway, as well as inflammation and apoptosis-related proteins, was examined to explore the mechanisms through which YQCM modulates inflammation and improves tear film stability. In vitro experiments employed serum containing YQCM, agomiR-146a, and the IRAK1 inhibitor (AZ1495) to further investigate the regulatory effects of YQCM on the miR-146a/IRAK1/JNK1 pathway and its impact on inflammation and apoptosis in human corneal epithelial cells (HCECs) under hypertonic conditions. RESULTS In vivo experimental results demonstrated that YQCM significantly restored tear film stability. Treatment with varying doses of YQCM improved corneal epithelial damage in rats, with the medium dose exhibiting the most pronounced effect. YQCM increased the SⅠT and BUT levels, effectively reduced FL levels, and inhibited apoptosis and inflammatory damage in corneal epithelial cells. Additionally, scanning electron microscopy, hematoxylin-eosin (HE) staining, TUNEL assays, and Western blot (WB) and qPCR analyses revealed that YQCM significantly ameliorated corneal damage in dry eye rats and reduced the expression levels of MMP-9, TNF-α, IL-1β, Caspase-3, IL-6, and IFN-γ. Moreover, YQCM modulated the expression of proteins in the miR-146a/IRAK1/JNK1 pathway. In vitro experiments demonstrated that YQCM regulated the levels of miR-146a/IRAK1/JNK1 in HCECs under hypertonic conditions and enhanced cell viability by reducing the expression of MMP-9, TNF-α, IL-1β, Caspase-3, IL-6, and IFN-γ. Using Annexin V-FITC/PI double staining and other techniques, it was further confirmed that YQCM alleviated apoptosis in HCECs under hypertonic conditions. CONCLUSION YQCM protects tear film stability in a rat model of dry eye by suppressing corneal epithelial inflammatory responses and reducing apoptosis through modulation of the miR-146a/IRAK1/JNK1 pathway.
Collapse
Affiliation(s)
- Yulin Liu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China; Liaoning University Of Traditional Chinese Medicine, Shenyang, 110847, China
| | - Yanhua Jiang
- Department of Ophthalmology, China Medical University the Fourth People's Hospital of Shenyang, Shenyang, 110031, China
| | - Caiqiu Song
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| | - Tao Zuo
- Department of Ophthalmology, The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110034, China
| | - Jinghan Zhang
- Liaoning University Of Traditional Chinese Medicine, Shenyang, 110847, China
| | - Lei Zhao
- Liaoning University Of Traditional Chinese Medicine, Shenyang, 110847, China; Department of Ophthalmology, The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110034, China.
| |
Collapse
|
16
|
Pai SK, Chakraborty K, Pai AA, Dhara S. Therapeutic promise of a sulfated (1 → 4) galactan from edible sea grape Caulerpa racemosa: modulation of cytokine expression in lipopolysaccharide-induced CALU-1 cells. Int J Biol Macromol 2025; 313:144117. [PMID: 40354863 DOI: 10.1016/j.ijbiomac.2025.144117] [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: 12/20/2024] [Revised: 04/19/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Sulfated polysaccharides from edible marine algae have gained significant attention for their remarkable nutritional and therapeutic potential. Caulerpa racemosa (sea grape), a widely consumed edible algae, is a source of bioactive compounds with significant medicinal potential. This study focuses on CRP-2, a sulfated (1 → 4) galactan derived from C. racemosa, and its anti-inflammatory effects in lipopolysaccharide (LPS)-induced CALU-1 cells. CRP-2, at a concentration of 125 μg/mL, reduced the expression of pro-inflammatory cytokines interleukin-1β and tumor necrosis factor-α by 41-49 % in CALU-1 cells. Additionally, it significantly downregulated transforming growth factor-β levels, as indicated by a decrease in mean fluorescence intensity from 20 to 4 units, compared to cells treated with LPS alone. Concurrently, downregulated expressions level of interleukin-33, interferon-α and interleukin-10 in LPS-induced CALU-1 cells were substantially elevated after the treatment with CRP-2 (mean fluorescence intensities 25-35-fold), in a dose-dependent manner. Gene expression analysis by qRT-PCR revealed downregulation of interferon-γ levels by 2 folds (at 125 μg/mL) from an overly expressed levels of 9-fold in LPS-induced cells. Structure-activity and molecular docking analyses emphasized the role of sulfate moieties in its bioactivity. These findings highlight the potential of CRP-2 as a natural anti-inflammatory agent and suggest its promise as a functional food ingredient with notable health benefits.
Collapse
Affiliation(s)
- Shilpa Kamalakar Pai
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala State, India; Department of Applied Chemistry, Cochin University of Science and Technology, South Kalamassery, Cochin 682022, Kerala State, India
| | - Kajal Chakraborty
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala State, India.
| | - Ashwin Ashok Pai
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala State, India; Department of Chemistry, Mangalore University, Mangalagangothri, 574199, Karnataka State, India
| | - Shubhajit Dhara
- Marine Biotechnology, Fish Nutrition and Health Division, ICAR-Central Marine Fisheries Research Institute, Ernakulam North P.O., P.B. No. 1603, Cochin 682018, Kerala State, India; Department of Chemistry, Mangalore University, Mangalagangothri, 574199, Karnataka State, India
| |
Collapse
|
17
|
Liu R, Zhang Z, Zhou C, Wang B, Zhang M, Sun Y, Yao Y, Zhang Y, He Y, Yu J, Xia Y, Liu Y, Ning S, Feng B. Inhibitior of Bcl6 by FX1 protects DSS induced colitis mice through anti-inflammatory effects. Front Immunol 2025; 16:1558845. [PMID: 40416976 PMCID: PMC12098098 DOI: 10.3389/fimmu.2025.1558845] [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: 01/11/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Inflammatory bowel disease (IBD) is a complex immune-mediated condition, and biologics are the most commonly used drugs for its treatment. However, there are still cases of ineffective treatment. B-cell lymphoma 6 (Bcl6), a transcriptional suppressor, is known to have regulatory effects on multiple immune-associated cell subsets. FX1, a novel specific BCL6 Bric-à-brac (BTB) inhibitor, has shown positive effects in many disease models, but its effects and mechanisms in IBD control remain unclear. Methods We observed colon length and DAI score of colitis mice after treatment. HE staining section was used to evaluate colonic injury, while the expression of colonic pro-inflammatory cytokines by RT-qPCR. And differences in immune cell subsets between the two groups was analyzed by flow cytometry. Additionally, IHC and RT-qPCR were employed to evaluate the expression of colonic tight junction proteins. Furthermore, RAW264.7 cells and co-cultured Caco2 cells were detected by ELISA and RT-qPCR. Results In the treat group, colitis symptoms in mice were significantly improved, and there was a decrease in proportion of macrophages and protection of intestinal mucosal integrity-indicating anti-inflammatory effects of FX1. In cell experiments, we found that FX1 decreased secretion of pro-inflammatory factors by macrophages and increased expression of tight junction proteins in Caco2 cells after co-culture. Discussion The experimental findings demonstrate the inhibitory effect of FX1 on inflammation in murine colitis model as well as its potential mechanism. BCL6 is a potential target for treating IBD.
Collapse
Affiliation(s)
| | - Zhe Zhang
- *Correspondence: Zhe Zhang, ; Baisui Feng,
| | | | | | | | | | | | | | | | | | | | | | | | - Baisui Feng
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Zhang H, Zhang X, Chai Y, Wang Y, Zhang J, Chen X. Astrocyte-mediated inflammatory responses in traumatic brain injury: mechanisms and potential interventions. Front Immunol 2025; 16:1584577. [PMID: 40406119 PMCID: PMC12094960 DOI: 10.3389/fimmu.2025.1584577] [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: 02/27/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Astrocytes play a pivotal role in the inflammatory response triggered by traumatic brain injury (TBI). They are not only involved in the initial inflammatory response following injury but also significantly contribute to Astrocyte activation and inflammasome release are key processes in the pathophysiology of TBI, significantly affecting the progression of secondary injury and long-term outcomes. This comprehensive review explores the complex triggering mechanisms of astrocyte activation following TBI, the intricate pathways controlling the release of inflammasomes from activated astrocytes, and the subsequent neuroinflammatory cascade and its multifaceted roles after injury. The exploration of these processes not only deepens our understanding of the neuroinflammatory cascade but also highlights the potential of astrocytes as critical therapeutic targets for TBI interventions. We then evaluate cutting-edge research aimed at targeted therapeutic approaches to modulate pro-inflammatory astrocytes and discuss emerging pharmacological interventions and their efficacy in preclinical models. Given that there has yet to be a relevant review elucidating the specific intracellular mechanisms targeting astrocyte release of inflammatory substances, this review aims to provide a nuanced understanding of astrocyte-mediated neuroinflammation in TBI and elucidate promising avenues for therapeutic interventions that could fundamentally change TBI management and improve patient outcomes. The development of secondary brain injury and long-term neurological sequelae. By releasing a variety of cytokines and chemokines, astrocytes regulate neuroinflammation, thereby influencing the survival and function of surrounding cells. In recent years, researchers have concentrated their efforts on elucidating the signaling crosstalk between astrocytes and other cells under various conditions, while exploring potential therapeutic interventions targeting these cells. This paper highlights the specific mechanisms by which astrocytes produce inflammatory mediators during the acute phase post-TBI, including their roles in inflammatory signaling, blood-brain barrier integrity, and neuronal protection. Additionally, we discuss current preclinical and clinical intervention strategies targeting astrocytes and their potential to mitigate neurological damage and enhance recovery following TBI. Finally, we explore the feasibility of pharmacologically assessing astrocyte activity post-TBI as a biomarker for predicting acute-phase neuroinflammatory changes.
Collapse
Affiliation(s)
- Haifeng Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Xian Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Yan Chai
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Yuhua Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| |
Collapse
|
19
|
Hasnat S, Metsäniitty M, Nurmi K, Eklund KK, Salem A. Intracellular bacterial LPS drives pyroptosis and promotes aggressive phenotype in oral squamous cell carcinoma. Med Oncol 2025; 42:205. [PMID: 40338411 PMCID: PMC12062154 DOI: 10.1007/s12032-025-02766-6] [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: 02/11/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
Intracellular bacterial components represent an emerging tumor element that has recently been documented in multiple cancer types, yet their biological functions remain poorly understood. Oral squamous cell carcinoma (OSCC) is a particularly aggressive malignancy lacking highly effective targeted treatments. Here, we explored the functional significance of intracellular bacterial lipopolysaccharide (LPS) in OSCC. Normal human oral keratinocytes (HOKs), HPV-transformed oral keratinocytes (IHGK), and three OSCC cell lines were transfected with ultrapure bacterial LPS. Cytotoxicity was assessed via lactate dehydrogenase (LDH) release assays. Production of interleukin (IL)-1β and IL-18 was measured using ELISA. Impact on tumor progression was evaluated using cell proliferation, migration, invasion, and tubulogenesis assays. Intracellular LPS-induced significant LDH release and increased secretion of IL-18 and IL-1β in IHGK and cancer cells, but not in normal HOKs, indicating selective cytotoxicity and pyroptosis. Notably, metastatic cancer cells exhibited enhanced invasive and vessel-like structures upon LPS exposure, while IHGK cells exhibited increased proliferation without changes in migration. Our findings suggest that intracellular LPS may not merely reside passively within the tumor milieu, but could contribute to OSCC progression by triggering noncanonical inflammasome activation and pyroptosis. This process may enhance pro-inflammatory signaling and more aggressive cellular phenotypes, especially in metastatic settings. Targeting intracellular LPS or its downstream inflammasome pathways may thus represent a promising therapeutic strategy for OSCC, warranting further in vivo and clinical investigations.
Collapse
Affiliation(s)
- Shrabon Hasnat
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Marjut Metsäniitty
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Katariina Nurmi
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, 00014, Helsinki, Finland
| | - Kari K Eklund
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, 00014, Helsinki, Finland
- Department of Rheumatology, University of Helsinki and Helsinki University Hospital, 00014, Helsinki, Finland
| | - Abdelhakim Salem
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, 00014, Helsinki, Finland.
- Head and Neck Oncobiome Group, Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
20
|
Li M, Lou S, Chen K, Dong Y, Wang S, Yu T, Deng X, Li S. Oxidative stress induced by combined glyphosate and TBBPA exposure promotes gill autophagy and inflammation via the PI3K/AKT/mTOR pathway. FISH & SHELLFISH IMMUNOLOGY 2025; 160:110190. [PMID: 40020950 DOI: 10.1016/j.fsi.2025.110190] [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: 12/20/2024] [Revised: 01/25/2025] [Accepted: 02/10/2025] [Indexed: 03/03/2025]
Abstract
Glyphosate and tetrabromobisphenol A (TBBPA) are pollutants that pose a serious threat to the ecological safety of aquatic environments. However, there has been no report on the effects of combined exposure on the toxicity of carp fish gills in water. Therefore, we constructed a model of carp gill tissue and the carp epithelioma cells (EPC) cells exposed to glyphosate and/or TBBPA in vitro and in vivo, established a control group, a glyphosate group, a TBBPA group, and a glyphosate + TBBPA group, and added PI3K/AKT pathway activator musk ketone in vitro to verify the relationship between toxins and pathways. qRT-PCR and western blotting methods were used to detect the expression of oxidative stress-related indicators (CAT, GSH-Px, T-AOC, H2O2) and related genes. In vitro and in vivo results showed that glyphosate and/or TBBPA exposure resulted in overproduction of ROS, decreased activity of CAT, GSH-Px, T-AOC, and increased H2O2 content. Glyphosate and/or TBBPA exposure inhibited the PI3K/AKT/mTOR signaling pathway, further resulting in increased autophagy related genes LC3, ATG-5, Beclin-1, and decreased p62 expression. Inflammation related genes TNF-α, IL-1β, IL-6, IL-18 increased. And it was more significant when exposed in combination than when exposed alone. The addition of PI3K/AKT signaling pathway activator musk ketone in vitro can significantly alleviate the changes of autophagy and inflammation-related indicators. In summary, glyphosate and/or TBBPA induce oxidative stress by promoting gill autophagy and inflammation via the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Mingyue Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Shuang Lou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Kai Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yuting Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Shize Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tingting Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xinrui Deng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
21
|
Deghrigue M, Cherif D, Lajili S, Mesmia HB, Muller CD, Majdoub H, Bouraoui A. Structural characterizations and bioactivities of fucoidans from Dyctyopteris membranaceae and Padina pavonica with in silico investigations. Int J Biol Macromol 2025; 307:142133. [PMID: 40090661 DOI: 10.1016/j.ijbiomac.2025.142133] [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: 06/11/2024] [Revised: 02/23/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Fucoidans, a complex water-soluble sulfated polysaccharide is regarded as a valuable source of new drug development. The aim of this study was to characterize the chemical properties of fucoidans isolated from two brown algae Dyctyopteris membranaceae and Padina pavonica and to evaluate their anti-inflammatory, gastroprotective, antioxidant and immunomodulatory activities. The characterization of fucoidans was investigated with colorimetric techniques and Fourier transform infrared spectroscopy. The different macromolecular characteristics of fucoidans were determined by size exclusion chromatography. The immunomodulatory activity was evaluated using cytometric bead array technology to follow up the secretion of TNF-α in lipopolysaccharide activated THP-1 cells. The antioxidant effect was determined using the stable radical 2,2-diphenyl-1-picrylhydrazyl (DPPH). The anti-inflammatory activity was evaluated using the carrageenan-induced rat paw edema model. The gastroprotective activity was determined using HCl/EtOH induced gastric ulcers in rats. Pharmacokinetic and molecular docking analysis was conducted. As a result, only fucoidan from D. membranaceae showed an effect on the synthesis of TNF-α in THP-1 cells induced by LPS with IC50 of 77 μg/mL. Fucoidans from both algae showed antioxidant properties with EC50 of 0.2 mg/mL for fucoidan from D. membranaceae, and 0.21 mg/mL for fucoidan from P. pavonica. Furthermore, isolated fucoidans from D. membranaceae and P. pavonica showed important anti-inflammatory activity with percentages of inhibition of oedema of 75 % and 57 %, respectively, at dose of 50 mg/kg, associated with significant gastroprotective activity with percentages of ulcer inhibition of 97 % and 88 %, respectively, at the same dose. Docking study showed the reactivity of this fucoidans. The study highlights the potential pharmacological importance of D. membranaceae and P. pavonica as sources of natural compounds with biological activities.
Collapse
Affiliation(s)
- Monia Deghrigue
- Laboratory of Pharmaceutical, Chemical and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, Université de Monastir, Monastir, Tunisia.
| | - Dora Cherif
- Laboratory of Pharmaceutical, Chemical and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, Université de Monastir, Monastir, Tunisia
| | - Sirine Lajili
- Laboratory of Pharmaceutical, Chemical and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, Université de Monastir, Monastir, Tunisia
| | - Hela Ben Mesmia
- Laboratory of Pharmaceutical, Chemical and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, Université de Monastir, Monastir, Tunisia
| | - Christian D Muller
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS, Faculté de Pharmacie, Université de Strasbourg, France
| | - Hatem Majdoub
- Laboratoire des Interfaces et des Matériaux Avancés (LIMA), Faculté des Sciences de Monastir, Université de Monastir, Monastir, Tunisia
| | - Abderrahman Bouraoui
- Laboratory of Pharmaceutical, Chemical and Pharmacological Drug Development LR12ES09, Faculty of Pharmacy, Université de Monastir, Monastir, Tunisia
| |
Collapse
|
22
|
Hu Y, Li S, Chen Y, Luo X, Chen YH, Kang Y, Hu W, Chen L, Ye S, Zhou X, Liu Y, Wang F, Li Y. Smoking disrupts the relationship between cerebrospinal fluid IL-1β and multiple subdimensions of sleep. Brain Behav Immun Health 2025; 45:100987. [PMID: 40235833 PMCID: PMC11998107 DOI: 10.1016/j.bbih.2025.100987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 02/13/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Objective Inflammatory factors and cigarette smoking have been associated with sleep disorders but molecular mechanisms that regulate sleep, specifically the role of interleukin-1β (IL-1β), remain controversial. Individuals' cerebrospinal fluid (CSF) IL-1β, smoking behavior, and sleep data were collected to investigate how smoking may influence the relationship between CSF IL-1β and sleep via moderation analysis. Methods A total of 191 Chinese male patients, including 104 non-smokers and 87 active smokers, scheduled for anterior cruciate ligament reconstructive surgery, were recruited. Pittsburgh Sleep Quality Index (PSQI) scores were collected prior to surgery, and CSF samples were obtained during preoperative lumbar puncture. Results Active smokers compared to non-smokers exhibited higher scores across various subdimensions of PSQI, specifically poorer sleep quality, increased sleep latency, reduced sleep efficiency, and heightened sleep disturbance (all p < 0.05). Positive correlations were observed between CSF IL-1β levels and PSQI total scores, as well as several subdimensions of sleep (all p < 0.05) in non-smokers. In contrast, a negative correlation was observed between CSF IL-1β levels and sleep efficiency scores (p < 0.05) among active smokers. Moderation analysis revealed that smoking negatively moderated the relationship between CSF IL-1β and sleep, particularly in PSQI total scores (β = -0.95, p < 0.001), sleep latency scores (β = -1.05, p < 0.001), and sleep disturbance scores (β = -0.45, p < 0.05). Conclusions The current study found that smoking disrupts multiple subdimensions of sleep and moderates the effect of the neuroinflammatory factor CSF IL-1β on PSQI sleep latency and sleep disturbance scores.
Collapse
Affiliation(s)
- Yueling Hu
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Siyuan Li
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yingjie Chen
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yu-Hsin Chen
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yimin Kang
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Hohhot, China
| | - Weiming Hu
- Department of Psychiatry, The Third Hospital of Quzhou, Quzhou, China
| | - Li Chen
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Siling Ye
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Xinchen Zhou
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Fan Wang
- Beijing Huilongguan Hospital, Peking University, Beijing, China
| | - Yuying Li
- Ruian People's Hospital, Wenzhou Medical University Affiliated Third Hospital, Wenzhou, China
| |
Collapse
|
23
|
Ying-Li S, Hua F, Wen-Qian L, Dan L, Ke-Fei B, Lu-Jiao D, Hui-Ning Z, Jian-Liang L, Zhi-Jing X. Proteome profiling of the lung tissues of the mice co-infected with H9N2 influenza A virus and Pseudomonas aeruginosa. Vet Microbiol 2025; 304:110499. [PMID: 40187293 DOI: 10.1016/j.vetmic.2025.110499] [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: 06/17/2024] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
H9N2 Influenza A virus (IAV) can contribute to Pseudomonas aeruginosa (P. aeruginosa) superinfection, causing severe pneumonia. But the underlying mechanisms remain unclear. In this study, to investigate molecular ecology of the lung tissues co-infected with H9N2 IAV and P. aeruginosa, the mouse models were developed to analyze proteome of the lung tissues. As a result, the differential proteins of the lungs sampled from the mice with single H9N2 IAV infection, single P. aeruginosa infection and co-infection with H9N2 IAV and P. aeruginosa were related to immune responses, cell proliferation and apoptosis, which were involved in NF-κB pathway, Toll-like signaling pathway, RIG-I signaling pathway and JAK-STAT signaling pathway. It implied that the different infection combinations of H9N2 IAV and P. aeruginosa influenced the protein expression levels. Based on the proteomics assays, the three proteins, NLRX1, ISG15 and IRF9, were screened to be further characterized using the in vitro MH-S cell models with H9N2 IAV and P. aeruginosa co-infection. The findings demonstrated that NLRX1, IRF9 and ISG15 might play the important roles in immune response against H9N2 IAV and P. aeruginosa co-infection and are potential targets for the development of drugs to prevent and treat the diseases.
Collapse
Affiliation(s)
- Sun Ying-Li
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Taian, Shandong Province 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong Province 271018, China
| | - Fan Hua
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Taian, Shandong Province 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong Province 271018, China
| | - Li Wen-Qian
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Taian, Shandong Province 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong Province 271018, China
| | - Li Dan
- Shandong Medicine Technician College, Taian City, Shandong Province 271016, China
| | - Bai Ke-Fei
- Bathurst Future Agri-Tech Institute, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Dong Lu-Jiao
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Taian, Shandong Province 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong Province 271018, China
| | - Zhang Hui-Ning
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Taian, Shandong Province 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong Province 271018, China
| | - Li Jian-Liang
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Taian, Shandong Province 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong Province 271018, China
| | - Xie Zhi-Jing
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, Taian, Shandong Province 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong Province 271018, China.
| |
Collapse
|
24
|
Liu X, Li Z, Xie H, Cui S, Li X, Lang M, Liu M, Shi L. Immunomodulatory effects of Sanghuangporus flavonoids: Key insights into enhancing immunity and restoring immune function. Int Immunopharmacol 2025; 153:114474. [PMID: 40117809 DOI: 10.1016/j.intimp.2025.114474] [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: 09/02/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
The immune system is vital for maintaining homeostasis, defending against external threats, and regulating inflammation, forming the cornerstone of human health. Sanghuangporus, a natural medicinal mushroom, contains flavonoids that may increase immune cell activity, potentially improving human health. This study investigated the immunomodulatory effects of Sanghuangporus flavonoids (PBF) via network pharmacology and in vitro and in vivo experiments. Network pharmacology identified PBF compounds targeting 46 immunosuppression-related targets, with rutin emerging as a key component. Molecular docking confirmed the strong binding affinity of rutin for the core targets IL6, TNF, and IL1B. In vitro, PBF activated mouse macrophages, promoting their proliferation, phagocytic activity, NO production, and cytokine regulation. In vivo, PBF enhanced immune function in normal mice by promoting thymus and spleen growth, increasing cellular and humoral immunity, and restoring immune function in immunosuppressed mice. These findings highlight the potential of PBFs in increasing immunity and treating immunosuppressive diseases.
Collapse
Affiliation(s)
- Xue Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhinan Li
- School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China.
| | - Hongqing Xie
- Institute of Industrial Crops, Shandong Academy of Agricultral Sciences, Jinan 250100, China.
| | - Shiyao Cui
- College of Life Sciences, Westlake University, Hangzhou 310058, China.
| | - Xiaotong Li
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Mingzi Lang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Mingming Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Liangen Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
25
|
Park JH, Jeong EY, Kim YH, Cha SY, Kim HY, Nam YK, Park JS, Kim SY, Lee YJ, Yoon JH, So B, Kim D, Kim M, Byun Y, Lee YH, Shin SS, Park JT. Epigallocatechin Gallate in Camellia sinensis Ameliorates Skin Aging by Reducing Mitochondrial ROS Production. Pharmaceuticals (Basel) 2025; 18:612. [PMID: 40430436 DOI: 10.3390/ph18050612] [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: 03/14/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Reactive oxygen species (ROS) generated by mitochondrial dysfunction damage cellular organelles and contribute to skin aging. Therefore, strategies to reduce mitochondrial ROS production are considered important for alleviating skin aging, but no effective methods have been identified. Methods: In this study, we evaluated substances utilized as cosmetic ingredients and discovered Camellia sinensis (C. sinensis) as a substance that reduces mitochondrial ROS levels. Results:C. sinensis extracts were found to act as senolytics that selectively kill senescent fibroblasts containing dysfunctional mitochondria. In addition, C. sinensis extracts facilitated efficient electron transport in the mitochondrial electron transport chain (ETC) by increasing the efficiency of oxidative phosphorylation (OXPHOS), thereby reducing mitochondrial ROS production, a byproduct of the inefficient ETC. This novel mechanism of C. sinensis extracts led to the restoration of skin aging and the skin barrier. Furthermore, epigallocatechin gallate (EGCG) was identified as an active ingredient that plays a key role in C. sinensis extract-mediated skin aging recovery. Indeed, similar to C. sinensis extracts, EGCG reduced ROS and improved skin aging in an artificial skin model. Conclusions: Our data uncovered a novel mechanism by which C. sinensis extract reverses skin aging by reducing mitochondrial ROS production via selective senescent cell death/increased OXPHOS efficiency. Our results suggest that C. sinensis extract or EGCG may be used as a therapeutic agent to reverse skin aging in clinical and cosmetic applications.
Collapse
Affiliation(s)
- Ji Ho Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Eun Young Jeong
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - Ye Hyang Kim
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - So Yoon Cha
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - Ha Yeon Kim
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - Yeon Kyung Nam
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - Jin Seong Park
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - So Yeon Kim
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - Yoo Jin Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jee Hee Yoon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Byeonghyeon So
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Duyeol Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Minseon Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Yun Haeng Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Song Seok Shin
- Hyundai Bioland Co., Ltd., 22, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28162, Republic of Korea
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
26
|
Saris J, Li Yim AYF, Bootsma S, Lenos KJ, Franco Fernandez R, Khan HN, Verhoeff J, Poel D, Mrzlikar NM, Xiong L, Schijven MP, van Grieken NCT, Kranenburg O, Wildenberg ME, Logiantara A, Jongerius C, Garcia Vallejo JJ, Gisbertz SS, Derks S, Tuynman JB, D'Haens GRAM, Vermeulen L, Grootjans J. Peritoneal resident macrophages constitute an immunosuppressive environment in peritoneal metastasized colorectal cancer. Nat Commun 2025; 16:3669. [PMID: 40246872 PMCID: PMC12006467 DOI: 10.1038/s41467-025-58999-6] [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: 04/22/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Patients with peritoneal metastasized colorectal cancer (PM-CRC) have a dismal prognosis. We hypothesized that an immunosuppressive environment in the peritoneal cavity underlies poor prognosis. We define the composition of the human peritoneal immune system (PerIS) using single-cell technologies in 18 patients with- and without PM-CRC, as well as in matched peritoneal metastases (n = 8). Here we show that the PerIS contains abundant immunosuppressive C1Q+VSIG4+ and SPP1+VSIG4+ peritoneal-resident macrophages (PRMs), as well as monocyte-like cavity macrophages (mono-CMs), which share features with tumor-associated macrophages, even in homeostasis. In PM-CRC, expression of immunosuppressive cytokines IL10 and VEGF increases, while simultaneously expression of antigen-presenting molecules decreases in PRMs. These intratumoral suppressive PRMs originate from the PerIS, and intraperitoneal depletion of PRMs in vivo using anti-CSF1R combined with anti-PD1 significantly reduces tumor burden and improves survival. Thus, PRMs define a metastatic site-specific immunosuppressive niche, and targeting PRMs is a promising treatment strategy for PM-CRC.
Collapse
Affiliation(s)
- J Saris
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - A Y F Li Yim
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
| | - S Bootsma
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - K J Lenos
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - R Franco Fernandez
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - H N Khan
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - J Verhoeff
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
- Molecular Cell Biology & Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - D Poel
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - N M Mrzlikar
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - L Xiong
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - M P Schijven
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Digital Health, Amsterdam, The Netherlands
| | - N C T van Grieken
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - O Kranenburg
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, The Netherlands
- Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, The Netherlands
| | - M E Wildenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A Logiantara
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - C Jongerius
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - J J Garcia Vallejo
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
- Molecular Cell Biology & Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S S Gisbertz
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - S Derks
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J B Tuynman
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - G R A M D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - L Vermeulen
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
- Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - J Grootjans
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam, The Netherlands.
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Oncode Institute, Amsterdam, The Netherlands.
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Carnazzo V, Rigante D, Restante G, Basile V, Pocino K, Basile U. The entrenchment of NLRP3 inflammasomes in autoimmune disease-related inflammation. Autoimmun Rev 2025; 24:103815. [PMID: 40233890 DOI: 10.1016/j.autrev.2025.103815] [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: 02/17/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Autoinflammation and autoimmunity are almost "opposite" phenomena characterized by chronic activation of the immune system, 'innate' in the first and 'adaptive' in the second, leading to inflammation of several tissues with specific protean effectors of tissue damage. The mechanism of involvement of multiprotein complexes called 'inflammasomes' within autoimmune pictures, differently from autoinflammatory conditions, is yet undeciphered. In this review we provide a comprehensive overview on NLRP3 inflammasome contribution into the pathogenesis of some autoimmune diseases. In response to autoantibodies against nucleic acids or tissue-specific antigens the NLRP3 inflammasome is activated within dendritic cells and macrophages of patients with systemic lupus erythematosus. Crucial is NLRP3 inflammasome to amplify tissue inflammation with interleukin-1 overexpression and matrix metalloproteinase production at the joint level in rheumatoid arthritis. A deregulated NLRP3 inflammasome activation occurs in the serous acini of salivary and lacrimal glands prone to Sjogren's syndrome, but also in the inflammatory process involving endothelial cells, leucocyte recruitment, and platelet plugging of vasculitides. Furthermore, organ-specific autoimmune diseases such as thyroiditis and hepatitis may display hyperactive NLRP3 inflammasomes at the level of resident immune cells within thyroid or liver, respectively. Therefore, it is not unexpected that preclinical studies have shown how specific inflammasome inhibitors may significantly overthrow the severity of different autoimmune diseases and slow down their trend towards an ominous progression. Specific markers of inflammasome activation could also reveal subclinical inflammatory components escaping conventional diagnostic approaches or improve monitoring of autoimmune diseases and personalizing their treatment.
Collapse
Affiliation(s)
- Valeria Carnazzo
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica Sacro Cuore, Rome, Italy.
| | - Giuliana Restante
- Department of Experimental Medicine, University "La Sapienza", Rome, Italy
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Krizia Pocino
- Unit of Clinical Pathology, Ospedale San Pietro Fatebenefratelli, Rome, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| |
Collapse
|
28
|
Sun H, Zhao Q, Liang X, He Y, Li Y, Yu J, Ding J, Yu C. Effects of cold environment exposure on female reproductive health and its regulatory mechanisms. Front Genet 2025; 16:1570053. [PMID: 40270542 PMCID: PMC12014596 DOI: 10.3389/fgene.2025.1570053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Objective To investigate the effects of cold environment exposure on female reproductive capacity and explore its potential regulatory mechanisms. Methods Female mice were subjected to cold water immersion to simulate cold environment exposure. Weight changes during cold exposure were recorded. Serum levels of anti-Müllerian hormone (AMH), estradiol (E2), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) were measured using enzyme-linked immunosorbent assay (ELISA). Ovarian and uterine tissues were collected via surgical procedures, and transcriptomic sequencing was performed to explore potential regulatory mechanisms. ELISA was used to assess the levels of inflammatory cytokines, including interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-18 (IL-18), and tumor necrosis factor-alpha (TNF-α) in peritoneal fluid. Furthermore, immunohistochemistry was used to detect the expression levels of IL-1, IL-6, and IL-18 in ovarian tissues, as well as IL-6 and IL-18 in uterine tissues. Results Compared with the control group, female mice exposed to cold environments exhibited a significant increase in body weight and elevated serum levels of AMH, E2, FSH, and LH. Transcriptomic sequencing of ovarian and uterine tissues indicated that differentially expressed genes were primarily enriched in inflammation-related pathways, including the cAMP signaling pathway, cytokine-cytokine receptor interaction, and PI3K-Akt signaling pathway. Additionally, levels of inflammatory cytokines in the peritoneal fluid, including IL-1β, IL-6, IL-18, and TNF-α, were significantly elevated. Immunohistochemical analysis showed that the expression levels of IL-1, IL-6, and IL-18 were markedly increased in ovarian tissue, while IL-6 and IL-18 expression levels were significantly elevated in uterine tissue. These differences were statistically significant (P < 0.05). Conclusion Cold environment exposure may induce inflammatory responses in the uterus and ovaries, contributing to the formation of an inflammatory microenvironment in the reproductive system. This process may lead to disruptions in sex hormone levels and ultimately impair female reproductive capacity.
Collapse
Affiliation(s)
- Hongyi Sun
- Basic Medicine School, Naval Medical University, Shanghai, China
| | - Qianqian Zhao
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaolan Liang
- Basic Medicine School, Naval Medical University, Shanghai, China
| | - Yalun He
- Department of Physiotherapy of Traditional Chinese Medicine, Beidaihe Rehabilitation and Recuperation Center, The People’s Liberation Army Joint Logistic Support Force, Qinhuangdao, China
| | - Yangshuo Li
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jin Yu
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jie Ding
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chaoqin Yu
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
29
|
Issa F, Abdulla M, Retnowati FD, Al-Khawaga H, Alhiraky H, Al-Harbi KM, Al-Haidose A, Maayah ZH, Abdallah AM. Cardio-Rheumatic Diseases: Inflammasomes Behaving Badly. Int J Mol Sci 2025; 26:3520. [PMID: 40331999 PMCID: PMC12026794 DOI: 10.3390/ijms26083520] [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: 01/11/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Cardio-rheumatology is an evolving and interdisciplinary field lying at the intersection of rheumatology and cardiovascular medicine that recognizes that individuals with autoimmune and inflammatory rheumatic complications have a much higher likelihood of developing cardiovascular diseases (CVDs). Inflammasomes are multiprotein complexes stimulated by the immune system after the detection of pathogens or cellular injury. Inflammasomes undergo a two-stage activation process initiated by nuclear factor (NF)-κB, subsequently playing a crucial role in innate immunity through activation of caspase 1 and the consequent release of proinflammatory cytokines such as IL-18 and IL-1β. However, a loss of control of inflammasome activation can cause inflammatory diseases in humans. Recent studies have focused on the role of inflammasomes in inflammatory cascades implicated in the pathogenesis of several diseases. Here, we review inflammasome activation, its mechanism of action, and its role in CVD. In particular, we describe the role of inflammasomes in rheumatic heart disease, Kawasaki disease, familial Mediterranean fever, ankylosing spondylitis, and rheumatoid arthritis as exemplars to illustrate pathobiological mechanisms and the potential for targeting inflammasomes for therapeutic benefit.
Collapse
Affiliation(s)
- Farah Issa
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Marah Abdulla
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Faizah D. Retnowati
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Huda Al-Khawaga
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Hanin Alhiraky
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Khalid M. Al-Harbi
- Department of Pediatric, College of Medicine, Taibah University, Madinah 41477, Saudi Arabia;
| | - Amal Al-Haidose
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Zaid H. Maayah
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health Sector, Qatar University, Doha 2713, Qatar;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| |
Collapse
|
30
|
Sun X, Hu X, Wei J, An H. Uncovering leading compounds for alzheimer's disease treatment: mendelian randomization and virtual screening insights into plasma protein modulation. Biol Res 2025; 58:19. [PMID: 40186323 PMCID: PMC11971886 DOI: 10.1186/s40659-025-00598-2] [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: 11/07/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder influenced by both genetic and environmental factors. Identifying therapeutic targets and interventions remains challenging. This study utilized Mendelian Randomization (MR) to investigate causal relationships between plasma proteins, lifestyle factors, and AD, along with virtual screening to identify potential drug compounds. A two-sample MR analysis assessed associations between plasma proteins, identified through genome-wide association studies (GWAS), and AD risk. Co-localization analysis (CA) confirmed the overlap between protein expression and AD susceptibility loci, and reverse MR ruled out reverse causality. A protein-protein interaction (PPI) network was constructed to explore therapeutic targets, followed by virtual screening to identify small-molecule inhibitors for selected proteins. The analysis found significant associations between eight plasma proteins and AD, with five proteins (GSTP1, BIN1, Siglec-3, SERPINF2, and GRN) showing strong evidence of involvement in AD pathogenesis. Virtual screening identified six compounds as potential inhibitors of GSTP1 and four compounds as potential inhibitors of BIN1. Furthermore, MR analysis of lifestyle factors, such as dietary behaviors and smoking cessation, indicated they may influence AD risk through their effects on specific proteins. These findings offer novel insights into the genetic mechanisms underlying AD and highlight the potential of combining MR with virtual screening to identify therapeutic targets. The study also suggests that lifestyle modifications could offer alternative prevention and treatment strategies for AD. Future research should focus on the experimental validation of the identified compounds and further explore the mechanisms linking lifestyle factors to AD.
Collapse
Affiliation(s)
- Xiaohan Sun
- School of Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Xiaofei Hu
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Haoyu An
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
31
|
Park JY, Lee SJ. Myricetin alleviates the mechanism of IL-1β production caused by the endocrine-disrupting chemical Di(2-ethylhexyl) phthalate in RAW 264.7 cells. Tissue Cell 2025; 93:102683. [PMID: 39675255 DOI: 10.1016/j.tice.2024.102683] [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: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Myricetin, a flavonoid present in numerous fruits, vegetables, and medicinal plants, is recognized for its potent antioxidant, anti-inflammatory, and anti-cancer activities. Nevertheless, its involvement in mitigating inflammation caused by the endocrine-disrupting chemical Di(2-ethylhexyl) phthalate (DEHP), commonly used in polyvinyl chloride (PVC) manufacturing to improve flexibility, has not been investigated. Here, we found that DEHP markedly increased IL-1β production through inflammatory pathways in RAW 264.7 murine macrophages. Treatment with myricetin at a concentration of 10 μM significantly reduced the elevated IL-1β levels. Myricetin achieves this by inhibiting the activation of protein kinase C (PKC) and extracellular signal-regulated kinase (ERK), which are driven by reactive oxygen species (ROS), thereby suppressing IL-1β transcription via nuclear factor-kappa B (NF-κB). Additionally, myricetin prevents ROS-induced activation of the NLRP3 inflammasome and subsequent caspase-1 activation, further decreasing IL-1β production. These dual actions highlight myricetin's therapeutic potential in countering the oxidative stress-mediated inflammatory pathways triggered by environmental toxins like DEHP.
Collapse
Affiliation(s)
- Ji-Yeon Park
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
32
|
Peng Y, Demidchik V, Li Y, Shen Z. Comparison of terpenoids in Nauclea officinalis and Paederia scandens and their anti-inflammatory effects on RAW264.7 macrophages. Fitoterapia 2025; 182:106411. [PMID: 39909359 DOI: 10.1016/j.fitote.2025.106411] [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: 10/18/2024] [Revised: 01/14/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Terpenoids are important components that exert pharmacological effects in Rubiaceae plants. In this study, two Rubiaceae plants, Nauclea officinalis and Paederia scandens, which are widely distributed in Hainan, China, were collected. The extracts of these two plants were obtained through boiling water extraction and analyzed using UHPLC-ESI-QE-Orbitrap-MS. By comparing with the mzCloud database, terpenoids with a matching rate of 85 % were identified. The results revealed that the aqueous extracts of N. officinalis mainly contain six pentacyclic triterpenoids, one diterpenoid, and one iridoid. The aqueous extracts of P. scandens contains one monoterpenoid, one diterpenoid, two pentacyclic triterpenes, and 4 iridoids. To verify the anti-inflammatory efficacy of the two extracts, in this study, they were added to the lipopolysaccharide (LPS)-induced RAW264.7 macrophages inflammation model. The results showed that the two extracts can reduce the secretion of proinflammatory cytokines IL-1β, IL-6, and TNF-α (p < 0.05) and the mortality rate of cell inflammation (p < 0.05) by inhibiting the activation of the NF-κB/NLRP3 pathway (p < 0.05). The study identified the terpenoids in N. officinalis and P. scandens and verified their anti-inflammatory effects in the RAW264.7 macrophages inflammation model.
Collapse
Affiliation(s)
- Yuxuan Peng
- Hainan College of Vocation and Technique, 95 Nanhai Ave., 570100 Haikou, China; Department of Plant Cell Biology and Bioengineering, Biology Faculty, Belarusian State University, 4 Independence Ave., 220030 Minsk, Belarus.
| | - Vadim Demidchik
- Institute of Experimental Botany, National Academy of Sciences of Belarus, 27 Botanichskaya St., 220072 Minsk, Belarus; International Research Centre for Environmental Membrane Biology and Department of Horticulture, Foshan University, Foshan, China
| | - Yan Li
- International Sakharov Environmental Institute, Belarusian State University, 15 Botanichskaya St., 220030 Minsk, Belarus
| | - Zhenguo Shen
- Hainan College of Vocation and Technique, 95 Nanhai Ave., 570100 Haikou, China.
| |
Collapse
|
33
|
Serra D, Garroni G, Cruciani S, Coradduzza D, Pashchenko A, Amler E, Pintore G, Parisse P, Satta R, Martini F, Tognon M, Brunetti A, Ventura C, Maioli M. PVA and PVP nanofibers combined with Helichrysum italicum oil preserve skin cell interactions, elasticity and proliferation. Sci Rep 2025; 15:10864. [PMID: 40158043 PMCID: PMC11954863 DOI: 10.1038/s41598-025-95788-z] [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: 11/03/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Development of electrospun nanofibers with suitable properties to promote wound healing is an advantage in developing non-invasive skin treatments. We showed the potential application of Polyvinyl acetate (PVA) and Polyvinylpyrrolidone (PVP) combined with Helichrysum italicum oil (HO) in wound healing. During this process, Tight junctions (TJs) play a crucial role in maintaining skin integrity. TJs are intercellular junctions composed of a variety of transmembrane proteins, including Occludin (OCLN), observed also in migrating epithelial cells. Changes in OCLN expression affect epidermal permeability, indicating an active role in the healing process. Within this context, we studied the OCLN expression during healing after scratch assay on Keratinocytes (HaCaT), by a confocal microscopic analysis. In addition, we evaluated the effect of treatment after scratch on cell elasticity by Atomic Force Microscopy (AFM) analysis. All results show a positive trend in cell proliferation and viability on HaCaT treated with functionalized nanofibers. These results were confirmed by the expression of genes involved in the early stages of the regenerative process. Understanding the cell mechanisms involved in skin changes during repair process would allow future application of nanomaterials combined with HO in vivo.
Collapse
Affiliation(s)
- Diletta Serra
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
- R&D Laboratory Center, InoCure s.r.o., Politických Veziu 935/13, 110 00, Prague, Czech Republic
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Donatella Coradduzza
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Aleksei Pashchenko
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
- Department of Biophysics, Second Faculty of Medicine, Charles University, V Uvalu 84, 150 06, Prague, Czech Republic
- University Centre for Energy Efficient Buildings, Czech Technical University in Prague, Trinecka 1024, 273 43, Bustehrad, Czech Republic
| | - Evzen Amler
- University Centre for Energy Efficient Buildings, Czech Technical University in Prague, Trinecka 1024, 273 43, Bustehrad, Czech Republic
- Student Science, Národních hrdinů 279, 190 12, Praha 9, Czech Republic
| | - Giorgio Pintore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Pietro Parisse
- Institute of Materials (IOM-CNR), Area Science Park, 34149, Basovizza, Trieste, Italy
| | - Rosanna Satta
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonio Brunetti
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering - Eldor Lab Istituto Nazionale Biostrutture e Biosistemi, Via Di Corticella 183, 40128, Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
- Department of Biomedical Sciences, Center for Developmental Biology and Reprogramming-CEDEBIOR, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
| |
Collapse
|
34
|
Darwish SF, Moustafa YM, Abdel Mageed SS, Hassan GS, Mangoura SA, Aly SH, Mansour MA, Raouf AA, Sallam AAM, Fawzi SF, Atta AM, Elazazy O, El-Dakroury WA, El-Demerdash AA, Esmat EZM, Elrebehy MA, Doghish AS. Insecticides and testicular health: mechanisms of injury and protective natural products. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04016-y. [PMID: 40137965 DOI: 10.1007/s00210-025-04016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
In agriculture and public health, insecticides are vital chemicals that help manage diseases and control pests. However, their extensive use has raised concerns about their negative consequences on both humans and animals. Pesticide exposure impacts numerous human organs, including the reproductive system. Infertility is caused by reproductive system disorders, which is why they have received a lot of attention in recent decades. According to what is currently known, insecticides are among the substances that may lower the quality of the semen produced by exposed workers. The mechanisms of this action are still unclear, even though numerous underlying mechanisms have been suggested. With an emphasis on the harmful effects of insecticides on male reproductive processes, this review provides a thorough analysis of the toxicity profile of these substances. To reduce insecticides' negative impacts on human and animal health and to direct future research initiatives, it is essential to comprehend their harmful consequences.
Collapse
Affiliation(s)
- Samar F Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Yasser M Moustafa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ghaneya S Hassan
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Safwat Abdelhady Mangoura
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Shaza H Aly
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo, Cairo, 11829, Egypt
| | - Mai A Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ahmed Amr Raouf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Al-Aliaa M Sallam
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Sylvia F Fawzi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Asmaa M Atta
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Aya A El-Demerdash
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - El-Zahra M Esmat
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Galala University, New Galala City, 43713, Suez, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
35
|
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
|
36
|
Kołtun-Jasion M, Dudek MK, Kiss AK. Eucommiae cortex Comprehensive Phytochemical Analysis Connected with Its In Vitro Anti-Inflammatory Activity in Human Immune Cells. Molecules 2025; 30:1364. [PMID: 40142139 PMCID: PMC11944357 DOI: 10.3390/molecules30061364] [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] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Eucommia ulmoides Oliv., commonly known as "plant gold", is a species of the Eucommiaceae family, native to East Asia and widely utilized in medicine, food, and the chemical industry. In Traditional Chinese Medicine, the bark of E. ulmoides plays a special role, used to nourish the liver and kidneys and to strengthen tendons and bones. Due to its extensive pharmacological profile, including anti-inflammatory, antioxidant, hypoglycemic, hypotensive, and cardio- and neuroprotective effects, there has been growing interest in elucidating the molecular mechanisms underlying its biological effects. However, many of these mechanisms remain poorly understood to date. This study analyzed the phytochemical composition of E. ulmoides bark infusions and tinctures and their dominant compounds using the HPLC-DAD-MS/MS method, and evaluated their anti-inflammatory effects in human immune cell models. The analysis identified lignans, iridoids, and caffeic acid derivatives as the dominant constituents of the tested samples. The extracts significantly inhibited the secretion of pro-inflammatory cytokines (TNF-α, IL-6, IL-8, MCP-1) in neutrophils, PBMC-derived monocytes/macrophages, and THP-1 cells. The results presented herein offer significant insights into the detailed phytochemical composition of E. ulmoides bark, and contribute to a deeper understanding of its anti-inflammatory mechanisms in human immune cells.
Collapse
Affiliation(s)
- Małgorzata Kołtun-Jasion
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Marta Katarzyna Dudek
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza H. 112, 90-001 Łódź, Poland;
| | - Anna Karolina Kiss
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| |
Collapse
|
37
|
Li Y, Cheng T, Zhou S, Li F, Guo W, Li M, Liu T. Changes in aqueous humor cytokines and metabolomics in contralateral eye after unilateral cataract surgery. BMC Ophthalmol 2025; 25:137. [PMID: 40098128 PMCID: PMC11916942 DOI: 10.1186/s12886-025-03961-9] [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: 06/18/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND For patients with bilateral age-related cataracts, sequential phacoemulsification and intraocular lens implantation is a common treatment. However, it remains unclear whether surgery on the first eye has an impact on the second eye, as current research results are inconsistent. This study will explore whether surgery on one eye affects the non-operated eye through aqueous humor cytokines and metabolomic analyses in the second eye. METHODS A rabbit model of unilateral phacoemulsification and intraocular lens implantation was established. The experimental group consisted of 15 rabbits undergoing this procedure. Postoperatively, rabbits were divided into five subgroups (three rabbits per subgroup), and aqueous humor was collected from both the operated and non-operated eyes at 1 day, 3 days, 1 week, 2 weeks, and 3 weeks after surgery. Additionally, 5 rabbits were selected as a control group, from which aqueous humor was extracted. Levels of IL-1a, IL-1β, IL-2, IL-4, IL-6, IL-8, IFN-γ, TNF-α, MCP-1, and VEGF in the aqueous humor were compared. In the clinical study, preoperative aqueous humor samples were collected from 22 patients undergoing bilateral phacoemulsification and intraocular lens implantation. Among them, 11 patients were tested for the aforementioned 10 cytokines, while the other 11 patients underwent untargeted metabolomics research. RESULTS In the animal experiment, levels of all 10 cytokines in the operated eyes were significantly higher compared to both the control and non-operated eyes groups (P < 0.05). In the non-operated eyes, IL-1β and IL-2 levels were also elevated compared to the control (P < 0.05); however, no statistically significant differences were observed between the non-operated eyes and the control group at postoperative time points of 1 day, 3 days, 1 week, 2 weeks, and 3 weeks. In the clinical study, no significant differences were found in cytokine levels between the two eyes. In the untargeted metabolomics analysis, 354 metabolites showed differential expression, 280 were upregulated and 74 were downregulated. Notably, Adenine and 2-Aminopurine were significantly downregulated, highlighting Purine metabolism as the most impacted pathway. CONCLUSIONS Animal experiments showed a significant increase in IL-1β and IL-2 levels in the non-operated eyes postoperatively, reflecting systemic and local inflammatory responses. In clinical experiments, although no significant changes in cytokines were observed in the aqueous humor of both eyes, differential expression of metabolites indicated metabolic adjustments in the non-operated eye following surgery on the first eye. These findings reveal that unilateral cataract surgery may affect the stability of the intraocular environment in the contralateral eye, suggesting that in staged bilateral surgeries, potential metabolic changes in the non-operated eye and their clinical significance should be considered. This result provides important reference value for optimizing postoperative management strategies, reducing complications, and determining the timing for bilateral surgeries, warranting further investigation.
Collapse
Affiliation(s)
- Yang Li
- Second Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Taiying Cheng
- Second Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Sujun Zhou
- Second Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Fayuan Li
- Zunyi Medical University, Guizhou, 563000, China
| | - Wenjun Guo
- Second Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Mingbo Li
- Second Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China
| | - Taixiang Liu
- Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China.
| |
Collapse
|
38
|
Muramoto S, Shimizu S, Shirakawa S, Ikeda H, Miyamoto S, Jo M, Takemori U, Morimoto C, Wu Z, Tozaki-Saitoh H, Oda K, Inoue E, Nonaka S, Nakanishi H. Noradrenaline Synergistically Enhances Porphyromonas gingivalis LPS and OMV-Induced Interleukin-1 β Production in BV-2 Microglia Through Differential Mechanisms. Int J Mol Sci 2025; 26:2660. [PMID: 40141302 PMCID: PMC11942402 DOI: 10.3390/ijms26062660] [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: 02/17/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Infection with Porphyromonas gingivalis (Pg), which is a major periodontal pathogen, causes a large number of systemic diseases based on chronic inflammation such as diabetes and Alzheimer's disease (AD). However, it is not yet fully understood how Pg can augment local systemic immune and inflammatory responses during progression of AD. There is a strong association between depression and elevated levels of inflammation. Noradrenaline (NA) is a key neurotransmitter that modulates microglial activation during stress conditions. In this study, we have thus investigated the regulatory mechanisms of NA on the production of interleukin-1β (IL-1β) by microglia following stimulation with Pg virulence factors, lipopolysaccharide (LPS), and outer membrane vesicles (OMVs). NA (30-1000 nM) significantly enhanced the mRNA level, promoter activity, and protein level of IL-1β up to 20-fold in BV-2 microglia following treatment with Pg LPS (10 μg/mL) and OMVs (150 μg of protein/mL) in a dose-dependent manner. Pharmacological studies have suggested that NA synergistically augments the responses induced by Pg LPS and OMVs through different mechanisms. AP-1 is activated by the β2 adrenergic receptor (Aβ2R)-mediated pathway. NF-κB, which is activated by the Pg LPS/toll-like receptor 2-mediated pathway, is required for the synergistic effect of NA on the Pg LPS-induced IL-1β production by BV-2 microglia. Co-immunoprecipitation combined with Western blotting and the structural models generated by AlphaFold2 suggested that cross-coupling of NF-κB p65 and AP-1 c-Fos transcription factors enhances the binding of NF-κB p65 to the IκB site, resulting in the synergistic augmentation of the IL-1β promoter activity. In contrast, OMVs were phagocytosed by BV-2 microglia and then activated the TLR9/p52/RelB-mediated pathway. The Aβ2R/Epac-mediated pathway, which promotes phagosome maturation, may be responsible for the synergistic effect of NA on the OMV-induced production of IL-1β in BV-2 microglia. Our study provides the first evidence that NA synergistically enhances the production of IL-1β in response to Pg LPS and OMVs through distinct mechanisms.
Collapse
Affiliation(s)
- Sakura Muramoto
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Sachi Shimizu
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Sumika Shirakawa
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Honoka Ikeda
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Sayaka Miyamoto
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Misato Jo
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Uzuki Takemori
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Chiharu Morimoto
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan;
| | - Hidetoshi Tozaki-Saitoh
- Department of Pharmaceutical Sciences, School of Pharmacy at Fukuoka, International University of Health and Welfare, Okawa 831-8501, Japan;
| | - Kosuke Oda
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan
| | - Erika Inoue
- School of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan; (S.M.); (S.S.); ; (S.S.); (H.I.); (S.M.); (M.J.); (U.T.); (C.M.); (E.I.)
| | - Saori Nonaka
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan;
| | - Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan;
| |
Collapse
|
39
|
Xiong J, Wu Y, Luo L, Shen X, Zeng Y, Meng X, Zhang H. Network pharmacology integrated with experimental validation reveals the mechanism of Xanthii Fructus against allergic rhinitis via JAK2/STAT3/HIF-1α signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119461. [PMID: 39923957 DOI: 10.1016/j.jep.2025.119461] [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: 01/09/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a natural medicine, Xanthii Fructus (XF) is widely used in traditional Chinese medicine (TCM) and Tibetan medicine. It has been demonstrated to alleviate allergic rhinitis (AR) in modern research. However, the specific molecular mechanism underlying its treatment of AR is still unclear. AIM OF STUDY To elucidate the effect and mechanism of XF in treating AR through network pharmacology and experimental validation. METHODS In the present study, blood-entry components of XF were analyzed using UPLC-Orbitrap-HRMS. Then, we conducted pharmacodynamic studies in vitro and vivo. In vitro study, Human IL-4 was used to treat HNEpCs cells to establish a vitro model. Subsequently, HNEpCs cells were administrated with XF extracts (0.5, 1, 2 mg/ml). And ovalbumin (OVA) was employed to establish an allergic rhinitis model, and different doses of XF (8, 16, 32 mg/kg) were administered by gavage to BABL/c mice for in vivo experiments. Next, the Swiss Target Prediction database was employed to acquire blood-entry components targets. Meanwhile, from OMIM and GeneCards databases, AR-related targets were obtained. A protein-protein interaction (PPI) network was established through the STRING database, and potential pathways of XF were identified through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. In the end, the results of network pharmacology were experimental validation in vivo and vitro experiments. RESULTS Fifteen compounds were identified, most of which were phenolic acids. In pharmacodynamic studies, the vitro study revealed that XF-treated gave rise to a significant decline of iNOS and COX2 protein expression in inflammatory conditions, as evidenced by Western blot results, and there was a sharp decline in the mRNA levels of TNF-α, IL-1β and IL-6. Meanwhile, the vivo studies demonstrated that XF exhibited favorable therapeutic efficacy against AR, as evidenced by a decrease in IgE, TNF-α, IL-4, and IL-6 levels in mice serum, an improvement in nasal mucosal injury pathology. Based on these findings, through network pharmacology, we identified 14 core AR-related targets, including HIF-1α, STAT3, TLR4. Using KEGG pathway analysis, it has been revealed that XF can alleviate AR through JAK2/STAT3/HIF-1α signaling pathway. Therefore, further experiments were conducted to verify the molecular mechanism of the anti-AR effect of XF. A decline of the phosphorylation of JAK2, STAT3 and HIF-1α proteins was observed, which resulted in the suppression of JAK2/STAT3/HIF-1α signaling pathway. These findings were corroborated by the same results obtained through IF. The results were verified by RT-qPCR, which demonstrated that XF was capable of downregulating the mRNA levels of TSLP and CCL11. Then, the conclusions were further reinforced with the introduction of WP1066. It could be observed that XF inhibited the STAT3 nuclear translocation. Finally, a restoration of p-JAK2, p-STAT3, HIF-1α expression levels to normal levels in AR mice. CONCLUSION The combined findings led to the conclusion that XF play its therapeutic role in AR by suppressing the JAK2/STAT3/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Jinrui Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yu Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Liuling Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xiaofei Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Yong Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Hai Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
40
|
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
|
41
|
Challa SR, Levingston H, Fornal CA, Baker IM, Boston J, Shanthappa N, Unnam P, Klopfenstein JD, Veeravalli KK. Temporal mRNA Expression of Purinergic P2 Receptors in the Brain Following Cerebral Ischemia and Reperfusion: Similarities and Distinct Variations Between Rats and Mice. Int J Mol Sci 2025; 26:2379. [PMID: 40141023 PMCID: PMC11941906 DOI: 10.3390/ijms26062379] [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: 01/23/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Purinergic P2 receptors are crucial in energy utilization and cellular signaling, making them key targets for stroke therapies. This study examines the temporal mRNA expression of all P2 receptors in rats and mice. Both species exhibited a common subset of P2X and P2Y receptors with elevated expression following cerebral ischemia and reperfusion (I/R), highlighting conserved mechanisms across these species. The receptors with upregulated expression in both species were P2X3, P2X4, P2X7, P2Y2, and P2Y6. While these similarities were observed, notable differences in receptor expression emerged between rats and mice. Rats exhibited a broader receptor profile, with five additional receptors (P2X1, P2Y1, P2Y12, P2Y13, and P2Y14) significantly upregulated compared to only two receptors (P2X2 and P2Y4) in mice, highlighting species-specific regulation of receptor expression distinct from the shared receptors. Following cerebral I/R, P2Y12 was the most upregulated receptor in rats, while P2Y2 was the most upregulated in mice. These findings reveal both conserved and species-specific changes in P2 receptor expression following cerebral I/R. Targeting purinergic receptors, particularly those conserved and upregulated in response to stroke, may represent a promising therapeutic approach.
Collapse
Affiliation(s)
- Siva Reddy Challa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Hunter Levingston
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Casimir A. Fornal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Isidra M. Baker
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Joseph Boston
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Nidhi Shanthappa
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Pavani Unnam
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
| | - Jeffrey D. Klopfenstein
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Illinois Neurological Institute, OSF HealthCare, Peoria, IL 61603, USA
| | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA; (S.R.C.); (H.L.); (C.A.F.); (I.M.B.); (J.B.); (P.U.); (J.D.K.)
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Neurology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| |
Collapse
|
42
|
Kim D, Kim C, Lee SE, Kim S, Lee SI, Park MH, Kim M, Sung D, Lee K. Development of ROS-Sensitive Sulfasalazine-Loaded Ferrocene Nanoparticles and Evaluation of Their Antirheumatic Effects in a 3D Synovial Hyperplasia Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407813. [PMID: 39950417 DOI: 10.1002/smll.202407813] [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: 09/01/2024] [Revised: 12/03/2024] [Indexed: 05/09/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by joint inflammation, synovial hyperplasia, and bone and cartilage destruction, which significantly impairs physical function and quality of life. Disease-modifying antirheumatic drugs, such as sulfasalazine (SSZ), are crucial for altering the course and progression of RA; however, their clinical use is hampered by poor water solubility and lack of specificity for the reactive oxygen species (ROS)-rich environment typical of RA. To overcome these challenges, ROS-sensitive SSZ-loaded ferrocene nanoparticles are developed. The nanoparticles facilitate enhanced solubility and stability of SSZ and particularly enable precision targeting through the distinctive redox properties of ferrocene. Using a 3D synovial hyperplasia model with fibroblast-like synoviocytes derive from RA patients and validate at both the protein and gene levels, these nanoparticles significantly reduce lactate dehydrogenase, ROS, and inflammatory cytokine levels. Further validation using a collagen-induced arthritis model demonstrates therapeutic efficacy and cytokine modulation in vivo. These findings highlight the potential of ferrocene nanoparticles as a novel and effective therapeutic strategy for RA, offering improved drug delivery and reduced systemic toxicity.
Collapse
Affiliation(s)
- Dongwoo Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chaehyun Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
| | - So Eun Lee
- Division of Rheumatology, Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Sangwoo Kim
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sang-Il Lee
- Division of Rheumatology, Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
- Department of Internal Medicine, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Min Hee Park
- THEDONEE Inc., Research Center, Seoul, South Korea
| | - Mingyo Kim
- Division of Rheumatology, Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
- Department of Internal Medicine, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Daekyung Sung
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, 28160, Republic of Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
43
|
Zhang R, Sougawa N, Mao D, Inoue H, Goda S. Signaling pathways of pro-IL-1β production induced by mechanical stress in gingival epithelial cells. J Oral Biosci 2025; 67:100626. [PMID: 39921162 DOI: 10.1016/j.job.2025.100626] [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: 11/13/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVES Mechanical stress on the teeth and alveolar bone caused by bruxism, orthodontics, and implants affects the periodontal tissues, causing gingival recession and alveolar bone resorption, and entire body, including the heart and vascular system. Although the same forces exerted on the alveolar bone and teeth are exerted on gingival epithelial cells, little is known about the effects of mechanical stress on these cells. This study investigated the effects of mechanical stress on gingival epithelial cells. METHODS Ca9-22 cells (human gingival epithelial cells) were used. They were seeded onto the silicone and stretched cyclically. Mechanical stress-stimulated Ca9-22 cells were evaluated for pro-inflammatory interleukin (pro-IL)-1β production using Western blotting and analyzed to assess the phosphorylation level of intracellular signaling molecules. RESULTS Mechanical stress induced pro-IL-1β upregulation in Ca9-22 cells, which was significantly inhibited by ruthenium red. Ruthenium red significantly inhibited mechanical stress-induced phosphorylation of focal adhesion kinase (FAK), P130cas, and extracellular signal-regulated kinase 1 and 2 (ERK1/2) induced by mechanical stress. Additionally, Y15 significantly inhibited the upregulation of pro-IL-1β expression and phosphorylation of FAK, P130cas, and ERK1/2 stimulated by mechanical stress. CONCLUSIONS In Ca9-22 cells, mechanical stress may increase pro-IL-1β production via mechanosensitive ion channels and FAK. These findings revealed the mechanisms of inflammation in mechanically-stressed Ca9-22 cells and may aid in the development of therapeutic approaches to prevent bone resorption.
Collapse
Affiliation(s)
- Ruixuan Zhang
- Graduate School of Dentistry, Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Nagako Sougawa
- Department of Physiology, Osaka Dental University, Osaka, Japan.
| | - Dan Mao
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Hiroshi Inoue
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Seiji Goda
- Department of Physiology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
44
|
Wang HM, Shieh KR, Chang ET. Correlation of the expression of circadian-clock genes with the severity of obstructive sleep apnea in patients. Chronobiol Int 2025; 42:428-439. [PMID: 40113269 DOI: 10.1080/07420528.2025.2480120] [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: 05/08/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
This study investigates the connection of Obstructive Sleep Apnea (OSA) with the expression and daily oscillation patterns of core circadian clock genes and related genes. OSA, a sleep disorder characterized by repetitive airway occlusion leading to nocturnal arousals, sleep fragmentation, and intermittent hypoxemia (IH), shares sleep dysfunction as an overlapping phenotype with circadian clock genes. The research involved 40 subjects (30 OSA patients and 10 normal controls), categorized into four groups based on Polysomnography (PSG) results: normal, mild, moderate, and severe. Peripheral blood samples were collected twice from each participant in the evening before and the morning after PSG examination. Using real-time quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR), the study measured the expression levels of target genes in leukocytes. Results revealed changes in diurnal expression patterns of several genes (PER1, PER3, CRY1, BMAL1, CLOCK, HIF-1α, IL-1β, TNFα) in OSA groups compared to normal controls. While PER2, CRY2, and NPAS2 genes did not show diurnal patterns, their expression was significantly elevated in severe OSA. Notably, the expression levels of HIF-1α, IL-1β, and TNFα increased with OSA severity, consistent with the roles of IH and inflammation as clinical indicators in OSA. These findings not only demonstrate that circadian clock-related gene expression fluctuates with OSA but also provide potential molecular markers for early diagnosis and personalized treatment. By identifying biomarkers parallel to clinical indicators in OSA, this innovative study paves the way for future research and clinical applications in the field.
Collapse
Affiliation(s)
- Hsiu-Mei Wang
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Long-Term Care, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Kun-Ruey Shieh
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Physiology, Tzu Chi University, Hualien, Taiwan
- Institute of Biomedical Sciences, Tzu Chi University, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - En-Ting Chang
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| |
Collapse
|
45
|
Wu MC, Li YF, Lin CY, Lin NH, Lee CY, Su YJ, Chang HC, Gau SY. New-onset Bipolar Disorder in Hidradenitis Suppurativa Patients: A Multi-center, Propensity-score-matched Cohort Study. In Vivo 2025; 39:1067-1077. [PMID: 40011005 PMCID: PMC11884441 DOI: 10.21873/invivo.13911] [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: 12/11/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 02/28/2025]
Abstract
BACKGROUND/AIM Hidradenitis suppurativa (HS) may be linked to an elevated risk of bipolar disorder, though the precise mechanism remains unclear. This study investigated the likelihood of bipolar disorder in patients with HS. PATIENTS AND METHODS We analyzed the electronic health records of 60,850 patients with HS and 60,850 matched controls from the TriNetX network, excluding those with a prior bipolar disorder diagnosis. Propensity score matching was conducted (1:1 ratio), and hazard ratios (HRs) were calculated to assess the risk of new-onset bipolar disorder in patients with HS compared to controls. RESULTS After matching, the HR for developing bipolar disorder in patients with HS was 1.549 [95% confidence interval (CI)=1.270-1.889] after a 1-year follow-up, remaining significant in 3- and 5-year follow-ups and sensitivity analyses. Stratified by sex, female patients with HS showed a notably higher risk (HR=1.509, 95%CI=1.353-1.683), while no significant increase was seen in males. CONCLUSION Patients with HS have a significantly elevated risk of developing bipolar disorder, especially among females. Healthcare providers should be mindful of this association when treating patients with HS.
Collapse
Affiliation(s)
- Meng-Che Wu
- Division of Pediatric Gastroenterology, Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan, R.O.C
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Yun-Feng Li
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Chen-Yu Lin
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C
| | - Nung-Hsiang Lin
- Drug Abuse Prevention Center, Health Bureau of Taichung City Government, Taichung, Taiwan, R.O.C
| | - Chien-Ying Lee
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yu-Jung Su
- Orthopedics Department, Chi-Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Hui-Chin Chang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.;
- Evidence-based Medicine Center, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
- Library, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Shuo-Yan Gau
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan, R.O.C
- Orthopedics Department, Chi-Mei Medical Center, Tainan, Taiwan, R.O.C
- Department and Graduate Institute of Business Administration, National Taiwan University, Taipei, Taiwan, R.O.C
| |
Collapse
|
46
|
Campagno KE, Lu W, Sripinun P, Albalawi F, Cenaj A, Mitchell CH. Priming and release of cytokine IL-1β in microglial cells from the retina. Exp Eye Res 2025; 252:110246. [PMID: 39848558 DOI: 10.1016/j.exer.2025.110246] [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: 07/08/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
The P2X7 receptor (P2X7R) for extracellular ATP is implicated in several forms of retinal degeneration, including diabetic retinopathy, age-related macular degeneration, and glaucoma. P2X7R stimulation can trigger release of master cytokine IL-1β from microglia in the brain and from macrophages, but evidence of release from retinal microglia is indirect. Isolated mouse and rat retinal microglia, and wholemounts from Cx3CR1+/GFP mice, were examined to determine if ATP induced IL-1β release directly from retinal microglial cells and if it also primed expression of IL-1β on an mRNA and protein level. Isolated retinal microglia were ramified and expressed low levels of polarization markers unless provoked. Over 90% of isolated microglial cells expressed P2X7R, with cytoplasmic Ca2+ elevation following receptor stimulation. ATP induced a dose-dependent release of IL-1β from primed microglial cells that was blocked by P2X7R antagonist A839977 and emulated by agonist BzATP. P2X7R stimulation also primed Il1b mRNA in isolated microglia cells. BzATP increased IL-1β immunostaining and GFP fluorescence throughout lamina of retinal wholemounts from CX3CR1+/GFP mice. Some of the IL-1β and GFP signals colocalized, particularly in the outer retina, and in projections extending distally through photoreceptor layers. The inner retina had more microglia without IL-1β, and more IL-1β staining without microglia. Substantial IL-1β release was also detected from rat retinal microglial cells, but not optic nerve head astrocytes. In summary, this study implicates microglial cells as a key source of released IL-1β when levels of extracellular ATP are increased following retinal damage, and suggest a greater participation in the outer retina.
Collapse
Affiliation(s)
- Keith E Campagno
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Wennan Lu
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Puttipong Sripinun
- Department of Orthodontics, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Farraj Albalawi
- Department of Orthodontics, University of Pennsylvania, Philadelphia, PA, 19104, United States; Department of Preventive Dental Sciences, College of Dentistry, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Aurora Cenaj
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Claire H Mitchell
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, 19104, United States; Department of Physiology, University of Pennsylvania, Philadelphia, PA, 19104, United States.
| |
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
|
Kihel A, El Filaly H, Darif D, Assouab A, Riyad M, Nait Irahal I, Akarid K. Itaconate: A Nexus Metabolite Fueling Leishmania Survival Through Lipid Metabolism Modulation. Microorganisms 2025; 13:531. [PMID: 40142422 PMCID: PMC11944847 DOI: 10.3390/microorganisms13030531] [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: 01/22/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Leishmaniasis, caused by the Leishmania parasite, is a neglected public health issue. Leishmania mainly infects macrophages, where metabolic reprogramming shapes their plasticity (M1/M2), affecting the host's resistance or susceptibility to infection. The development of this infection is influenced by immune responses, with an excessive anti-inflammatory reaction linked to negative outcomes through the modulation of various mediators. Itaconate, produced by the Acod1 gene, is recognized for its anti-inflammatory effects, but its function in leishmaniasis is not well understood. This study aimed to investigate the potential role of itaconate in leishmaniasis. Using transcriptomic data from L. major-infected BMDMs, we assessed the expression dynamics of Il1b and Acod1 and performed pathway enrichment analysis to determine the profile of genes co-expressed with Acod1. Early Acod1 upregulation followed by later Il1b downregulation was noted, indicating a shift towards an anti-inflammatory response. Among the genes co-expressed with Acod1, Ldlr, Hadh, and Src are closely associated with lipid metabolism and the polarization of macrophages towards the M2 phenotype, thereby creating a favorable environment for the survival of Leishmania. Overall, these findings suggest that Acod1 and its co-expressed genes may affect the outcome of Leishmania infection by modulating host metabolism. Accordingly, targeting itaconate-associated pathways could provide a novel therapeutic strategy for leishmaniasis.
Collapse
Affiliation(s)
- Ayyoub Kihel
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca 20100, Morocco; (A.K.); (H.E.F.); (D.D.); (A.A.); (I.N.I.)
| | - Hajar El Filaly
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca 20100, Morocco; (A.K.); (H.E.F.); (D.D.); (A.A.); (I.N.I.)
| | - Dounia Darif
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca 20100, Morocco; (A.K.); (H.E.F.); (D.D.); (A.A.); (I.N.I.)
| | - Aicha Assouab
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca 20100, Morocco; (A.K.); (H.E.F.); (D.D.); (A.A.); (I.N.I.)
| | - Myriam Riyad
- Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca (UH2C), Casablanca 20000, Morocco;
| | - Imane Nait Irahal
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca 20100, Morocco; (A.K.); (H.E.F.); (D.D.); (A.A.); (I.N.I.)
| | - Khadija Akarid
- Biochemistry, Biotechnology and Immunophysiopathology Research Team, Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca 20100, Morocco; (A.K.); (H.E.F.); (D.D.); (A.A.); (I.N.I.)
| |
Collapse
|
49
|
Armanville S, Tocco C, Haj Mohamad Z, Clarke D, Robitaille R, Drouin-Ouellet J. Chemically Induced Senescence Prompts Functional Changes in Human Microglia-Like Cells. J Immunol Res 2025; 2025:3214633. [PMID: 40041406 PMCID: PMC11876530 DOI: 10.1155/jimr/3214633] [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: 03/26/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
In response to various stressors, cells can enter a state called cellular senescence which is characterized by irreversible cell cycle arrest and a senescence-associated secretory phenotype (SASP). The progressive accumulation of senescent glial cells in the central nervous system (CNS) with aging suggests a potential role for senescence as driver of aging and inflammation in the brain. As the main immune cell population residing in the CNS, microglia are thought to play a pivotal role in the progression of age-associated neuroinflammation. Furthermore, due to their slow turnover, microglia are highly susceptible to undergoing cellular senescence. However, current understanding of age-related changes in microglia and their impact on brain aging is limited. Due to the challenge in accessing human primary microglia and the lack of models to adequately recapitulate aging, this knowledge is predominantly limited to rodent studies. Here, we chemically induced senescence in a human immortalized microglia cell line with a cocktail of senescence-inducing molecules. We demonstrate that chemically induced senescent microglia adopt a proinflammatory phenotype, have reduced phagocytic activity, and impaired calcium activity. Our results show that chemically induced senescence can mimic features of cellular aging and can provide insight into the impact of aging and cellular senescence on human microglia.
Collapse
Affiliation(s)
- S. Armanville
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - C. Tocco
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - Z. Haj Mohamad
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - D. Clarke
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
| | - R. Robitaille
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
- Research Group on Neural Signalling and Circuits (SNC), University of Montreal, Montreal, Quebec, Canada
- Center for Interdisciplinary Research on Brain and Learning (CIRCA), Montreal, Quebec, Canada
| | - J. Drouin-Ouellet
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
- Research Group on Neural Signalling and Circuits (SNC), University of Montreal, Montreal, Quebec, Canada
- Center for Interdisciplinary Research on Brain and Learning (CIRCA), Montreal, Quebec, Canada
| |
Collapse
|
50
|
Hua K, Cummings M, Bernatik M, Brinkhaus B, Usichenko T, Willich SN, Scheibenbogen C, Dietzel J. Effects of Auricular Stimulation on Inflammatory Parameters: Results of a Systematic Review and Meta-Analysis of Randomized Controlled Trials. Neuromodulation 2025:S1094-7159(25)00008-X. [PMID: 39998453 DOI: 10.1016/j.neurom.2024.12.007] [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: 09/12/2024] [Revised: 12/02/2024] [Accepted: 12/19/2024] [Indexed: 02/26/2025]
Abstract
BACKGROUND The number of randomized controlled trials (RCTs) using auricular stimulation (AS) techniques, such as transauricular vagus nerve stimulation, auricular electrostimulation, auricular acupuncture, and acupressure in experimental and clinical settings has increased markedly over the last three decades. This systematic review evaluates the effects of AS on biomarkers of inflammation and stress responses. MATERIALS AND METHODS The following data bases were searched: MEDLINE (PubMed), EMBASE, Cochrane Central Register of Controlled Trials (CENTRAL), ISI Web of Science, and Scopus Data base. Data collection and analysis were conducted independently by two reviewers. Quality and risk assessments of the included studies were performed, and a meta-analysis of the effects of the most frequently assessed biomarkers was conducted using RevMan statistical software. RESULTS A total of 1122 patients and healthy volunteers from 27 RCTs were included in this systematic review; 81% of the participants were female, with a median age of 51 years. Pooled data of 18 studies showed a significant effect of AS regarding a reduction of serum C-reactive protein, tumor necrosis factor-α, interleukin (IL)-6, and IL-10. Although IL-4, IL1β, cortisol, substance P and calcitonin gene-related peptide, and adrenocorticotropic hormone did not show any changes, salivary amylase increased under AS. CONCLUSIONS The influence of inflammatory cytokines seems to be mediated by AS. More research is needed to investigate the effects of AS on the immunologic system in addition to its clinical significance in high-quality RCT.
Collapse
Affiliation(s)
- Kevin Hua
- Institute for Social Medicine, Epidemiology and Health Economics, Charité-University Medicine, Corporate Member of Freie Universität Berlin, Humboldt-University Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | | | - Benno Brinkhaus
- Institute for Social Medicine, Epidemiology and Health Economics, Charité-University Medicine, Corporate Member of Freie Universität Berlin, Humboldt-University Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Taras Usichenko
- Department for Anaesthesiology, University Hospital Greifswald, Greifswald, Germany; Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
| | - Stefan N Willich
- Institute for Social Medicine, Epidemiology and Health Economics, Charité-University Medicine, Corporate Member of Freie Universität Berlin, Humboldt-University Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Joanna Dietzel
- Institute for Social Medicine, Epidemiology and Health Economics, Charité-University Medicine, Corporate Member of Freie Universität Berlin, Humboldt-University Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|