1
|
Shi L, Liu S, Chen J, Wang H, Wang Z. Microglial polarization pathways and therapeutic drugs targeting activated microglia in traumatic brain injury. Neural Regen Res 2026; 21:39-56. [PMID: 39665832 PMCID: PMC12094552 DOI: 10.4103/nrr.nrr-d-24-00810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/03/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Traumatic brain injury can be categorized into primary and secondary injuries. Secondary injuries are the main cause of disability following traumatic brain injury, which involves a complex multicellular cascade. Microglia play an important role in secondary injury and can be activated in response to traumatic brain injury. In this article, we review the origin and classification of microglia as well as the dynamic changes of microglia in traumatic brain injury. We also clarify the microglial polarization pathways and the therapeutic drugs targeting activated microglia. We found that regulating the signaling pathways involved in pro-inflammatory and anti-inflammatory microglia, such as the Toll-like receptor 4 /nuclear factor-kappa B, mitogen-activated protein kinase, Janus kinase/signal transducer and activator of transcription, phosphoinositide 3-kinase/protein kinase B, Notch, and high mobility group box 1 pathways, can alleviate the inflammatory response triggered by microglia in traumatic brain injury, thereby exerting neuroprotective effects. We also reviewed the strategies developed on the basis of these pathways, such as drug and cell replacement therapies. Drugs that modulate inflammatory factors, such as rosuvastatin, have been shown to promote the polarization of anti-inflammatory microglia and reduce the inflammatory response caused by traumatic brain injury. Mesenchymal stem cells possess anti-inflammatory properties, and clinical studies have confirmed their significant efficacy and safety in patients with traumatic brain injury. Additionally, advancements in mesenchymal stem cell-delivery methods-such as combinations of novel biomaterials, genetic engineering, and mesenchymal stem cell exosome therapy-have greatly enhanced the efficiency and therapeutic effects of mesenchymal stem cells in animal models. However, numerous challenges in the application of drug and mesenchymal stem cell treatment strategies remain to be addressed. In the future, new technologies, such as single-cell RNA sequencing and transcriptome analysis, can facilitate further experimental studies. Moreover, research involving non-human primates can help translate these treatment strategies to clinical practice.
Collapse
Affiliation(s)
- Liping Shi
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shuyi Liu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Jialing Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Hong Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| |
Collapse
|
2
|
Zhao X, Qiu Y, Liang L, Fu X. Interkingdom signaling between gastrointestinal hormones and the gut microbiome. Gut Microbes 2025; 17:2456592. [PMID: 39851261 PMCID: PMC11776477 DOI: 10.1080/19490976.2025.2456592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/12/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The interplay between the gut microbiota and gastrointestinal hormones plays a pivotal role in the health of the host and the development of diseases. As a vital component of the intestinal microecosystem, the gut microbiota influences the synthesis and release of many gastrointestinal hormones through mechanisms such as modulating the intestinal environment, producing metabolites, impacting mucosal barriers, generating immune and inflammatory responses, and releasing neurotransmitters. Conversely, gastrointestinal hormones exert feedback regulation on the gut microbiota by modulating the intestinal environment, nutrient absorption and utilization, and the bacterial biological behavior and composition. The distributions of the gut microbiota and gastrointestinal hormones are anatomically intertwined, and close interactions between the gut microbiota and gastrointestinal hormones are crucial for maintaining gastrointestinal homeostasis. Interventions leveraging the interplay between the gut microbiota and gastrointestinal hormones have been employed in the clinical management of metabolic diseases and inflammatory bowel diseases, such as bariatric surgery and fecal microbiota transplantation, offering promising targets for the treatment of dysbiosis-related diseases.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ye Qiu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Tfilin Samuel M, Rostovsky I, Kuzmina A, Taube R, Sal-Man N. Engineering non-pathogenic bacteria for auto-transporter-driven secretion of functional interferon. Gut Microbes 2025; 17:2474146. [PMID: 40032826 PMCID: PMC11881866 DOI: 10.1080/19490976.2025.2474146] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/30/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
In recent years, various strategies have been developed to enable the oral administration of protein-based drugs (biologics) with the aim of overcoming the degradation and inactivation of these drugs that can occur as they traverse the gastrointestinal tract (GIT). In this study, we investigated bacteria as a delivery vehicle for biologics, harnessing their ability to withstand the harsh gastric environment and deliver therapeutic drugs directly to the intestine. Specifically, we explored using the type 5 secretion system (T5SS) to secrete therapeutic cargoes under simulated gut conditions. Our research focused on EspC, a T5SS protein from enteropathogenic Escherichia coli, and its potential to secrete interferon-α (IFNα), a cytokine with immunomodulatory and antiviral properties widely used in the clinic. We demonstrated that EspC can facilitate the secretion of IFNα variant when expressed in nonpathogenic bacteria. Moreover, this EspC-secreted IFN was able to activate the JAK-STAT pathway, upregulate IFN-stimulated genes, and induce a robust antiviral response in cells. Collectively, these findings provide proof of concept supporting the utilization of the EspC protein as a novel delivery platform for protein-based therapeutics.
Collapse
Affiliation(s)
- May Tfilin Samuel
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Irina Rostovsky
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alona Kuzmina
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ran Taube
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Neta Sal-Man
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
4
|
Zhou Y, Zhou L, Wang M, Xu L, Li T, Wang C. Shen Shuai II Recipe alleviates renal fibrosis in chronic kidney disease by improving of hypoxia and inhibition of TLR4/MyD88/NF-κB/NLRP3 pathway. Ren Fail 2025; 47:2502875. [PMID: 40384385 PMCID: PMC12090266 DOI: 10.1080/0886022x.2025.2502875] [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: 01/10/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
OBJECTIVES To investigate the anti-fibrotic mechanisms of Shen Shuai II Recipe (SSR) in chronic kidney disease (CKD), focusing on its modulation of hypoxia-associated inflammatory pathways and the TLR4/MyD88/NF-κB/NLRP3 axis. METHODS A 5/6 nephrectomy-induced chronic renal failure (CRF) rat model and hypoxia-exposed human renal tubular epithelial (HK-2) cells were utilized. In vivo, renal function was assessed via serum creatinine, urea nitrogen, and creatinine clearance measurements, alongside histopathological evaluation of renal fibrosis and inflammation. In vitro, hypoxia-treated HK-2 cells were analyzed for fibrotic markers (fibronectin, collagen I, α-smooth muscle actin) and pro-inflammatory cytokines (IL-1β, IL-18). Molecular mechanisms were probed through protein expression analysis of HIF-1α and the TLR4/MyD88/NF-κB pathway, with NLRP3 inflammasome activity evaluated. RESULTS SSR treatment significantly improved renal function in CRF rats, reducing serum creatinine (Scr) and urea nitrogen (BUN) while enhancing creatinine clearance. Histopathology revealed preserved renal architecture with attenuated fibrosis and inflammatory infiltration. In hypoxic HK-2 cells, SSR downregulated fibrotic markers and suppressed IL-1β and IL-18 levels. Mechanistically, SSR reduced HIF-1α expression, inhibited TLR4/MyD88/NF-κB signaling, and suppressed NLRP3 inflammasome activation in both models. CONCLUSIONS SSR alleviates renal fibrosis and CKD progression by mitigating hypoxia-driven inflammation and blocking the TLR4/MyD88/NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Zhou
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Xu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingting Li
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Zhang Y, Ji X, Chang K, Yin H, Zhao M, Zhao L. The regulatory effect of chitooligosaccharides on islet inflammation in T2D individuals after islet cell transplantation: the mechanism behind Candida albicans abundance and macrophage polarization. Gut Microbes 2025; 17:2442051. [PMID: 39694919 DOI: 10.1080/19490976.2024.2442051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
Islet cell transplantation (ICT) represents a promising therapeutic approach for addressing diabetes mellitus. However, the islet inflammation during transplantation significantly reduces the surgical outcome rate, which is related to the polarization of macrophages. Chitooligosaccharides (COS) was previously reported which could modulate the immune system, alleviate inflammation, regulate gut microecology, and repair the intestinal barrier. Therefore, we hypothesized COS could relieve pancreatic inflammation by regulating macrophage polarization and gut microbiota. First, 18S rDNA gene sequencing was performed on fecal samples from the ICT population, showing abnormally increased amount of Candida albicans, possibly causing pancreatic inflammation. Functional oligosaccharides responsible for regulating macrophage polarization and inhibiting the growth of Candida albicans were screened. Afterwards, human flora-associated T2D (HMA-T2D) mouse models of gut microbiota were established, and the ability of the selected oligosaccharides were validated in vivo to alleviate inflammation and regulate gut microbiota. The results indicated that ICT significantly decreased the alpha diversity of gut fungal, altered fungal community structures, and increased Candida albicans abundance. Moreover, Candida albicans promoted M1 macrophage polarization, leading to islet inflammation. COS inhibited Candida albicans growth, suppressed the MyD88-NF-κB pathway, activated STAT6, inhibited M1, and promoted M2 macrophage polarization. Furthermore, COS-treated HMA-T2D mice displayed lower M1 macrophage differentiation and higher M2 macrophage numbers. Additionally, COS also enhanced ZO-1 and Occludin mRNA expression, reduced Candida albicans abundance, and balanced gut microecology. This study illustrated that COS modulated macrophage polarization via the MyD88/NF-κB and STAT6 pathways, repaired the intestinal barrier, and reduced Candida albicans abundance to alleviate islet inflammation.
Collapse
Affiliation(s)
- Yayu Zhang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Xiaoguo Ji
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, China
| | - Kunlin Chang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Hao Yin
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, China
| | - Mengyao Zhao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai, China
| | - Liming Zhao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, China
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai, China
| |
Collapse
|
6
|
Yin X, Wang Y, Li M, Zhang E, Huang L, Yang C. Yi-Qi-Qing-Shi-Hua-Yu method improves uterine inflammation in rats with sequelae of pelvic inflammatory disease through the TLR4/NF-κB signaling pathway and regulates intestinal flora. Tissue Cell 2025; 95:102918. [PMID: 40253799 DOI: 10.1016/j.tice.2025.102918] [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: 11/07/2024] [Revised: 03/04/2025] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Sequelae of pelvic inflammatory disease (SPID) is a common gynecological disease, which is often accompanied by pathological changes and inflammation, and may even lead to infertility. The Yi-Qi-Qing-Shi-Hua-Yu method (YQQSHY), as a traditional Chinese medicine treatment method, is considered to have potential therapeutic effects on SPID. This study will explore the efficacy and potential mechanism of YQQSHY on SPID. This study established a SPID rat model by mixed bacteria and evaluated the regulatory effect of YQQSHY on uterine tissue pathology, immune status, and intestinal flora in SPID rats through histopathology, molecular experiments, immunology, and intestinal flora sequencing analysis. H&E staining showed that YQQSHY significantly reduced the pathological changes and fibrosis in the uterine tissue of SPID rats. ELISA results showed that YQQSHY could significantly reduce the levels of pro-inflammatory cytokines in the serum and vaginal secretions of SPID rats and increase the expression of anti-inflammatory factors. Flow cytometry analysis showed that the YQQSHY treatment group significantly adjusted the proportion of T cells in the peripheral blood of SPID rats. Western blot showed that YQQSHY can regulate TLR4, MyD88, p-NF-KB p65, and induce the transcription of p65 into the nucleus. Immunofluorescence examination of T cell subsets in uterine tissue. In addition, intestinal flora sequencing results showed that YQQSHY significantly modulated the composition and diversity of the intestinal flora of SPID rats. In conclusion, YQQSHY inhibits inflammatory response, regulates T cell ratio, and improves intestinal flora structure through the TLR4/NF-κB signaling pathway, which is the main mechanism for improving uterine inflammation and fibrosis in SPID rats. These findings provide experimental basis and theoretical support for further exploring the application of YQQSHY in clinical treatment.
Collapse
Affiliation(s)
- Xiaolan Yin
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yan Wang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Maoya Li
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Enfeng Zhang
- Chengdu University of Traditional Chinese Medicine, China
| | - Li Huang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Chengcheng Yang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
7
|
Utama K, Khamto N, Janthong A, Thiraphatchotiphum C, Roytrakul S, Kantapan J, Meepowpan P, Sangthong P. Discovery of cinnamoyl-flavonoid hybrid derivatives as inhibitors of SARS-CoV-2 M pro and anti-inflammatory agents: Experimental and in silico insights into their efficacy against lipopolysaccharide-induced lung injury. Eur J Pharmacol 2025; 998:177636. [PMID: 40252899 DOI: 10.1016/j.ejphar.2025.177636] [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: 11/06/2024] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
The chemical structures of the parental compounds of flavonoids from Boesenbergia rotunda were modified by conjugation with cinnamic acid to form cinnamoyl-flavonoid hybrid derivatives with enhanced anti-inflammatory and SARS-CoV-2 Mpro-inhibitory properties. Cinnamoyl-flavonoid hybrid derivatives 6 and 10 showed the potential to inhibit SARS-CoV-2 Mpro with IC50 values of 52.49 and 22.62 μM. Compounds 6 and 10 showed lower cytotoxicity in the human lung cell lines MRC-5 and A549 at concentrations greater than 50 μM. The effects of compounds 6 and 10 on cell viability were studied in a 3D co-culture model of A549 and MRC-5 treated with lipopolysaccharide (LPS) and observed through confocal microscopy. Compounds 6 and 10 downregulated p65 mRNA expression, resulting in a reduction of pro-inflammatory cytokines, including Interleukin 8 (IL-8) and Monocyte Chemoattractant Protein-1 (MCP-1/CCL2), leading to an anti-inflammatory response through Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signalling pathways. Compound 6 showed potential anti-inflammatory activity, downregulating Bcl-2 Associated X gene (BAX), which resulted in inhibition of apoptotic cell death when compared to compound 10. In silico molecular dynamic simulation shed light on how these cinnamoyl-flavonoid hybrid derivatives interact with myeloid differentiation factor 2 (MD-2), which is involved in the inflammatory response. Our findings suggest that cinnamoyl-flavonoid hybrid derivatives show potential as anti-inflammatory drugs and anti-SARS-CoV-2 drugs.
Collapse
Affiliation(s)
- Kraikrit Utama
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand; Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nopawit Khamto
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Atchara Janthong
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai, 50200, Thailand
| | | | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok, 12120, Thailand
| | - Jiraporn Kantapan
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Puttinan Meepowpan
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Materials Science and Technology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Chiang Mai University, 239 Huay Kaew Road, Chiang Mai, 50200, Thailand
| | - Padchanee Sangthong
- Center of Excellence in Materials Science and Technology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
8
|
Zhang J, Wang W, Liu Y, Wan X, Zhang L. Linarin Relieves Apoptosis, Inflammation and Oxidative Stress in LPS-Induced Acute Kidney Injury by Modulating COX2. Clin Exp Pharmacol Physiol 2025; 52:e70048. [PMID: 40491178 DOI: 10.1111/1440-1681.70048] [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: 10/29/2024] [Revised: 04/14/2025] [Accepted: 05/06/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Acute kidney injury (AKI) is a common clinical disease with a rapid decline of renal function. Linarin (LIN) is a flavonoid compound with wide application in different diseases. However, the role and relevant mechanism of LIN in AKI are not fully clear. This study aimed to investigate the function of LIN in modulating the inflammatory response and oxidative stress in lipopolysaccharide (LPS)-induced AKI models and further focused on the interaction between LIN and cyclooxygenase-2 (COX2). METHODS AKI model in vivo was established in mice with 10 mg/kg LPS and 60 mg/kg/day LIN. Also, the AKI model in vitro was constructed in HK2 cells with 10 μg/mL LPS and 40 μM LIN. The TUNEL assay was used for apoptosis detection in tissues. Cell viability and apoptosis were examined using the CCK-8 assay and flow cytometry. Inflammatory factors and oxidative indicators were determined via ELISA and commercial kits. Target screening was carried out using the PPI network and molecular docking. Expression analysis was performed by RT-qPCR and western blot. RESULTS LIN protected kidney tissues from LPS-induced kidney dysfunction and pathological damage in mice. Apoptosis, inflammatory reaction and oxidative stress in LPS-induced mice were restored by LIN. LIN treatment also mitigated kidney cell apoptosis, inflammation and oxidative injury caused by LPS. COX2 (PTGS2) was selected as a hub gene for LIN and AKI through PPI network, and its interaction with LIN was proved by molecular docking. LIN could decrease the COX2 protein expression in LPS-treated HK2 cells. The protective function of LIN from LPS-induced cell injury was achieved by downregulating COX2 level. CONCLUSION These evidences demonstrated that LIN alleviated inflammation and oxidative stress in LPS-stimulated AKI through reducing COX2 protein level.
Collapse
Affiliation(s)
- Jia Zhang
- TCM Pharmacy, Zhejiang Hospital, Hangzhou, China
| | - Wenqi Wang
- Department of Traditional Chinese Medicine Classics, The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou, China
| | - Yafen Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Xiaoqing Wan
- TCM Pharmacy, Zhejiang Hospital, Hangzhou, China
| | - Lin Zhang
- TCM Pharmacy, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
9
|
Fang X, Zhao J, Wu S, Liao P, Guan G. The intestinal toxicity mechanisms of triclosan and triclocarban and their possible clinical nutritional intervention mechanisms. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126396. [PMID: 40345375 DOI: 10.1016/j.envpol.2025.126396] [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: 03/13/2025] [Revised: 04/20/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Triclosan (TCS) and triclocarban (TCC) are widely used as antimicrobial agents in personal care products. Their widespread use has become a potential environmental contaminant. This review reviews the mechanisms of intestinal toxicity of TCS and TCC and their potential nutritional intervention strategies. TCS and TCC can be metabolized to glucuronic acid conjugates in the host and subsequently uncoupled by microorganisms in the intestine to regenerate free forms of TCS and TCC. TCS and TCC are unique metabolic pathways that lead to accumulation in the gut, altering the structure of intestinal flora, increasing the relative abundance of pathogenic bacteria, while reducing the abundance of beneficial bacteria, thereby disrupting the balance of intestinal flora. In addition, they can interfere with the self-renewal and differentiation of ISCs, thereby weakening intestinal barrier function. TCS and TCC can also activate the TLR4-NFκB signaling pathway, inducing and exacerbating inflammatory responses. These mechanisms together lead to intestinal toxicity and have a significant negative impact on intestinal health. In order to cope with the intestinal toxicity caused by these mechanisms of action, this paper believes that prebiotics, probiotics, vitamins, minerals and herbal extracts can be used as potential nutritional interventions to reduce the intestinal toxicity of TCS and TCC by regulating intestinal microbiota, enhancing intestinal barrier function and inhibiting inflammatory response. Although preliminary studies have shown the potential benefits of these interventions, their specific efficacy and safety still need further study.
Collapse
Affiliation(s)
- Xinyu Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Jinfeng Zhao
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Simin Wu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China; Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China
| | - Peng Liao
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan, 410125, China; Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, 410219, China.
| | - Guiping Guan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, 410128, China.
| |
Collapse
|
10
|
Chen G, Wang W, Guan B, Zhang G, Zhang Z, Lin L, Han X, Xu T, Hu C, Pang M, Zhao X, Zhou Y, Li R. Cycloastragenol reduces inflammation in CLP-induced septic MICE by suppressing TLR4 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156645. [PMID: 40319834 DOI: 10.1016/j.phymed.2025.156645] [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: 04/10/2024] [Revised: 06/26/2024] [Accepted: 01/05/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Aggressive systemic inflammation due to activation of macrophage-derived excessive immune responses is a critical cause of sepsis leading to clinical death. The effect of cycloastragenol (CAG) on cecal ligation and puncture (CLP)-induced systemic inflammation in mice with sepsis and the underlying mechanism are still unknown. PURPOSE Here, we firstly investigated the ameliorative functions of CAG in CLP-induced systemic inflammation in sepsis and LPS-mediated inflammatory response, and the impact of Toll-like receptor 4 (TLR4) pathway on the anti-inflammatory effects of CAG. METHODS The in vitro effect of CAG on RAW264.7 cells and THP-1-derived macrophages induced by LPS was detected with quantitative polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA), and Western blotting (WB) assays. In addition, the association of TLR4-MD2 complex with CAG was measured through molecular docking, molecular dynamics (MD) simulation, surface plasmon resonance imaging (SPRi), cellular thermal shift assay (CETSA), immunofluorescence and WB. A specific inhibitor of TLR4 receptor TAK-242 and a TLR4-encoding adenovirus were adopted for verifying the functions of CAG. Meanwhile, the in vivo effects of CAG on cardiopulmonary structure, inflammatory factors and survival of CLP-induced septic mice were analyzed through hematoxylin and eosin staining, qPCR, ELISA, and survival analysis. RESULTS CAG hindered the LPS-induced production of inflammatory mediators like TNF-α, IL-6 and IL-1β within macrophages in vitro. It also inhibited MAPK and NF-κB pathway activation induced by binding of LPS to TLR4 receptor. As suggested by molecular docking results, the MD2-CAG binding energy was -9.53 kcal/mol. During the MD simulation, CAG could tightly bind to the binding pocket of MD2. SPRi revealed that the equilibrium dissociation constant (KD) value for CAG and TLR4 was 5.24× 10-9 M. Moreover, CAG enhanced the thermal stability of TLR4 by approximately 2.68 °C. It further inhibited the binding between LPS-488 and cell membrane receptors. These inhibitory effects of CAG could be partly reversed by TLR4 overexpression and could not increase by specifically blocking TLR4. In vivo, CAG attenuated cardiopulmonary injury and inflammation and improved survival in septic mice dose-dependently. CONCLUSION CAG exerts its anti-inflammatory activity through suppressing MAPK and NF-κB pathway activation caused by TLR4 activation and inhibiting inflammatory factor production dose-dependently.
Collapse
Affiliation(s)
- Guanghong Chen
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Postdoctoral Research Station of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Wanyu Wang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Baoyi Guan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Postdoctoral Research Station of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Guoyong Zhang
- school of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Zhimin Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Liwen Lin
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Xin Han
- school of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Tong Xu
- school of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Changlei Hu
- school of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Mingjie Pang
- school of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China
| | - Xinjun Zhao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China.
| | - Yingchun Zhou
- school of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, 510515, China.
| | - Rong Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China.
| |
Collapse
|
11
|
Wang YX, Ge P, Chen HL. Induction of hyperlipidemic pancreatitis by different fatty acids: A narrative review. World J Gastroenterol 2025; 31:106575. [PMID: 40539203 PMCID: PMC12175850 DOI: 10.3748/wjg.v31.i22.106575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/09/2025] [Accepted: 05/26/2025] [Indexed: 06/12/2025] Open
Abstract
Epidemiological evidence suggests that there is a direct relationship between the degree of obesity and acute pancreatitis severity. Intake of different fatty acids leads to different types of hyperlipidemias. Adipose degradation by pancreatic lipase generates different free fatty acids, which can exacerbate pancreatitis. Saturated fatty acids (SFAs) play an inflammatory role in human metabolic syndrome and obesity, whereas unsaturated fatty acids (UFAs) are "good fats" that are thought to enhance overall health status. However, it appears that serum UFAs correlate with severe acute pancreatitis. Additionally, the "obesity paradox" suggests that UFAs potentially minimize direct harm to the organ. This review provides an in-depth overview of the role of SFAs and UFAs in acute pancreatitis of hyperlipidemia and discusses potential prevention targets for severe acute pancreatitis.
Collapse
Affiliation(s)
- Yu-Xi Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, United States
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian 116027, Liaoning Province, China
| | - Peng Ge
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| | - Hai-Long Chen
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| |
Collapse
|
12
|
Zeng F, Zhu Y, Li R, Chen M, Liang Y, Zhang L, Li Y, Peng S, Li J, Zhou W. Phenolic profiles in the peels of three wampee (Clausena lansium (Lour.) Skeels) fruit cultivars and immune-modulatory activity through the cluster of differentiation 14-mitogen activated protein kinase/nuclear factor kappa B pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119924. [PMID: 40324700 DOI: 10.1016/j.jep.2025.119924] [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: 11/24/2024] [Revised: 03/08/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The peels of wampee (Clausena lansium (Lour.) Skeels) fruits have been ethnomedicinally used to cure malarial fever, coughs, bronchitis, viral hepatitis, and gastrointestinal disorders, suggesting potential immunoprotective and anti-inflammatory properties. AIM OF THE STUDY This study aimed to compare the phenolic profiles in the peels of three wampee fruit cultivars and initially reveal the comprehensive immune-modulatory mechanisms of the phenolic fraction. MATERIALS AND METHODS The fruits of Golden wampee (GLW), Jixin wampee (JXW) and Guifei wampee (GFW) were collected. Their peels (GLP, JXP, GFP) were used to extract crude phenols (GLPE, JXPE, GFPE). JXPF was a phenolic fraction purified from JXPE using preparative high-performance liquid chromatography, which contained the mixture of various phenolic compounds in JXP. Phenolic contents were quantified using HPLC. The binding receptors and immune-modulatory pathways were investigated in LPS-induced RAW 264.7 cells. RESULTS JXP exhibited higher proportions of phenolic compounds, including myricetin-3-O-galactoside, nicotiflorin, rutin, isorhamnetin-3-O-neohesperidoside, isoquercitrin, quercitrin, and 8-hydroxypsoralen. GLPE, JXPE and GFPE reduced LPS-induced overexpression of NO, IL-6 and TNF-α, maintaining these mediators at moderate levels, which indicated immune-modulatory activity. The phenolic compounds in JXPF could occupy the CD14 receptor on macrophage surfaces and finally suppress the phosphorylation of ERK 1/2, p38, JNK, IκBα, and p65 in the MAPK and NF-κB signaling pathways. CONCLUSIONS JXW was identified as a superior cultivar with a higher abundance of phenolic compounds in its peels. The phenols derived from wampee peels demonstrated potential as immune-modulatory agents.
Collapse
Affiliation(s)
- Fanke Zeng
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Yuxiang Zhu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China
| | - Ruyi Li
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China.
| | - Mianhong Chen
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Yuwei Liang
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Li Zhang
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Yingying Li
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Shaodan Peng
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Jihua Li
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China
| | - Wei Zhou
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
13
|
Wang S, Wu Y, Zong W, Wang ZQ. "Yin-Yang" of PARP1 in genotoxic and inflammatory response. DNA Repair (Amst) 2025; 152:103858. [PMID: 40527058 DOI: 10.1016/j.dnarep.2025.103858] [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: 05/05/2025] [Revised: 06/09/2025] [Accepted: 06/09/2025] [Indexed: 06/19/2025]
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1), the founding member of the PARP superfamily, is an enzyme with poly-ADP-ribosyltransferase activity that conducts the majority of poly-ADP-ribosylation (PARylation). PARP1 is the most extensively studied member of the PARP family. It plays a role in various biological processes, particularly in DNA damage repair, including base excision repair, single-strand break repair, double-strand break repair, and maintenance of replication fork stability. Besides DNA damage repair, PARP1 is also involved in the inflammatory response, including, but not limited to, the NF-κB, JAK-STAT, inflammasome assembly, and cGAS-STING signaling pathways. As a scaffold and enzyme, PARP1 and its mediated PARylation induce genotoxic and inflammatory responses to various intracellular and extracellular stressors. Thus, PARP1 has been a target as a pharmaceutical intervention for a range of pathological conditions, including cancer and inflammatory diseases. This review article attempts to provide a comprehensive view of PARP1 as a bridging point between genotoxic and inflammatory responses.
Collapse
Affiliation(s)
- Sijia Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Yukang Wu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Wen Zong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Zhao-Qi Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China; Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Jena 07743, Germany; Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena 07745, Germany.
| |
Collapse
|
14
|
Sun Y, Guo Z, Huo Y, Zhang H, Li T, Wang P, Han W. CMTM7 inhibits TLR4 signaling pathway via promoting Rab5 activation and alleviates acute liver injury. Cell Mol Life Sci 2025; 82:229. [PMID: 40490565 DOI: 10.1007/s00018-025-05748-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/26/2025] [Accepted: 05/09/2025] [Indexed: 06/11/2025]
Abstract
The activation of macrophages mediated by TLR4 is crucial for innate immune responses, while the regulatory mechanisms of TLR4 are still under investigation. This study demonstrates that CMTM7 inhibits TLR4 pathway activation in macrophages and exerts a protective role in acute liver injury (ALI). CMTM7 is highly expressed in monocytes/macrophages, which is downregulated upon LPS stimulation. CMTM7 inhibits LPS/HMGB1-induced activation of the TLR4 pathway in macrophages. Mechanistically, CMTM7 promotes the binding between Rab5 and Gapex5, leading to the generation of GTP-Rab5, which facilitates the internalization and degradation of TLR4, thereby inhibiting TLR4 signaling activation. Utilizing Cmtm7 myeloid conditional knockout mice, we confirmed the protective role of CMTM7 in ALI and highlighted its therapeutic potential through the adoptive transfer of CMTM7-overexpressing macrophages. This study elucidates a novel regulatory mechanism of TLR4 signaling transduction and provides a novel therapeutic strategy for ALI treatment.
Collapse
Affiliation(s)
- Yingzhe Sun
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
- Key Laboratory of Geriatrics (Hepatobiliary Diseases), China General Technology Group, Beijing Electric Power Hospital, State Grid Corporation of China, Capital Medical University, Beijing, China
| | - Zixia Guo
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Yangbo Huo
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Hanxiao Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Ting Li
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China
- Peking University Center for Human Disease Genomics, Beijing, China
- Key Laboratory of Geriatrics (Hepatobiliary Diseases), China General Technology Group, Beijing Electric Power Hospital, State Grid Corporation of China, Capital Medical University, Beijing, China
| | - Wenling Han
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University Health Science Center, Beijing, China.
- Peking University Center for Human Disease Genomics, Beijing, China.
- Key Laboratory of Geriatrics (Hepatobiliary Diseases), China General Technology Group, Beijing Electric Power Hospital, State Grid Corporation of China, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Zhao W, Wang H, Dai C, Li B, Fu Y, Cheng J, Li H. The Erk1/2-EGR1 signaling pathway is involved in lipopolysaccharide-induced transforming growth factor-beta 1 expression in mouse macrophages. Microb Pathog 2025; 203:107453. [PMID: 40057004 DOI: 10.1016/j.micpath.2025.107453] [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: 09/26/2024] [Revised: 02/20/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Numerous studies have demonstrated that lipopolysaccharide (LPS) stimulates TGF-β1 expression. Although studies have implicated the NF-κB/METTL3/METTL14 transactivation/m6A-dependent and AMPK-dependent signaling pathways are engaged in this process in a variety of cell types, the underlying regulatory mechanism in murine macrophages is still not fully understood. To address this issue, in vitro studies were performed using the murine macrophage cell line, RAW264.7. The results showed that LPS challenge resulted in a significant increase in TGF-β1 expression at both mRNA and protein levels. Subsequent studies revealed that the MAPK (including p38, Erk1/2, and JNK) and NF-κB signaling pathways were activated in response to LPS stimulation, but only blocking the Erk1/2 singling pathway completely abolished LPS-induced TGF-β1 expression. Further studies revealed that the levels of a downstream regulator of the Erk1/2 pathway, EGR1, were significantly increased after LPS treatment, and its knockdown significantly reduced LPS-induced Tgf-β1 expression levels. Finally, dual luciferase reporter and ChIP-PCR assays confirmed that EGR1 is a key transcription factor in the regulation of Tgf-β1 expression by binding to its promoter region in response to LPS stimulation. In conclusion, we elucidated the molecular events by which LPS regulates TGF-β1 expression in murine macrophages through the Erk1/2-EGR1 signaling pathway. These findings provide a conceptually novel pathway for LPS-induced TGF-β1 expression beyond the known NF-κB/METTL3/METTL14 transactivation/m6A-dependent and AMPK-dependent signaling pathways.
Collapse
Affiliation(s)
- Weimin Zhao
- Jiangsu Province Engineering Research Center of Precision Animal Breeding, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Hong Wang
- Jiangsu Province Engineering Research Center of Precision Animal Breeding, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chaohui Dai
- Jiangsu Province Engineering Research Center of Precision Animal Breeding, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Bixia Li
- Jiangsu Province Engineering Research Center of Precision Animal Breeding, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Yanfeng Fu
- Jiangsu Province Engineering Research Center of Precision Animal Breeding, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Jinhua Cheng
- Jiangsu Province Engineering Research Center of Precision Animal Breeding, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China.
| | - Hui Li
- Jiangsu Province Engineering Research Center of Precision Animal Breeding, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
16
|
Zhang Y, Ni P, Chen H, Tang L, Song H, Wen H, Miao Y, Li W, Li X. Vitamin D 3 ameliorates hyperglycemia by modulating gut microbiota and metabolites in prediabetic KKay mice. Food Res Int 2025; 211:116369. [PMID: 40356099 DOI: 10.1016/j.foodres.2025.116369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/16/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Prediabetes represents a pivotal stage in the development and pathogenesis of diabetes, during which notable alterations in the gut microbiota can be observed. Vitamin D (VD) showed anti-diabetic properties, but it is unknown whether the improvement of VD on hyperglycemia is associated with gut microbiota. Thus, our objective was to investigate and verify the effects of VD3 on glucose metabolism in prediabetes, as well as to elucidate the underlying mechanisms. In this study, different concentrations of VD3 were intraperitoneally administered to prediabetic mice induced by high fat diet for 16 weeks. Biochemical analyses, oral glucose tolerance test, 16S rRNA and untargeted metabolomics were used, the mechanism was explored. Then, fecal suspensions collected from the above donors were transplanted into KKay mice for 6 weeks, and the relevant indicators were measured. The results showed that VD3 intervention alleviated glucose metabolism in KKay mice. It increased the protein expression of colon tight junction proteins, alleviated metabolic endotoxemia and inflammation, so that reduced tumor necrosis factor alpha (TNFα) induced toll-like receptor 4/nuclear factor kappa-B (TLR4/NFκB) and improvement of phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) insulin signaling pathway. VD3 affected the structure of gut microbiota and metabolites, and functional prediction analysis suggested that VD3 may affect carbohydrate. Besides, the effect of VD3 could be delivered by fecal microbiota transplantation (FMT). Consequently, VD3 ameliorate glucose metabolism by modulating gut microbiota and metabolites in KKay mice, and this ability could be transferred by FMT.
Collapse
Affiliation(s)
- Yujing Zhang
- Department of Food Hygiene and Nutrition, School of Medicine, Henan University of Chinese Medicine, Zhengzhou 450000, Henan, China; Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Peng Ni
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Hao Chen
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Lulu Tang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Hanlu Song
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Hongwei Wen
- Department of Public Health, Zhengzhou Shuqing Medical College, Zhengzhou 450000, Henan, China
| | - Yufan Miao
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Wenjie Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China.
| | - Xing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China.
| |
Collapse
|
17
|
Gao M, Meng X, Cui Y, Deng L, Zhang X, Liu C, Fu R. Role of Macrophage TLR4 Expression in the Immunopathogenesis of Severe Aplastic Anemia. J Clin Lab Anal 2025; 39:e70055. [PMID: 40481763 DOI: 10.1002/jcla.70055] [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: 03/04/2025] [Revised: 04/16/2025] [Accepted: 05/06/2025] [Indexed: 06/22/2025] Open
Abstract
BACKGROUND Severe aplastic anemia (SAA) is a life-threatening hematologic disorder characterized by bone marrow failure and impaired immunity. PURPOSE Investigating the role of Toll-like receptor 4 (TLR4) highly expressing macrophages in the immunopathogenesis of SAA. METHODS Macrophage TLR4 expression levels were detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting (WB). Knocked down or inhibited macrophage TLR4 expression, detected the pyroptosis (IL-1β, IL-18, NLRP3, caspase-1, gasdermin D) levels by RT-qPCR and WB. Using RNA sequencing to find differential genes and pathways. Co-cultured CD8+ T cells with macrophages that knocked down or inhibited TLR4, and the levels of perforin and granzyme B expression in CD8+ T cells were detected by flow cytometry. CD8+ T cells were further co-cultured with K562, and the apoptosis rate of K562 was detected. RESULTS The TLR4 in the bone marrow macrophages of patients with untreated SAA were significantly higher than those in the remission and control groups, and were negatively correlated with clinical indicators. RNA sequencing of macrophages with TLR4 knockdown showed that differentially expressed genes were enriched in the innate immune and inflammatory chemotaxis signaling pathways. After TLR4 knockout or TLR4 inhibitor addition in bone marrow macrophages of patients with untreated SAA, the mRNA and protein expression levels of pyroptosis markers interleukin (IL)-1β, IL-18, NLRP3, caspase-1, and gasdermin D were significantly lower than those in the control group. When CD8+ T cells were co-cultured with TLR4-knocked-down or inhibitor-added macrophages, the expression of perforin and granzyme B in CD8+ T cells was significantly reduced, and CD8+ T cell cytotoxicity decreased. CONCLUSIONS Inhibition of macrophage TLR4 expression in SAA patients could alleviate the over-activated cellular immune response in SAA patients by decreasing the level of pyroptosis.
Collapse
Affiliation(s)
- Mengran Gao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Cancer Hematopoietic Cloning Prevention and Treatment, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Xiaorui Meng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Cancer Hematopoietic Cloning Prevention and Treatment, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Yi Cui
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Cancer Hematopoietic Cloning Prevention and Treatment, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Ling Deng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Cancer Hematopoietic Cloning Prevention and Treatment, Tianjin, China
| | - Xinrui Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Cancer Hematopoietic Cloning Prevention and Treatment, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Chunyan Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Cancer Hematopoietic Cloning Prevention and Treatment, Tianjin, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Cancer Hematopoietic Cloning Prevention and Treatment, Tianjin, China
| |
Collapse
|
18
|
Wang Q, Xu F, Cao Y, Li B, Zhang S, Zhang Y. Peptide GTSFTTTAER From Rapana venosa Alleviates TNBS-Induced Inflammatory Bowel Disease in a Zebrafish Model via Multi-Pathway Regulation. Food Sci Nutr 2025; 13:e70427. [PMID: 40521083 PMCID: PMC12162357 DOI: 10.1002/fsn3.70427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/13/2025] [Accepted: 05/31/2025] [Indexed: 06/18/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a refractory chronic intestinal disease caused by immune dysfunction, with an unknown pathogenesis. In this study, the peptide GTSFTTTAER was isolated from Rapana venosa for treatment of IBD for the first time. We examined its protective effects on a zebrafish model of 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced IBD. The results indicate that the peptide GTSFTTTAER ameliorates intestinal inflammatory injury by reducing the number of immune cells at the intestinal site and increasing the frequency of intestinal peristalsis. Besides, in order to predict and verify the potential mechanism of the anti-inflammatory effects of peptide GTSFTTTAER, we performed transcriptome and reverse transcription-quantitative polymerase chain reaction analysis. The transcriptome analysis revealed that the key pathways for the potential protective effects of GTSFTTTAER were the Toll-like receptor signaling pathway and the necroptosis pathway. Lastly, molecular docking technology further confirmed the action target of peptide GTSFTTTAER. In conclusion, GTSFTTTAER has a beneficial effect on IBD in TNBS-induced zebrafish. Our study will provide a valuable reference for the utilization of peptide GTSFTTTAER from Rapana venosa, and it may also be helpful in developing therapeutic agents for IBD.
Collapse
Affiliation(s)
- Qifei Wang
- Biology InstituteQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Fenghua Xu
- Biology InstituteQilu University of Technology (Shandong Academy of Sciences)JinanChina
- Research Center of Translational MedicineCentral Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Yongna Cao
- Biology InstituteQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Bin Li
- Biology InstituteQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Shanshan Zhang
- Biology InstituteQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Yun Zhang
- Biology InstituteQilu University of Technology (Shandong Academy of Sciences)JinanChina
| |
Collapse
|
19
|
Baral H, Kaundal RK. Novel insights into neuroinflammatory mechanisms in traumatic brain injury: Focus on pattern recognition receptors as therapeutic targets. Cytokine Growth Factor Rev 2025; 83:18-34. [PMID: 40169306 DOI: 10.1016/j.cytogfr.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/14/2025] [Indexed: 04/03/2025]
Abstract
Traumatic brain injury (TBI) is a major global health concern and a leading cause of morbidity and mortality. Neuroinflammation is a pivotal driver of both the acute and chronic phases of TBI, with pattern recognition receptors (PRRs) playing a central role in detecting damage-associated molecular patterns (DAMPs) and initiating immune responses. Key PRR subclasses, including Toll-like receptors (TLRs), NOD-like receptors (NLRs), and cGAS-like receptors (cGLRs), are abundantly expressed in central nervous system (CNS) cells and infiltrating immune cells, where they mediate immune activation, amplify neuroinflammatory cascades, and exacerbate secondary injury mechanisms. This review provides a comprehensive analysis of these PRR subclasses, detailing their distinct structural characteristics, expression patterns, and roles in post-TBI immune responses. We critically examine the molecular mechanisms underlying PRR-mediated signaling and explore their contributions to neuroinflammatory pathways and secondary injury processes. Additionally, preclinical and clinical evidence supporting the therapeutic potential of targeting PRRs to mitigate neuroinflammation and improve neurological outcomes is discussed. By integrating recent advancements, this review offers an in-depth understanding of the role of PRRs in TBI pathobiology and underscores the potential of PRR-targeted therapies in mitigating TBI-associated neurological deficits.
Collapse
Affiliation(s)
- Harapriya Baral
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| |
Collapse
|
20
|
Ko B, Son J, In Won J, Kang BM, Choi CW, Kim R, Sung JH. Gut microbe-skin axis on a chip for reproducing the inflammatory crosstalk. LAB ON A CHIP 2025; 25:2609-2619. [PMID: 40042226 DOI: 10.1039/d4lc01010h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
The gut-skin axis has emerged as a crucial mediator of skin diseases, with mounting evidence highlighting the influence of gut microbiota on skin health. However, investigating these mechanisms has been hindered by the lack of experimental systems that enable direct study of gut microbiota-skin interactions. Here, we present the gut microbe-skin chip (GMS chip), a novel microfluidic platform designed to model microbiome-gut-skin axis interactions. The GMS chip allows the coculture of intestinal epithelial cells (Caco-2), human epidermal keratinocytes (HEKa), and gut microbes with fluidic connection mimicking the blood flow. We validated that the gut compartment, with a self-sustaining oxygen gradient, enabled coculturing gut bacteria such as Escherichia coli (E. coli) and Lactobacillus rhamnosus GG (LGG), and the skin cells properly differentiated in the chip in the presence of fluid flow. Disruption of intestinal epithelial integrity by dextran sodium sulfate (DSS) combined with lipopolysaccharides (LPS) selectively decreased skin cell viability while sparing gut cells. Notably, pretreatment with LGG showed a protective effect against the skin cell damage by enhancing the intestinal barrier function. The GMS chip effectively recapitulates the influence of gut microbiota on skin health, representing a pivotal step forward in studying gut-skin axis mechanisms and the role of the gut microbiome in skin diseases.
Collapse
Affiliation(s)
- Byungho Ko
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Jimin Son
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Jong In Won
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Bo Mi Kang
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Chong Won Choi
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong, 30016, Republic of Korea.
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| |
Collapse
|
21
|
Dou JY, Wu YN, Gao C, Zheng S, Wang CY, Dai X, Lian LH, Cui ZY, Nan JX, Wu YL. Ginseng and Platycodon grandiflorum ameliorated pulmonary fibrosis and inflammation targeting TLR4-P2X7r/NLRP3 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119913. [PMID: 40318772 DOI: 10.1016/j.jep.2025.119913] [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: 03/23/2025] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng and Platycodon grandiflorum (Jacq.) A. DC. (PG) are traditional Chinese herb medicine and classified into the lung meridian, which traditionally used to treat respiratory disorders. AIM OF THE STUDY This study investigated the protective mechanisms of ginseng and PG against pulmonary fibrosis. MATERIALS AND METHODS Panax ginseng C.A.Mey. (GS), Ginseng Radix et Rhizoma Rubra (RGR), PG and GS + PG extracts were prepared using aqueous or ethanol extraction methods and analyzed by HPLC. Cigarette smoke (CS)-induced pulmonary fibrosis mice were administrated with GS, RGR, PG, GS + PG aqueous extracts or platycodin D (PD, the major active component of PG), respectively. A549 were stimulated with different stimulators TGF-β, LPS + ATP or conditioned medium from LPS-primed THP-1 (CM), then cultured with PG, PD or A438079 (P2X7r antagonist), respectively. RESULTS In CS-exposed mice, GS, RGR, PG, or GS + PG extracts significantly reduced lung index elevation without effects on kidney, cardiac or liver indices. These extracts ameliorated CS-induced alveolar wall thickening, extracellular matrix (ECM) accumulation, inflammation, and inhibited P2X7r/NLRP3, TLR4/IRAK4, and NF-κB/IκB-α. PG or PD significantly alleviated lung injury and ECM deposition in CS-exposed mice. PG or PD inhibited CS-induced inflammatory cytokine secretion and immune cell recruitment by TLR4-P2X7r/NLRP3 blockade. In CM-stimulated A549, PG or PD significantly reduced ECM accumulation and inflammatory factors release. PG blocked CM-triggered TLR4-P2X7r/NLRP3 activation in A549, with similar functioning as A438079. CONCLUSIONS GS and PG ameliorated pulmonary fibrosis via TLR4-P2X7r/NLRP3. PG and PD regulated cell crosstalk in alveolar microenvironment against pulmonary injury, which might be novel therapeutic strategy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Jia-Yi Dou
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Yu-Nuo Wu
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Chong Gao
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Shuang Zheng
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Chen-Yu Wang
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Xu Dai
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Li-Hua Lian
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Zhen-Yu Cui
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Ji-Xing Nan
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| | - Yan-Ling Wu
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), Key Laboratory of Natural Medicines of the Changbai Mountain (Ministry of Education), College of Pharmacy, Yanbian University, Yanji, Jilin Province, 133002, PR China.
| |
Collapse
|
22
|
Chen Y, Zhang XW, Zhao MM, Li L, Liu Y, Wei TT, Yu W, Han B, Liu ZP, Zeng KW. Chlorogenic acid targets SLC37A2 to inhibit macrophage activation via ER-dependent NF-κB and NLRP3 signaling pathways against sepsis-induced acute lung injury. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-22. [PMID: 40423577 DOI: 10.1080/10286020.2025.2506181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 05/10/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025]
Abstract
Sepsis-induced acute lung injury (SI-ALI) requires urgent treatment due to severe inflammation. Our study found chlorogenic acid (CGA) suppressed LPS-induced macrophage activation by lowering NO, TNF-α, and IL-6. TPP-based strategies identified SLC37A2 as the direct target of CGA, validated by CETSA/MST. Molecular docking indicated CGA-SLC37A2 hydrogen bonding. CGA alleviated endoplasmic reticulum stress via SLC37A2, inhibiting TLR4/NF-κB and NLRP3 pathways to reduce inflammation. In SI-ALI mice and zebrafish models, CGA mitigated lung injury through these mechanisms taken together. This work highlights the therapeutic potential of CGA for SI-ALI and the critical role of SLC37A2 in combating infectious pneumonia.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization Ministry of Education, Shihezi 832003, China
| | - Xiao-Wen Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mei-Mei Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ling Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yang Liu
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Tian-Tian Wei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wei Yu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization Ministry of Education, Shihezi 832003, China
| | - Bo Han
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization Ministry of Education, Shihezi 832003, China
| | - Zheng-Ping Liu
- Shandong Academy of Pharmaceutical Sciences, Jinan 250101, China
- Shandong Engineering Research Center of New sustained and controlled release formulations and drug targeted delivery systems, Jinan 250101, China
| | - Ke-Wu Zeng
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization Ministry of Education, Shihezi 832003, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
23
|
Li Y, Tang T, Sun Y, Chen G, Yuan X, Cai D. The role of TLR-4 in chemoresistance of cancer. Discov Oncol 2025; 16:865. [PMID: 40404908 PMCID: PMC12098248 DOI: 10.1007/s12672-025-02509-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/25/2025] [Indexed: 05/24/2025] Open
Abstract
Chemotherapy, which aims to eradicate tumor cells and enhance patient survival, is a prevalent approach for tumor treatment. Nevertheless, recurrence and drug resistance resulting from consecutive chemotherapy regimens have emerged as significant factors contributing to the high fatality rates among cancer patients. Numerous studies have revealed that chemicals discharged by injured and deceased cells can trigger the host repair program mediated by toll-like receptor-4 (TLR-4), enhancing tumor resistance. TLR-4 is not only expressed in immune cells but also in various malignant tumor cells, especially inflammation-associated tumor cells, and plays a crucial role in tumor formation, development, and chemoresistance. Endogenous ligands are released upon the killing of tumor cells by chemotherapy drugs, binding to and activating TLR-4, subsequently activating downstream NF-κB and other essential molecules, leading to the release of multiple factors associated with tumor proliferation and invasion, creating a microenvironment conducive to local recurrence and metastasis, and promoting tumor progression and drug resistance. This review assessed studies on the resistance of several tumor cells to commonly utilized anticancer treatments induced by TLR-4 to better comprehend the phenomena and mechanism of TLR-4-dependent resistance, as well as to put forward suggestions and insights for overcoming tumor resistance.
Collapse
Affiliation(s)
- Yuhua Li
- Department of Pharmacy, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, People's Republic of China
- Department of Pharmacy, The First Naval Force Hospital of Southern Theatre Command, Zhanjiang, 524005, Guangdong, People's Republic of China
| | - Tianle Tang
- Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Yang Sun
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Gui'e Chen
- Department of Pharmacy, The First Naval Force Hospital of Southern Theatre Command, Zhanjiang, 524005, Guangdong, People's Republic of China
| | - Xinrong Yuan
- Department of Gynecology and Obstetrics, The First Naval Force Hospital of Southern Theatre Command, Zhanjiang, 524005, Guangdong, People's Republic of China.
| | - De Cai
- Department of Pharmacy, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, People's Republic of China.
| |
Collapse
|
24
|
Zitta K, Berndt R, Hess K, Fändrich F, Gurvich O, Sirviö K, Kekarainen T, Hummitzsch L, Wong YL, Sattler O, Braem S, Krebs M, Fudickar A, Engels S, Flack N, Steinfath M, Albrecht M. Transcriptomic characterization of GMP-compliant regulatory macrophages (TRI-001) under inflammatory and hypoxic conditions: a comparative analysis across macrophage subtypes. J Transl Med 2025; 23:551. [PMID: 40380234 DOI: 10.1186/s12967-025-06548-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/29/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Regulatory macrophages (Mreg) represent a unique subset of macrophages known for their angiogenic and anti-inflammatory properties, positioning them as promising candidates for cell-based therapies. Recently, we have differentiated and characterized a distinct Mreg subtype (TRI-001), which is currently being produced in accordance with good manufacturing practice (GMP) for a multicenter study aimed at treating patients with peripheral arterial occlusive disease (PAOD). AIM OF THE STUDY To compare the transcriptome of TRI-001 with various in vitro differentiated macrophage subtypes to provide a comprehensive context for TRI-001 within the macrophage landscape. Additionally, we aimed to develop a detailed transcriptome profile of TRI-001 under transient hypoxic and inflammatory conditions, mimicking the microenvironment in PAOD patients. METHODS Mreg were differentiated from human CD14 + monocytes using a GMP-compliant protocol and identified as TRI-001 by flow cytometry. Hypoxia was induced via an enzymatic model, while LPS treatment of TRI-001 was employed as inflammatory stimulus. Transcriptomic profiling was conducted using the Illumina HiSeq 4000 platform. In vitro cell migration assays (Oris assays) were conducted using human umbilical vein endothelial cells (HUVEC) cultured with supernatants derived from normoxia and hypoxia treated TRI-001. RESULTS TRI-001 demonstrated significant transcriptomic similarities with Mreg and Mreg_UKR but were different from M0, M1, M2a, and PCMO subtypes. Under hypoxic conditions and LPS stimulation, TRI-001 displayed distinct gene expression profiles compared to TRI-001 under control conditions, with hypoxic and LPS-stimulated profiles showing notable overlap. Pathway enrichment analysis suggested the activation of chemotaxis and migration-associated pathways especially under hypoxic conditions. Findings from functional in vitro cell migration assays were inconclusive, as the secretome of TRI-001, whether cultured under hypoxic or normoxic conditions, did not elicit a significant effect on endothelial cell migration. CONCLUSION TRI-001 represents a novel type of regulatory macrophages (Mreg). The distinctive transcriptional responses to hypoxia and inflammatory stimuli highlight its potential as a cell therapy for the treatment of PAOD patients.
Collapse
Affiliation(s)
- Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig- Holstein, Schwanenweg 21, Kiel, Germany
| | - Rouven Berndt
- Clinic of Vascular Medicine, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Vascular Research Center, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Katharina Hess
- ICRSM Institute for Clinical Research and Systems Medicine, HMU Health and Medical University, Potsdam, Germany
| | - Fred Fändrich
- Department for Applied Cell Therapy, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | | | | | - Lars Hummitzsch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig- Holstein, Schwanenweg 21, Kiel, Germany
| | - Yuk Lung Wong
- Vascular Research Center, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Ole Sattler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig- Holstein, Schwanenweg 21, Kiel, Germany
| | | | - Mark Krebs
- 3D-PharmXchange, Tilburg, The Netherlands
| | - Axel Fudickar
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig- Holstein, Schwanenweg 21, Kiel, Germany
| | | | | | - Markus Steinfath
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig- Holstein, Schwanenweg 21, Kiel, Germany
| | - Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig- Holstein, Schwanenweg 21, Kiel, Germany.
| |
Collapse
|
25
|
Mafe AN, Büsselberg D. The Effect of Microbiome-Derived Metabolites in Inflammation-Related Cancer Prevention and Treatment. Biomolecules 2025; 15:688. [PMID: 40427581 PMCID: PMC12109317 DOI: 10.3390/biom15050688] [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: 03/05/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Chronic inflammation plays a crucial role in cancer development, yet the mechanisms linking the microbiome to inflammation-related carcinogenesis remain unclear. Emerging evidence suggests that microbiome-derived metabolites influence inflammatory pathways, presenting both challenges and opportunities for therapy. However, a deeper understanding of how these metabolites regulate inflammation and contribute to cancer prevention is still needed. This review explores recent advances in microbiome-derived metabolites and their roles in inflammation-related carcinogenesis. It highlights key molecular mechanisms, emerging therapies, and unresolved challenges. Synthesizing current research, including clinical trials and experimental models, bridges the gap between microbiome science and cancer therapy. Microbial metabolites such as short-chain fatty acids (SCFAs), polyamines, indoles, and bile acids play vital roles in regulating inflammation and suppressing cancer. Many metabolites exhibit potent anti-inflammatory and immunomodulatory effects, demonstrating therapeutic potential. Case studies show promising results, but challenges such as metabolite stability, bioavailability, and individual variability remain. Understanding microbiome-metabolite interactions offers novel strategies for cancer prevention and treatment. This review identifies knowledge gaps and proposes future research directions to harness microbiome-derived metabolites for innovative cancer therapies. Addressing these issues may pave the way for microbiome-targeted cancer interventions.
Collapse
Affiliation(s)
- Alice N. Mafe
- Department of Biological Sciences, Faculty of Sciences, Taraba State University, Main Campus, Jalingo 660101, Taraba State, Nigeria;
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha Metropolitan Area, Al Rayyan P.O. Box 22104, Qatar
| |
Collapse
|
26
|
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
|
27
|
Tsai YS, Niemietz I, Sundqvist M, Brown KL. No-lyse no-wash flow cytometry method for ex vivo study of human neutrophils. J Leukoc Biol 2025; 117:qiaf050. [PMID: 40267231 DOI: 10.1093/jleuko/qiaf050] [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: 10/30/2024] [Revised: 02/04/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025] Open
Abstract
Neutrophils are vital immune cells involved in immunity via their effector roles and death. While their rapid response to the environment is crucial for immune function, it also makes them difficult to study. Here, we provide a no-lyse no-wash (NLNW) flow cytometry protocol to quantify neutrophil viability and death in whole blood with minimal sample handling. Viability of neutrophils was measured by Annexin V/7AAD staining in whole blood at 0 and 20 h. Associated cost, sample volume, and neutrophil survival were compared with traditional density-gradient isolated neutrophils. The NLNW analysis of whole blood immediately after collection showed ∼85% viable neutrophils. After 20 h of incubation, viability dropped to ∼70%, yet was significantly greater compared with isolated neutrophils, where half the population (∼44%) entered early-stage apoptosis. The NLNW flow cytometry method is an efficient and cost-effective method to measure neutrophil viability in small volumes of whole blood with minimal sample handling.
Collapse
Affiliation(s)
- Yun-Shan Tsai
- Department of Microbiology & Immunology, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| | - Iwona Niemietz
- Department of Microbiology & Immunology, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| | - Martina Sundqvist
- Department of Pediatrics, The University of British Columbia, 4480 Oak Street, Vancouver, BC, Canada V6H 0B3
| | - Kelly L Brown
- Department of Pediatrics, The University of British Columbia, 4480 Oak Street, Vancouver, BC, Canada V6H 0B3
- Centre for Blood Research, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
- British Columbia Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| |
Collapse
|
28
|
Burns-Ray EC, Dasinger JH, Cherian-Shaw M, Walton SD, Baldwin KE, Cormier AB, Hasan S, Fehrenbach DJ, Abais-Battad JM, Mattson DL. Impact of Hematopoietic CD14 on Oxidative Stress during Salt-Sensitive Hypertension and Kidney Injury. J Am Soc Nephrol 2025:00001751-990000000-00642. [PMID: 40327837 DOI: 10.1681/asn.0000000717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/29/2025] [Indexed: 05/08/2025] Open
Abstract
Key Points
Deletion of CD14 in Dahl salt-sensitive females increased NADPH oxidase 2 oxidative stress in macrophages, exacerbated hypertension, and worsened kidney injury.Double knockout of CD14 and functional NADPH oxidase 2 via
Ncf2 deletion ameliorated the exacerbated salt-sensitive phenotype of female salt-sensitiveCD14−/−.Immune-specific double knockout of CD14 and p67phox via bone marrow transfer markedly attenuated hypertension and kidney damage.
Background
In high-salt diet-fed Dahl salt-sensitive rats, increased Cd14 expression by infiltrating macrophages accompanies hypertension and kidney damage. Interestingly, genetic deletion of Cd14 in the Dahl salt-sensitive rat model (SSCD14−/−) conferred a significant exacerbation of salt-induced hypertension and associated kidney disease in females. We speculated that CD14 may function to modulate the production of reactive oxygen species in macrophages, since previous findings implicated hematopoietic NADPH oxidase 2 (NOX2)–derived superoxide in the pathogenesis of salt-sensitive hypertension. This study tested the hypothesis that the amplification of salt-sensitive hypertension and kidney damage in salt-sensitive females lacking CD14 is dependent on NOX2.
Methods
A double knockout salt-sensitive rat was bred to lack both CD14 and the critical p67phox subunit of NOX2 (SSCD14−/−p67phox−/−). In vivo and ex vivo experiments explored the consequences of CD14 deletion in addition to nonfunctional NOX2 on salt-induced BP, kidney damage, and inflammation. Complementary experiments using diseased human kidneys correlated our findings to humans.
Results
We found that SSCD14−/− peritoneal macrophages demonstrated increased NOX2 subunit mRNA and a greater capacity to produce NOX2-superoxide versus genetically intact Dahl salt-sensitive rats, whereas SSCD14−/−p67phox−/− macrophages did not produce superoxide. We also showed that SSCD14−/−p67phox−/− females had significantly attenuated salt-induced hypertension, kidney damage, and renal inflammation compared with SSCD14−/−, indicating that NOX2 mediates the exacerbation of disease in CD14 absence. A subsequent total body irradiation/bone marrow transfer approach using Dahl salt-sensitive female recipients and transfer of SSCD14−/−p67phox−/− bone marrow recapitulated these findings, demonstrating a clear interplay of CD14 and NOX2 specifically in immune cells. Finally, consistent with observations in the Dahl salt-sensitive rats, immunohistochemical observations in human tissue demonstrated a positive correlation between kidney damage, CD68+ macrophages, and CD14 in the kidney.
Conclusions
We highlight a novel regulatory pathway whereby hematopoietic CD14 influences NOX2-derived oxidative stress and therefore modulates the progression of hypertension and kidney disease.
Collapse
Affiliation(s)
- Emily C Burns-Ray
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - John Henry Dasinger
- Department of Physiology, School of Medicine at Tulane University, New Orleans, Louisiana
| | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Samuel D Walton
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Kaitlyn E Baldwin
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ann B Cormier
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Sadaf Hasan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Daniel J Fehrenbach
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
29
|
Pan L, Xie L, Yang W, Feng S, Mao W, Ye L, Cheng H, Wu X, Mao X. The role of brain-liver-gut Axis in neurological disorders. BURNS & TRAUMA 2025; 13:tkaf011. [PMID: 40321299 PMCID: PMC12048006 DOI: 10.1093/burnst/tkaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 05/08/2025]
Abstract
In recent years, with the increasing volume of related research, it has become apparent that the liver and gut play important roles in the pathogenesis of neurological disorders. Considering the interactions among the brain, liver, and gut, the brain-liver-gut axis has been proposed and gradually recognized. In this article, we summarized the complex network of interactions within the brain-liver-gut axis, encompassing the vagus nerve, barrier permeability, immunity and inflammation, the blood-brain barrier, gut microbial metabolites, the gut barrier, neurotoxic metabolites, and beta-amyloid (Aβ) metabolism. We also elaborated on the impact of the brain-liver-gut axis on various neurological disorders. Furthermore, we outline several therapies aimed at modulating the brain-liver-gut axis, including antibiotics, probiotics and prebiotics, fecal microbiota transplantation (FMT), vagus nerve stimulation (VNS), and dietary interventions. The focus is on elucidating possible mechanisms underlying neurological disorders pathogenesis and identifying effective treatments that are based on our understanding of the brain-liver-gut axis.
Collapse
Affiliation(s)
- Li Pan
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Lizheng Xie
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| | - Wenpei Yang
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| | - Shi Feng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Wenbao Mao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Lei Ye
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Hongwei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xiao Wu
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xiang Mao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| |
Collapse
|
30
|
Wang X, Kong X, Ding Y, An M, Zhu X, Guan Y, Niu Y. Inverted day-night feeding during pregnancy affects the brain health of both maternal and fetal brains through increasing inflammation levels associated with dysbiosis of the gut microbiome in rats. J Neuroinflammation 2025; 22:130. [PMID: 40317047 PMCID: PMC12048959 DOI: 10.1186/s12974-025-03447-x] [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: 12/24/2024] [Accepted: 04/15/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND In both humans and rodents, inappropriate feeding times during pregnancy can cause maternal metabolic abnormalities, increasing the risk of neurodevelopmental disorders in both the mother and offspring. Using a rat model, this study investigates whether feeding only during the inactive phase in rats leads to anxiety-like behaviors and abnormal brain development in fetuses through gut microbiota imbalance. METHODS 10-week-old female rats in the inactive-phase feeding group (IF group) were first trained for daytime feeding, ensuring that energy intake was statistically insignificant and different from that of the normal diet feeding group (ND group). They were then paired with male rats, and the previous feeding regimen was continued after pregnancy. Anxiety-like behavior was evaluated using the open-field test. Maternal caecal microbiota was analyzed using 16S rRNA sequencing. Enzyme-linked immunosorbent assay (ELISA) measured serum inflammation factors. RT-PCR was employed to assess mRNA levels of integrity genes and inflammatory cytokines in the maternal hippocampi, intestines, fetal brains, and placentae. RESULTS There were no statistically significant differences in energy intake or body weight gain between the IF and ND groups. In the open field test, dams in the IF group exhibited anxiety-like behavior, as indicated by fewer entries into and shorter duration in the central zone. Active-phase fasting elevated maternal serum inflammatory cytokine levels and impaired antioxidant capacity. It also increased intestinal permeability and induced gut microbiota dysbiosis, characterized by a decrease in Akkermansia and an increase in Dubosiella. Changes in the expression of intestinal circadian genes and elevated intestinal inflammatory cytokines were observed. Lipopolysaccharide (LPS) translocated into the maternal circulation, activated Toll-like receptor 4 (TLR 4), and passed through the compromised placental barrier into the fetal brain, leading to increased expression of inflammatory cytokines in the fetal brain. CONCLUSIONS The misalignment between maternal feeding time and the biological clock during pregnancy disrupts the balance of the gut microbiota and peripheral rhythms. The impaired intestinal and placental barriers allow LPS from the gut to infiltrate the maternal hippocampus and fetal brain, increasing inflammation and impacting both maternal and fetal brain health.
Collapse
Affiliation(s)
- Xinyue Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Xiangju Kong
- Department of Gynaecology and Obstetrics, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yibo Ding
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Mengqing An
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Xuan Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Yue Guan
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China.
| | - Yucun Niu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China.
| |
Collapse
|
31
|
Schultz TE, Mathmann CD, Domínguez Cadena LC, Muusse TW, Kim H, Wells JW, Ulett GC, Hamerman JA, Brooks AJ, Kobe B, Sweet MJ, Stacey KJ, Blumenthal A. TLR4 endocytosis and endosomal TLR4 signaling are distinct and independent outcomes of TLR4 activation. EMBO Rep 2025; 26:2740-2766. [PMID: 40204912 PMCID: PMC12116916 DOI: 10.1038/s44319-025-00444-2] [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: 08/10/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Toll-like receptor 4 (TLR4) signaling at the plasma membrane and in endosomes results in distinct contributions to inflammation and host defence. Current understanding indicates that endocytosis of cell surface-activated TLR4 is required to enable subsequent signaling from endosomes. Contrary to this prevailing model, our data show that endosomal TLR4 signaling is not reliant on cell surface-expressed TLR4 or ligand-induced TLR4 endocytosis. Moreover, previously recognized requirements for the accessory molecule CD14 in TLR4 endocytosis and endosomal signaling are likely attributable to CD14 binding as well as trafficking and transferring lipopolysaccharide (LPS) to TLR4 at different subcellular localizations. TLR4 endocytosis requires the TLR4 intracellular signaling domain, contributions by phospholipase C gamma 2, spleen tyrosine kinase, E1/E2 ubiquitination enzymes, but not canonical TLR signaling adaptors and cascades. Thus, our study identifies independently operating TLR4 signaling modes that control TLR4 endocytosis, pro-inflammatory cell surface-derived, as well as endosomal TLR4 signaling. This revised understanding of how TLR4 functions within cells might be harnessed to selectively amplify or restrict TLR4 activation for the development of adjuvants, vaccines and therapeutics.
Collapse
Affiliation(s)
- Thomas E Schultz
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Carmen D Mathmann
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | | | - Timothy W Muusse
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Hyoyoung Kim
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - James W Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4215, Australia
| | - Jessica A Hamerman
- Immunology Program, Benaroya Research Institute, Seattle, WA, 98101, USA
| | - Andrew J Brooks
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Katryn J Stacey
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Antje Blumenthal
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
32
|
Varughese A, Balnadupete A, Ramesh P, Prasad TSK, Nidha AB, Bhandary Y. Guardians Turned Culprits: NETosis and Its Influence on Pulmonary Fibrosis Development. Mol Biotechnol 2025; 67:1752-1764. [PMID: 38717537 DOI: 10.1007/s12033-024-01171-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating, life-threatening irreversible lung disease characterized by the excessive accumulation of fibrotic tissue in the lungs, impairing their function. The exact mechanisms underlying Pulmonary fibrosis (PF) are multifaceted and not yet fully understood. Reports show that during COVID-19 pandemic, PF was dramatically increased due to the hyperactivation of the immune system. Neutrophils and macrophages are the patrolling immune cells that keep the microenvironment balanced. Neutrophil extracellular traps (NETs) are a normal protective mechanism of neutrophils. The chief components of the NETs include DNA, citrullinated histones, and anti-microbial peptides which are released by the activated neutrophils. However, it is becoming increasingly evident that hyperactivation of immune cells can also turn into criminals when it comes to pathological state. Dysregulated NETosis may contribute to sustained inflammation, overactivation of fibroblasts, and ultimately promoting collagen deposition which is the characteristic feature of PF. The role of NETs along with inflammation is attaining greater attention. However, seldom researches are related to the relationship between NETs causing PF. This review highlights the cellular mechanism of NETs-induced pulmonary fibrosis, which could give a better understanding of molecular targets which may be helpful for treating NETs-induced PF.
Collapse
Affiliation(s)
- Aleena Varughese
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India
| | - Akarsha Balnadupete
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India
| | - Poornima Ramesh
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India
| | | | | | - Yashodhar Bhandary
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India.
| |
Collapse
|
33
|
Maurício T, Guerra IMS, Pinho M, Melo T, Bonciarelli S, Goracci L, Neves B, Domingues R, Domingues P. Phosphatidylethanolamine species with n-3 and n-6 fatty acids modulate macrophage lipidome and attenuate responses to LPS stimulation. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159614. [PMID: 40254048 DOI: 10.1016/j.bbalip.2025.159614] [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: 01/21/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Phospholipids are increasingly recognized as key regulators of biological processes, including macrophage polarization and function. Among these, phosphatidylethanolamine (PE), a major constituent of cell membranes, is pivotal in maintaining cellular structure and function, yet the mechanisms through which native PE species influence macrophage immunometabolism remain largely unexplored. This study investigates the effects of two native PE species, PE 18:0/22:6 and PE 18:0/20:4, on the lipidome of resting and LPS-activated macrophages. Using C18 HPLC-MS/MS, we identified 337 lipid molecular species across 15 lipid subclasses, 332 of which varied significantly among conditions. Both PE 18:0/22:6 and PE 18:0/20:4 supplementation modulated the macrophage lipidome without inducing a pro-inflammatory phenotype under basal conditions. Notably, supplementation with PE 18:0/22:6 and PE 18:0/20:4 significantly increased lipid classes such as PE, PE O-, SM, CL, PG, LPE and PS, producing unique lipid profiles. Pre-treatment with PE 18:0/22:6 and PE 18:0/20:4 partially attenuated LPS-induced lipidomic changes, significantly reducing lipid classes like PC, including PC O- and PC P-, and Cer, which are typically linked to inflammation. While PE 18:0/20:4, from an individual lipid species perspective, may promote certain lipid profiles compatible with pro-inflammatory signaling pathways, particularly under inflammatory conditions, PE 18:0/22:6 seems to foster a lipid profile more supportive of inflammation resolution. This behaviour of PE 18:0/22:6 and PE 18:0/20:4 highlights the intricate complexity of lipid-mediated immunomodulation and emphasizes the critical role of cellular context in determining the functional outcomes of phospholipid supplementation in macrophage lipid metabolism and immune responses.
Collapse
Affiliation(s)
- Tatiana Maurício
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; Department of Medical Sciences and Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Inês M S Guerra
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Marisa Pinho
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Tânia Melo
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | | | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Bruno Neves
- Department of Medical Sciences and Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rosário Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal.
| |
Collapse
|
34
|
Zhang Q, Li Q, Wang X, Wu Y, Chen R, Sun Y, Pan Y, Li S, Wang Z. Comprehensive analysis through multi-omics integration to compare and elucidate the specific substances in milk of donkey、horse、camel、human and pig. Food Chem X 2025; 28:102592. [PMID: 40520710 PMCID: PMC12167115 DOI: 10.1016/j.fochx.2025.102592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 05/18/2025] [Accepted: 05/26/2025] [Indexed: 06/18/2025] Open
Abstract
Featured milk generally refers to less common dairy products with unique sources or specific nutritional components and functional characteristics. We used omics methods to study the lipids, metabolites, and proteins in donkey milk, mare milk, and camel milk and compared them with human milk and pig milk. The phospholipid content in camel milk is relatively high. Donkey milk contains metabolites that have potential therapeutic effects on mental disorders and chronic inflammatory diseases. Camel milk exhibits potential neuroprotective effects. Compared with other species, human milk contains more receptor protein-tyrosine kinase and galectin-3-binding protein; camel milk contains more monocyte differentiation antigen CD14 and fibrinogen beta chain; while pig milk contains more aminopeptidase. This research provides a theoretical basis for the application of featured milk in the field of functional foods and other food sectors.
Collapse
Affiliation(s)
| | | | | | - Yanzhi Wu
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Rui Chen
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Yinggang Sun
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Yuan Pan
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Siyi Li
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Zeying Wang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
35
|
Mahmoud YS, Hassanin IA, Sabra SA, Shehat MG, Abdel-Mohsen MA, Khattab SN, Hussein AA. Lipopolysaccharide nanomedicine-based reversion of chemotherapy-induced metastatic potential of breast cancer via hampering tumoral TLR4/SIRT2 axis and IL6 secretion from tumor-associated macrophages. Int J Biol Macromol 2025; 306:141396. [PMID: 39993692 DOI: 10.1016/j.ijbiomac.2025.141396] [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: 11/21/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
Triple-negative breast cancer (TNBC) is a metastatic disease. Targeted approaches, as implementing nanoliposomes, e.g., liposomal doxorubicin (DOX), did not exhibit significantly improved survival. Therefore, we aimed at reducing the metastatic potential of TNBC through a double punch to cancer cells and tumor-associated macrophages (TAMs). Databases' analyses showed that targeting TLR4/SIRT2 axis might be a possible option. Inspired by the emergence of lipopolysaccharide (LPS) in clinical trials, we developed bioactive copolymeric nanomicelles, originating from the self-assembly of our synthesized LPS-pectin conjugate (LPS-PEC) for the delivery of DOX (DOX@LPS-PEC). Targeting TLR4 via DOX@LPS-PEC micelles enhanced cellular uptake, however, it failed to significantly improve the cytotoxic potential of DOX. Alternatively, co-targeting SIRT2 via Sirtinol at a specific ratio (DOX@LPS-PEC: Sirtinol 1:5 w/w) elevated cellular oxidative stress, improved cytotoxic potential on 2D-monolayer and 3D-spheroid models, and significantly reduced migratory potential of MDA-MB-231 cells compared to DOX@LPS-PEC alone. Finally, DOX@LPS-PEC plus Sirtinol at the same ratio exhibited an ability to hamper TAM-secreted IL6, which contribute to the metastatic potential of TNBC. In conclusion, targeting TLR4/SIRT2 axis in TNBC synergizes with the effect of chemotherapeutics, e.g. DOX, reduce the metastatic potential of TNBC cells via down-regulating TLR4 and hampering tumor-microenvironment IL6.
Collapse
Affiliation(s)
- Yosra S Mahmoud
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, 21526, Egypt
| | - Islam A Hassanin
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, 21526, Egypt; Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Sally A Sabra
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, 21526, Egypt
| | - Michael G Shehat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mohamed A Abdel-Mohsen
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria 21526, Egypt
| | - Sherine N Khattab
- Department of Chemistry, Faculty of Science, Alexandria University, Alexandria 21321, Egypt.
| | - Ahmed A Hussein
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, 21526, Egypt.
| |
Collapse
|
36
|
BharathwajChetty B, Kumar A, Deevi P, Abbas M, Alqahtani A, Liang L, Sethi G, Liu L, Kunnumakkara AB. Gut microbiota and their influence in brain cancer milieu. J Neuroinflammation 2025; 22:129. [PMID: 40312370 PMCID: PMC12046817 DOI: 10.1186/s12974-025-03434-2] [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: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Microbial communities are not simply remnants of the past but dynamic entities that continuously evolve under the selective pressures of nature, reflecting the intricate and adaptive processes of evolution. The microbiota residing in the various regions of the human body has numerous roles in different physiological processes such as nutrition, metabolism, immune regulation, etc. In the zeal of achieving empirical insights into the ambit of the gut microbiome, the research over the years led to the revelation of reciprocal interaction between the gut microbiome and the cognitive functioning of the human body. Dysbiosis in the gut microbial composition disturbs the homeostatic cognitive functioning of the human body. This dysbiosis has been associated with various chronic diseases, including brain cancer, such as glioma, glioblastoma, etc. This review explores the mechanistic role of dysbiosis-mediated progression of brain cancers and their subtypes. Moreover, it demonstrates the regulatory role of microbial metabolites produced by the gut microbiota, such as short-chain fatty acids, amino acids, lipids, etc., in the tumour progression. Further, we also provide valuable insights into the microbiota mediating the efficiency of therapeutic regimens, thereby leveraging gut microbiota as potential biomarkers and targets for improved treatment outcomes.
Collapse
Affiliation(s)
- Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Pranav Deevi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin Scool of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
37
|
Wang Y, Zhou Y, Liu J, Liu C, Li Z, Sun X. Temporal and spatial expression of Phosphodiesterase-4B after sciatic nerve compression in rats and its mechanism of action on sciatic nerve repair. Neurochem Int 2025; 185:105940. [PMID: 39914666 DOI: 10.1016/j.neuint.2025.105940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Macrophage phenotype transformation is vital in sciatic nerve injury. The study of biomolecule expression and its impact on macrophage phenotype transformation is a current research focus. MATERIAL AND METHODS We created a rat model of sciatic nerve compression injury to examine the expression of PDE4B and the distribution of M1 and M2 macrophages over time and their relationship. We confirmed the effect of inhibiting PDE4B expression on macrophage phenotype changes and its role in sciatic nerve injury repair. The experiments consisted of immunofluorescence, western blotting, HE staining, TEM, and behavioral evaluation. Investigate in vivo experiment results with RAW264.7 cells in vitro. PDE4B knockdown lentivirus was transfected into RAW264.7 cells and stimulated with LPS and IFN-γ. We assessed CD86 and CD206 expression using flow cytometry and western blot. The relationship between PDE4B and the TLR4/NF-κB pathway was studied. RESULTS PDE4B peaked on day 7 after surgery, alongside the highest M1 macrophages count. PDE4B and M1 macrophages decreased, and M2 macrophages increased. PDE4B inhibition reduced M1 macrophages, increased M2 macrophages, suppressed inflammation, and promoted sciatic nerve repair while alleviating pain. In vitro experiments confirmed that PDE4B regulated macrophage phenotype via the TLR4/NF-κB pathway. Inhibiting PDE4B disrupted this pathway and promoted M2 macrophage transformation. CONCLUSIONS In the sciatic nerve injury, PDE4B expression is linked to the M1 macrophage phenotype. Low PDE4B expression facilitates the M1 to M2 macrophage transformation and supports sciatic nerve repair. The TLR4/NF-κB pathway is involved in this process.
Collapse
Affiliation(s)
- Yufei Wang
- Pain Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Youfei Zhou
- Pain Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Jinhao Liu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, China
| | - Chen Liu
- Pain Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Zirui Li
- Pain Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xuehua Sun
- Pain Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China.
| |
Collapse
|
38
|
Zapico D, Espinosa J, Criado M, Gutiérrez D, Ferreras MDC, Benavides J, Pérez V, Fernández M. Immunohistochemical expression of TLR1, TLR2, TLR4, and TLR9 in the different types of lesions associated with bovine paratuberculosis. Vet Pathol 2025; 62:305-318. [PMID: 39720873 DOI: 10.1177/03009858241302850] [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] [Indexed: 12/26/2024]
Abstract
The factors that determine the appearance of the different pathologic forms associated with bovine paratuberculosis are not fully understood, but new research suggests a critical role of innate immunity. Toll-like receptors (TLRs) trigger the recognition of invading pathogens by innate immune cells and the onset of specific immune responses. The aim of this work was to assess, immunohistochemically, the expression of TLR1, TLR2, TLR4, and TLR9 in intestinal samples of 20 cows showing different types of paratuberculous lesions: uninfected controls, focal lesions, paucibacillary, and multibacillary diffuse forms. The majority of labeled cells were morphologically consistent with macrophages. A differential cell count was performed in the intestinal lamina propria, gut-associated lymphoid tissue, and mesenteric lymph node. TLR9 immunolabeling between the different types of lesions was compared using a complete H-score. Focal and diffuse paucibacillary forms contained significantly increased TLR2-expressing macrophages outside of the lesions compared with the controls and diffuse multibacillary forms, and moderate TLR9 immunolabeling within granulomas. In the multibacillary granulomatous lesions, the expression of TLR1 and TLR4 was observed as well as increased TLR9 expression compared with the rest of the groups. Differences in the predominance of one type or another of TLR allows us to elucidate the importance of the innate immune response and its possible role in the development of the different types of paratuberculosis lesions.
Collapse
Affiliation(s)
- David Zapico
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - José Espinosa
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - Miguel Criado
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - Daniel Gutiérrez
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | | | | | - Valentín Pérez
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | | |
Collapse
|
39
|
Zeng Z, Wei N, Cai X, Xiao J. A magnetic bifunctional endotoxin removal nano-agent for the efficient elimination of endotoxins in recombinant protein preparation. Int J Biol Macromol 2025; 311:143663. [PMID: 40311983 DOI: 10.1016/j.ijbiomac.2025.143663] [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: 02/21/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Endotoxin contamination, as one of the most significant challenges in recombinant protein production by Escherichia coli, represents a critical biosafety concern and greatly hinders the biomedical application of recombinant proteins. Conventional methods, such as extreme-condition inactivation and chromatography-based separation, are plagued by issues including protein denaturation, low efficiency, and operational complexity in endotoxin removal. In this work, we developed a novel magnetic bifunctional endotoxin removal nano-agent (MagBER) with a multi-layered structure, consisting of a superparamagnetic Fe3O4 core, a mesoporous TiO2 intermediary layer, and an outer shell functionalized with boronic acid groups. This dual-functional design significantly enhances endotoxin removal efficiency through the TiO2 layer and boronic acid groups, ensuring stable endotoxin clearance performance even in high-salt environments and complex biological matrices. MagBER exhibits reusability while maintaining protein structural integrity. Moreover, MagBER has been successfully employed for endotoxin removal in various proteins, establishing it as a promising and sustainable solution for endotoxin clearance in biopharmaceutical applications.
Collapse
Affiliation(s)
- Zibing Zeng
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China
| | - Nannan Wei
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China
| | - Xiangdong Cai
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China.
| | - Jianxi Xiao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou, Gansu 730000, PR China.
| |
Collapse
|
40
|
Jasim MH, Mukhlif BAM, Uthirapathy S, Zaidan NK, Ballal S, Singh A, Sharma GC, Devi A, Mohammed WM, Mekkey SM. NFĸB and its inhibitors in preeclampsia: mechanisms and potential interventions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04211-x. [PMID: 40299024 DOI: 10.1007/s00210-025-04211-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
Preeclampsia (PE), which affects between 2 and 15% of pregnancies, is one of the most often reported prenatal problems. It is defined as gestational hypertension beyond 20 weeks of pregnancy, along with widespread edema or proteinuria and specific types of organ damage. PE is characterized by increased levels and activation of nuclear factor kappa B (NF-κB) in the mother's blood and placental cells. This factor controls over 400 genes linked to inflammatory, apoptotic, angiogenesis, and cellular responses to hypoxia and oxidative stress. In the final stages of physiological pregnancy, NF-κB levels need to be lowered to favor maternal immunosuppressive events and continue gestation to prevent hypoxia and inflammation, which are advantageous for implantation. Pharmacotherapy is thought to be a potential treatment for PE by downregulating NF-κB activation. NF-κB activity has been discovered to be regulated by several medications used for both prevention and treatment of PE. However, in order to guarantee treatment safety and effectiveness, additional creativity is desperately required. This article provides an overview of the current understanding of the defined function of NF-κB in PE progression. According to their effect on the cellular control of NF-κB pathways, newly proposed compounds for preventing and treating PE have also been emphasized.
Collapse
Affiliation(s)
- Mohannad Hamid Jasim
- Biology Department, College of Education, University of Fallujah, Fallujah, Iraq
| | - Bilal Abdul Majeed Mukhlif
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Noor Khalid Zaidan
- Department of Applied Chemistry, College of Applied Science, University of Fallujah, Fallujah, Iraq
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Anita Devi
- Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Wisam Mahmood Mohammed
- Department of Applied Chemistry, College of Applied Science, University of Fallujah, Fallujah, Iraq
| | - Shereen M Mekkey
- College of Pharmacy, Al-Mustaqbal University, 51001 Hilla, Babylon, Iraq
| |
Collapse
|
41
|
Guo L, Zeng M, Zhang G, Wang Z, Zhao A, Yang L, Gan J, Jiang X, Yu B. Targeting to miR-130b-5p/TLR4: How sodium danshensu suppresses inflammatory response of microglia in cerebral ischemia-reperfusion injury. Int Immunopharmacol 2025; 153:114497. [PMID: 40121745 DOI: 10.1016/j.intimp.2025.114497] [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: 11/18/2024] [Revised: 02/25/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Cerebral ischemia reperfusion injury (CIRI) is a crucial process in the inflammatory response. Sodium danshensu (SDSS) is of protective effects in cardiovascular and cerebrovascular diseases due to its anti-inflammatory properties. Studies have demonstrated that SDSS administration reduces infarct volume, attenuates neurological impairment, and inhibits microglia activation in rat models of CIRI. While it is well established that miRNAs play roles in a wide range of diseases through multiple pathways. However, the mechanism by which SDSS alleviates inflammatory injury after CIRI and its potential interaction with miRNAs remain unclear. Thus, we aimed to investigate the effectiveness and mechanism of SDSS in CIRI, and to verify whether it exerts anti-inflammatory effects by affecting miRNA. Through bioinformatics analysis and experimental validation, we identified miR-130b-5p is a key gene in the CIRI process, with SDSS administration leading to an upregulation of miR-130b-5p that is indispensable for its anti-inflammatory effects. Moreover, both SDSS and miR-130b-5p reduced the expression of TLR4. Overall, the beneficial effects of SDSS on CIRI can be attributed to the up-regulation of miR-130b-5p and the inhibition of TLR4, resulting in the attenuation of the inflammatory response.
Collapse
Affiliation(s)
- Lin Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China
| | - Miao Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China
| | - Ziyu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China
| | - Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China
| | - Lin Yang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town,Jinghai District, Tianjin 301617, People's Republic of China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China.
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town,Jinghai District, Tianjin 301617, People's Republic of China.
| |
Collapse
|
42
|
Sachetto ATA, Archibald SJ, Perkins M, Zhang G, Zhang Y, Ye D, Grover SP, Wu C, Li Z, Mackman N. Pathways regulating the levels of tissue factor-positive extracellular vesicles and activation of coagulation in endotoxemic mice. J Thromb Haemost 2025:S1538-7836(25)00262-4. [PMID: 40286911 DOI: 10.1016/j.jtha.2025.04.012] [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: 01/07/2025] [Revised: 03/03/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Sepsis and endotoxemia are associated with activation of coagulation as part of the host response to infection, but this can lead to disseminated intravascular coagulation. Lipopolysaccharide (LPS) is detected by the cell surface receptor toll-like receptor (TLR)4 and the intracellular receptor caspase 11. OBJECTIVES This study aimed to determine the roles of TLR4, caspase 11, and the NOD-, LRR-, and pyrin domain-containing protein (NLRP)3 inflammasome in increases of extracellular vesicle (EV) tissue factor (TF) activity and activation of coagulation in a mouse endotoxemia model. METHODS LPS was injected intraperitoneally into control mice and Tlr4-/-, Casp11-/-, Nlrp3-/-, or Casp1-/- mice or wild-type mice treated with the TLR4 inhibitor TAK-242 or the NLRP3 inhibitor MCC950. Blood samples were collected at 3 and 8 hours for analysis of cells, tumor necrosis factor α, interleukin (IL)-6, IL-1β, soluble intercellular adhesion molecule 1, EV TF activity, and thrombin-antithrombin (TAT) complexes. RESULTS LPS induced IL-1β at 3 and 8 hours, indicating inflammasome activation at these times. Tlr4 deficiency was associated with a significant decrease in tumor necrosis factor α and IL-6 but not soluble intercellular adhesion molecule 1 in endotoxemic mice. LPS induction of EV TF activity and TAT reduced significantly in Tlr4-/- mice at both 3 and 8 hours postinjection. In contrast, EV TF activity and TAT were only reduced in Casp11-/- mice at 8 hours post-LPS injection. CONCLUSION Our results indicate that TLR4 plays a major role whereas caspase 11 and the NLRP3 inflammasome play minor roles in the generation of TF-positive EVs and activation of coagulation in a mouse model of endotoxemia.
Collapse
Affiliation(s)
- Ana T A Sachetto
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sierra J Archibald
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Megan Perkins
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guoying Zhang
- Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, Texas, USA
| | - Yan Zhang
- Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, Texas, USA
| | - Dien Ye
- SAHA Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Steven P Grover
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Congqing Wu
- SAHA Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Zhenyu Li
- Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, Texas, USA
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
43
|
Dai L, Huang L, Li L, Tang L, Yao J, Shi Y, Han X. Pretreatment plasma sCD14 as a prognostic indicator in advanced non-small cell lung cancer patients undergoing immunotherapy. BMC Cancer 2025; 25:763. [PMID: 40269765 PMCID: PMC12016321 DOI: 10.1186/s12885-025-14148-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND This study aims to evaluate cytokines as a prognostic biomarker in patients with advanced non-small cell lung cancer (aNSCLC) undergoing immunotherapy. METHODS A comprehensive analysis was conducted to assess the prognostic significance of sCD14 and other cytokines in aNSCLC patients receiving immune checkpoint inhibitors (ICIs) using flow fluorescence. A discovery cohort (n = 42) was used to evaluate the differential expression of 41 cytokines between durable clinical benefit (DCB) and no durable benefit (NDB) groups in Cancer Hospital, Chinese Academy of Medical Sciences (CHCAMS). The prognostic value was further validated in multiple independent cohorts, including plasma protein measurements (n = 109), multiplex immunofluorescence (mIF) (n = 22), and messenger RNA datasets (n = 403) of NSCLC in CHCAMS. RESULTS In the discovery cohort, 7 cytokines (CD14, CCL27, IL-17 A, EGF, TNFR1, GFAP, CHI3L1) exhibited differential expression between the DCB and NDB groups. Among these, CD14, CCL27, IL-17 A, and TNFR1 were significantly elevated in the DCB group, while EGF, CHI3L1, and CCL5 were higher in the NDB group. CD14 showed a high area under the curve (AUC = 0.84) for predicting clinical benefit. Functional enrichment analysis indicated that these cytokines are involved in key immune pathways, including the inflammatory response and MAPK signaling. Univariate COX for progression-free survival (PFS) analysis demonstrated prognostic value for CD14 (p < 0.001, HR = 0.054 [0.014-0.219]), CCL27 (p < 0.001, HR = 0.054 [0.015-0.196]), IL-17 A (p < 0.001, HR = 0.110 [0.041-0.298]), and CCL5 (p < 0.05, HR = 2.387 [1.023-5.570]). Validation in the CHCAMS cohort confirmed that CD14 expression, measured via mIF, was a predictor of PFS (p < 0.05). Furthermore, high CD14 expression was consistently associated with superior PFS across multiple external datasets (GSE126044, GSE135222, GSE136961, and GSE218989). CD14 expression was found to be elevated in various normal tissue types, particularly in lung adenocarcinoma and lung squamous cell carcinoma, compared to tumors, indicating its potential role in immune surveillance. CONCLUSION sCD14 is a promising prognostic biomarker for aNSCLC patients undergoing immunotherapy. Elevated plasma sCD14 levels are associated with improved PFS and a favorable immune response.
Collapse
Affiliation(s)
- Liyuan Dai
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Liling Huang
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1, Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
44
|
Smail SW, Albarzinji N, Salih RH, Taha KO, Hirmiz SM, Ismael HM, Noori MF, Azeez SS, Janson C. Microbiome dysbiosis in SARS-CoV-2 infection: implication for pathophysiology and management strategies of COVID-19. Front Cell Infect Microbiol 2025; 15:1537456. [PMID: 40330025 PMCID: PMC12052750 DOI: 10.3389/fcimb.2025.1537456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/27/2025] [Indexed: 05/08/2025] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of coronavirus disease 2019 (COVID-19), in late 2019 initiated a global health crisis marked by widespread infection, significant mortality, and long-term health implications. While SARS-CoV-2 primarily targets the respiratory system, recent findings indicate that it also significantly disrupts the human microbiome, particularly the gut microbiota, contributing to disease severity, systemic inflammation, immune dysregulation, and increased susceptibility to secondary infections and chronic conditions. Dysbiosis, or microbial imbalance, exacerbates the clinical outcomes of COVID-19 and has been linked to long-COVID, a condition affecting a significant proportion of survivors and manifesting with over 200 symptoms across multiple organ systems. Despite the growing recognition of microbiome alterations in COVID-19, the precise mechanisms by which SARS-CoV-2 interacts with the microbiome and influences disease progression remain poorly understood. This narrative review investigates the impact of SARS-CoV-2 on host-microbiota dynamics and evaluates its implications in disease severity and for developing personalized therapeutic strategies for COVID-19. Furthermore, it highlights the dual role of the microbiome in modulating disease progression, and as a promising target for advancing diagnostic, prognostic, and therapeutic approaches in managing COVID-19.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- College of Pharmacy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | | | - Rebaz Hamza Salih
- Department of Respiratory Medicine, PAR Private Hospital, Erbil, Kurdistan Region, Iraq
| | - Kalthum Othman Taha
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Sarah Mousa Hirmiz
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Hero M. Ismael
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Marwa Fateh Noori
- College of Pharmacy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Sarkar Sardar Azeez
- Department of Medical Laboratory Technology, Soran Technical College, Erbil Polytechnic University, Erbil, Kurdistan Region, Iraq
| | - Christer Janson
- Department of Medical Science, Respiratory Medicine, and Allergology, Uppsala University and University Hospital, Uppsala, Sweden
| |
Collapse
|
45
|
Jin S, Wang H, Gong H, Guo L, Zhang H, Zhang J, Chang Q, Li J, Zhang R, Bao J. Music intervention mitigates LPS-induced gut barrier disruption and immune stress in broilers via TLR4/NF-κB regulation. Poult Sci 2025; 104:105189. [PMID: 40294553 PMCID: PMC12059385 DOI: 10.1016/j.psj.2025.105189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/02/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025] Open
Abstract
Immune stress induced by harsh environment in intensive farming can impair broiler intestinal health. Although music as an environmental intervention can alleviate short-term stress injury, its long-term regulatory mechanism on intestinal inflammation has not been clarified. In this study, we investigated the effects of a music-enriched environment on growth performance, intestinal barrier function, and inflammatory responses in lipopolysaccharide (LPS)-induced immunostressed broilers. AA broilers were randomly divided into four groups: control group (CON), music-enriched environment group (MUC), LPS-induced immune stress group (LPS) and music-enriched environment + LPS group (MUC+LPS). On the 14th, 16th and 18th days, the LPS and MUC+LPS groups were injected intraperitoneally with 500 μg of LPS to construct an immune stress model, and the CON and MUC groups were injected with an equal amount of saline. On day 28, the birds were sacrificed to detect the indicators associated with intestinal barrier and inflammation. The LPS group showed a significant decrease in performance from 14 to 28 days, with elevated serum levels of CORT, ACTH, DAO, and d-LA, and a decrease in the activity of intestinal mucosal SOD/GSH-Px, and impaired gut morphology. impaired; music remission significantly alleviated the decline in production performance, reduced the levels of stress hormones and markers of intestinal barrier damage, while elevating jejuno-ileal GSH-Px activity and improving intestinal morphology. Significant inflammatory gene expression characteristics were observed in jejunum and ileum tissues after LPS injection: upregulation of TLR4, NF-κB, TNF-α, IL-1β, and IL-6, and significant suppression of jejunal IL-10 expression. Notably, IL-10 and IFN-γ expression in the ileum did not show statistical differences. Inflammation-related gene expression showed an overall down-regulation trend after the music intervention, but was still significantly different from the control group. Music intervention on the regulation of jejunal MYD88 and ileal TNF-α - the LPS group did not show statistically significant differences in the expression of these two key inflammatory nodes with the LPS+MUS group. Mechanistic studies have shown that LPS triggers an oxidative stress cascade through activation of the TLR4/NF-κB signaling axis, leading to disruption of intestinal barrier integrity. In contrast, music exposure exerts a protective effect through a dual mechanism: on the one hand, it helps to enhance the expression of the tight junction protein ZO-1/Occludin to repair the physical barrier; on the other hand, it inhibits the activation of the TLR4/NF-κB pathway, which can effectively alleviate LPS-induced immunopathological damage.
Collapse
Affiliation(s)
- Shengzi Jin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Haowen Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Haiyue Gong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Lu Guo
- Department of Basic Medical Sciences, Heilongjiang Nursing College, Harbin, Heilongjiang 150086, China
| | - Haoran Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiaqi Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Qingqing Chang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, PR China
| | - Runxiang Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Jun Bao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
46
|
Kanewska A, Lackner I, Friedrich A, Winkelmann M, Rojewski M, Weber B, Preßmar J, Perl M, Schrezenmeier H, Kalbitz M. Immunomodulatory and cardio-protective effects of differentially originated multipotent mesenchymal stroma cells during polymicrobial sepsis in mice. Eur J Trauma Emerg Surg 2025; 51:178. [PMID: 40253667 PMCID: PMC12009780 DOI: 10.1007/s00068-025-02862-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/03/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE Sepsis is a life-threatening condition with cardiac complications being an independent predictor of poor outcome. Although their mechanisms have been widely investigated, therapeutic options remain limited. One promising therapeutic tool are mesenchymal stromal cells (MSCs). The aim of this study is to investigate the immunomodulatory effects of human MSCs from two different sources (bone marrow/BMMSC and adipose tissue/ASC) and to evaluate their cardioprotective potential. METHODS 60 adult male C57BL/6 mice were divided into sham, sepsis (cecal ligation puncture (CLP)) and two i.v. treatment groups CLP + human BMMSC and CLP + human ASC with 5 animals in each group. The observation periods were 8, 24 and 72 h. Left ventricular tissue was analyzed histologically, by qPCR (C3ar, C5ar1, Il-1b, Il-6, Il-10, Tlr2, Tlr4, Tnfa, and Nlrp3) and western blot. Cardiac damage markers troponin I and heart fatty acid binding protein (HFABP) were detected in serum by ELISA. RESULTS Troponin I and HFABP were significantly increased in CLP group after 8 h compared to sham. In cardiac tissue the expression of C3ar, C5ar1, Il-1b, Il-6, Il-10, Tlr2, Tlr4, Tnfa and Nlrp3 inflammasome was upregulated up to 24h after CLP compared to sham. After BMMSC treatment, C3ar as well as C5ar, Tlr2 and Il-10 mRNA expression in left ventricle was downregulated compared to CLP, whereas ASC treatment was associated with the downregulation of Il-6 and Nlrp3. CONCLUSIONS CLP-induced polymicrobial sepsis in mice was associated with cardiac damage and increased inflammation in left ventricular tissue. Therapeutic systemic application of human BMMSC and ASC ameliorated damage and inflammation in the heart.
Collapse
Affiliation(s)
- Anna Kanewska
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Ina Lackner
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Traumatology, Hand, Plastic and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Anne Friedrich
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Martina Winkelmann
- Institute for Transfusion Medicine, University Medical Center Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service, Ulm, Germany
| | - Markus Rojewski
- Institute for Transfusion Medicine, University Medical Center Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service, Ulm, Germany
| | - Birte Weber
- Department of Trauma Surgery and Orthopedics, University Hospital Frankfurt, Goethe-University, Frankfurt Am Main, Germany
| | - Jochen Preßmar
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Traumatology, Hand, Plastic and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
| | - Mario Perl
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Medical Center Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service, Ulm, Germany
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Traumatology, Hand, Plastic and Reconstructive Surgery, Center of Surgery, University of Ulm, Ulm, Germany
- Military Medical City Hospital (MMCH), Doha, Qatar
| |
Collapse
|
47
|
Burge KY, Georgescu C, Zhong H, Wilson AP, Gunasekaran A, Yu Z, Franca A, Eckert JV, Wren JD, Chaaban H. Spatial transcriptomics delineates potential differences in intestinal phenotypes of cardiac and classical necrotizing enterocolitis. iScience 2025; 28:112166. [PMID: 40201118 PMCID: PMC11978348 DOI: 10.1016/j.isci.2025.112166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/20/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal gastrointestinal disease, often resulting in multi-organ failure and death. While classical NEC is strictly associated with prematurity, cardiac NEC is a subset of the disease occurring in infants with comorbid congenital heart disease. Despite similar symptomatology, the NEC subtypes vary slightly in presentation and may represent etiologically distinct diseases. We compared ileal spatial transcriptomes of patients with cardiac and classical NEC. Epithelial and immune cells cluster well by cell-type segment and NEC subtype. Differences in metabolism and immune cell activation functionally differentiate the cell-type makeup of the NEC subtypes. The classical NEC phenotype is defined by dysbiosis-induced inflammatory signaling and metabolic acidosis, while that of cardiac NEC involves reduced angiogenesis and endoplasmic reticulum stress-induced apoptosis. Despite subtype-associated clinical and demographic variability, spatial transcriptomics has substantiated pathway and network differences within immune and epithelial segments between cardiac and classical NEC.
Collapse
Affiliation(s)
- Kathryn Y. Burge
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Constantin Georgescu
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hua Zhong
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Adam P. Wilson
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Aarthi Gunasekaran
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zhongxin Yu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Addison Franca
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jeffrey V. Eckert
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jonathan D. Wren
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hala Chaaban
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
48
|
Zelkoski AE, Lu Z, Sukumar G, Dalgard C, Said H, Alameh MG, Mitre E, Malloy AMW. Ionizable lipid nanoparticles of mRNA vaccines elicit NF-κB and IRF responses through toll-like receptor 4. NPJ Vaccines 2025; 10:73. [PMID: 40246950 PMCID: PMC12006303 DOI: 10.1038/s41541-025-01124-x] [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: 05/02/2024] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Ionizable lipid nanoparticles (LNP) that have enabled the success of messenger RNA (mRNA) vaccines have been shown to be immunostimulatory in the absence of mRNA. However, the mechanisms through which they activate innate immune cells is incompletely understood. Using a monocyte cell line, we compared the ability of three LNP formulations to activate transcription factors Nuclear Factor-kappa B (NF-κB) and Interferon Regulatory Factor (IRF). Comparison of signaling in knockout cell lines illustrated a role for Toll-like receptor (TLR) 4 in initiation of this signaling cascade and the contribution of the ionizable lipid component. Activation induced by empty LNPs was similar to that induced by LNPs containing mRNA, indicating that LNPs may provide the majority of innate stimulation for the mRNA vaccine platform. Our findings demonstrate that ionizable lipids within LNPs signal through TLR4 to activate NF-κB and IRF, identifying a mechanism for innate activation that can be optimized for adjuvant design.
Collapse
Affiliation(s)
- Amanda E Zelkoski
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Zhongyan Lu
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Clifton Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Hooda Said
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of Health Sciences, Bethesda, PA, USA
| | - Allison M W Malloy
- Department of Pediatrics, Uniformed Services University of Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
49
|
Rivi V, Rigillo G, Alboni S, Koene JM, Pani L, Lukowiak K, Tascedda F, Blom JMC, Benatti C. Unraveling lipopolysaccharide-induced behavioral and molecular effects in Lymnaea stagnalis, an emerging model organism for translational neuroscience. Int Immunopharmacol 2025; 152:114418. [PMID: 40090086 DOI: 10.1016/j.intimp.2025.114418] [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: 11/10/2024] [Revised: 02/05/2025] [Accepted: 03/02/2025] [Indexed: 03/18/2025]
Abstract
In this study, we employed a reductionist (yet not simplistic) approach utilizing the established invertebrate model system of the pond snail, Lymnaea stagnalis, to investigate the behavioral and molecular effects of systemic administration of lipopolysaccharide (LPS)-a bacterial endotoxin-on the snails' central ring ganglia. Snails received injections of either a low dose (2.5 μg) or a high dose (25 μg) of LPS, and their behavioral and molecular responses were assessed at 2, 6, and 24 h post-injection. With the high dose, snails exhibited a significant increase in homeostatic aerial respiration lasting for at least 24 h, consistent with a sickness-like state induced by the immune challenge. Additionally, we found that when administered 2, 6, or 24 h before operant conditioning training, the high dose of LPS, impaired memory formation. To further explore the underlying molecular mechanisms, we examined the transcriptional effects of the two doses of LPS in the snails' central ring ganglia. Our analysis showed a dose- and time-dependent upregulation of immune and stress-related genes, including key enzymes involved in the kynurenine pathway (KP), toll-like receptor 4 (TLR4), and heat shock protein 70 (HSP70). Metabolomic analysis suggested that the high LPS dose shifted KP metabolism toward the production of neurotoxic metabolites within the ganglia, indicating a LPS-induced neuroinflammatory state. Together, our findings provide valuable insight into the conserved mechanisms of neuroinflammation in this invertebrate model, offering a simplified yet effective tool to further explore the molecular interactions between the immune and central nervous systems.
Collapse
Affiliation(s)
- Veronica Rivi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanna Rigillo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Joris M Koene
- Ecology & Evolution, Amsterdam Institute for Life and Environment (A-LIFE), Faculty of Science, Vrije Universiteit, 1081, BT, Amsterdam, The Netherlands
| | - Luca Pani
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, USA
| | - Ken Lukowiak
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy; CIB, Consorzio Interuniversitario Biotecnologie, 34148 Trieste, Italy
| | - Johanna M C Blom
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cristina Benatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| |
Collapse
|
50
|
He Z, Tan X, Yuan M, Chen L, Meng Y, Wang Q, Hu J, Qiu Z, Yang Y. Anethole trithione mitigates LPS/D-Gal-induced acute liver injury by suppressing ROS production and NF-κB activity. Int Immunopharmacol 2025; 152:114371. [PMID: 40054324 DOI: 10.1016/j.intimp.2025.114371] [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: 09/04/2024] [Revised: 01/14/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Acute liver injury (ALI) is a prevalent form of hepatic disease associated with significant morbidity and mortality due to medical treatments, exposure to toxins or viral infections. Anethole trithione (ATT) is a heterocyclic sulfur compound recognized for its chemoprotective properties against cancer and drug-induced toxicity. This study aimed to evaluate the effectiveness of ATT in the treatment of ALI. The therapeutic effects of ATT on hepatic injury were evaluated in vivo by inducing ALI in mice through the administration of lipopolysaccharide (LPS) and D-galactosamine (D-Gal). Additionally, HepG2 and Huh7 cells exposed to LPS were utilized to investigate the underlying mechanisms in vitro. The results indicated that ATT significantly reduced the production of reactive oxygen species (ROS), mitigated oxidative stress-related biochemical markers, and inhibited hepatocyte apoptosis in vivo, resulting in marked improvement in ALI in the murine model. Mechanistic studies conducted both in vivo and in vitro demonstrated that ATT alleviates LPS/D-Gal-induced ALI by inhibiting ROS production and the activity of nuclear factor-kappa B (NF-κB). Collectively, these findings underscore the potential therapeutic benefits of ATT in the management of ALI.
Collapse
Affiliation(s)
- Zhen He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xiangyun Tan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Yuan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Liang Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yan Meng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Qi Wang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Junjie Hu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China.
| | - Yuan Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, China.
| |
Collapse
|