1
|
Liu L, Ma C, Ji J, Gao R, Li D. Role of antidiarrheal agents nifuroxazide in antitumor multi‑target anticancer, multi‑mechanism anticancer drug (Review). Oncol Lett 2025; 29:260. [PMID: 40230426 PMCID: PMC11995686 DOI: 10.3892/ol.2025.15006] [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: 10/04/2024] [Accepted: 01/07/2025] [Indexed: 04/16/2025] Open
Abstract
Nifuroxazide (NFZ) is an antimicrobial drug, which has been found to be a promising antitumor agent in recent years. In addition to being a classic STAT3 inhibitor, NFZ can also act on IL-6 and exert an anti-tumor role through inflammatory factor pathways. It can also bind to target proteins of aldehyde dehydrogenase 1, one of the families of E-twenty-six transcription factors and ubiquitin-specific protease 21 to play an anti-tumor role in different pathways. NFZ is able to act on the tumor cell microenvironment to inhibit tumor angiogenesis and tumor cell migration, enhance tumor immune cells, increase the cytotoxicity of tumor cells and enhance the anti-tumor effect of other drugs. Furthermore, it has high safety with few toxic side effects. The anti-tumor mechanisms of NFZ were described in the current review, aiming to provide insight and a reference for future studies promoting the implementation of NFZ as an anti-tumor drug in the clinic.
Collapse
Affiliation(s)
- Liping Liu
- Oncology Department, Qingdao Endocrine and Diabetes Hospital, Qingdao, Shandong 266000, P.R. China
| | - Chengshan Ma
- Department of Orthopedic Surgery, Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250000, P.R. China
| | - Jinfeng Ji
- Oncology Department, Qingdao Endocrine and Diabetes Hospital, Qingdao, Shandong 266000, P.R. China
| | - Rong Gao
- Oncology Department, Qingdao Endocrine and Diabetes Hospital, Qingdao, Shandong 266000, P.R. China
| | - Deliang Li
- Emergency Department, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| |
Collapse
|
2
|
Xu R, Gao Y. CD28 + CD45RA - CD8br AC mediated the effects of Interleukin- 6 on Alzheimer's disease: A Mendelian Randomization Study. BMC Neurol 2025; 25:203. [PMID: 40369481 PMCID: PMC12077015 DOI: 10.1186/s12883-025-04194-5] [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: 05/06/2024] [Accepted: 04/14/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND IL-6 has garnered significant attention as a potential factor in AD pathogenesis. The association between peripheral immune cells and IL-6 is evident, yet how peripheral immune cells mediate IL-6's effects on AD remains enigmatic regarding the precise pathophysiological processes. To address these uncertainties, we employed genetic evidence to investigate their impact. Our current study explores further the intricate relationship between IL-6, peripheral immune cells, and AD by using extensive publicly available genetic data, aiming to provide novel insights into this critical area of medical research. METHODS The relevant data regarding IL-6, 731 peripheral immune cells, and AD were screened and retrieved from the GWAS database. Subsequently, we predominantly utilized the IVW approach to carry out bi-directional MR analyses between IL-6, 731 peripheral immune cells, and AD. We utilized two-step, two-sample MR analyses to determine three key factors: (i) IL-6 exhibits associations with both AD and specific peripheral immune cells, respectively, and there is an absence of inverse causality. (ii) Specific peripheral immune cells exhibit associations with AD, and there is an absence of inverse causality. (iii) to identify which peripheral immune cells mediate the effects of IL-6 on AD. Then we employed the MVMR approach to verify whether the mediating relationships obtained from the two-step, two-sample MR analyses remained valid. Furthermore, we calculated their respective mediating effects, the combined mediating effects, and the proportions of their mediating effect shares. All of the aforementioned steps were utilized to verify the reliability of causality employing sensitivity analysis, heterogeneity analysis, and horizontal pleiotropy analysis. RESULTS Our findings indicate a significant correlation between increased IL-6 levels and a reduced risk of AD (P = 0.009, OR = 0.941, 95%CI = 0.899- 0.985), along with elevated levels of CD28+ CD45RA- CD8br AC (P = 0.007, OR = 1.159, 95%CI = 1.007- 1.333). Also indicates a significant correlation between increased CD28+ CD45RA- CD8br AC (P = 0.005, OR = 0.983, 95%CI = 0.971- 0.995) levels and a reduced risk of AD. Therefore, through MVMR analysis, the effect of IL-6 on AD increased from -0.061 to -0.046 (95% CI: -0.090, -0.002) after genetic adjustment for CD28+ CD45RA- CD8br AC. CONCLUSIONS Increased CD28+ CD45RA- CD8br AC levels appear to partially mediate the effect of IL-6 on reducing AD risk.
Collapse
Affiliation(s)
- Rui Xu
- Chengdu Sixth People's Hospital, Chengdu City, Sichuan Province, China.
| | - YongJun Gao
- The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, China
| |
Collapse
|
3
|
Wu X, Chang Y, Kong C, Ding Z, Pan D, Lin P, Wang S, Mao G. Inhibitory effect of Alantolactone against varicella-zoster virus in vitro. Virol J 2025; 22:138. [PMID: 40346667 PMCID: PMC12063397 DOI: 10.1186/s12985-025-02759-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Varicella-zoster virus (VZV), a member of the α-herpesvirus family, is known for causing two distinct diseases: chickenpox (varicella) during the primary infection and shingles (zoster) due to reactivation of the virus later in life. Although there are vaccines available to prevent VZV infection, it is still not universally effective, and antiviral treatments for VZV are limited and may come with significant side effects. Thus, development of novel therapeutics is urgently needed. METHODS We identified a naturally occurring Alantolactone (ALT) that inhibited replication of recombinant VZV in human diploid fibroblast (WI-38 cells) and Adult Retinal Pigment Epithelial cell line-19 (ARPE-19 cells) through Western blotting, qPCR and plaque assays. Subsequently, we explored the mechanism underlying the anti-VZV activity of ALT using time-of-addition experiments and transcriptomic analyses. RESULTS A screening model was established for anti-VZV compounds, and we screened ALT was with good anti-VZV efficacy. Our findings revealed that ALT alleviated cytopathic changes, reduced viral titres, and inhibited the expression of viral genes and proteins in WI-38 cells and ARPE-19 cells. Furthermore, our data showed that ALT inhibited VZV infection in intracellular viral replication. Finally, multiple inflammatory pathways were involved in the antiviral role of ALT, and IL-6 was one of the most critical hub genes. CONCLUSION Together, our findings identify ALT as an anti-VZV agent that may prove useful in the treatment of VZV replication.
Collapse
Affiliation(s)
- Xinna Wu
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, No.1229 Gudun Road, Xihu District, Hangzhou, 310030, China
| | - Yunchuang Chang
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, No.1229 Gudun Road, Xihu District, Hangzhou, 310030, China
| | - Chengcheng Kong
- Geriatric Department of the 3rd Hospital of Hangzhou, Hangzhou, 310009, China
| | - Zhiwei Ding
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, No.1229 Gudun Road, Xihu District, Hangzhou, 310030, China
| | - Dongli Pan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, 310030, China
| | - Ping Lin
- Geriatric Department of the 3rd Hospital of Hangzhou, Hangzhou, 310009, China.
- Hangzhou Third People's Hospital, No.38, Xihu Avenue, Shangcheng District, Hangzhou, Email, China.
| | - Sanying Wang
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, No.1229 Gudun Road, Xihu District, Hangzhou, 310030, China.
| | - Genxiang Mao
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, No.1229 Gudun Road, Xihu District, Hangzhou, 310030, China.
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, No.1229 Gudun Road, Xihu District, Hangzhou, 310030, China.
| |
Collapse
|
4
|
Cao Y, Xiao S, He B, Shi X, Xiao N, Liu X, Liu D, Zhou Z, Wang P. Chronic Exposure to Fluxapyroxad Exacerbated Susceptibility to Colitis in Mice via a Gut Microbiota-Indole Derivatives-Th17/Treg Cell Balance Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10172-10185. [PMID: 40244699 DOI: 10.1021/acs.jafc.5c02749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Fluxapyroxad is the most commonly used succinate dehydrogenase inhibitor fungicide. This work investigated its adverse effects on colitis susceptibility and explored the underlying mechanisms based on a mouse model. After 13 weeks of exposure at the acceptable daily intake (ADI) level, fluxapyroxad exacerbated the susceptibility to colitis, impaired the intestinal barrier, and elevated proinflammatory cytokines and chemokines of the colon in mice. It was found that this toxic effect was caused by the disruption of the gut microbiome. Specifically, the abundance of Lachnospiraceae and Muribaculaceae decreased, while Desulfovibrionaceae and Eggerthellaceae increased. Altered microbiota reduced fecal indole derivatives, including indole-3-lactic acid (ILA), indole-3-acetic acid (IAA), and indole-3-acrylic acid (IArA), inhibiting aryl hydrocarbon receptor (AHR) activation, disrupting immune homeostasis by overactivating Th17 cells and insufficient Treg cell differentiation, and causing mild colonic inflammation. Oral antibiotic-treated mice and fecal transfer experiments validated the pathway. Susceptibility to colitis induced by fluxapyroxad was not detected in the oral antibiotic-treated mice. Fecal transfer of the disordered gut microbiota caused by fluxapyroxad could aggravate the severity of colitis in recipient oral antibiotic-treated mice that did not receive fluxapyroxad exposure. In conclusion, chronic fluxapyroxad exposure at the ADI level exacerbated colitis via a gut microbiota-indole derivatives-Treg/Th17 cell balance axis, offering a new risk assessment perspective of fluxapyroxad.
Collapse
Affiliation(s)
- Yue Cao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Shouchun Xiao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Bingying He
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Xinlei Shi
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Nan Xiao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Xueke Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Donghui Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Zhiqiang Zhou
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| | - Peng Wang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, P.R. China
| |
Collapse
|
5
|
Liu J, Xia W, Cheng J, Geng Y, Li W, Fan Y. Escherichia coli aggravates inflammatory response in mice oral mucositis through regulating Th17/Treg imbalance. Front Cell Infect Microbiol 2025; 15:1585020. [PMID: 40365536 PMCID: PMC12069327 DOI: 10.3389/fcimb.2025.1585020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Microbial dysbiosis links to mucosal immune dysregulation, but the specific bacterial contributions to oral mucosal inflammation remain unclear. Escherichia coli (E. coli), a pathogen well-characterized in mucosal immunity and immune regulation studies, has been observed to be enriched in chronic oral inflammatory lesions and was reported to modulate T helper 17 cells (Th17)/T regulatory cells (Treg) homeostasis. Here, we developed an oral mucositis mouse model via tongue scratch and E. coli topical application to investigate its role in Th17/Treg imbalance. Methods The inflammatory infiltration was evaluated by macroscopic photography and HE staining. The expression of inflammatory factors in tongue tissue and peripheral blood of mice were detected by immunohistochemical staining and enzyme-linked immunosorbent assay. The number of Th17 and Treg in mice spleen lymphocytes were evaluated with flow cytometry. Differential gene expression analysis, functional enrichment analysis and immune infiltration analysis were performed using RNA-seq data from oral lichen planus (OLP). Results E. coli stimulation aggravated inflammatory responses induced by scratching in lingual mucosa of mice, including increased local and systemic expression of interleukin 6 (IL6), interleukin 17 (IL17), chemokine receptor 6 (CCR6) and chemokine C-C motif ligand 20 (CCL20), increased proportions of Th17 cells and increased Th17/Treg ratio in spleen lymphocytes. Analysis of RNA-seq data from OLP revealed alterations in antimicrobial responses and inflammatory factors associated with upregulation of Th17/Treg balance. Conclusion This study supports the role of E. coli in promoting oral mucosal inflammation and provides an experimental basis for in vivo study of OLP from the perspective of microorganisms.
Collapse
Affiliation(s)
- Jia Liu
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Wenhui Xia
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
- Hefei Stomatological Hospital, Hefei Stomatology Clinical College of Anhui Medical University, Hefei, China
| | - Juehua Cheng
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yanlin Geng
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Weiping Li
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yuan Fan
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
6
|
Li L, Liu J, Lu J, Wu J, Zhang X, Ma T, Wu X, Zhu Q, Chen Z, Tai Z. Interventions in cytokine signaling: novel horizons for psoriasis treatment. Front Immunol 2025; 16:1573905. [PMID: 40303401 PMCID: PMC12037536 DOI: 10.3389/fimmu.2025.1573905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Intricate interactions between immune cells and cytokines define psoriasis, a chronic inflammatory skin condition that is immunological-mediated. Cytokines, including interleukins (ILs), interferons (IFNs), tumor necrosis factors (TNFs), chemokines, and transforming growth factor-β (TGF-β), are essential for controlling cellular activity and immunological responses, maintaining homeostasis and contributing to the pathogenesis of psoriasis. These molecules modulate the immune microenvironment by either promoting or suppressing inflammation, which significantly impacts therapeutic outcomes. Recent research indicates that treatment strategies targeting cytokines and chemokines have significant potential, offering new approaches for regulating the immune system, inhibiting the progression of psoriasis, and reducing adverse effects of traditional therapies. This review consolidates current knowledge on cytokine and chemokine signaling pathways in psoriasis and examines their significance in treatment. Specific attention is given to cytokines like IL-17, IL-23, and TNF-α, underscoring the necessity for innovative therapies to modulate these pathways and address inflammatory processes. This review emphasizes the principal part of cytokines in the -pathological process of psoriasis and explores the challenges and opportunities they present for therapeutic intervention. Furthermore, we examine recent advancements in targeted therapies, with a particular focus on monoclonal antibodies, in ongoing research and clinical trials.
Collapse
Affiliation(s)
- Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Jiaye Lu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Tianyou Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xiying Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Zhang M, Xu N, Cheng Q, Ye J, Wu S, Liu H, Zhao C, Yu L, Feng W. Immune status assessment based on plasma proteomics with meta graph convolutional networks. BMC Genomics 2025; 26:360. [PMID: 40211143 PMCID: PMC11983875 DOI: 10.1186/s12864-025-11537-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025] Open
Abstract
Plasma proteins, especially immune-related proteins, are vital for assessing immune health and predicting disease risks. Despite their significance, the link between these proteins and systemic immune function remains unclear. To bridge this gap, researchers developed ProMetaGCN, a model integrating meta-learning, graph convolutional networks, and protein-protein interaction (PPI) data to evaluate immune status via plasma proteomics. This framework identified 309 immune-related factors with associated biological functions and pathways. Using six machine learning methods, four algorithms (Random Forest, LightGBM, XGBoost, Lasso) were selected for immune profiling and aging analysis, revealing ADAMTS13, GDF15, and SERPINF2 as key biomarkers. Validation across two COVID-19 cohorts confirmed the model's robustness, showing immune status correlates with infection progression and recovery. Furthermore, the study proposed ImmuneAgeGap, a novel metric linking immune profiles to survival rates in non-small-cell lung cancer (NSCLC) patients. These insights advance personalized immune health strategies and disease prevention.
Collapse
Affiliation(s)
- Min Zhang
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Nan Xu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Qi Cheng
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Jing Ye
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Shiwei Wu
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Haoliang Liu
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Chengkui Zhao
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China.
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai, China.
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd., Shanghai, China.
| | - Weixing Feng
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China.
| |
Collapse
|
8
|
Lin X, Li X, Zhai Z, Zhang M. JAK-STAT pathway, type I/II cytokines, and new potential therapeutic strategy for autoimmune bullous diseases: update on pemphigus vulgaris and bullous pemphigoid. Front Immunol 2025; 16:1563286. [PMID: 40264772 PMCID: PMC12011800 DOI: 10.3389/fimmu.2025.1563286] [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: 01/19/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025] Open
Abstract
Autoimmune Bullous Diseases (AIBDs), characterized by the formation of blisters due to autoantibodies targeting structural proteins, pose significant therapeutic challenges. Current treatments, often involving glucocorticoids or traditional immunosuppressants, are limited by their non-specificity and side effects. Cytokines play a pivotal role in AIBDs pathogenesis by driving inflammation and immune responses. The JAK-STAT pathway is central to the biological effects of various type I and II cytokines, making it an attractive therapeutic target. Preliminary reports suggest that JAK inhibitors may be a promising approach in PV and BP, but further clinical validation is required. In AIBDs, particularly bullous pemphigoid (BP) and pemphigus vulgaris (PV), JAK inhibitors have shown promise in modulating pathogenic cytokine signaling. However, the safety and selectivity of JAK inhibitors remain critical considerations, with the potential for adverse effects and the need for tailored treatment strategies. This review explores the role of cytokines and the JAK-STAT pathway in BP and PV, evaluating the therapeutic potential and challenges associated with JAK inhibitors in managing these complex disorders.
Collapse
Affiliation(s)
| | | | - Zhifang Zhai
- Department of Dermatology, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Mingwang Zhang
- Department of Dermatology, The First Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Petraglia L, Iacotucci P, Ferrillo L, Cabaro S, Somma J, Lacava F, Amaranto I, Crucito S, Perrotti M, Formisano P, Rengo G, Leosco D, Carnovale V. Effects of Oral Amino Acid Supplementation on Physical Activity, Systemic Inflammation, and Quality of Life in Adult Patients with Cystic Fibrosis: A Single-Center, Randomized, Double-Blind, Placebo-Controlled Pilot Study. Nutrients 2025; 17:1239. [PMID: 40218996 PMCID: PMC11990193 DOI: 10.3390/nu17071239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objective: Cystic Fibrosis (CF) is a common, life-threatening genetic disorder that leads to progressive lung function decline, respiratory failure, and premature death. Musculoskeletal complications, affecting both peripheral and respiratory muscles, are major concerns in CF patients. Inflammatory cytokines seem to be responsible for the activation of the molecular pathways involved in the imbalance between protein synthesis and catabolism, with consequent loss of muscle mass and function. This study aims to assess the effects of amino acid supplements on functional status, muscle mass and strength, inflammation, and quality of life in adult CF patients. Methods: We conducted a randomized, double-blind, placebo-controlled pilot trial with 60 adult CF patients, aged 18 or older. Participants were randomly assigned to receive either amino acid supplementation or a placebo for 4 weeks. Physical function tests and self-assessment questionnaires on quality of life, global health, and sleep status, as well as blood samples to measure pro-inflammatory cytokines, were performed at baseline and after the treatment period. Results: The amino acid supplementation group showed a significant improvement in self-perceived physical performance and health status. Interleukin-6 serum levels were significantly reduced in this group compared to those who received the placebo (p = 0.042). Conclusions: Amino acid supplementation in adult CF patients improves self-perception of health status and may reduce systemic inflammation, significantly decreasing serum levels of Interleukin-6. This suggests potential benefits for the overall well-being of CF patients and a reduction in their inflammatory status.
Collapse
Affiliation(s)
- Laura Petraglia
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
- ANASTE Humanitas Foundation, 00192 Rome, RM, Italy
| | - Paola Iacotucci
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, NA, Italy;
| | - Lorenza Ferrillo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Serena Cabaro
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Jolanda Somma
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Francesca Lacava
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Ilaria Amaranto
- Emergency Department, AORN San Pio, 82100 Benevento, BN, Italy;
| | - Silvia Crucito
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Maria Perrotti
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
- Istituti Clinici Scientifici Maugeri IRCCS, Scientific Institute of Telese Terme, 82037 Telese Terme, BN, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| | - Vincenzo Carnovale
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, NA, Italy; (L.P.); (L.F.); (S.C.); (J.S.); (F.L.); (S.C.); (M.P.); (P.F.); (G.R.); (V.C.)
| |
Collapse
|
10
|
Metwally H. STAT Signature Dish: Serving Immunity with a Side of Dietary Control. Biomolecules 2025; 15:487. [PMID: 40305224 PMCID: PMC12024614 DOI: 10.3390/biom15040487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Immunity is a fundamental aspect of animal biology, defined as the host's ability to detect and defend against harmful pathogens and toxic substances to preserve homeostasis. However, immune defenses are metabolically demanding, requiring the efficient allocation of limited resources to balance immune function with other physiological and developmental needs. To achieve this balance, organisms have evolved sophisticated signaling networks that enable precise, context-specific responses to internal and external cues. These networks are essential for survival and adaptation in multicellular systems. Central to this regulatory architecture is the STAT (signal transducer and activator of Transcription) family, a group of versatile signaling molecules that govern a wide array of biological processes across eukaryotes. STAT signaling demonstrates remarkable plasticity, from orchestrating host defense mechanisms to regulating dietary metabolism. Despite its critical role, the cell-specific and context-dependent nuances of STAT signaling remain incompletely understood, highlighting a significant gap in our understanding. This review delves into emerging perspectives on immunity, presenting dynamic frameworks to explore the complexity and adaptability of STAT signaling and the underlying logic driving cellular decision-making. It emphasizes how STAT pathways integrate diverse physiological processes, from immune responses to dietary regulation, ultimately supporting organismal balance and homeostasis.
Collapse
Affiliation(s)
- Hozaifa Metwally
- Laboratory of Immune Regulation, The World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Wang L, Qu M, Li L, Mei W, Zhang F, Hu Z, Li G, Xu L, Liang H. Effects of glycyrrhetinic acid on production performance, serum biochemical indexes, ruminal parameters, and rumen microflora of beef cattle. Front Vet Sci 2025; 12:1529383. [PMID: 40206252 PMCID: PMC11979252 DOI: 10.3389/fvets.2025.1529383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
This research was carried out to assess the impact of supplementing with glycyrrhetinic acid (GA) on production performance, serum biochemical indexes, ruminal parameters, and rumen bacterial flora of beef cattle. Twenty-four Simmental bulls were randomly assigned to two dietary treatments (n = 12 per treatment): the control treatment (basal ration, CON) and the GA treatment (basal ration supplemented with GA at 0.1% DM). After an 87-day feeding trial (7-day adaptation period and 80-day period dedicated to data and sample collection), feces, blood, and rumen fluid samples were collected on day 87. The GA addition significantly increased the average daily gain of beef cattle (p < 0.05). The GA treatment exhibited significantly greater apparent digestibility of crude protein, neutral detergent fiber, and acid detergent fiber than the control treatment (p < 0.05). Total volatile fatty acid concentration, microbial protein concentration, and propionic acid concentration in the rumen fluid were significantly increased by GA addition (p < 0.05). Compared with the control group, the interleukin-4 concentration was significantly higher in GA treatment (p < 0.05). The indices, including operational taxonomic units (OTUs), Sobs, Shannon, Ace, and Chao1, were found to be greater in the GA treatment. At the phyla level, GA addition (p < 0.05) significantly decreased the relative abundance of Bacteroidetes and increased the relative abundance of Firmicutes, while also significantly decreasing the Bacteroidetes:Firmicutes ratios. At the genera level, the relative abundance of Prevotella, NK4A214_group, norank_f_UCG-011, Prevotellaceae_UCG-003, Christensenellaceae_R-7_treatment, Prevotellaceae_UCG-001, norank_f_Bacteroidales_UCG-001, Pseudobutyrivibrio, and Butyrivibrio significantly differed due to GA addition (p < 0.05). Carbohydrate and amino acid transport and metabolism, as well as energy production and conversion, were significantly enriched in the GA treatment (p < 0.05). In summary, the findings indicated that adding glycyrrhetinic acid to the diet could improve growth performance and modify the rumen microbial composition and diversity of beef cattle.
Collapse
Affiliation(s)
- Long Wang
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| | - Mingren Qu
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| | - Lin Li
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| | - Wenliang Mei
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| | - Fengwei Zhang
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| | - Ziyu Hu
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| | - Geping Li
- Shenglong Cattle Industry Group Co., Ltd, Pingxiang Jiangxi, China
| | - Lanjiao Xu
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| | - Huan Liang
- Jiangxi Key Laboratory of Animal Nutrition, Nanchang, Jiangxi, China
| |
Collapse
|
12
|
Ye X, Ren D, Chen Q, Shen J, Wang B, Wu S, Zhang H. Resolution of inflammation during rheumatoid arthritis. Front Cell Dev Biol 2025; 13:1556359. [PMID: 40206402 PMCID: PMC11979130 DOI: 10.3389/fcell.2025.1556359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes synovial joint inflammation as well as bone destruction and erosion, typically characterized by joint pain, swelling, and stiffness, with complications and persistent pain after remission posing a significant health burden for RA patients. The etiology of RA has not yet been fully elucidated, but a large number of studies have shown that the initiation of inflammation in RA is closely related to T-cell activation, the production of a variety of pro-inflammatory cytokines, macrophage M1/M2 imbalance, homeostatic imbalance of the intestinal flora, fibroblast-like synoviocytes (FLSs) and synovial tissue macrophages (STMs) in the synovial lumen of joints that exhibit an aggressive phenotype. While the resolution of RA is less discussed, therefore, we provided a systematic review of the relevant remission mechanisms including blocking T cell activation, regulating macrophage polarization status, modulating the signaling pathway of FLSs, modulating the subpopulation of STMs, and inhibiting the relevant inflammatory factors, as well as the probable causes of persistent arthritis pain after the remission of RA and its pain management methods. Achieving resolution in RA is crucial for improving the quality of life and long-term prognosis of patients. Thus, understanding these mechanisms provide novel potential for further drug development and treatment of RA.
Collapse
Affiliation(s)
- Xiaoou Ye
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Dan Ren
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Qingyuan Chen
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Jiquan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Bo Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| |
Collapse
|
13
|
Li J, Han WH, Zhang MY, Fan JQ, Li GD, Li JY, Chen X. Hyper-IgE Syndrome: A Case Report with Insights from Bioinformatics Analysis of Key Pathways and Genes. Infect Drug Resist 2025; 18:1567-1580. [PMID: 40162035 PMCID: PMC11952148 DOI: 10.2147/idr.s507797] [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: 11/21/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose This study reports on a patient with High IgE Syndrome(HIES), focusing on clinical manifestations and pathogenic mechanisms through bioinformatics to enhance understanding and treatment. Patients and Methods The patient received appropriate interventions and was currently undergoing treatment with close monitoring. Additionally, bioinformatics analyses were conducted to investigate potential signaling pathways and key genes associated with HIES. Results A 28-year-old woman presented with a 6-month history of cough, worsening dyspnea, and eczema was diagnosed with HIES after elevated immunoglobulin levels and a STAT3 mutation. Initially, she declined immunoglobulin therapy, but showed improvement with sulfamethoxazole-trimethoprim and subsequently required intravenous immunoglobulin therapy for ongoing management. KEGG pathway analysis revealed that these genes were primarily associated with infection-related signaling pathways, consistent with the susceptibility to infections observed in HIES patients. Protein-protein interaction (PPI) network analysis highlighted the importance of key genes such as IL6, CDH2, and CLDN1. Conclusion Increased HIES awareness among healthcare providers is crucial for patients with recurrent infections, requiring a multidisciplinary approach. Our study identified IL6, CDH2, and CLDN1 as key factors in HIES progression, suggesting naive B cells and dormant mast cells may be involved.
Collapse
Affiliation(s)
- Juan Li
- Department of Comprehensive Ward, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an, People’s Republic of China
| | - Wei-Hua Han
- Department of Intensive Care Unit,The Affiliated Taian City Central Hospital of Qingdao University, Tai’an, People’s Republic of China
| | - Meng-Yu Zhang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Jia-Qi Fan
- Jining Medical University, Jining, People’s Republic of China
| | - Guo-Dong Li
- Department of Pulmonary and Critical Care Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an, People’s Republic of China
| | - Jun-Yi Li
- The Second Clinical Medical College of Nanchang University, Nanchang, People’s Republic of China
| | - Xiao Chen
- Department of Pulmonary and Critical Care Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an, People’s Republic of China
| |
Collapse
|
14
|
Qin G, Song R, Sun J, Dai J, Wang W, Meng F, Wang D, Liu Z, Sun B, Li C. Unveiling the Therapeutic Potential of Banxia Xiexin Decoction in Alzheimer's Disease: Insights From Network Pharmacology and Experimental Validation. Drug Des Devel Ther 2025; 19:2133-2155. [PMID: 40134954 PMCID: PMC11934878 DOI: 10.2147/dddt.s499852] [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: 10/23/2024] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Background Alzheimer's disease (AD) is associated with various pathological states for which there is no effective treatment. First documented in the Eastern Han Dynasty's medical classic, "Treatise on Febrile and Miscellaneous Diseases" (200-210 Anno Domini), Banxia Xiexin Decoction (BXD) stands as a quintessential approach to treating spleen ailments. Recent studies have shown BXD's effectiveness in mitigating memory impairment associated with AD. Yet, the precise mechanisms underlying BXD's action against AD require further exploration. Aim of the Study To explore the important components of BXD in exerting anti-AD effects and the underlying molecular mechanisms using network pharmacology, metabolomics analysis, and in vitro and in vivo validation strategies. Initially, candidates for BXD's application in AD therapy were identified through extensive database searches, followed by an analysis of protein-protein interactions (PPI). To elucidate BXD's therapeutic pathways in AD, we engaged in Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) assessments. Further, we delved into BXD's primary constituents through ultra-high-pressure liquid chromatography coupled with Q Exactive mass spectrometry and molecular docking techniques. Finally, AD-associated Aβ42-SY5Y cells and APPswe/PS1dE9 (APP/PS1) transgenic mice models were utilized to further determine the activity and mechanisms of BXD through various molecular or phenotypic assays and metabolomics analysis. Results Our findings identified the PI3K/Akt signaling pathways as central to BXD's effects. Using in vitro and in vivo models, we found the activity of BXD against AD to be mediated by the suppression of neuroinflammation and apoptosis, accompanied by activation of the PI3K/Akt pathway. Finally, we observed robust changes in metabolite levels in the plasma of BXD-treated APP/PS1 mice. Conclusion Through systematic data analysis and experimental validation, the therapeutic advantages and fundamental molecular mechanisms of BXD in treating AD were revealed. These findings underscore the promising prospects and compelling potential of BXD, which targets the PI3K/Akt signaling pathway and inflammation, apoptosis, as a therapeutic strategy for improving AD.
Collapse
Affiliation(s)
- Gaofeng Qin
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
- Postdoctoral Research Mobile Station, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Rongqiang Song
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Jingyi Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Juanjuan Dai
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Wentao Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Dan Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Zhe Liu
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Baoliang Sun
- Second AfFIliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, People’s Republic of China
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| |
Collapse
|
15
|
Czajkowski M, Wierzbicki PM, Dolny M, Matuszewski M, Hakenberg OW. Inflammation in Penile Squamous Cell Carcinoma: A Comprehensive Review. Int J Mol Sci 2025; 26:2785. [PMID: 40141426 PMCID: PMC11943298 DOI: 10.3390/ijms26062785] [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: 02/24/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammation appears to play a crucial role in the development and progression of penile cancer (PeCa). Two molecular pathways of PeCa are currently described: HPV-dependent and HPV-independent. The tumor immune microenvironment (TIME) of PeCa is characterized by the presence of tumor-associated macrophages, cancer-associated fibroblasts, and tumor-infiltrating lymphocytes. The components of the TIME produce pro-inflammatory cytokines and chemokines, which have been found to be overexpressed in PeCa tissues and are associated with tumor progression and unfavorable prognoses. Additionally, the nuclear factor kappa B (NF-κB) pathway and secreted phosphoprotein 1 (SPP1) have been implicated in PeCa pathogenesis. Elevated C-reactive protein (CRP) levels and the neutrophil-to-lymphocyte ratio (NLR) have been identified as potential prognostic biomarkers in PeCa. This overview presents the complex contribution of the inflammatory process and collates projects aimed at modulating TIME in PeCa.
Collapse
Affiliation(s)
- Mateusz Czajkowski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Piotr M. Wierzbicki
- Department of Histology, Medical University of Gdańsk, Dębinki, 80-211 Gdansk, Poland;
| | - Maciej Dolny
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Marcin Matuszewski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Oliver W. Hakenberg
- Department of Urology, University Medical Center Rostock, 18055 Rostock, Germany;
- Department of Urology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
16
|
Liu Y, Kong X, Sun Q, Cui T, Xu S, Ding C. Identification and validation of the common pathogenesis and hub biomarkers in Papillary thyroid carcinoma complicated by rheumatoid arthritis. PLoS One 2025; 20:e0317369. [PMID: 40063597 PMCID: PMC11892850 DOI: 10.1371/journal.pone.0317369] [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: 06/14/2024] [Accepted: 12/19/2024] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Papillary thyroid carcinoma coexisting with rheumatoid arthritis is frequently observed in clinical patients, yet its pathogenesis has not been fully elucidated. This investigation sought to further explore the molecular underpinnings of these two diseases. METHODS Gene expression profiles for thyroid papillary carcinoma and rheumatoid arthritis patients were obtained from the Comprehensive Gene Expression Database (GEO). Following the discovery of shared differentially expressed genes (DEGs) between these two conditions, three separate analyses were conducted. These included functional annotation, the establishment of a protein‒protein interaction (PPI) network and module, and the identification of hub genes via coexpression analysis. The final step involved the validation of target genes via clinical specimens. RESULTS This study analyzed datasets from four GEO databases and identified 64 common DEGs. Functional enrichment analysis revealed that these genes are predominantly associated with pathways related to immunity and signal transduction. Protein‒protein interaction (PPI) network analysis revealed complex interactions among these differentially expressed genes and highlighted several genes that may play pivotal roles in shared pathological mechanisms, namely, CCR5, CD4, IL6, CXCL13, FOXM1, CXCL9, and CXCL10. CONCLUSION Our study highlights the shared pathogenesis between papillary thyroid cancer and rheumatoid arthritis. Shared pathways and crucial genes could offer novel perspectives for subsequent investigations into the mechanisms of these diseases.
Collapse
MESH Headings
- Humans
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/complications
- Thyroid Cancer, Papillary/pathology
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/complications
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Protein Interaction Maps/genetics
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/complications
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Databases, Genetic
- Transcriptome
Collapse
Affiliation(s)
- Yingming Liu
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangjun Kong
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qianshu Sun
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianxing Cui
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shengnan Xu
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chao Ding
- General Surgery Ward four, Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Wang X, Guo Z, Xia Y, Wang X, Wang Z. Research Progress on the Immune Function of Liver Sinusoidal Endothelial Cells in Sepsis. Cells 2025; 14:373. [PMID: 40072101 PMCID: PMC11899273 DOI: 10.3390/cells14050373] [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: 01/12/2025] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025] Open
Abstract
Sepsis is a complex clinical syndrome closely associated with the occurrence of acute organ dysfunction and is often characterized by high mortality. Due to the rapid progression of sepsis, early diagnosis and intervention are crucial. Recent research has focused on exploring the pathological response involved in the process of sepsis. Liver sinusoidal endothelial cells (LSECs) are a special type of endothelial cell and an important component of liver non-parenchymal cells. Unlike general endothelial cells, which mainly provide a barrier function within the body, LSECs also have important functions in the clearance and regulation of the immune response. LSECs are not only vital antigen-presenting cells (APCs) in the immune system but also play a significant role in the development of infectious diseases and tumors through their specific immune regulatory pathways. However, in certain disease states, the functions of LSECs may be impaired, leading to immune imbalance and the development of organ failure. Investigating the immune pathways of LSECs in sepsis may provide new solutions for the prevention and treatment of sepsis and is crucial for maintaining microcirculation and improving patient outcomes.
Collapse
Affiliation(s)
- Xinrui Wang
- School of Clinical Medicine, Tsinghua University, Beijing 100190, China; (X.W.); (Y.X.)
| | - Zhe Guo
- Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 100084, China;
| | - Yuxiang Xia
- School of Clinical Medicine, Tsinghua University, Beijing 100190, China; (X.W.); (Y.X.)
| | - Xuesong Wang
- School of Clinical Medicine, Tsinghua University, Beijing 100190, China; (X.W.); (Y.X.)
- Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 100084, China;
| | - Zhong Wang
- School of Clinical Medicine, Tsinghua University, Beijing 100190, China; (X.W.); (Y.X.)
- Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 100084, China;
| |
Collapse
|
18
|
Yan S, Zhao Y, Yang Y, Liu B, Xu W, Ma Z, Yang Q. Progress of ADAM17 in Fibrosis-Related Diseases. Mediators Inflamm 2025; 2025:9999723. [PMID: 40224489 PMCID: PMC11986189 DOI: 10.1155/mi/9999723] [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: 07/27/2024] [Accepted: 01/09/2025] [Indexed: 04/15/2025] Open
Abstract
Fibrosis leads to structural damage and functional decline and is characterized by an accumulation of fibrous connective tissue and a reduction in parenchymal cells. Because of its extremely poor prognosis, organ fibrosis poses a significant economic burden. In order to prevent and treat fibrosis more effectively, potential mechanisms need to be investigated. A disintegrin and metalloprotease 17 (ADAM17) is a membrane-bound protein. It regulates intracellular signaling and membrane protein degradation. Fibrosis mediated by ADAM17 has been identified as an important contributor, although the specific relationship between its multiple regulatory functions and the pathogenesis is unclear. This article describes ADAM17 activation, function, and regulation, as well as the role of ADAM17 mediated fibrosis injury in kidney, liver, heart, lung, skin, endometrium, and retina. To develop new therapeutic approaches based on ADAM17 related signal pathways.
Collapse
Affiliation(s)
- Suyan Yan
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Yaqi Zhao
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| | - Yuyu Yang
- UCL School of Pharmacy, University College London, London, UK
| | - Baocheng Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Wei Xu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| | - Zhenzhen Ma
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
- Shandong University of Traditional Chinese Medicine, Jinan 250021, Shandong, China
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| |
Collapse
|
19
|
Oliver MA, Alward KJ, Rhoads ML, Ealy AD. Human Recombinant Interleukin-6 and Leukemia Inhibitory Factor Improve Inner Cell Mass Cell Number but Lack Cryoprotective Activities on In Vitro-Produced Bovine Blastocysts. Animals (Basel) 2025; 15:668. [PMID: 40075953 PMCID: PMC11899334 DOI: 10.3390/ani15050668] [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: 12/14/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
This work explored whether supplementing recombinant human interleukin-6 (IL6), interleukin-11 (IL11), or leukemia inhibitory factor (LIF) improves IVP bovine embryo development, morphology, and cryosurvivability. Embryos were treated from day 5 to 8 post-fertilization with either the carrier only (control) or 100 ng/mL of IL6, IL11, or LIF. Blastocyst formation and stage were determined on day 7 and 8. A subset of day 8 blastocysts was processed for immunofluorescence to count trophectoderm (TE) and inner cell mass (ICM) cell numbers and another subset was slow frozen and stored in liquid nitrogen until thawing. No differences in the blastocyst rate or blastocyst stage of development were detected. Increases in ICM cell numbers were observed for IL6 and LIF but not the IL11 treatment. None of the cytokine treatments applied before freezing affected post-thaw survival, TE or ICM cell number, or cell death 24 h after thawing. In conclusion, supplementing IL6 and LIF improves ICM cell numbers in non-frozen blastocysts, but there was no evidence that any of these cytokine treatments contain cryoprotective properties in bovine embryos.
Collapse
Affiliation(s)
| | | | | | - Alan D. Ealy
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (M.A.O.); (K.J.A.); (M.L.R.)
| |
Collapse
|
20
|
D’Amico RC, Nagashima S, Carstens LB, Bertoldi KDG, Mataruco S, Honório D’Agostini JC, Hlatchuk EC, da Silva SB, de Noronha L, Baena CP. COVID-19 Induces Greater NLRP3 Inflammasome Activation in Obese Patients than Other Chronic Illnesses: A Case-Control Study. Int J Mol Sci 2025; 26:1541. [PMID: 40004007 PMCID: PMC11855377 DOI: 10.3390/ijms26041541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity has been identified as an independent risk factor for severe COVID-19 unfavorable outcomes. Several factors, such as increased ACE2 receptor expression and chronic inflammation, can contribute to this relationship, yet the activation of the NLRP3 inflammasome pathway is also a key element. Our primary goal was to determine whether chronic NLRP3 inflammasome activation in people with obesity is different in critical COVID-19 and in critical chronic conditions. A retrospective analysis was conducted using clinical data and post-mortem lung tissue samples from 14 COVID-19 patients with obesity (group A) and 9 patients with obesity who died from non-COVID-19 causes (group B). Immunohistochemical analysis assessed twelve markers related to the NLRP3 inflammasome pathway. Group A showed a significantly higher expression of ASC (p = 0.0387) and CASP-1 (p = 0.0142). No significant differences were found for IL-8, TNF-α, NF-kB, NLRP3, IL-1β, and gasdermin-D. Group B had higher levels of IL-6 (p < 0.0001), IL-18 (p = 0.002), CASP-9 (p < 0.0001), and HIF (p = 0.0327). We concluded that COVID-19 activates the NLRP3 inflammasome pathway, possibly leading to pyroptotic cell death mediated by caspase-1. In contrast, people with obesity without COVID-19, despite exhibiting some markers of the NLRP3 inflammasome, are more likely to experience necroptosis mediated by caspase-9.
Collapse
Affiliation(s)
- Raíssa Campos D’Amico
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Seigo Nagashima
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Lucas Baena Carstens
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Karina de Guadalupe Bertoldi
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
| | - Sabrina Mataruco
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | | | - Elisa Carolina Hlatchuk
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Sofia Brunoro da Silva
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| | - Lucia de Noronha
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
- School of Medicine, Universidade Federal do Paraná, Curitiba 80060-240, Paraná, Brazil; (J.C.H.D.); (E.C.H.)
| | - Cristina Pellegrino Baena
- Post-Graduate Program in Health Sciences (PPGCS), Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (R.C.D.); (S.N.); (L.B.C.); (K.d.G.B.); (C.P.B.)
- School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil; (S.M.); (S.B.d.S.)
| |
Collapse
|
21
|
Yang X, Wang X, Yang J. Single-cell analysis reveals cellular heterogeneity, gene expression profiles, and pathway dynamics in acne vulgaris. Arch Dermatol Res 2025; 317:362. [PMID: 39920471 DOI: 10.1007/s00403-025-03894-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/12/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
Acne vulgaris is a common dermatological condition, particularly in adolescents, and is associated with significant physical and psychological impacts. Its pathogenesis involves genetic factors, sebaceous gland dysfunction, inflammatory responses, and alterations in skin microbiota. Despite advancements in treatment, a comprehensive understanding of its cellular and molecular mechanisms remains limited, making therapeutic strategies challenging. Single-cell RNA sequencing (scRNA-seq) data from six acne vulgaris patients were obtained from the GEO database (GSE175817), filtered, and integrated. Differentially expressed genes (DEGs) were identified and subjected to functional enrichment analysis to explore the underlying pathogenesis. Gene set variation analysis (GSVA) was used to investigate metabolic pathway alterations, and ligand-receptor interactions were analyzed to examine cell-to-cell communication between lesional and non-lesional skin tissues. The integration of scRNA-seq data yielded the identification of eight distinct cell clusters, including endothelial cells, myeloid cells, lymphocytes, melanocytes, sebaceous gland cells, smooth muscle cells, keratinocytes and fibroblasts. The proportions of lymphoid and myeloid cells were found to be significantly different between the lesional and non-lesional sites. The differential expression of genes (DEGs) was found to be significantly specific to the different cell clusters. Abnormal intercellular communication was found to result in a substantial increase in the number and intensity of communications in the area of acne vulgaris lesions. Moreover, specific ligand-receptor pairs for SPP1 and IL6 associated with acne vulgaris were identified. Furthermore, the presence of specific alterations in metabolic pathways, including riboflavin metabolism, niacin metabolism, and lipoic acid metabolism, was observed. Our findings demonstrate the cellular heterogeneity and dysfunction of intercellular communication and metabolic signaling in the lesional skin tissues of patients with acne vulgaris. These findings have important implications for understanding the complex biological processes of acne vulgaris.
Collapse
Affiliation(s)
- Xiaoyi Yang
- School of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| | - Xiaoyan Wang
- Department of Experimental Teaching Center, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, Guangxi, China.
- Key Laboratory of Biochemistry and Molecular Biology, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, Guangxi, China.
| | - Jiankang Yang
- School of Basic Medical Sciences, Dali University, Dali, Yunnan, China.
| |
Collapse
|
22
|
Li B, Dong B, Xie L, Li Y. Exploring Advances in Natural Plant Molecules for Allergic Rhinitis Immunomodulation in Vivo and in Vitro. Int J Gen Med 2025; 18:529-565. [PMID: 39911299 PMCID: PMC11796455 DOI: 10.2147/ijgm.s493021] [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: 08/25/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
Allergic rhinitis (AR) is a prevalent allergic disease that imposes significant economic burdens and life pressures on individuals, families, and society, particularly in the context of accelerating globalization and increasing pathogenic factors. Current clinical therapies for AR include antihistamines, glucocorticoids administered via various routes, leukotriene receptor antagonists, immunotherapy, and several decongestants. These treatments have demonstrated efficacy in alleviating clinical symptoms and pathological states. However, with the growing awareness of AR and rising expectations for improvements in quality of life, these treatments have become associated with a higher incidence of side effects and an elevated risk of drug resistance. Furthermore, the development of AR is intricately associated with dysregulation of the immune system, yet the underlying pathogenetic mechanisms remain incompletely understood. In contrast, widely available natural plant molecules offer multiple targeting pathways that uniquely modify the typical pathophysiology of AR through immunomodulatory processes. This review presents a comprehensive analysis of both in vivo and in vitro studies on natural plant molecules that modulate immunity for treating AR. Additionally, we examine their specific mechanisms of action in animal models to provide new insights for developing safe and effective targeted therapies while guiding experimental and clinical applications against AR.
Collapse
Affiliation(s)
- Bingquan Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Boyang Dong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Liangzhen Xie
- Ear-Nose-Throat Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Yan Li
- Ear-Nose-Throat Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
23
|
Weitz HT, Ettich J, Rafii P, Wittich C, Schultz L, Frank NC, Heise D, Krusche M, Lokau J, Garbers C, Behnke K, Floss DM, Kolmar H, Moll JM, Scheller J. Interleukin-11 receptor is an alternative α-receptor for interleukin-6 and the chimeric cytokine IC7. FEBS J 2025; 292:523-536. [PMID: 39473075 PMCID: PMC11796321 DOI: 10.1111/febs.17309] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/30/2024] [Accepted: 10/16/2024] [Indexed: 02/06/2025]
Abstract
The cytokine interleukin 6 (IL-6) signals via the IL-6 α-receptor (IL-6Rα or IL-6R) in complex with the gp130 β-receptor. Cell type restricted expression of the IL-6R limits the action of IL-6 mainly to hepatocytes and some immune cells. Here, we show that IL-6 also binds to the IL-11 α receptor (IL-11Rα or IL-11R) and induces signaling via IL-11R:gp130 complexes, albeit with a lower affinity compared to IL-11. Antagonistic antibodies directed against IL-11R, but not IL-6R, inhibit IL-6 signaling via IL-11R:gp130 receptor complexes. Notably, IL-11 did not cross-react with IL-6R. IL-11R has also been identified as an alternative α receptor for the CNTF/IL-6-derived chimeric cytokine IC7, which has recently been shown to induce weight loss in mice. Accordingly, the effects of therapeutic monoclonal antibodies against IL-6 or IL-6R, which both block IL-6 signaling, may be slightly different. These findings provide new insights into IL-6 signaling and therefore offer new potential therapeutic intervention options in the future.
Collapse
Affiliation(s)
- Hendrik T. Weitz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Christoph Wittich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Laura Schultz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Nils C. Frank
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Denise Heise
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Matthias Krusche
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Juliane Lokau
- Institute of Clinical BiochemistryHannover Medical SchoolGermany
| | | | - Kristina Behnke
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Doreen M. Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Harald Kolmar
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtGermany
- Centre of Synthetic BiologyTechnical University of DarmstadtGermany
| | - Jens M. Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| |
Collapse
|
24
|
Marczak M, Krejner-Bienias A, Jasińska A, Kulus M, Miklis P, Grzela K, Grzela T. The High Levels of Soluble Receptors for Tumor Necrosis Factor and Heart Injury in Children with the Pediatric Inflammatory Multisystem Syndrome Associated with Coronavirus Infection: Is This Just a Coincidence? A Proof-of-Concept Study. Int J Mol Sci 2025; 26:924. [PMID: 39940694 PMCID: PMC11817279 DOI: 10.3390/ijms26030924] [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: 11/27/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
(1) Pediatric inflammatory multisystem syndrome (PIMS) is a relatively rare complication of coronavirus disease (COVID-19). So far, it is unclear why COVID-19 in children has usually mild or asymptomatic courses, whereas PIMS, which develops several weeks after COVID-19, is a serious life-threatening condition. (2) In this proof-of-concept study, using the ELISA method, we compared selected clinical and immunological parameters in small groups of children with PIMS and COVID-19. Children with various inflammatory diseases were included as a control. (3) Patients with PIMS revealed significantly higher levels of pro-inflammatory molecules (C-reactive protein and IL-6) and markers of heart injury (troponin I and N-terminal prohormone of brain natriuretic peptide) as compared to other groups. Moreover, these markers correlated with increased levels of soluble receptors for tumor necrosis factor (sTNF-R1 and sTNF-R2). (4) Our observation may be a step forward to better understand the phenomenon of mild COVID-19 in children and its severe complications in PIMS. It is hypothesized that the delayed inflammation results in excessive cardiomyocyte damage and the release of sTNF-R1 and -R2. Therefore, possibly the involvement of the TNF pathway in PIMS could be explored as a potential therapeutic target. However, further studies are required to validate this approach.
Collapse
Affiliation(s)
- Maciej Marczak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Alicja Krejner-Bienias
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Agnieszka Jasińska
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Marek Kulus
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł Miklis
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Katarzyna Grzela
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Tomasz Grzela
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
25
|
Zhao Z, Chen T, Liu Q, Hu J, Ling T, Tong Y, Han Y, Zhu Z, Duan J, Jin Y, Fu D, Wang Y, Pan C, Keyoumu R, Sun L, Li W, Gao X, Shi Y, Dou H, Liu Z. Development and Validation of a Diagnostic Model for Stanford Type B Aortic Dissection Based on Proteomic Profiling. J Inflamm Res 2025; 18:533-547. [PMID: 39816951 PMCID: PMC11734266 DOI: 10.2147/jir.s494191] [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: 09/02/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
Purpose Stanford Type B Aortic Dissection (TBAD), a critical aortic disease, has exhibited stable mortality rates over the past decade. However, diagnostic approaches for TBAD during routine health check-ups are currently lacking. This study focused on developing a model to improve the diagnosis in a population. Patients and Methods Serum biomarkers were investigated in 88 participants using proteomic profiling combined with machine learning. The findings were validated using ELISA in other 80 participants. Subsequently, a diagnostic model for TBAD integrating biomarkers with clinical indicators was developed and assessed using machine learning. Results Six differentially expressed proteins (DEPs) were identified through proteomic profiling and machine learning in discovery and derivation cohorts. Five of these (GDF-15, IL6, CD58, LY9, and Siglec-7) were further verified through ELISA validation within the validation cohort. In addition, ten blood-related indicators were selected as clinical indicators. Combining biomarkers and clinical indicators, the machine learning-based models performed well (AUC of the biomarker model = 0.865, AUC of the clinical model = 0.904, and AUC of the combined model = 0.909) using relative quantitation. The performance of the three models was verified (AUC of biomarker model = 0.866, AUC of clinical model = 0.868, and AUC of combined model = 0.886) using absolute quantitation. Crucially, the combined models outperformed individual biomarkers and clinical models, demonstrating superior efficacy. Conclusion Using proteomic profiling, we identified serum IL-6, GDF-15, CD58, LY9, and Siglec-7 as TBAD biomarkers. The machine-learning-based diagnostic model exhibited significant potential for TBAD diagnosis using only blood samples within the population.
Collapse
Affiliation(s)
- Zihe Zhao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Taicai Chen
- The State Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, People’s Republic of China
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, People’s Republic of China
| | - Qingyuan Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jianhang Hu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Tong Ling
- The State Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, People’s Republic of China
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, People’s Republic of China
| | - Yuanhao Tong
- Department of Thoracic Surgery, BenQ Medical Center, Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yuexue Han
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Zhengyang Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Jianfeng Duan
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Yi Jin
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Dongsheng Fu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Yuzhu Wang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Chaohui Pan
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Reyaguli Keyoumu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Lili Sun
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Wendong Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Xia Gao
- Department of Otolaryngology, Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
- Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Yinghuan Shi
- The State Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, People’s Republic of China
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, People’s Republic of China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, People’s Republic of China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, People’s Republic of China
| | - Zhao Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
26
|
Varanasi SK, Chen D, Liu Y, Johnson MA, Miller CM, Ganguly S, Lande K, LaPorta MA, Hoffmann FA, Mann TH, Teneche MG, Casillas E, Mangalhara KC, Mathew V, Sun M, Jensen IJ, Farsakoglu Y, Chen T, Parisi B, Deota S, Havas A, Lee J, Chung HK, Schietinger A, Panda S, Williams AE, Farber DL, Dhar D, Adams PD, Feng GS, Shadel GS, Sundrud MS, Kaech SM. Bile acid synthesis impedes tumor-specific T cell responses during liver cancer. Science 2025; 387:192-201. [PMID: 39787217 DOI: 10.1126/science.adl4100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/26/2024] [Accepted: 10/30/2024] [Indexed: 01/12/2025]
Abstract
The metabolic landscape of cancer greatly influences antitumor immunity, yet it remains unclear how organ-specific metabolites in the tumor microenvironment influence immunosurveillance. We found that accumulation of primary conjugated and secondary bile acids (BAs) are metabolic features of human hepatocellular carcinoma and experimental liver cancer models. Inhibiting conjugated BA synthesis in hepatocytes through deletion of the BA-conjugating enzyme bile acid-CoA:amino acid N-acyltransferase (BAAT) enhanced tumor-specific T cell responses, reduced tumor growth, and sensitized tumors to anti-programmed cell death protein 1 (anti-PD-1) immunotherapy. Furthermore, different BAs regulated CD8+ T cells differently; primary BAs induced oxidative stress, whereas the secondary BA lithocholic acid inhibited T cell function through endoplasmic reticulum stress, which was countered by ursodeoxycholic acid. We demonstrate that modifying BA synthesis or dietary intake of ursodeoxycholic acid could improve tumor immunotherapy in liver cancer model systems.
Collapse
Affiliation(s)
- Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Yingluo Liu
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Melissa A Johnson
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Cayla M Miller
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Souradipta Ganguly
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, School of Medicine, University of California, San Diego, CA, USA
| | - Kathryn Lande
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael A LaPorta
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Filipe Araujo Hoffmann
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Thomas H Mann
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marcos G Teneche
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Eduardo Casillas
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kailash C Mangalhara
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Varsha Mathew
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ming Sun
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Isaac J Jensen
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Timothy Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Bianca Parisi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Shaunak Deota
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Aaron Havas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - H Kay Chung
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, New York, NY, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - April E Williams
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Debanjan Dhar
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, School of Medicine, University of California, San Diego, CA, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Gen-Sheng Feng
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gerald S Shadel
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mark S Sundrud
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Center for Digestive Health, Dartmouth Health, Lebanon, NH, USA
- Dartmouth Cancer Center, Dartmouth Health, Lebanon, NH, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
27
|
Lokau J, Garbers Y, Vicente MM, Dittrich A, Meltendorf S, Lingel H, Münster-Kühnel AK, Brunner-Weinzierl M, Garbers C. Long-term increase in soluble interleukin-6 receptor levels in convalescents after mild COVID-19 infection. Front Immunol 2025; 15:1488745. [PMID: 39835136 PMCID: PMC11743636 DOI: 10.3389/fimmu.2024.1488745] [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: 08/30/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Serum levels of interleukin-6 (IL-6) are increased in COVID-19 patients. IL-6 is an effective therapeutic target in inflammatory diseases and tocilizumab, a monoclonal antibody that blocks signaling via the IL-6 receptor (IL-6R), is used to treat patients with severe COVID-19. However, the IL-6R exists in membrane-bound and soluble forms (sIL-6R), and the sIL-6R in combination with soluble glycoprotein 130 (sgp130) forms an IL-6-neutralizing buffer system capable of neutralizing small amounts of IL-6. Methods In this study, we analyzed serum levels of IL-6, sIL-6R and sgp130 in the serum of COVID-19 convalescent individuals with a history of mild COVID-19 disease and in acute severely ill COVID-19 patients compared to uninfected control subjects. Furthermore, we used single cell RNA sequencing data in order to determine which immune cell types are sources and targets of the individual cytokines and whether their expression is altered in severe COVID-19 patients. Results We find that sIL-6R levels are not only increased in acute severely ill patients, but also in convalescents after a mild COVID-19 infection. We show that this increase in sIL-6R results in an enhanced capacity of the sIL-6R/sgp130 buffer system, but that significantly enhanced free IL-6 is still present due to an overload of the buffer. Further, we identify IL-6 serum levels, age and the number of known pre-existing medical conditions as crucial determinants of disease outcome for the patients. We also show that IL-11 has no major systemic role in COVID-19 patients and that sCD25 is only increased in acute severely ill COVID-19 patients, but not in mild convalescent individuals. Discussion In conclusion, our study shows long-lasting alterations of the IL-6 system after COVID-19 disease, which might be relevant when applying anti-IL-6 or anti-IL-6R therapy.
Collapse
Affiliation(s)
- Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Yvonne Garbers
- Faculty of Management, Culture and Technology (Lingen campus), Osnabrück University of Applied Sciences, Lingen, Germany
| | - Manuel M. Vicente
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Anna Dittrich
- Department of Systems Biology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Stefan Meltendorf
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | - Monika Brunner-Weinzierl
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| |
Collapse
|
28
|
Peng Y, Liu C, Liu W, Gan R. Causal Relationship Between Gluten-Free Diet and Autoimmune-Related Disease Risk: A Comprehensive Mendelian Randomization Study. Int J Med Sci 2025; 22:432-440. [PMID: 39781523 PMCID: PMC11704703 DOI: 10.7150/ijms.104928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/14/2024] [Indexed: 01/12/2025] Open
Abstract
While the gluten-free diet (GFD) is primarily used to treat celiac disease (CD), recent research suggests it may also offer benefits for autoimmune-related diseases (ARDs), though findings remain inconsistent. This study aimed to investigate the potential protective effect of a GFD against ARDs by Mendelian Randomization (MR) analysis. Utilizing data from over 500,000 samples from the UK Biobank and other publicly available genome-wide association studies (GWAS), MR analysis revealed a significant negative causal relationship between GFD and the risk of developing rheumatoid arthritis (RA) (OR = 0.782, 95% CI = [0.727-0.841], p < 0.001). Mediation analysis identified immune cells such as CD14+ CD16+ monocyte absolute count (mediating 2.441% of the effect), CD14+ CD16+ monocyte percentage (2.346%), and CD20 on IgD+ CD38^dim B cells (3.119%) as potential mediators in the protective effect of GFD on RA. These findings suggest that GFD may help reduce RA risk by modulating specific immune cell populations. However, further research is necessary to clarify the exact mechanisms underlying these associations.
Collapse
Affiliation(s)
- Yi Peng
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chenxi Liu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, 410008, China
| | - Runxin Gan
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, 410008, China
| |
Collapse
|
29
|
Zhang Y, Li R, Chen X, Li Y, Zhang Q, Yang L, Wang L, Sun Y, Mao F, Zhuo CJ. Clozapine Induces Agranulocytosis via Inflammatory and Hematopoietic Cytokine Induction of the JAK-STAT Signaling Pathway: Evidence From Network Pharmacology and Molecular Docking. CNS Neurosci Ther 2025; 31:e70206. [PMID: 39776289 PMCID: PMC11707432 DOI: 10.1111/cns.70206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/09/2024] [Accepted: 12/01/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Clozapine exhibits significant therapeutic efficacy in schizophrenia, especially treatment-resistant schizophrenia. However, clozapine can cause agranulocytosis, a fatal adverse effect, and the aim of this study is to explore this mechanism based on network pharmacology and molecular docking. METHOD Six and two databases were used to identify targets associated with clozapine and agranulocytosis, respectively. The bioinformatics online platform was used to identify overlaps between the drug and disease targets. The protein-protein interaction (PPI) network was characterized using Cystoscope 3.10.1 and STRING. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) were analyzed using the DAVID online platform. A drug-target-pathway-disease network was constructed utilizing Cystoscope 3.10.1. The Auto Dock Vina and PyMOL software were used to verify the molecular docking of clozapine and core targets. RESULTS The analysis revealed 188 overlapping targets. The PPI and KEGG enrichment pathway analyses demonstrated that clozapine induces agranulocytosis by modulating the hematopoietic cell lineage and JAK-STAT signaling pathways via interleukin-3 (IL3), IL6, IL2 receptor subunit alpha (IL2RA), and granulocyte colony-stimulating factor. Binding energies between clozapine and core targets were favorable (< -7.0 kcal/mol). CONCLUSION Clozapine-induced agranulocytosis may be linked to the JAK-STAT inflammatory signaling pathway through inflammatory and hematopoietic-related cytokines. Our findings enhance our comprehension of the potential mechanisms underlying clozapine-induced agranulocytosis.
Collapse
Affiliation(s)
- Ying Zhang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Department of Psychiatry and Psychology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Ranli Li
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Ximing Chen
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Yachen Li
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Qiuyu Zhang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Lei Yang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Lina Wang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Yun Sun
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Fuqiang Mao
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Department of Psychiatry and Psychology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Chuan Jun Zhuo
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
30
|
Kiatsoonthon K, Phimthong N, Potikanond S, Wikan N, Nimlamool W. Panduratin A Inhibits TNF Alpha-Stimulated Endothelial Cell Activation Through Suppressing the NF-κB Pathway. Biomolecules 2024; 15:34. [PMID: 39858429 PMCID: PMC11762725 DOI: 10.3390/biom15010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Upon exposure to inflammatory stimuli including TNF-α, endothelial cells are activated leading to the adhesion of monocytes to their surface. These events are involved in the pathophysiology of atherosclerosis. Since TNF-α activates the NF-κB pathway, which contributes to atherosclerosis, targeting this signaling pathway may help prevent the risk of developing the disease. The current study elucidated the inhibitory effect of panduratin A (PA) on TNF-α-induced endothelial activation and monocyte adhesion. We discovered that PA reduced the level of pro-inflammatory cytokine IL-6 and chemokine MCP-1 in the media collected from endothelial cells stimulated with TNF-α. In addition, PA inhibited the expression of ICAM-1 and VCAM-1 on the surface of TNF-α-induced endothelial cells resulting in a decrease in the number of monocytes attached to endothelial cell surface. Mechanistically, PA prevented IκB degradation and specifically suppressed NF-κB phosphorylation and nuclear translocation in endothelial cells. However, PA had no inhibitory effect on the phosphorylation of AKT, ERK1/2, p38, and JNK. Taken together, PA blocked the production of cytokine and chemokine, adhesion molecules, and monocyte adhesion in response to TNF-α stimulation, in part, through NF-κB inhibition. Our study suggests that PA may possibly be effective in blocking the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Kriangkrai Kiatsoonthon
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Nitchakarn Phimthong
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
- PhD’s Degree Program in Pharmacology, Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| |
Collapse
|
31
|
Li B, Jiang X, Liu C, Ma Y, Zhao R, Zhang H. Exploring the preventive effects of Jie Geng Tang on pulmonary fibrosis induced in vitro and in vivo: a network pharmacology approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:10005-10016. [PMID: 38961002 DOI: 10.1007/s00210-024-03262-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Pulmonary fibrosis is a debilitating lung disease marked by excessive fibrotic tissue accumulation, which significantly impairs respiratory function. Given the limitations of current therapies, there is an increasing interest in exploring traditional herbal formulations like Jie Geng Tang (JGT) for treatment. This study examines the potential of JGT and its bioactive component, quercetin, in reversing bleomycin (BLM)-induced pulmonary fibrosis in mice. We employed a BLM-induced MLE-12 cell damage model for in vitro studies and a bleomycin-induced fibrosis model in C57BL/6 mice for in vivo experiments. In vitro assessments showed that JGT significantly enhanced cell viability and reduced apoptosis in MLE-12 cells treated with BLM. These findings underscore JGT's potential for cytoprotection against fibrotic agents. In vivo, JGT was effective in modulating the expression of E-cadherin and vimentin, key markers of the epithelial-mesenchymal transition (EMT) pathway, indicating its role in mitigating EMT-associated fibrotic changes in lung tissue. Quercetin, identified through network pharmacology analysis as a potential key bioactive component of JGT, was highlighted for its role in the regulatory mechanisms underlying fibrosis progression, particularly through the modulation of the IL-17 pathway and Il6 expression. By targeting inflammatory pathways and key processes like EMT, JGT and quercetin offer a potent alternative to conventional therapies, meriting further clinical exploration to harness their full therapeutic potential in fibrotic diseases.
Collapse
Affiliation(s)
- Bingxin Li
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Xiaojie Jiang
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Chang Liu
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Yun Ma
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Ruining Zhao
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China
| | - Haijun Zhang
- School of Life Science, Huaibei Normal University, Dongshan Road 100, Huaibei, 235000, China.
| |
Collapse
|
32
|
Wang ZX, Qin RJ, Yu LL, Nurlan A, Jing XH, Li M. Acupuncture Treats Sepsis through Immune Modulation and Organ Protection. Curr Med Sci 2024; 44:1185-1192. [PMID: 39673001 DOI: 10.1007/s11596-024-2957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/14/2024] [Indexed: 12/15/2024]
Abstract
Sepsis is a secondary condition resulting from severe systemic infections. It is a significant contributor to mortality in critically ill patients with rapid onset and severe symptoms. Acupuncture is a traditional Chinese medical treatment. Recent clinical studies have demonstrated that acupuncture, as an important synergistic therapy, has promising therapeutic effects in the treatment of sepsis. This paper reviews the mechanisms of immunomodulation and target organ protection associated with acupuncture and synergistic drug acupuncture in the treatment of sepsis. It also integrates existing studies to elucidate the modulation of the immune system and the protective effect of acupuncture on target organs.
Collapse
Affiliation(s)
- Zi-Xiao Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ren-Jie Qin
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling-Ling Yu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Arman Nurlan
- College of Acupuncture and Massage, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330000, China
| | - Xiang-Hong Jing
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100000, China.
| | - Man Li
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
33
|
Stajer M, Horacek JM, Kupsa T, Zak P. The role of chemokines and interleukins in acute lymphoblastic leukemia: a systematic review. J Appl Biomed 2024; 22:165-184. [PMID: 40033805 DOI: 10.32725/jab.2024.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/15/2024] [Indexed: 03/05/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood hematological malignancy, but it also affects adult patients with worse prognosis and outcomes. Leukemic cells benefit from protective mechanisms, which are mediated by intercellular signaling molecules - cytokines. Through these signals, cytokines modulate the biology of leukemic cells and their surroundings, enhancing the proliferation, survival, and chemoresistance of the disease. This ultimately leads to disease progression, refractoriness, and relapse, decreasing the chances of curability and overall survival of the patients. Targeting and modulating these pathological processes without affecting the healthy physiology is desirable, offering more possibilities for the treatment of ALL patients, which still remains unsatisfactory in certain cases. In this review, we comprehensively analyze the existing literature and ongoing trials regarding the role of chemokines and interleukins in the biology of ALL. Focusing on the functional pathways, genetic background, and critical checkpoints, we constructed a summary of molecules that are promising for prognostic stratification and mainly therapeutic use. Targeted therapy, including chemokine and interleukin pathways, is a new and promising approach to the treatment of cancer. With the expansion of our knowledge, we are able to uncover a spectrum of new potential checkpoints in order to modulate the disease biology. Several cytokine-related targets are advancing toward clinical application, offering the hope of higher disease response rates to treatment.
Collapse
Affiliation(s)
- Martin Stajer
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Jan M Horacek
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Tomas Kupsa
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Pavel Zak
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| |
Collapse
|
34
|
Li Z, Gan H, Li S, Xue Y, Luo K, Huang K, Zhang Y, Wang Y, Jiang L, Zhang H. Bioinformatics Identification and Validation of Ferroptosis-Related Key Genes and Therapeutic Compounds in Septic Lung Injury. J Inflamm Res 2024; 17:9215-9230. [PMID: 39600675 PMCID: PMC11589777 DOI: 10.2147/jir.s476522] [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: 05/02/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Background Septic lung injury (SLI) is a severe condition with high mortality, and ferroptosis, a form of programmed cell death, is implicated in its pathogenesis. However, the explicit mechanisms underlying this condition remain unclear. This study aimed to elucidate and validate key ferroptosis-related genes involved in the pathogenesis of SLI through bioinformatics analysis and experimental validation. Methods Microarray data related to SLI from the GSE130936 dataset were downloaded from the Gene Expression Omnibus (GEO) database. These data were then intersected with the FerrDb database to obtain ferroptosis-related differentially expressed genes (DEGs). Protein-protein interaction (PPI) networks and functional enrichment analysis were employed to identify key ferroptosis-related DEGs. The Connectivity Map (c-MAP) tool was used to search for potential compounds or drugs that may inhibit ferroptosis-related DEGs. The transcriptional levels of the key genes and potential therapeutic compounds were verified in an LPS-induced mouse model of lung injury. The expression of these key genes was further verified using the GSE60088 and GSE137342 datasets. Results 38 ferroptosis-related DEGs were identified between the septic and control mice. PPI network analysis revealed four modules, the most significant of which included eight ferroptosis-related DEGs. Functional enrichment analysis showed that these genes were enriched in the HIF-1 signaling pathway, including IL-6 (Interleukin-6), TIMP1 (Tissue Inhibitor of Metalloproteinase 1), HIF-1α (Hypoxia-Inducible Factor-1α), and HMOX1 (Heme Oxygenase-1). Phloretin, a natural compound, was identified as a potential inhibitor of these genes. Treatment with phloretin significantly reduced the expression of these genes (p < 0.05), mitigated lung injury, improved inflammatory profiles by approximately 50%, and ferroptosis profiles by nearly 30% in the SLI models. Conclusion This study elucidates the significant role of ferroptosis in SLI and identifies phloretin as a potential therapeutic agent. However, further research, particularly involving human clinical trials, is necessary to validate these findings for clinical use.
Collapse
Affiliation(s)
- Zhile Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Han Gan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Siyuan Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuchen Xue
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kai Luo
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kai Huang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yunqian Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
35
|
Wang X, Song X, Fang K, Chang X. CD38 modulates cytokine secretion by NK cells through the Sirt1/NF-κB pathway, suppressing immune surveillance in colorectal cancer. Sci Rep 2024; 14:28702. [PMID: 39562615 PMCID: PMC11577103 DOI: 10.1038/s41598-024-79008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
Tregs and M2-type macrophages are essential for immune surveillance. CD38 + NK cells are involved in immunoregulation by modulating cytokine secretion. This study investigated how CD38 + NKs affect Tregs and macrophages in colorectal cancer (CRC). Higher proportions of CD38 + NKs and Tregs were detected in bloods and tumor tissues of CRC patients than that in the samples from healthy controls (HCs). Compared with CD38 + NKs from HCs, the NK cells from CRC promoted the differentiation of Tregs from CD4 + T cells, and secreted increased levels of IL-10, TGF-β and TNF-α and decreased levels of IFN-γ. CD38 + NKs from CRC expressed higher levels of CD38, NF-κB and acetyl-NF-κB and lower levels of Sirt1. When CRC CD38 + NK cells were treated with anti-CD38 monoclonal antibody, the above trends were reversed. CRC CD38 + NKs with treatment of NF-κB inhibitor also showed opposite effects on cytokine secretion and CD4 + T-cell differentiation. After treatment with a Sirt1 activator, NF-κB signaling was inhibited in these CD38 + NKs, whereas treatment with a Sirt1 inhibitor activated NF-κB signaling. The supernatants of CRC CD38 + NK culture promoted M0 macrophage polarization to M2-type. We suggest that CD38 modulates cytokine secretion by NK cells through Sirt1/NF-κB signaling pathway, thereby suppressing immune surveillance in tumorigenesis.
Collapse
Affiliation(s)
- Xueling Wang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, People's Republic of China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Jiaozhou road 1, Qingdao, 266000, People's Republic of China
| | - Xianqin Song
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, People's Republic of China
| | - Kehua Fang
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, Shandong, People's Republic of China.
| | - Xiaotian Chang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, People's Republic of China.
| |
Collapse
|
36
|
Eggers B, Seher L, Marciniak J, Pauck T, Deschner J, Eick S, Stope MB, Kramer FJ, Küchler EC, Kirschneck C, Nokhbehsaim M, Beisel-Memmert S. Beneficial effects of non-invasive physical plasma on human periodontal ligament cells in vitro. Front Med (Lausanne) 2024; 11:1443368. [PMID: 39629237 PMCID: PMC11611554 DOI: 10.3389/fmed.2024.1443368] [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: 06/03/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Periodontitis is a chronic inflammatory disease of the periodontium that can lead to the loss of affected teeth if left untreated. It is induced by a multifactorial process centered on microbial pathogens such as Fusobacterium nucleatum (F.n.). Non-invasive physical plasma (NIPP), a highly reactive gas, has become a focus of research, not only for its hemostatic, proliferation-enhancing and apoptotic properties, but also for its antimicrobial potential. The objective of this study was to examine the impact of NIPP on human periodontal ligament (PDL) cells that had been induced into a state of periodontal infection in vitro. Methods Initially, the solitary effect of NIPP was evaluated by measuring temperature and pH and analyzing reactive oxygen species (ROS). Additionally, DAPI and phalloidin staining were employed to investigate possible cytotoxic effects. The cells were pre-incubated with F.n. and treated with NIPP after 24 hours. Interleukin (IL)-6 and IL-8 were analyzed at mRNA and protein levels, respectively, by real-time PCR and ELISA. Results NIPP alone had no significant effect on PDL cells. However, the F.n.-induced upregulation of IL-6 and IL-8 was counteracted by NIPP. Discussion Thus, the utilization of NIPP may be regarded as a promising therapeutic strategy for the treatment of periodontal diseases.
Collapse
Affiliation(s)
- Benedikt Eggers
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, Bonn, Germany
| | - Lennard Seher
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, Bonn, Germany
- Department of Orthodontics, University Hospital Bonn, Bonn, Germany
| | - Jana Marciniak
- Department of Orthodontics, University Hospital Bonn, Bonn, Germany
| | - Tristan Pauck
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sigrun Eick
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Matthias Bernhard Stope
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Bonn, Germany
| | - Franz-Josef Kramer
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, Bonn, Germany
| | | | | | - Marjan Nokhbehsaim
- Section of Experimental Dento-Maxillo-Facial Medicine, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
37
|
Tsuruno Y, Nagano A, Sugita K, Onishi S, Tabata Y, Kedoin C, Murakami M, Yano K, Kawano T, Hasuzawa N, Nomura M, Kaji T, Bitoh Y, Ieiri S. Favorable inhibitory effect of clodronate on hepatic steatosis in short bowel syndrome model rats. Pediatr Surg Int 2024; 40:307. [PMID: 39537943 PMCID: PMC11561034 DOI: 10.1007/s00383-024-05858-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE This study investigated the anti-inflammatory effect of clodronate, a vesicular nucleotide transporter (VNUT) inhibitor, on intestinal-failure-associated liver disease (IFALD) in a rat model of short bowel syndrome (SBS). METHODS The rats underwent jugular vein catheterization for continuous total parenteral nutrition (TPN) and 90% small bowel resection. The animals were divided into the following groups: TPN/SBS (Control group), TPN/SBS/intravenous administration of low-dose clodronate (20 mg/kg twice per week; Low group), or TPN/SBS/intravenous administration of high-dose clodronate (60 mg/kg twice per week; High group). On day 7, the rats were euthanized. Hepatic steatosis and hepatocellular injury were also assessed. RESULTS Hepatic steatosis and lobular inflammation in the liver were observed in all groups. The High group showed histologically reduced hepatic steatosis compared with the Control group. IL-6 and Nlrp3 expression in the High group was significantly suppressed compared to that in the Control group. The expression of other inflammatory cytokines tended to be lower in the High dose group than in the control group. The lipid metabolism gene expression in the liver specimens showed no significant differences among the groups. CONCLUSION The high-dose administration of clodronate may, therefore, inhibit hepatic steatosis and inflammation associated with IFALD in patients with SBS.
Collapse
Affiliation(s)
- Yudai Tsuruno
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
- Division of Pediatric Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ayaka Nagano
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Koshiro Sugita
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan.
| | - Shun Onishi
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Yumiko Tabata
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Chihiro Kedoin
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Masakazu Murakami
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Keisuke Yano
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Takafumi Kawano
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | - Nao Hasuzawa
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Masatoshi Nomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuru Kaji
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Yuko Bitoh
- Division of Pediatric Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Ieiri
- Department of Pediatric Surgery, Medical and Dental Area, Research and Education Assembly, Research Field in Medical and Health Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| |
Collapse
|
38
|
Zhou Y, Stevis PE, Cao J, Ehrlich G, Jones J, Rafique A, Sleeman MW, Olson WC, Franklin MC. Structures of complete extracellular assemblies of type I and type II Oncostatin M receptor complexes. Nat Commun 2024; 15:9776. [PMID: 39532904 PMCID: PMC11557873 DOI: 10.1038/s41467-024-54124-1] [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: 12/06/2023] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Oncostatin M (OSM) is a unique Interleukin 6 (IL-6) family cytokine that plays pivotal roles in numerous biological events by signaling via two types of receptor complexes. While type I OSM receptor complex is formed by glycoprotein 130 (gp130) heterodimerization with Leukemia Inhibitory Factor receptor (LIFR), type II OSM receptor complex is composed of gp130 and OSM receptor (OSMR). OSM is an important contributor to multiple inflammatory diseases and cancers while OSM inhibition has been shown to be effective at reducing symptoms, making OSM an attractive therapeutic target. Using cryogenic electron microscopy (cryo-EM), we characterize full extracellular assemblies of human type I OSM receptor complex and mouse type II OSM receptor complex. The juxtamembrane domains of both complexes are situated in close proximity due to acute bends of the receptors. The rigid N-terminal extension of OSM contributes to gp130 binding and OSM signaling. Neither glycosylation nor pro-domain cleavage of OSM affects its activity. Mutagenesis identifies multiple OSM and OSMR residues crucial for complex formation and signaling. Our data reveal the structural basis for the assemblies of both type I and type II OSM receptor complexes and provide insights for modulation of OSM signaling in therapeutics.
Collapse
Affiliation(s)
- Yi Zhou
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA.
| | | | - Jing Cao
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - George Ehrlich
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Jennifer Jones
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | - Mark W Sleeman
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | | | | |
Collapse
|
39
|
Lu L, Huang C, Zhou Y, Jiang H, Chen C, Du J, Zhou T, Wen F, Pei J, Wu Q. Tinosporae Radix attenuates acute pharyngitis by regulating glycerophospholipid metabolism and inflammatory responses through PI3K-Akt signaling pathway. Front Pharmacol 2024; 15:1491321. [PMID: 39568590 PMCID: PMC11576305 DOI: 10.3389/fphar.2024.1491321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction With the onset of the COVID-19 pandemic, the incidence and prevalence of acute pharyngitis (AP) have increased significantly. Tinosporae Radix (TR) is a vital medication utilized in the treatment of pharyngeal and laryngeal ailments, especially AP. The study endeavors to explore unclear molecular mechanisms of TR in addressing AP. Methods Network pharmacology and metabolomics analyses of effect of TR on AP were conducted, and apossible pathway was validated both in vivo using the acute pharyngitis rat model and in vitro using the LPS-induced RAW264.7 cells model, through techniques such as histopathological examinations, immunohistochemical technology, ELISA, RT-qPCR, and Western blotting to systematically explore the possible mechanisms underlying the inhibition of AP by TR. Results and discussion Network pharmacology analysis identified several key targets, including PIK3CA, IL6, AKT1, TNF, and PTGS2, alongside pivotal signaling pathways such as IL-17, TNF, Hepatitis B, nuclear factor kappa B (NF-κB), Influenza A, and the PI3K-Akt pathway. Most of them are closely associated with inflammation. Then, wide-target metabolomics analysis showed that TR downregulated substances within the glycerophospholipid metabolic pathway, and modulated the PI3K-Akt pathway. The integrated findings from network pharmacology and metabolomics underscored the pivotal role of the PI3K-Akt signaling pathway and the attenuation of inflammatory responses. Finally, in vitro and in vivo experiments have shown that TR can inhibit inflammatory factors such as IL-6, TNF - α, and COX-2, downregulate targets such as PI3K and AKT on the PI3K-Akt signaling pathway, and thereby alleviate the inflammatory response of AP. Our study demonstrated that TR exerts an anti-AP effect through suppression of release of inflammatory factors and modulation of glycerophospholipid metabolism via suppressing the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Lijie Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chengfeng Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongfeng Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Huajuan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cuiping Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinyu Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feiyan Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin Pei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinghua Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
40
|
Hösel K, Chasan B, Tode J, Rose-John S, Roider JB, Ehlken C. Effect of intravitreal injection of anti-interleukin (IL)-6 antibody in experimental autoimmune uveitis in mice. J Ophthalmic Inflamm Infect 2024; 14:57. [PMID: 39497001 PMCID: PMC11535092 DOI: 10.1186/s12348-024-00441-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: 03/22/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
PURPOSE The aim of this study was to assess the functional and clinical impact of intravitreal administration of a neutralizing anti-IL-6 antibody in the treatment of experimental autoimmune uveitis (EAU) in mice. METHODS EAU was induced in 17 female B10.RIII mice by administering Inter-Photoreceptor-Binding-Protein (IRBP) in complete Freund's adjuvant, followed by a boost with Pertussis toxin. Intravitreal injections of anti-Interleukin (IL)-6 antibody were administered on days 10, 13, and 16 after EAU induction (day 0) into the randomized treatment eye, with an isotype antibody similarly injected into the fellow control eye. Visual acuity was assessed using the optomotor reflex via OptoDrum, and clinical scoring was performed via fundus imaging (utilizing 6 EAU grades) in a single-blinded manner on days 0, 10, 13, 16, and 18. RESULTS Uveitis developed in all 17 mice. Significantly higher visual acuity was observed in treated eyes compared to control eyes on days 13, 16, and 18. The most pronounced effect was noted on days 16 and 18 (p < 0.001). On days 13, 16, and 18 the number of eyes with lower EAU-score was significantly higher in the treatment group, with the most notable effect observed on day 18 (p < 0.003). CONCLUSION Intravitreal administration of anti-IL-6 treatment notably mitigates experimental autoimmune uveitis in mice, both functionally and clinically. Further investigations are warranted to assess the potential of intravitreal anti-IL-6 therapy as a treatment option for non-infectious uveitis in humans.
Collapse
Affiliation(s)
- Kristin Hösel
- Department of Ophthalmology, UKSH Kiel, Kiel, Germany.
| | - Büsra Chasan
- Department of Ophthalmology, UKSH Kiel, Kiel, Germany
| | - Jan Tode
- Department of Ophthalmology, MHH Hannover, Hannover, Germany
| | - Stefan Rose-John
- Department of Biochemistry, Klinik für Augenheilkunde, CAU Kiel, Haus B2, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | | | | |
Collapse
|
41
|
Zhu X, Wang X, Wang J, Du L, Zhang Z, Zhou D, Han J, Luan B. Intermittent Fasting-Induced Orm2 Promotes Adipose Browning via the GP130/IL23R-p38 Cascade. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407789. [PMID: 39248328 PMCID: PMC11558143 DOI: 10.1002/advs.202407789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/14/2024] [Indexed: 09/10/2024]
Abstract
Intermittent fasting (IF) plays a critical role in mitigating obesity, yet the precise biological mechanisms require further elucidation. Here Orosomucoid 2 (Orm2) is identified as an IF-induced hepatokine that stimulates adipose browning. IF induced Orm2 expression and secretion from the liver through peroxisome proliferator-activated receptor alpha (PPARα). In adipose tissue, Orm2 bound to glycoprotein 130/interleukin 23 receptor (GP130/IL23R) and promoted adipose browning through the activation of p38 mitogen-activated protein kinases (p38-MAPK). In obese mice, Orm2 led to a significant induction of adipose tissue browning and subsequent weight loss, an effect that is not replicated by a mutant variant of Orm2 deficient in GP130/IL23R binding capability. Crucially, genetic association studies in humans identified an obesity-associated Orm2 variant (D178E), which shows decreased GP130/IL23R binding and impaired browning capacity in mice. Overall, the research identifies Orm2 as a promising therapeutic target for obesity, mediating adipose browning through the GP130/IL23R-p38 signalling pathway.
Collapse
Affiliation(s)
- Xuejuan Zhu
- Department of EndocrinologyTongji Hospital Affiliated to Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Xinran Wang
- Department of EndocrinologyTongji Hospital Affiliated to Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
- Department of Breast and Thyroid SurgeryShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Jingang Wang
- Department of EndocrinologyTongji Hospital Affiliated to Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Lei Du
- Department of Breast and Thyroid SurgeryShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Zhen‐Ning Zhang
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative MedicineShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Donglei Zhou
- Department of Gastric SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Junfeng Han
- Department of EndocrinologyTongji Hospital Affiliated to Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Bing Luan
- Department of EndocrinologyTongji Hospital Affiliated to Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| |
Collapse
|
42
|
Zhou K, Lu J. Progress in cytokine research for ARDS: A comprehensive review. Open Med (Wars) 2024; 19:20241076. [PMID: 39479463 PMCID: PMC11524396 DOI: 10.1515/med-2024-1076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/16/2024] [Accepted: 10/06/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction Acute respiratory distress syndrome (ARDS) is a critical form of acute respiratory failure characterized by diffuse alveolar damage, refractory hypoxemia, and non-cardiogenic pulmonary edema, resulting in high mortality. Dysregulated inflammation, driven by cytokines, is central to ARDS pathogenesis, progression, and prognosis. Objective This review synthesizes current knowledge on the role of cytokines in ARDS and evaluates their potential as therapeutic targets, offering new insights for clinical management. Methods A comprehensive analysis of recent studies was conducted to explore the roles of pro-inflammatory cytokines (e.g., IL-1β, IL-6, IL-8) and anti-inflammatory cytokines (e.g., IL-10, IL-22) in ARDS pathogenesis and to assess current and emerging therapies targeting these cytokines. Results Pro-inflammatory cytokines are crucial in initiating inflammatory responses and lung injury in early ARDS, while anti-inflammatory cytokines help regulate and resolve inflammation. Targeted therapies, such as IL-1 and IL-6 inhibitors, show potential in managing ARDS, particularly in COVID-19, but their clinical efficacy is still debated. Combination therapy strategies may enhance outcomes, but further large-scale, multicenter randomized controlled trials are required to establish their safety and efficacy. Conclusion Understanding cytokine regulation in ARDS could lead to innovative therapeutic approaches. Future research should focus on cytokine roles across ARDS subtypes and stages and develop biomarker-driven, individualized treatments.
Collapse
Affiliation(s)
- Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxuedong Road, Nanning, Guangxi 530007, China
| |
Collapse
|
43
|
Zhang Y, Guo N, Zhu H, Liu M, Hao J, Wang S, Guo T, Mamun MAA, Pang J, Liu Q, Zheng Y, Liu H, Si P, Zhao L. Unlocking the dual role of LSD1 in tumor immunity: innate and adaptive pathways. Theranostics 2024; 14:7054-7071. [PMID: 39629133 PMCID: PMC11610140 DOI: 10.7150/thno.102037] [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: 08/07/2024] [Accepted: 10/10/2024] [Indexed: 12/07/2024] Open
Abstract
The roles of innate and adaptive immunity are crucial in both the development of cancer and its response to treatment. Numerous studies have demonstrated that histone lysine-specific demethylase 1 (LSD1) is overexpressed in various cancers. Elevated levels of LSD1 intricately modulate immune checkpoints, the function of immune cells, and the expression of immunomodulators, impacting both innate and adaptive immunity. Moreover, compelling evidence suggests that inhibiting LSD1 enhances tumor immunity, suppresses tumor growth, and improves the effectiveness of immunotherapy. However, a comprehensive classification of LSD1's role in both innate and adaptive immunity is lacking. In this review, we outline the role of LSD1 in tumor immunity in terms of both innate and adaptive immunity, summarizing the mechanisms associated with LSD1-mediated tumor immunity and its potential regulatory capacity in tumor immune escape. Finally, we summarize the research status of LSD1 inhibitors in tumor immunotherapy, which be valuable for promoting the development of effective LSD1-targeted agents used as combination immunotherapy drugs.
Collapse
Affiliation(s)
- Yu Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
- XNA platform, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ningjie Guo
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Haoyi Zhu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mengyang Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jiahui Hao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Shoukai Wang
- Department of Breast Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University; Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan 450003, China
| | - Ting Guo
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| | - MAA Mamun
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Jingru Pang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qi Liu
- Department of Breast Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University; Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan 450003, China
| | - Yichao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
- XNA platform, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Pilei Si
- Department of Breast Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University; Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan 450003, China
| | - Lijuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; School of Pharmaceutical Sciences; Academy of Medical Sciences; Tianjian Laboratory of Advanced Biomedical Sciences; Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
44
|
Covino DA, Farina I, Catapano L, Sozzi S, Spadaro F, Cecchetti S, Purificato C, Gauzzi MC, Fantuzzi L. Induction of the antiviral factors APOBEC3A and RSAD2 upon CCL2 neutralization in primary human macrophages involves NF-κB, JAK/STAT, and gp130 signaling. J Leukoc Biol 2024; 116:876-889. [PMID: 38798090 DOI: 10.1093/jleuko/qiae123] [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: 10/20/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
The CCL2/CC chemokine receptor 2 axis plays key roles in the pathogenesis of HIV-1 infection. We previously reported that exposure of monocyte-derived macrophages to CCL2 neutralizing antibody (αCCL2 Ab) restricted HIV-1 replication at postentry steps of the viral life cycle. This effect was associated with induction of transcripts coding for innate antiviral proteins, including APOBEC3A and RSAD2. This study aimed at identifying the signaling pathways involved in induction of these factors by CCL2 blocking in monocyte-derived macrophages. Through a combination of pharmacologic inhibition, quantitative reverse transcription polymerase chain reaction, Western blotting, and confocal laser-scanning microscopy, we demonstrated that CCL2 neutralization activates the canonical NF-κB and JAK/STAT pathways, as assessed by time-dependent phosphorylation of IκB, STAT1, and STAT3 and p65 nuclear translocation. Furthermore, pharmacologic inhibition of IκB kinase and JAKs strongly reduced APOBEC3A and RSAD2 transcript accumulation elicited by αCCL2 Ab treatment. Interestingly, exposure of monocyte-derived macrophages to αCCL2 Ab resulted in induction of IL-6 family cytokines, and interference with glycoprotein 130, the common signal-transducing receptor subunit shared by these cytokines, inhibited APOBEC3A and RSAD2 upregulation triggered by CCL2 neutralization. These results provide novel insights into the signal transduction pathways underlying the activation of innate responses triggered by CCL2 neutralization in macrophages. Since this response was found to be associated with protective antiviral effects, the new findings may help design innovative therapeutic approaches targeting CCL2 to strengthen host innate immunity.
Collapse
Affiliation(s)
- Daniela Angela Covino
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Iole Farina
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Laura Catapano
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Silvia Sozzi
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Francesca Spadaro
- Core Facilities, Microscopy Unit, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Serena Cecchetti
- Core Facilities, Microscopy Unit, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Cristina Purificato
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Cristina Gauzzi
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Laura Fantuzzi
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
45
|
Mazurek-Mochol M, Bonsmann T, Malinowski D, Serwin K, Czerewaty M, Safranow K, Pawlik A. Interleukin-6 Receptor Gene rs1800795 Polymorphism and Expression of Interleukin-6 in Gingival Tissue in Patients with Periodontitis. Microorganisms 2024; 12:1954. [PMID: 39458264 PMCID: PMC11509941 DOI: 10.3390/microorganisms12101954] [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: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Periodontitis is a multifactorial inflammatory disease. This chronic periodontal disease is caused by a bacterial infection in the gums, which triggers a host inflammatory response. To eliminate the bacterial infection, immune response mechanisms are activated, leading to inflammation and damage to the periodontal tissues. This process involves many cytokines, including IL-6, a cytokine with antibacterial properties. An ongoing bacterial infection in the periodontal tissues leads to its excessive production, which increases inflammation. In this study, we examined IL-6 receptor gene rs1800795 polymorphism in patients with periodontitis in comparison with healthy subjects, as well as the correlation between rs1800795 genotypes and clinical parameters. Additionally we examined the expression of IL-6 in gingival tissue in patients with periodontitis and control subjects, as well as the correlation between gingival expression of IL-6 and clinical parameters. This study included 200 patients with periodontitis and 158 healthy subjects as the control group. Biopsy specimens of gingival tissue in which IL-6 expression was detected were taken from 14 patients with periodontitis and 8 controls who had undergone minor surgery. There were no statistically significant differences in the distribution of IL-6 rs1800795 genotypes and alleles between patients with periodontitis and control subjects. There were also no statistically significant correlations between IL-6 rs1800795 genotypes and clinical parameters in patients with periodontitis. There were no differences in IL-6 expression in the gingival tissue between patients with periodontitis and controls. There was also no correlation between IL-6 expression in the gingival tissue of patients with periodontitis and clinical parameters. In the control group, IL-6 expression in gingival tissue correlated negatively with the approximal plaque index, which reflects the size of bacterial plaques. The results of our study suggest a protective role for IL-6 against bacterial growth in the periodontal tissue. However, it should be noted that several parameters directly or indirectly affect the accumulation of bacterial plaque.
Collapse
Affiliation(s)
- Małgorzata Mazurek-Mochol
- Department of Periodontology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland; (M.M.-M.); (T.B.)
| | - Tobias Bonsmann
- Department of Periodontology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland; (M.M.-M.); (T.B.)
| | - Damian Malinowski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Karol Serwin
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Michał Czerewaty
- Department of Physiology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| |
Collapse
|
46
|
Calcaterra V, Magenes VC, Bianchi A, Rossi V, Gatti A, Marin L, Vandoni M, Zuccotti G. How Can Promoting Skeletal Muscle Health and Exercise in Children and Adolescents Prevent Insulin Resistance and Type 2 Diabetes? Life (Basel) 2024; 14:1198. [PMID: 39337980 PMCID: PMC11433096 DOI: 10.3390/life14091198] [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: 08/31/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Skeletal muscle secretome, through its paracrine and endocrine functions, contributes to the maintenance and regulation of overall physiological health. We conducted a narrative review on the role of skeletal muscle and exercise in maintaining glucose homeostasis, driving insulin resistance (IR), and preventing type 2 diabetes in pediatric populations, especially in the context of overweight and obesity. Myokines such as interleukin (IL)-6, IL-8, and IL-15, as well as irisin, myonectin, and myostatin, appear to play a crucial role in IR. Skeletal muscle can also become a target of obesity-induced and IR-induced inflammation. In the correlation between muscle, IR, and inflammation, the role of infiltration of the immune cells and the microvasculature may also be considered. It remains unclear which exercise approach is the best; however, combining aerobic exercise with resistance training seems to be the most effective strategy for managing IR, with high-intensity activities offering superior metabolic benefits and long-term adherence. Encouraging daily participation in enjoyable and engaging exercise is key for long-term commitment and effective glucose metabolism management. Promoting physical activity in children and adolescents must be a top priority for public health, not only in terms of individual quality of life and well-being but also for community health.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Vittoria Carlotta Magenes
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Alice Bianchi
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Virginia Rossi
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Alessandro Gatti
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy; (A.G.); (L.M.); (M.V.)
| | - Luca Marin
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy; (A.G.); (L.M.); (M.V.)
| | - Matteo Vandoni
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy; (A.G.); (L.M.); (M.V.)
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milano, Italy
| |
Collapse
|
47
|
Pang S, Li X. Early use of SGLT2 inhibitors reduces the progression of diabetic kidney disease: a retrospective cohort study. Am J Transl Res 2024; 16:4967-4978. [PMID: 39398587 PMCID: PMC11470299 DOI: 10.62347/arya8831] [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: 05/29/2024] [Accepted: 07/29/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To evaluate the potential of sodium-glucose cotransporter 2 (SGLT2) inhibitors in preventing the progression of diabetic kidney disease and to provide guidance for clinical practice to improve renal health management strategies for diabetic patients. METHODS A retrospective analysis was conducted on 178 patients with diabetic kidney disease admitted to Baoji High Tech Hospital from March 2023 to March 2024. Of these, 88 patients who received early treatment with the SGLT2 inhibitor dapagliflozin were included in the early SGLT2-i group, while 90 patients receiving later treatment with SGLT2 inhibitor dapagliflozin were included in the late SGLT2-i group. Clinical data, overall effectiveness, adverse reactions, blood glucose, renal function, lipid levels, and inflammatory markers were compared between the two groups. RESULTS Prior to treatment, there were no differences in blood glucose indicators between the two groups (all P > 0.05). Following treatment, both groups showed reductions in 2-hour postprandial blood glucose (2hPG), fasting plasma glucose (FPG), and glycosylated hemoglobin (HbA1c), with the early SGLT2-i group demonstrating significantly lower values compared to the late SGLT2-i group (all P < 0.05). Similarly, there were no differences in renal function indicators between the two groups before treatment (all P > 0.05). However, following treatment, the early SGLT2-i group showed more noticeable improvements compared to the late SGLT2-i group (P < 0.05). Inflammatory markers and lipid levels followed similar patterns. The overall effectiveness of the early SGLT2-i group was higher than that of the late SGLT2-i group (92.05% vs. 78.89%, P < 0.05), while the incidence of adverse reactions did not differ statistically between the two groups (6.82% vs. 10.00%, P > 0.05). CONCLUSION Early use of SGLT2 inhibitors in diabetic kidney disease patients effectively controls blood glucose and lipid levels, improves renal function, reduces inflammatory responses, and exhibits a low incidence of adverse reactions. This demonstrates high safety and an important role in delaying disease progression. Therefore, it is worth considering clinical promotion and use for this patient population.
Collapse
Affiliation(s)
- Shaowei Pang
- Nephrology Department, Baoji High Tech Hospital Baoji 721000, Shaanxi, China
| | - Xiaoli Li
- Nephrology Department, Baoji High Tech Hospital Baoji 721000, Shaanxi, China
| |
Collapse
|
48
|
Pang L, Zhao Y, Xu Y, Gao C, Wang C, Yu X, Wang F, He K. Mechanisms Underlying the Therapeutic Effects of JianPiYiFei II Granules in Treating COPD Based on GEO Datasets, Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulations. BIOLOGY 2024; 13:711. [PMID: 39336138 PMCID: PMC11428342 DOI: 10.3390/biology13090711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND JianPiYiFei (JPYF) II granules are a Chinese medicine for the treatment of chronic obstructive pulmonary disease (COPD). However, the main components and underlying mechanisms of JPYF II granules are not well understood. This study aimed to elucidate the potential mechanism of JPYF II granules in the treatment of COPD using network pharmacology, molecular docking, and molecular dynamics simulation techniques. METHODS The active compounds and corresponding protein targets of the JPYF II granules were found using the TCMSP, ETCM, and Uniport databases, and a compound-target network was constructed using Cytoscape3.9.1. The COPD targets were searched for in GEO datasets and the OMIM and GeneCards databases. The intersection between the effective compound-related targets and disease-related targets was obtained, PPI networks were constructed, and GO and KEGG enrichment analyses were performed. Then, molecular docking analysis verified the results obtained using network pharmacology. Finally, the protein-compound complexes obtained from the molecular docking analysis were simulated using molecular dynamics (MD) simulations. RESULTS The network pharmacological results showed that quercetin, kaempferol, and stigmasterol are the main active compounds in JPYF II granules, and AKT1, IL-6, and TNF are key target proteins. The PI3K/AKT signaling pathway is a potential pathway through which the JPYF II granules affect COPD. The results of the molecular docking analysis suggested that quercetin, kaempferol, and stigmasterol have a good binding affinity with AKT1, IL-6, and TNF. The MD simulation results showed that TNF has a good binding affinity with the compounds. CONCLUSIONS This study identified the effective compounds, targets, and related underlying molecular mechanisms of JPYF II granules in the treatment of COPD through network pharmacology, molecular docking, and MD simulation techniques, which provides a reference for subsequent research on the treatment of COPD.
Collapse
Affiliation(s)
- Liyuan Pang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yongjuan Zhao
- Department of Pulmonary and Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun 130021, China
| | - Yang Xu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Chencheng Gao
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Chao Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiao Yu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
49
|
Giacco A, Petito G, Silvestri E, Scopigno N, Vigliotti M, Mercurio G, de Lange P, Lombardi A, Moreno M, Goglia F, Lanni A, Senese R, Cioffi F. Comparative effects of 3,5-diiodo-L-thyronine and 3,5,3'-triiodo-L-thyronine on mitochondrial damage and cGAS/STING-driven inflammation in liver of hypothyroid rats. Front Endocrinol (Lausanne) 2024; 15:1432819. [PMID: 39301315 PMCID: PMC11410700 DOI: 10.3389/fendo.2024.1432819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024] Open
Abstract
Maintaining a well-functioning mitochondrial network through the mitochondria quality control (MQC) mechanisms, including biogenesis, dynamics and mitophagy, is crucial for overall health. Mitochondrial dysfunction caused by oxidative stress and further exacerbated by impaired quality control can trigger inflammation through the release of the damage-associated molecular patterns (mtDAMPs). mtDAMPs act by stimulating the cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) pathway. Recently, aberrant signalling of the cGAS-STING axis has been recognised to be closely associated with several sterile inflammatory diseases (e.g. non-alcoholic fatty liver disease, obesity). This may fit the pathophysiology of hypothyroidism, an endocrine disorder characterised by the reduction of thyroid hormone production associated with impaired metabolic fluxes, oxidative balance and inflammatory status. Both 3,5,3'-triiodo-L-tyronine (T3) and its derivative 3,5-diiodo-L-thyronine (3,5-T2), are known to mitigate processes targeting mitochondria, albeit the underlying mechanisms are not yet fully understood. Therefore, we used a chemically induced hypothyroidism rat model to investigate the effect of 3,5-T2 or T3 administration on inflammation-related factors (inflammatory cytokines, hepatic cGAS-STING pathway), oxidative stress, antioxidant defence enzymes, mitochondrial DNA (mtDNA) damage, release and repair, and the MQC system in the liver. Hypothyroid rats showed: i) increased oxidative stress, ii) accumulation of mtDNA damage, iii) high levels of circulating cytokines, iv) hepatic activation of cGAS-STING pathways and v) impairment of MQC mechanisms and autophagy. Both iodothyronines restored oxidative balance by enhancing antioxidant defence, preventing mtDNA damage through the activation of mtDNA repair mechanisms (OGG1, APE1, and POLγ) and promoting autophagy progression. Concerning MQC, both iodothyronines stimulated mitophagy and dynamics, with 3,5-T2 activating fusion and T3 modulating both fusion and fission processes. Moreover, only T3 enhanced mitochondrial biogenesis. Notably, 3,5-T2, but not T3, reversed the hypothyroidism-induced activation of the cGAS-STING inflammatory cascade. In addition, it is noteworthy that 3,5-T2 seems more effective than T3 in reducing circulating pro-inflammatory cytokines IL-6 and IL-1B and in stimulating the release of IL-10, a known anti-inflammatory cytokine. These findings reveal novel molecular mechanisms of hepatic signalling pathways involved in hypothyroidism, which could be targeted by natural iodothyronines, particularly 3,5-T2, paving the way for the development of new treatment strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Antonia Giacco
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Giuseppe Petito
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Elena Silvestri
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Nicla Scopigno
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Michela Vigliotti
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Giovanna Mercurio
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Pieter de Lange
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Assunta Lombardi
- Department of Biology, University of Naples Federico II, Napoli, Italy
| | - Maria Moreno
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Fernando Goglia
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Antonia Lanni
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Rosalba Senese
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Federica Cioffi
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| |
Collapse
|
50
|
Gomes BS, Gonçalves AB, Lanza SZ, Esquisatto MAM, Bomfim FRCD, Lopes Filho GDJ. Effects of Photobiomodulation Associated with Platelet-Rich Plasma in Acute Rheumatoid Arthritis Induced in Female Wistar Rats' Knee. Photobiomodul Photomed Laser Surg 2024; 42:585-592. [PMID: 39069887 DOI: 10.1089/pho.2024.0060] [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: 07/30/2024] Open
Abstract
Objective: Rheumatoid arthritis causes inflammation, pain, and joint degradation, necessitating treatment with anti-inflammatory drugs and corticosteroids, posing various challenges. We aimed to evaluate the effects of photobiomodulation (PBM) at two different doses associated to platelet-rich plasma (PRP) in an in vivo model of induced acute arthritis in Wistar rats' knee. Methods: Eighty-four Wistar rats were assigned into seven groups, including animals treated with PBM and/or PRP. On day 0, arthritis was induced in sham and treated groups through the intra-articular injection of zymosan (200 μg). Twenty-four hours after induction, the PBM groups were treated with an AsGaAl laser, whereas the PRP-treated groups received intra-articular injections with a concentration of 8 × 105 platelets obtained from another four animals. After 3 days, the animals were euthanized, and the interleukin (IL)-6 and complement C3 gene and protein expression levels were analyzed. Statistical analysis was performed using the mean ± SD with analysis of variance and Tukey's posttest, with a significance level set at 5% (p < 0.05). Results: Synovial inflammation decreased in PBM-treated groups; however, PRP alone showed no significant difference. Gene expression analysis revealed a significant difference in IL-6 and C3 levels in the PBM and PBM+PRP-treated groups. Meanwhile, the PRP alone group exhibited significance for IL-6. Moreover, the PBM and PBM+PRP-treated groups showed a significant difference in C3 protein expression levels, whereas the PRP alone group showed no difference. Conclusion: The increase in cellular activity in the synovial membrane and the decrease protein expression levels are owing to the reduction in proinflammatory mediators following PBM therapy.
Collapse
Affiliation(s)
- Bruna Silva Gomes
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | - Fernando Russo Costa do Bomfim
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, São Paulo, Brazil
- Centro Universitário da Fundação Hermínio Ometto, Araras, Brazil
| | - Gaspar de Jesus Lopes Filho
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|