1
|
He W, Yang H, Yang X, Huang J, Wu Z. Global research trends in biological therapy for ankylosing spondylitis: A comprehensive visualization and bibliometric study (2004-2023). Hum Vaccin Immunother 2025; 21:2445900. [PMID: 39813123 PMCID: PMC11740677 DOI: 10.1080/21645515.2024.2445900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025] Open
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disease primarily affecting the spine and pelvic bones. Recently, many researchers have confirmed that biological therapy is effective for AS patients, which provides a new perspective for the treatment of AS. This study aimed to evaluate the characteristics of scientific research on AS and biological therapy worldwide and investigate research hotspots and the direction of future trends. Global literature on AS and biological therapy published from 2004 to 2023 was searched in the Web of Science, Scopus, and PubMed databases. Visualization and bibliometric analysis were carried out using the VOSviewer and CiteSpace software with the retrieved data regarding countries, institutions, journals, authors, and keywords. A total of 2,243 related articles were included, showing that the number of articles in this field has increased annually. The highest number of articles were from the USA (24.39%), followed by Italy (14.36%), England (12.19%), Germany (10.66%), and Spain (7.86%). Braun J was the most prolific author, with a h-index of 16. The institution with the most articles was Charite Universitatsmedizin Berlin, and the Rheumatology journal had the highest number of publications. "janus kinase inhibitor" and "secukinumab" displayed a notable citation burst in recent years, indicating IL-17i and JAKi are research hotspots. More and more attention has been paid to the association between AS and biological therapy in the past two decades. The USA plays a leading role, and China has made remarkable progress. This study has provided a valuable reference for future research in this field.
Collapse
Affiliation(s)
- Weiliang He
- Institute of Orthopaedics, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Haicheng Yang
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Xuanzhe Yang
- Institute of Orthopaedics, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - JinFeng Huang
- Institute of Orthopaedics, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Zixiang Wu
- Institute of Orthopaedics, Xijing Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
2
|
Zoheir KMA, Ali NI, Ashour AE, Kishta MS, Othman SI, Rudayni HA, Rashad AA, Allam AA. Lipoic acid improves wound healing through its immunomodulatory and anti-inflammatory effects in a diabetic mouse model. J Diabetes Metab Disord 2025; 24:56. [PMID: 39868353 PMCID: PMC11759746 DOI: 10.1007/s40200-025-01559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/28/2024] [Indexed: 01/28/2025]
Abstract
Objectives Diabetes mellitus is a chronic disease that has become more prevalent worldwide because of lifestyle changes. It leads to serious complications, including increased atherosclerosis, protein glycosylation, endothelial dysfunction, and vascular denervation. These complications impair neovascularization and wound healing, resulting in delayed recovery from injuries and an elevated risk of infections. The present study aimed to investigate the effect of lipoic acid (LA) on the key mediators involved in the wound healing process, specifically CD4 + CD25 + T cell subsets, CD4 + CD25 + Foxp3 + regulatory T (Treg) cells, T-helper-17 (Th17) cells that generate IL-17 A, glucocorticoid-induced tumor necrosis factor receptor (GITR) expressing cells, as well as cytokines such as IL-2, IL-1β, IL-6, and TNF-α and IFN-γ. These mediators play crucial roles in epidermal and dermal proliferation, hypertrophy, and cell migration. Methods We divided mice into 5 groups: the non-diabetic (normal control; NC), wounded non-diabetic mice (N + W), wounded diabetic mice (D + W), wounded diabetic mice treated with 50 mg/kg lipoic acid (D + W + L50) for 14 days, and wounded diabetic mice treated with 100 mg/kg lipoic acid (D + W + L100) for 14 days. Results Flow cytometric analysis indicated that lipoic acid-treated mice exhibited a significant decrease in the frequency of intracellular cytokines (IL-17 A, TNF-α and IFN-γ) in CD4 + T cells, as well as a reduction in the number of GITR-expressing cells. Conversely, a significant upregulation in the number CD4+, CD25+, FOXp3 + and CD4 + CD25 + Foxp3 + regulatory T (Treg) cells was observed in this group compared to both the normal + wounded (N + W) and diabetic + wounded (D + W) groups. Additionally, the mRNA Levels of inflammatory mediators (IL-2, IL-1β, IL-6, and TNF-α) were downregulated in lipoic acid-treated mice compared to other groups. T thereby he histological findings of diabetic skin wounds treated with lipoic acid showed well-healed surgical wounds. Conclusions These findings support the beneficial role of lipoic acid in fine-tuning the balance between anti-inflammatory and pro-inflammatory cytokines, influencing both their release and gene expression.
Collapse
Affiliation(s)
- Khairy M. A. Zoheir
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza 12622 Egypt
| | - Neama I. Ali
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza 12622 Egypt
| | - Abdelkader E. Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Salman International University, Ras Sudr, South Sinai Egypt
| | - Mohamed S. Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, and Stem Cell Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622 Egypt
| | - Sarah I. Othman
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, 11671 Riyadh, Saudi Arabia
| | - Hassan A. Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623 Saudi Arabia
| | - Ahmed A. Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829 Egypt
| | - Ahmed A. Allam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623 Saudi Arabia
| |
Collapse
|
3
|
Zhou Y, Yu Z, Lu Y. To explore the influencing factors of clinical failure of anti-tumor necrosis factor-α (TNF-α) therapy in sepsis. Life Sci 2025; 369:123556. [PMID: 40068733 DOI: 10.1016/j.lfs.2025.123556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/30/2025]
Abstract
Sepsis, a condition of significant clinical concern, is characterized by life-threatening organ dysfunction that arises from an infection and is exacerbated by a dysregulated host response. Targeting immune modulation, particularly against tumor necrosis factor-alpha (TNF-α), has emerged as a promising anti-inflammatory therapeutic strategy. However, approaches such as blood purification to eliminate inflammatory mediators or the use of anti-TNF-α therapies have shown limited efficacy in clinical practice. This literature review aims to elucidate the pathogenesis of sepsis and dissect the factors contributing to unfavorable outcomes in TNF-α-targeted treatments. Our analysis highlights several potential reasons for therapeutic failure. Complete blockade of TNF-α may adversely affect both TNFR1 and TNFR2 signaling, thereby reducing the efficacy of TNF-α inhibitors. Additionally, the complex heterogeneity of sepsis, including the etiology of infection, patient-specific factors (e.g., immune responsiveness, body mass index, and obesity), the development of anti-drug antibodies, and treatment duration, significantly influences therapeutic outcomes. Based on these insights, we emphasize the need for precision medicine in sepsis management. This includes stratifying patients into subgroups, using TNFR2 agonists or TNFR1-specific antagonists, refining drug design, implementing multi-target combination therapies, and considering the patient's physiological state at the time of treatment. Collectively, these strategies could enhance the efficacy of sepsis management. This review underscores the multifaceted nature of sepsis treatment and highlights the imperative for personalized, multimodal therapeutic approaches to improve clinical outcomes.
Collapse
Affiliation(s)
- Yonghong Zhou
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China; Department of Pharmacy, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Zhaoran Yu
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yiming Lu
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China; Department of Pharmacy, School of Medicine, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
4
|
Raavi, Koehler AN, Vegas AJ. At The Interface: Small-Molecule Inhibitors of Soluble Cytokines. Chem Rev 2025; 125:4528-4568. [PMID: 40233276 DOI: 10.1021/acs.chemrev.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytokines are crucial regulators of the immune system that orchestrate interactions between cells and, when dysregulated, contribute to the progression of chronic inflammation, cancer, and autoimmunity. Numerous biologic-based clinical agents, mostly monoclonal antibodies, have validated cytokines as important clinical targets and are now part of the standard of care for a number of diseases. These agents, while impactful, still suffer from limitations including a lack of oral bioavailability, high cost of production, and immunogenicity. Small-molecule cytokine inhibitors are attractive alternatives that can address these limitations. Although targeting cytokine-cytokine receptor complexes with small molecules has been a challenging research endeavor, multiple small-molecule inhibitors have now been identified, with a number of them undergoing clinical evaluation. In this review, we highlight the recent advancements in the discovery and development of small-molecule inhibitors targeting soluble cytokines. The strategies for identifying these novel ligands as well as the structural and mechanistic insights into their activity represent important milestones in tackling these challenging and clinically important protein-protein interactions.
Collapse
Affiliation(s)
- Raavi
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Arturo J Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02115, United States
| |
Collapse
|
5
|
Zhao S, Liu H, Zhong H, Qin Y, Duan J, Liu A. Systemic Inflammation Mediates the Association Between Blood Trihalomethane Concentrations and Cardiovascular Disease in U.S. Individuals Over 45: Insights from NHANES 2005-2012. Cardiovasc Toxicol 2025:10.1007/s12012-025-10008-4. [PMID: 40366518 DOI: 10.1007/s12012-025-10008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
Trihalomethanes (THMs), the major byproducts of water chlorination which are associated with various adverse health outcomes. However, the relationship of THMs with cardiovascular disease (CVD) in aging populations remains underexplored. We analyzed data from 5,400 participants in the National Health and Nutrition Examination Survey (NHANES) 2005-2012. Associations between blood THM concentrations and CVD were evaluated using weighted multivariable logistic regression. Weighted quantile sum (WQS) regression was applied to identify the most relevant THM components. We also performed mediation analysis to evaluate the role of inflammatory markers, including neutrophil-to-lymphocyte ratio (NLR), white blood cell count (WBC), and systemic inflammation response index (SIRI). Network toxicology analysis was used to explore the biological pathways linking THM exposure, CVD, and aging. Elevated blood concentrations of THMs, particularly chloroform (TCM) and total THMs (TTHMs), were significantly associated with increased odds of CVD. Stratified analyses revealed stronger associations among older adults, males, individuals with higher BMI, and those with hypertension. WQS regression identified TCM as the predominant contributor to the THM-CVD association, accounting for 58.0% of the mixture's effect. Mediation analysis showed that NLR partially mediated the association between TTHMs and CVD, explaining 7.12% of the total effect. Network toxicology analysis highlighted inflammation-related pathways, including the IL-17 signaling pathway, as key mechanisms linking THM exposure, CVD, and aging. Our study revealed elevated blood TCM and TTHM concentrations are associated with increased prevalence of CVD among U.S. adults aged 45 years and older. Network toxicology and mediation analysis suggest that systemic inflammation may play a mediating role in this relationship.
Collapse
Affiliation(s)
- Songfeng Zhao
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Hunan, China
| | - Hongyi Liu
- Beijing Tiantan Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hailong Zhong
- Beijing Tiantan Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yongkai Qin
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Hunan, China
| | - Jiayue Duan
- Key Laboratory of Endocrinology, Department of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Aihua Liu
- Beijing Tiantan Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
6
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Study on the comorbid mechanisms of sarcopenia and late-life depression. Behav Brain Res 2025; 485:115538. [PMID: 40122287 DOI: 10.1016/j.bbr.2025.115538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The increasing global aging population has brought greater focus to age-related diseases, particularly muscle-brain comorbidities such as sarcopenia and late-life depression. Sarcopenia, defined by the gradual loss of muscle mass and function, is notably prevalent among older individuals, while late-life depression profoundly affects their mental health and overall well-being. Epidemiological evidence suggests a high co-occurrence of these two conditions, although the precise biological mechanisms linking them remain inadequately understood. This review synthesizes the existing body of literature on sarcopenia and late-life depression, examining their definitions, prevalence, clinical presentations, and available treatments. The goal is to clarify the potential connections between these comorbidities and offer a theoretical framework for the development of future preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Jiale Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Jun Tang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Di Huang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yu Wang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Enyuan Zhou
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
7
|
Chen S, Fu P, Rastegar-Kashkooli Y, Zhu L, Zong Y, Huang M, Gao C, Wang J, Zhang J, Wang J, Jiang C. AX-024 Inhibits Antigen-Specific T-Cell Response and Improves Intracerebral Hemorrhage Outcomes in Mice. Stroke 2025; 56:1253-1265. [PMID: 40143825 DOI: 10.1161/strokeaha.124.048507] [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: 07/11/2024] [Revised: 12/26/2024] [Accepted: 02/03/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Stroke-induced opposite T-cell responses in the peri-lesion area and periphery worsen stroke outcomes by aggravating brain injury or increasing infectious complications, respectively. Despite their well-known role in T lymphocyte activation, the impact of TCRs (T-cell receptors) on stroke remains poorly understood. In this study, we investigated the causal link between TCRs and the opposite T-cell responses observed in intracerebral hemorrhage (ICH). METHODS We established the ICH model by injecting the collagenase VII-S into the left striatum of young adult (10-12 weeks) male and female and aged (18-20 months) male C57BL/6 mice. We intraperitoneally administered AX-024, a small molecule inhibitor of TCR signaling, and evaluated the results using flow cytometry, Western blotting, immunofluorescence staining, histological staining, bacterial culture, and behavioral tests. RESULTS Our findings in young adult male mice indicate that administering AX-024 within 48 hours suppressed the activation of nonspecific and antigen-specific CD3 (cluster of differentiation 3)+CD4+ and CD3+CD8+ cells in the brain 36 hours and 3 days after ICH but not 7 days after. Additionally, it temporarily inhibited antigen-specific T-cell activation in the periphery at the above 2 time points. It also reduced molecular and cellular neuroinflammation in the hemorrhagic brain early after ICH. These effects in the brain and periphery of young adult male mice ultimately improved ICH outcomes while having no impact on lung bacterial loads. This can be further supported by similar findings in young adult female and aged male mice with ICH. CONCLUSIONS AX-024 may represent a promising option for mitigating the detrimental effects of T cells entering the damaged brain without increasing bacterial loads in the lung in ICH. The potential of AX-024 as a potent immunosuppressive treatment for ICH is an exciting prospect that warrants further investigation.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Neurology, The People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, People's Republic of China (S.C., P.F., Y.Z., M.H., C.G., J.Z., C.J.)
- Department of Neurology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Peiji Fu
- Department of Neurology, The People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, People's Republic of China (S.C., P.F., Y.Z., M.H., C.G., J.Z., C.J.)
- Department of Neurology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, People's Republic of China (Y.R.-K., Junmin Wang, Jian Wang)
| | - Li Zhu
- Department of Neurology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Yan Zong
- Department of Neurology, The People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, People's Republic of China (S.C., P.F., Y.Z., M.H., C.G., J.Z., C.J.)
- Department of Neurology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Maosen Huang
- Department of Neurology, The People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, People's Republic of China (S.C., P.F., Y.Z., M.H., C.G., J.Z., C.J.)
- Department of Neurology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
| | - Chenhao Gao
- Department of Neurology, The People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, People's Republic of China (S.C., P.F., Y.Z., M.H., C.G., J.Z., C.J.)
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, People's Republic of China (Y.R.-K., Junmin Wang, Jian Wang)
| | - Jiewen Zhang
- Department of Neurology, The People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, People's Republic of China (S.C., P.F., Y.Z., M.H., C.G., J.Z., C.J.)
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, People's Republic of China (Y.R.-K., Junmin Wang, Jian Wang)
| | - Chao Jiang
- Department of Neurology, The People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, People's Republic of China (S.C., P.F., Y.Z., M.H., C.G., J.Z., C.J.)
- Department of Neurology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (S.C., P.F., L.Z., Y.Z., M.H., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, People's Republic of China
| |
Collapse
|
8
|
Pan D, Xia J, Qu Y, Shi C, Du H, Zhang J, Peng F, Wang X, Liu R, Wu L, Hong Q, Chen X. Oleanolic acid inhibits mesangial cell proliferation and inflammatory response in mesangial proliferative glomerulonephritis through IL-17/ERK/AKT pathway. Int Immunopharmacol 2025; 152:114459. [PMID: 40088868 DOI: 10.1016/j.intimp.2025.114459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 02/13/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Mesangial proliferative glomerulonephritis (MsPGN) is a common form of glomerulonephritis characterized by mesangial cell proliferation and inflammatory responses. However, current clinical treatment options for MsPGN are rather limited. Oleanolic acid (OA), a natural pentacyclic triterpenoid compound, exhibits anti-tumor and anti-inflammatory properties and has been proven to have renal protective effects. We speculate that OA could potentially serve as an alternative therapy for MsPGN. OBJECTIVE This study aimed to investigate the therapeutic efficacy and mechanism of OA against MsPGN. METHODS Tail vein injection of anti-Thy1 antibody was used to establish the MsPGN model, followed by a comprehensive assessment of the effects of OA on renal function, histopathological changes, and inflammatory responses in anti-Thy1 nephritis rats. Subsequently, network pharmacology was employed to predict the key targets and pathways of OA in treating MsPGN. Finally, in vivo and in vitro experiments were conducted to validate the results of network pharmacology. RESULTS OA significantly improved renal function, and attenuated mesangial cell proliferation and inflammatory reactions in anti-Thy1 nephritis rats. Network pharmacology analysis identified TNF-α, IL-6, IL-1β, MAPK3, and AKT1 as key targets of OA in the treatment of MsPGN, and involved the IL-17 signaling pathway. Additionally, we observed increased phosphorylation levels of ERK and AKT, as well as activation of downstream inflammatory responses, in both anti-Thy1 nephritis rats and mesangial cells stimulated with IL-17. In contrast, treatment with OA and the ERK inhibitor PD98059 reversed these effects. Furthermore, we identified IL17RA within this pathway as a potential target of OA. CONCLUSIONS Our study demonstrates that OA can modulate the IL-17/ERK/AKT signaling pathway, thereby improving anti-Thy1 antibody-induced MsPGN. This establishes a theoretical basis for OA to potentially serve as a therapeutic agent for treating MsPGN.
Collapse
Affiliation(s)
- Dan Pan
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; The College Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jikai Xia
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Yilun Qu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Chunru Shi
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; The College Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hongjian Du
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Jie Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; School of Medicine, Nankai University, Tianjin, 300191, China
| | - Fei Peng
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; School of Medicine, Nankai University, Tianjin, 300191, China.
| | - Xu Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Ran Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Lingling Wu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China.
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China.
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China; The College Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
9
|
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
|
10
|
Sunzini F, Vossou D, McInnes IB. Novel options to treat psoriatic arthritis and spondyloarthritis - Interleukin-17 gives up its family secrets. Joint Bone Spine 2025; 92:105905. [PMID: 40239760 DOI: 10.1016/j.jbspin.2025.105905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/18/2025]
Affiliation(s)
- Flavia Sunzini
- College of Medical Veterinary and Life Sciences, University of Glasgow, Wolfson Medical School Building, University Avenue, G12 8QQ Glasgow, United Kingdom
| | - Danai Vossou
- College of Medical Veterinary and Life Sciences, University of Glasgow, Wolfson Medical School Building, University Avenue, G12 8QQ Glasgow, United Kingdom
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Wolfson Medical School Building, University Avenue, G12 8QQ Glasgow, United Kingdom.
| |
Collapse
|
11
|
Xie A, Qian W, Ye D, Deng X, Ma Y, Wang R, Zhou Q, Bao Z, Yu R. Sodium propionate protects against bronchopulmonary dysplasia by inhibiting IL-17-mediated apoptosis of alveolar epithelial cells. Sci Rep 2025; 15:11722. [PMID: 40188136 PMCID: PMC11972331 DOI: 10.1038/s41598-025-94794-5] [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: 11/18/2024] [Accepted: 03/17/2025] [Indexed: 04/07/2025] Open
Abstract
Sodium propionate (SP) has been shown to enhance alveolar growth retardation in Bronchopulmonary Dysplasia (BPD), but the mechanism remains unclear. The aim of this study is to explore the potential mechanism of SP in the treatment of BPD by utilizing animal and cell models along with bioinformation analysis. Neonatal mice were exposed to either air (21% O2) or hyperoxia (85% O2) from the first day after birth to establish the BPD model. The neonatal mice were intraperitoneally injected with normal saline (control group) or SP (500 mg/kg, SP group) from day 8 to day 14. SP significantly reduced the inflammatory condition of alveolar septal thickening, and decreased the alveolar fusion and mitigated weight loss in BPD mice. ELISA results demonstrated that SP significantly inhibited the secretion of IL-17, IL-6 and TNFα. Transcriptome analysis confirmed that IL-17 signaling pathway is closely related to the therapeutic effects of SP on BPD. In addition, MX2, MMP10, IL-11, ZMAT4 and SEC1 genes were identified as key and potential targets involved in the mechanism of SP treating BPD. Meanwhile, in mouse alveolar epithelial cells, apoptosis was induced by hyperoxia, but it was reduced following SP intervention. The expression of IL-17 pathway related genes: IL-17A, IL-6, TNFα and cox2 was decreased in hyperoxia treated cells after SP intervention. In conclusion, through transcriptome analysis, animal and cell experiments, we explored the role of sodium propionate in attenuating apoptosis in a BPD model through IL-17 pathway.
Collapse
Affiliation(s)
- Anni Xie
- Department of Neonatology, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, China
| | - Weilin Qian
- Department of Neonatology, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, China
| | - Danni Ye
- Department of Neonatology, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, China
| | - Xianhui Deng
- Department of Neonatology, Jiangyin People's Hospital of Nantong University, Jiangyin, 214400, China
| | - Yizhe Ma
- Department of Neonatology, Jiangyin People's Hospital of Nantong University, Jiangyin, 214400, China
| | - Ran Wang
- Department of Neonatology, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, China
| | - Qin Zhou
- Department of Pediatric, Wuxi Yihe Gynaecology and Obstetrics Hospital, Wuxi, 214124, China.
| | - Zhidan Bao
- Department of Neonatology, Jiangyin People's Hospital of Nantong University, Jiangyin, 214400, China.
| | - Renqiang Yu
- Department of Neonatology, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, 214002, China.
| |
Collapse
|
12
|
Louis K, Tabib T, Macedo C, Wang J, Cantalupo P, Chandran U, Gu X, Lucas M, Randhawa P, Abundis M, Das J, Singh H, Lefaucheur C, Metes D. High-dimensional profiling of immune responses to kidney transplant reveals heterogeneous T helper 1 and B cell effectors associated with rejection. Am J Transplant 2025; 25:706-719. [PMID: 39419342 PMCID: PMC11972895 DOI: 10.1016/j.ajt.2024.10.009] [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: 07/12/2024] [Revised: 09/20/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Rejection is a primary cause of allograft dysfunction after kidney transplantation. The diversity of immune subpopulations involved in the different endotypes of rejection remains to be delineated at single-cell resolution. In a cohort of 76 kidney transplant recipients, we conducted high-dimensional immune phenotyping of blood CD4 T and B cells, single-cell RNA and T/B cell receptor sequencing, and plasma cytokine profiling. Phenotypic, transcriptional, and clonal states of CD4T and B cells could significantly distinguish stable allograft states from rejection. Patients undergoing T cell-mediated rejection displayed accumulation of clonally expanded cytotoxic T helper (Th)1 cells and Th17-like cells, associated with predominant naive B cell responses. In contrast, antibody-mediated rejection was characterized by clonal expansion of Th1-polarized T follicular helper cells and effector T-bet+ memory B cells, both of which strongly expressed interleukin 12 and tumor necrosis factor-signaling pathways. Plasma cytokine analysis confirmed mixed Th1/Th17 and Th1/T follicular helper cell-driven inflammatory profiles distinguishing T cell-mediated rejection and antibody-mediated rejection, respectively. CD4T and B cell subpopulations and signatures were validated using bulk RNA-seq analysis of matched kidney allografts and using an independent single-cell RNA-seq data set. These data improve mechanistic understanding of the immune pathogenesis of rejection and support the development of more specific immunosuppressive therapies to treat allograft rejection.
Collapse
Affiliation(s)
- Kevin Louis
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, Paris, France.
| | - Tracy Tabib
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Camila Macedo
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jiefei Wang
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Paul Cantalupo
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Uma Chandran
- Department of Biomedical Informatics and UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Xinyan Gu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michelle Lucas
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Parmjeet Randhawa
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marisa Abundis
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jishnu Das
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Harinder Singh
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carmen Lefaucheur
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, Paris, France
| | - Diana Metes
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
13
|
Bai L, Li Y, Lu C, Yang Y, Zhang J, Lu Z, Huang K, Xian S, Yang X, Na N, Huang F, Zeng Z. Anti-IL-17 Inhibits PINK1/Parkin Autophagy and M1 Macrophage Polarization in Rheumatic Heart Disease. Inflammation 2025; 48:870-884. [PMID: 38977539 PMCID: PMC12052801 DOI: 10.1007/s10753-024-02094-3] [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: 04/13/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
Rheumatic heart disease (RHD) is an important and preventable cause of cardiovascular death and disability, but the lack of clarity about its exact mechanisms makes it more difficult to find alternative methods or prevention and treatment. We previously demonstrated that increased IL-17 expression plays a crucial role in the development of RHD-related valvular inflammatory injury. Macrophage autophagy/polarization may be a pro-survival strategy in the initiation and resolution of the inflammatory process. This study investigated the mechanism by which IL-17 regulates autophagy/polarization activation in macrophages. A RHD rat model was generated, and the effects of anti-IL-17 and 3-methyladenine (3-MA) were analyzed. The molecular mechanisms underlying IL-17-induced macrophage autophagy/polarization were investigated via in vitro experiments. In our established RHD rat model, the activation of the macrophage PINK1/Parkin autophagic pathway in valve tissue was accompanied by M1 macrophage infiltration, and anti-IL-17 treatment inhibited autophagy and reversed macrophage inflammatory infiltration, thereby attenuating endothelial-mesenchymal transition (EndMT) in the valve tissue. The efficacy of 3-MA treatment was similar to that of anti-IL-17 treatment. Furthermore, in THP-1 cells, the pharmacological promotion of autophagy by IL-17 induced M1-type polarization, whereas the inhibition of autophagy by 3-MA reversed this process. Mechanistically, silencing PINK1 in THP-1 blocked autophagic flux. Moreover, IL-17-induced M1-polarized macrophages promoted EndMT in HUVECs. This study revealed that IL-17 plays an important role in EndMT in RHD via the PINK1/Parkin autophagic pathway and macrophage polarization, providing a potential therapeutic target.
Collapse
Affiliation(s)
- Ling Bai
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Yuan Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Yiping Yang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Jie Zhang
- Emergency Office, Nanning Center for Disease Control and Prevention, Nanning , Guangxi, China
| | - Zirong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Keke Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Xi Yang
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Na Na
- Department of Neuroscience, The Scripps Research Institute, La Jolla, USA
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China.
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6, Nanning, 530021, Guangxi, China.
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China.
| |
Collapse
|
14
|
Ma X, Guo S, Liu F, Li C, Shi X, Liu W, Qi L, Yuan Y, Xie X, Wang P, Borish L, Feng X. Unveiling the prevalence and impact of silent rhinovirus infection in chronic rhinosinusitis with nasal polyps. Ann Allergy Asthma Immunol 2025; 134:420-430.e1. [PMID: 39892505 PMCID: PMC11972899 DOI: 10.1016/j.anai.2025.01.026] [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: 10/19/2024] [Revised: 01/06/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNPs) involves persistent sinus inflammation, with emerging evidence suggesting a potential role of rhinovirus (RV) in its pathophysiology. However, whether RV exists in nasal tissues and affects the nasal mucosa after the resolution of infection symptoms remains unknown. OBJECTIVE To investigate the prevalence and impact of silent RV infection in nasal tissues. METHODS RV loads were detected in the nasal tissues of 47 controls and 101 patients with CRSwNP without respiratory infection. Participants were categorized into RV-positive (+), RV-negative (-), and the "gray zone" groups. Quantitative polymerase chain reaction, Western blotting, and immunofluorescence assays were used to analyze the impact of silent RV infection on the immune status of nasal tissues. RESULTS Silent RV infection was prevalent in both control (34%) and CRSwNP (30.7%) tissues, with higher viral loads observed in the nasal polyps. In controls, it was associated with high expression of types 1 and 2 interferon (IFN), type 2 inflammation, interleukin (IL)-17A, and IL-10. In patients with CRSwNP, silent RV infection was associated with lower levels of type 1 IFN, IL-17A, type 2 inflammation, and IL-10 but higher levels of type 2 IFN compared with those without RV infection. Meanwhile, RV (+) nasal polyps exhibited fewer tissue eosinophils and neutrophils than RV (-) nasal polyps. CONCLUSION Silent RV infection was prevalent in the nasal tissues, with a higher viral load detected in the nasal polyps. This silent RV infection is associated with distinct immune responses in healthy controls and patients with CRSwNP, involving differential modulation of IFNs, TH2 cytokines, IL-17A, IL-10, and eosinophil and neutrophil levels.
Collapse
Affiliation(s)
- Xinyi Ma
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Shu Guo
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Fangying Liu
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Changqing Li
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Xueyun Shi
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Weiyuan Liu
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Lijie Qi
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Ye Yuan
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Xinyu Xie
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Pin Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Larry Borish
- Departments of Medicine and Microbiology, University of Virginia Health System, Charlottesville, Virginia
| | - Xin Feng
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology (Shandong University), Shandong Provincial Key Medical and Health Discipline of Qilu Hospital of Shandong University, Jinan, People's Republic of China.
| |
Collapse
|
15
|
Emmanuel T, Ben Abdallah H, Baez E, Rather IM, Steiniche T, Bregnhøj A, Iversen L, Johansen C. Early Neutrophil Activation in Psoriatic Skin at Relapse Following Dead Sea Climatotherapy. Exp Dermatol 2025; 34:e70094. [PMID: 40181552 PMCID: PMC11969059 DOI: 10.1111/exd.70094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/19/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
Psoriasis, a chronic inflammatory skin disorder characterised by erythematous and scaly plaques, can be both physically and emotionally distressing for patients. Dead Sea climatotherapy (DSC), a treatment modality combining sun exposure, mineral-rich water and mud therapy during 4 weeks at Ein Gedi, Israel, is used for a small group of patients with psoriasis. This study aimed to investigate the cellular composition of psoriatic skin lesions at relapse after complete clearance from DSC. Skin biopsies from baseline, end of treatment and relapse were collected from eight patients with plaque psoriasis who achieved complete clearance from Dead Sea climatotherapy treatment. These biopsies were subjected to immunohistochemistry, RNA sequencing and quantitative polymerase chain reaction analysis (qPCR). Our findings demonstrate that DSC effectively reduces inflammatory markers to levels comparable to baseline non-lesional skin in the short term. The differential expression analysis identified several upregulated differentially expressed genes, including OSM, CXCL8, TREM1, CXCL1, CSF3R, BCL2A1 and CXCL2, in relapsed psoriasis skin compared with baseline lesional skin. These findings were confirmed by qPCR analysis. Pathway enrichment analysis indicated a marked upregulation of neutrophil-associated pathways in relapse skin compared with baseline lesional skin. Immunohistochemical staining for neutrophil markers, such as CD11b, CD15, CD66b, CD207, MPO and NE, showed a non-significant trend towards enhanced neutrophil infiltration and activation at relapse. In conclusion, while DSC provides short-term effectiveness in managing psoriasis, the initial relapse phase is associated with neutrophil activation and migration. Thus, targeting neutrophils early in the psoriasis disease course may disturb the evolution of psoriasis, potentially preventing disease chronicity.
Collapse
Affiliation(s)
- Thomas Emmanuel
- Department of DermatologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
| | - Hakim Ben Abdallah
- Department of DermatologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
| | - Elena Baez
- Department of DermatologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
| | - Ida Maja Rather
- Department of DermatologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
| | | | - Anne Bregnhøj
- Department of DermatologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
| | - Lars Iversen
- Department of DermatologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
- MC2 Therapeutics A/SHoersholmDenmark
| | - Claus Johansen
- Department of DermatologyAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
| |
Collapse
|
16
|
Palazzo E, Lotti R, Quadri M, Pincelli C, Marconi A. IL-17 Ligand and Receptor Family Members Are Differentially Expressed by Keratinocyte Subpopulations and Modulate Their Differentiation and Inflammatory Phenotype. Int J Mol Sci 2025; 26:2989. [PMID: 40243580 PMCID: PMC11988555 DOI: 10.3390/ijms26072989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by dysregulation of the interleukin 17 (IL-17) signaling axis. Given that psoriasis development depends on keratinocyte stem cells and early progenitors' sensitivity to differentiation, we analyzed IL-17 ligands and the expression and function of in a novel subset of keratinocyte subpopulations: keratinocyte stem cells (KSC) and early and late Transit Amplifying (ETA or LTA, respectively) cells. We found that all subpopulations expressed all IL-17 variants, predominantly in ETA and LTA. Conversely, IL-17 receptor expression resulted in more heterogeneity, with IL-17RA, -C, and -E being the most differentially regulated. Stimulus with IL-17A, IL-17-F, IL-17-A/F, and IL-17C promotes the upregulation of CXCL1, CXCL8, and DEFB4 mRNAs expression in both KSC and ETA. Moreover, IL-17A and IL-17A/F mainly decrease KSC proliferation and promote cell cycle block. Globally, IL-17A and IL-17A/F modulated the expression of proliferation, differentiation, and psoriasis-associated markers. Furthermore, KSC- and ETA-derived 3D reconstructions displayed increased epidermal thickness and upregulated KRT16 expression after treatment with IL-17A or IL-17A/F. Therefore, our data demonstrated that IL-17 family members perform distinctive functions in a specific keratinocyte subpopulation and define IL-17 signaling as a critical modulator of KSC behavior, proving its role in epidermal homeostasis dysregulation of psoriasis.
Collapse
Affiliation(s)
- Elisabetta Palazzo
- DermoLab, Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.L.); (M.Q.); (C.P.); (A.M.)
| | | | | | | | | |
Collapse
|
17
|
Sun N, Wang Y. RORγt inhibitors in clinical development for the treatment of autoimmune diseases: challenges and opportunities. Expert Opin Ther Pat 2025:1-13. [PMID: 40110872 DOI: 10.1080/13543776.2025.2482936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Nuclear receptor retinoid-related orphan receptor gamma-t (RORγt) is a major transcription factor for Th17 cell differentiation and IL-17 production. RORγt has been considered as a promising drug target for the treatment of IL-17-mediated inflammatory diseases. Numerous small molecule inhibitors have been discovered, and more than 20 of RORγt inhibitors have been advanced to clinical trials. However, none of these compounds has yet achieved market approval. AREAS COVERED This manuscript summarizes the development of 22 clinical-stage RORγt inhibitors, including their structures, patent applications, and clinical trial status, based on publications and patents available up to November 2024. EXPERT OPINION The discovery of RORγt inhibitors was considered as an exciting field for the development of small molecular treatments, which has gone through a boom period in the past 10 years. However, some of the leading RORγt inhibitors recently failed in clinical trials due to lack of efficacy or having some safety concerns, although a few small molecule candidates targeting RORγt are still in trials and more in preclinical studies. Realizing the challenge, researchers started to develop different approaches such as dual targeting or exploring new indications, utilizing the potential value of RORγt inhibitors.
Collapse
Affiliation(s)
- Nannan Sun
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, China
| | - Yonghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Liu Y, Li L, Sun Y, Wang S, Jiang P, Chen Q, Zhang L, Hao Z, Yang X, Yan J, Pan J, Guan W, Xu Z, Zhou Y, Lv S, Kuang H, Yang B. Dictamnus dasycarpus turcz. Attenuates collagen-induced rheumatoid arthritis in DBA/1J mice through inhibiting IL-17 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119458. [PMID: 39929403 DOI: 10.1016/j.jep.2025.119458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 03/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rheumatoid Arthritis (RA), a chronic inflammatory autoimmune condition, presents a substantial challenge to public health. Dictamnus dasycarpus Turcz. (D. dasycarpus) is a traditional Chinese medicinal plant recognized for its anti-inflammatory properties and is increasingly being utilized as a potential anti-RA agent, but the underlying mechanism is still unclear. AIM OF THE STUDY The objective of this research is to elucidate the potential active components and therapeutic properties of D. dasycarpus in experimental RA-induced DBA/1J mice, and to uncover the pharmacological basis of its action. MATERIALS AND METHODS Surface plasmon resonance (SPR) was employed to ascertain the specificity of interactions between protein targets and D. dasycarpus active ingredients for treating RA. The extract of D. dasycarpus was obtained by HP-20 microresin column chromatography, and its chemical composition was assessed using UPLC-Orbitrap-MS. This study utilized a collagen-induced arthritis (CIA) mouse model for in vivo experimentation. Body weight, foot thickness measurements, arthritis scores, immune organ index, and serum antibody levels of mice were used as indicators to evaluate the effects of D. dasycarpus components in treating RA. The serum levels of inflammatory factors in mice were measured using a cytokine antibody microarray assay. Additionally, this study quantified the protein expression levels associated with inflammatory responses through a combination of immunohistochemical staining and western blotting analyses. RESULTS This research investigated the interaction between D. dasycarpus active components and target proteins, including PTPN14, using a BIACORE system. The screened active components were identified as alkaloids through mass spectrometry. The UPLC-Orbitrap-MS analysis revealed that alkaloids were the predominant constituents in the 60% EtOH extract of D. dasycarpus. Alkaloid components significantly reduced the arthritis index, foot swelling, and serum antibody levels of IgG1, IgG 2a, and IgG 2b in CIA mice. Histological staining results indicated that alkaloid components mitigate disease exacerbations in CIA mice. Bioinformatics analysis and protein level detection results show that the therapeutic mechanism of D. dasycarpus in managing RA could be attributed to the suppression of the IL-17 signaling pathway. CONCLUSION This study was based on clarifying the therapeutic effect of D. dasycarpus on RA, identifies its effective chemical components as alkaloids. It systematically elucidates the pharmacological mechanisms of alkaloids in treating RA, thereby laying a crucial theoretical foundation for further exploration of the active constituents of D. dasycarpus.
Collapse
Affiliation(s)
- Yan Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Luxin Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Ye Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Siyi Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Peng Jiang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Qingshan Chen
- College of Agriculture, Northeast Agricultural University, Harbin, 150038, PR China.
| | - Lili Zhang
- College of Agriculture, Northeast Agricultural University, Harbin, 150038, PR China.
| | - Zhichao Hao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Xu Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Jiujiang Yan
- Heilongjiang Zbd Pharmaceutical Co., Ltd, Harbin, 150046, PR China.
| | - Juan Pan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Wei Guan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Zhenpeng Xu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Yuanyuan Zhou
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Shaowa Lv
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, PR China; Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), PR China.
| |
Collapse
|
19
|
Zhang J, Shen M. The Role of IL-17 in Systemic Autoinflammatory Diseases: Mechanisms and Therapeutic Perspectives. Clin Rev Allergy Immunol 2025; 68:27. [PMID: 40074883 DOI: 10.1007/s12016-025-09042-5] [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] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Interleukin (IL)-17, a pro-inflammatory cytokine, plays a pivotal role in immune regulation by bridging innate and adaptive responses. Beyond its canonical involvement in T helper-17 cells-mediated immunity, IL-17 contributes significantly to the pathogenesis of systemic autoinflammatory diseases (SAIDs) including Familial Mediterranean Fever (FMF), nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-associated autoinflammatory diseases, and synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome. Dysregulated IL-17 signaling drives inflammasome activation, neutrophil recruitment, and chronic tissue inflammation. IL-17 inhibitors have demonstrated efficacy in refractory SAIDs, though challenges such as increased infection risks, paradoxical inflammatory reactions, and uncertainties regarding long-term safety persist. Currently, there is insufficient data to support the use of IL-17 inhibitors as first-line treatments, and their role in managing SAIDs is yet to be fully defined. This review highlights the mechanistic role of IL-17 in SAIDs and emerging therapeutic strategies, including IL-17-targeted monotherapies and combination approaches with IL-1 or tumor necrosis factor (TNF) inhibitors. Future research should focus on biomarker development, combination therapies, and long-term studies to optimize the safety and efficacy of IL-17-targeted therapies in SAIDs.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Min Shen
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
20
|
Kokubu E, Kikuchi Y, Yonezawa H, Sasaki H, Matsuzaka K, Ishihara K. Effect of Porphyromonas Gingivalis Infection on Epithelial Rests of Malassez. THE BULLETIN OF TOKYO DENTAL COLLEGE 2025; 66:13-23. [PMID: 39956574 DOI: 10.2209/tdcpublication.2024-0028] [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: 02/18/2025]
Abstract
The epithelial cell rests of Malassez (ERM) are located within the periodontal ligament. They are reportedly involved in maintaining homeostasis, particularly with regards to the thickness of the periodontal ligament. Their role in apical periodontitis lesions remains unclear, however. This study investigated the response of ERM to infection with Porphyromonas gingivalis. After being infected, the morphology of the P. gingivalis-infected cells was observed using confocal laser-scanning microscopy. The gene expression of P. gingivalis-infected and uninfected cells was investigated by RNA-sequencing analysis. Morphological observation showed the invasion and adhesion of P. gingivalis to the surface of ERM. The RNA analysis showed that the gene expression profile significantly differed between the infected and uninfected cells. At an expression level of ≥2 and false discovery rate of <0.1, the infected cells showed a decrease in 99 genes and an increase in 6 compared with in the non-infected cells. Most of the upregulated genes were unique to epithelial cells, such as endothelial cell-specific molecules and cytokeratin 5; the upregulated genes were associated with the immune response, however. These results indicate that ERM upregulate genes associated with epithelial cells and suppress those associated with the immune response following P. gingivalis infection.
Collapse
Affiliation(s)
- Eitoyo Kokubu
- Department of Microbiology, Tokyo Dental College
- Oral Health Science Center, Tokyo Dental College
| | - Yuichiro Kikuchi
- Department of Microbiology, Tokyo Dental College
- Oral Health Science Center, Tokyo Dental College
| | - Hideo Yonezawa
- Department of Microbiology, Tokyo Dental College
- Oral Health Science Center, Tokyo Dental College
| | - Hodaka Sasaki
- Department of Oral and Maxillofacial Implantology, Tokyo Dental College
- Oral Health Science Center, Tokyo Dental College
| | - Kenichi Matsuzaka
- Department of Pathology, Tokyo Dental College
- Oral Health Science Center, Tokyo Dental College
| | - Kazuyuki Ishihara
- Department of Microbiology, Tokyo Dental College
- Oral Health Science Center, Tokyo Dental College
| |
Collapse
|
21
|
Neuwirth T, Malzl D, Knapp K, Tsokkou P, Kleissl L, Gabriel A, Reininger B, Freystätter C, Marella N, Kutschat AP, Ponweiser E, Haschemi A, Seruggia D, Menche J, Wagner EF, Stary G. The polyamine-regulating enzyme SSAT1 impairs tissue regulatory T cell function in chronic cutaneous inflammation. Immunity 2025; 58:632-647.e12. [PMID: 40023161 DOI: 10.1016/j.immuni.2025.02.011] [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: 04/02/2024] [Revised: 11/08/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025]
Abstract
Regulatory T (Treg) cells are a critical immune component guarding against excessive inflammation. Treg cell dysfunction can lead to chronic inflammatory diseases with current therapies aimed at inhibiting effector T cells rather than rescuing Treg cell function. We utilized single-cell RNAsequencing data from patients with chronic inflammation to identify SAT1, the gene encoding spermidine/spermine N1-acetyltransferase (SSAT), as a driver of skin-resident Treg cell dysfunction. CRISPRa-driven SAT1 expression in human skin-derived Treg cells impaired their suppressive function and induced a pro-inflammatory phenotype. During cutaneous type-17 inflammation, keratinocyte 4-1BBL induces SAT1 on Treg cells. In a mouse model of psoriasis, pharmacological inhibition of SSAT rescued Treg cell number and function. Together, these data show that SAT1 expression has severe functional consequences on Treg cells and suggest a therapeutic target to treat chronic inflammatory disease.
Collapse
Affiliation(s)
- Teresa Neuwirth
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Daniel Malzl
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, Vienna, Austria; Center for Molecular Biology, Department of Structural and Computational Biology, University of Vienna, Vienna, Austria
| | - Katja Knapp
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Panagiota Tsokkou
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Lisa Kleissl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Anna Gabriel
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Baerbel Reininger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christian Freystätter
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Nara Marella
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Ana P Kutschat
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Elisabeth Ponweiser
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Davide Seruggia
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, Vienna, Austria; Center for Molecular Biology, Department of Structural and Computational Biology, University of Vienna, Vienna, Austria; Faculty of Mathematics, University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Network Medicine at the University of Vienna, Vienna, Austria
| | - Erwin F Wagner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria; Christian Doppler Laboratory for Chronic Inflammatory Skin Diseases, Vienna, Austria.
| |
Collapse
|
22
|
Zhu C, Boucheron N, Al-Rubaye O, Chung BK, Thorbjørnsen LW, Köcher T, Schuster M, Claudel T, Halilbasic E, Kunczer V, Muscate F, Cavanagh LL, Waltenberger D, Lercher A, Ohradanova-Repic A, Schatzlmaier P, Stojakovic T, Scharnagl H, Bergthaler A, Stockinger H, Huber S, Bock C, Kenner L, Karlsen TH, Ellmeier W, Trauner M. 24-Nor-ursodeoxycholic acid improves intestinal inflammation by targeting T H17 pathogenicity and transdifferentiation. Gut 2025:gutjnl-2024-333297. [PMID: 40032499 DOI: 10.1136/gutjnl-2024-333297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND 24-Nor-ursodeoxycholic acid (NorUDCA) is a novel therapeutic bile acid for treating immune-mediated cholestatic liver diseases, such as primary sclerosing cholangitis (PSC). OBJECTIVE Since PSC strongly associates with T helper-type-like 17 (TH17)-mediated intestinal inflammation, we explored NorUDCA's immunomodulatory potential on TH17 cells. DESIGN NorUDCA's impact on TH17 differentiation was assessed using a CD4+TNaive adoptive transfer mouse model, and on intraepithelial TH17 pathogenicity and transdifferentiation using an αCD3 stimulation model combined with interleukin-17A-fate-mapping. Mechanistic studies used molecular and multiomics approaches, flow cytometry and metabolic assays with pathogenic (p) TH17. Pathogenicity of pTH17 exposed to NorUDCA in vitro was evaluated following adoptive transfer in intestinal tissues or the central nervous system (CNS). Key findings were validated in an αCD3-stimulated humanised NSG mouse model reconstituted with peripheral blood mononuclear cells from patients with PSC. RESULTS NorUDCA suppressed TH17 effector function and enriched regulatory T cell (Treg) abundance upon CD4+TNaive cell transfer. NorUDCA mitigated intraepithelial TH17 pathogenicity and decreased the generation of proinflammatory 'TH1-like-TH17' cells, and enhanced TH17 transdifferentiation into Treg and Tr1 (regulatory type 1) cells in the αCD3-model. In vivo ablation revealed that Treg induction is crucial for NorUDCA's anti-inflammatory effect on TH17 pathogenicity. Mechanistically, NorUDCA restrained pTH17 effector function and simultaneously promoted functional Treg formation in vitro, by attenuating a glutamine-mTORC1-glycolysis signalling axis. Exposure of pTH17 to NorUDCA dampened their pathogenicity and expansion in the intestine or CNS upon transfer. NorUDCA's impact on TH17 inflammation was corroborated in the humanised NSG mouse model. CONCLUSION NorUDCA restricts TH17 inflammation in multiple mouse models, potentiating future clinical applications for treating TH17-mediated intestinal diseases and beyond.
Collapse
Affiliation(s)
- Ci Zhu
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Nicole Boucheron
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Osamah Al-Rubaye
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Brian K Chung
- Department of Transplantation Medicine, Clinic of Surgery and Specialized Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Liv Wenche Thorbjørnsen
- Department of Transplantation Medicine, Clinic of Surgery and Specialized Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Thomas Köcher
- Vienna BioCenter Core Facilities, Metabolomics, Vienna BioCenter, Vienna, Austria
| | - Michael Schuster
- Biomedical Sequencing Facility, Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Emina Halilbasic
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Victoria Kunczer
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Fanziska Muscate
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Lois L Cavanagh
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Darina Waltenberger
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alexander Lercher
- Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Philipp Schatzlmaier
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Andreas Bergthaler
- Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Bock
- Biomedical Sequencing Facility, Cemm, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lukas Kenner
- Division of Experimental and Translational Pathology, Department of Pathology, Medical University of Vienna, Vienna, Austria
- Unit of Laboratory Animal Pathology, Department for Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tom H Karlsen
- Department of Transplantation Medicine, Clinic of Surgery and Specialized Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Wilfried Ellmeier
- Institute of Immunology, Center for Pathophysiology Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Elzubeir A, High J, Hammond M, Shepstone L, Pond M, Walmsley M, Trivedi P, Culver E, Aithal G, Dyson J, Thorburn D, Alexandre L, Rushbrook S. Assessing brodalumab in the treatment of primary sclerosing cholangitis (SABR-PSC pilot study): protocol for a single-arm, multicentre, pilot study. BMJ Open Gastroenterol 2025; 12:e001596. [PMID: 40032516 PMCID: PMC11877274 DOI: 10.1136/bmjgast-2024-001596] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION Primary sclerosing cholangitis (PSC) is a rare immune-mediated hepatobiliary disease, characterised by progressive biliary fibrosis, cirrhosis, and end-stage liver disease. As yet, no licensed pharmacological therapy exists. While significant advancements have been made in our understanding of the pathophysiology, the exact aetiology remains poorly defined. Compelling evidence from basic science and translational studies implicates the role of T helper 17 cells (Th17) and the interleukin 17 (IL-17) pro-inflammatory signalling pathway in the pathogenesis of PSC. However, exploration of the safety and efficacy of inhibiting the IL-17 pathway in PSC is lacking. METHODS AND ANALYSIS This is a phase 2a, open-label, multicentre pilot study, testing the safety of brodalumab, a recombinant human monoclonal antibody that binds with high affinity to interleukin-17RA, in adults with PSC. This study will enrol 20 PSC patients across five large National Health Service tertiary centres in the UK. The primary outcome of the study relates to determining the safety and feasibility of administering brodalumab in early, non-cirrhotic PSC patients. Secondary efficacy outcomes include non-invasive assessment of liver fibrosis, changes in alkaline phosphatase values and other liver biochemical readouts, assessment of biliary metrics through quantitative MR cholangiography+, and quality of life evaluation on completion of follow-up (using the 5D-itch tool, the PSC-patient-reported outcome and PSC-specific Chronic Liver Disease Questionnaire). ETHICS AND DISSEMINATION Ethical approval for this study has been obtained from the London Bridge Research Ethics Committee (REC23/LO/0718). Written informed consent will be obtained from all trial participants prior to undertaking any trial-specific examinations or investigations. On completion of the study, results will be submitted for publication in peer-reviewed journals and presented at national and international hepatology meetings. A summary of the findings will also be shared with participants and PSC communities. TRIAL REGISTRATION NUMBER ISRCTN15271834.
Collapse
MESH Headings
- Humans
- Cholangitis, Sclerosing/drug therapy
- Cholangitis, Sclerosing/immunology
- Pilot Projects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/immunology
- Multicenter Studies as Topic
- Treatment Outcome
- Adult
- Clinical Trials, Phase II as Topic
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/administration & dosage
- Quality of Life
- United Kingdom
- Male
- Female
- Th17 Cells/immunology
- Th17 Cells/drug effects
- Receptors, Interleukin-17/antagonists & inhibitors
Collapse
Affiliation(s)
- Amera Elzubeir
- University of East Anglia Norwich Medical School, Norwich, UK
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | - Juliet High
- Norwich Clinical Trials Unit, University of East Anglia, Norwich, UK
| | - Matthew Hammond
- Norwich Clinical Trials Unit, University of East Anglia, Norwich, UK
| | - Lee Shepstone
- Norwich Clinical Trials Unit, University of East Anglia, Norwich, UK
| | - Martin Pond
- Norwich Clinical Trials Unit, University of East Anglia, Norwich, UK
| | | | - Palak Trivedi
- NIHR Birmingham Biomedical Research Centre, Centre for Liver and Gastrointestinal Research, University of Birmingham, Birmingham, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Emma Culver
- Translational Gastroenterology Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Guruprasad Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Jessica Dyson
- Liver Unit, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Douglas Thorburn
- University College London institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Leo Alexandre
- University of East Anglia Norwich Medical School, Norwich, UK
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | - Simon Rushbrook
- University of East Anglia Norwich Medical School, Norwich, UK
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| |
Collapse
|
24
|
Chakraborty C, Bhattacharya M, Das A, Saha A. Regulation of miRNA in Cytokine Storm (CS) of COVID-19 and Other Viral Infection: An Exhaustive Review. Rev Med Virol 2025; 35:e70026. [PMID: 40032584 DOI: 10.1002/rmv.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/29/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
In the initial stage of the COVID-19 pandemic, high case fatality was noted. The case fatality during this was associated with the cytokine storm (CS) or cytokine storm syndrome (CSS). Sometimes, virus infections are due to the excessive secretion of pro-inflammatory cytokines, leading to cytokine storms, which might be directed to ARDS, multi-organ failure, and death. However, it was noted that several miRNAs are involved in regulating cytokines during SARS-CoV-2 and other viruses such as IFNs, ILs, GM-CSF, TNF, etc. The article spotlighted several miRNAs involved in regulating cytokines associated with the cytokine storm caused by SARS-CoV-2 and other viruses (influenza virus, MERS-CoV, SARS-CoV, dengue virus). Targeting those miRNAs might help in the discovery of novel therapeutics, considering CS or CSS associated with different virus infections.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | | | - Arpita Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | - Abinit Saha
- Deparment of Zoology, J.K. College, Purulia, India
| |
Collapse
|
25
|
Yang X, Si M, Liu T, Yang J, Jiang L, Sun X, Yu H. The aryl hydrocarbon receptor affects the inflammatory response of bone marrow mesenchymal stem cell via the hippo-YAP pathway to exacerbate systemic lupus erythematosus. FASEB J 2025; 39:e70410. [PMID: 39985295 DOI: 10.1096/fj.202402784r] [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: 11/09/2024] [Revised: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
The impaired immune regulation of bone marrow mesenchymal stem cells (BM-MSCs) disrupts T-cell homeostasis and alters the immunological environment in individuals with systemic lupus erythematosus (SLE). However, the specific molecular mechanisms underlying the defective immune functions of BM-MSCs in patients with SLE remain unclear. Here, we report that BM-MSCs derived from MRL/lpr mice exhibit a diminished proliferative capacity, elevated levels of aryl hydrocarbon receptor (AhR) and increased levels of secreted proinflammatory cytokines, including IL-1β, IL-6, and TNF-α. These BM-MSCs can increase splenocyte proliferation and upregulate the expression of retinoic acid receptor-related orphan receptor gamma t (RORγt) in EL4 cells, which constitute a murine T-cell lymphoblastic leukemia cell line. Furthermore, MRL/lpr mice treated with FICZ (an AhR agonist) displayed splenomegaly and exacerbated renal pathology, alongside increased levels of AhR, and inflammatory cytokines. Notably, BM-MSCs isolated from FICZ-treated mice also facilitated splenocyte proliferation and increased the RORγt level in EL4 cells during coculture. Similar effects were observed when BM-MSCs were exposed to FICZ in vitro, but these effects were reversed by the administration of CH223191 (an AhR antagonist). Additionally, the expression of Yes-associated protein (YAP) was significantly increased in both MRL/lpr mice and FICZ-treated BM-MSCs. Importantly, verteporfin (a Hippo-YAP inhibitor) attenuated the elevated RORγt levels in EL4 cells and the increased splenocyte proliferation. This study advances our understanding of SLE pathogenesis by pinpointing AhR as a pivotal modulator of the inflammatory response of BM-MSCs through the Hippo-YAP pathway in individuals with SLE. This novel insight not only enriches the current knowledge of SLE mechanisms but also highlights new potential therapeutic targets for SLE.
Collapse
Affiliation(s)
- Xingzhi Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Mingjun Si
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Ting Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jingyu Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Lili Jiang
- School of Material Science and Technology, Lanzhou University of Technology, Lanzhou, Gansu, China
| | - Ximeng Sun
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Haitao Yu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
26
|
Sun X, Yang J, Wang Z, Nie Q, Yang Q, Zhang W, Liu M, Wang L, Zhu L. ZEB1 expression in Th17 cells correlated with p-STAT3 in human apical periodontitis. BMC Oral Health 2025; 25:315. [PMID: 40016707 PMCID: PMC11869427 DOI: 10.1186/s12903-025-05633-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND ZEB1, a zinc-finger E homeobox-binding transcription factor most frequently associated with developmental programs linked to epithelial-mesenchymal transition, has been demonstrated to regulate immune cell function. The study aimed to investigate the expression pattern of ZEB1 in Th17 cells and its colocalization with p-STAT3 in human apical periodontitis lesions. METHODS Thirty-nine human periapical tissues were collected for ex vivo study, including periapical granulomas (PGs, n = 14), radicular cysts (RCs, n = 12), and healthy control tissues (control group, n = 13). Inflammatory infiltration of the lesions was assessed using hematoxylin-eosin staining. The expression of ZEB1 was detected and analyzed by immunohistochemistry. The localization of ZEB1 in Th17 cells and its colocalization with p-STAT3 were assessed using fluorescence colocalization. RESULTS ZEB1 expression was significantly higher in PGs and RCs than in the healthy control group; however no significant difference between the two groups was observed. Immunofluorescence analysis revealed that ZEB1 expression was correlated with IL17 and CD4 double-positive cells in human periapical lesions. ZEB1/ p-STAT3 double-positive cells were predominant in RCs and PGs than in the healthy control group. CONCLUSIONS The expression of ZEB1 was significantly elevated in PGs and RCs, and correlated with Th17 cells and p-STAT3 expression. This study revealed that ZEB1 is a potential player correlated with STAT3 activation and Th17 cells in apical periodontitis pathogenesis.
Collapse
Affiliation(s)
- Xiaoyue Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jingwen Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zijun Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qing Nie
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qian Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Mingwen Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Li Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Lingxin Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
27
|
Cui Z, Wang P, Gao W. Microbial dysbiosis in periodontitis and peri-implantitis: pathogenesis, immune responses, and therapeutic. Front Cell Infect Microbiol 2025; 15:1517154. [PMID: 40007610 PMCID: PMC11850578 DOI: 10.3389/fcimb.2025.1517154] [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: 10/25/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
The oral microbiome comprises over 700 distinct species, forming complex biofilms essential for maintaining oral and systemic health. When the microbial homeostasis in the periodontium is disrupted, pathogens within the biofilm can cause periodontitis and peri-implantitis, inducing host immune responses. Understanding the role of microbial communities and the immune mechanisms in oral health and disease is crucial for developing improved preventive, diagnostic and therapeutic strategies. However, many questions remain about how changes in bacterial populations contribute to the development and progression of these conditions. An electronic and manual literature search was conducted using PubMed, Excerpta Medica, Frontiers Reports and the Wiley Online Library databases for relevant articles. Data from these publications were extracted and the overall findings were summarized in a narrative manner. The variations in microbial communities and immune responses of periodontitis and peri-implantitis are explored. Dysbiosis of the subgingival microbiome-characterized by an increase in pathogenic bacteria such as Porphyromonas gingivalis, Tannerella forsythia, and Aggregatibacter actinomycetemcomitans-plays a pivotal role in the initiation and progression of periodontitis. As for peri-implantitis, alterations include a higher abundance of opportunistic pathogens and reduced microbial diversity around implants. Moreover, oral dysbiosis potentially influencing systemic health through immune-mediated pathways. Regional immunity of periodontium involving neutrophils, T helper cells-17, and immune-related cytokines is crucial for maintaining periodontal homeostasis and responding to microbial imbalances. Additionally, the impact of non-mechanical treatments-such as probiotics and laser therapy-on the oral microbiome is discussed, demonstrating their potential in managing microbial dysbiosis. These findings underscore that bacterial dysbiosis is a central factor in the development of periodontitis and peri-implantitis. Maintaining microbial balance is essential for preventing these diseases, and interventions targeting the microbiome could enhance treatment outcomes. Strategies focusing on controlling pathogenic bacteria, modulating immune responses, and promoting tissue regeneration are key to restoring periodontal stability. Further research is needed to clarify the mechanisms underlying the transition from peri-implant mucositis to peri-implantitis and to optimize prevention and treatment approaches, considering the complex interactions between the microbiome and host immunity.
Collapse
Affiliation(s)
| | | | - Weiyue Gao
- Stomatology Center, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
28
|
Grunwell JR, Fitzpatrick AM. Asthma Phenotypes and Biomarkers. Respir Care 2025. [PMID: 40013975 DOI: 10.1089/respcare.12352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Asthma experienced by both adults and children is a phenotypically heterogeneous condition. Severe asthma, characterized by ongoing symptoms and airway inflammation despite high doses of inhaled and/or systemic corticosteroids, is the focus of research efforts to understand this underlying heterogeneity. Clinical phenotypes in both adult and pediatric asthma have been determined using supervised definition-driven classification and unsupervised data-driven clustering methods. Efforts to understand the underlying inflammatory patterns of severe asthma have led to the seminal discovery of type 2-high versus type 2-low phenotypes and to the development of biologics targeted at type 2-high inflammation to reduce the rates of severe asthma exacerbations. Type 2-high asthma is characterized by upregulation of T helper 2 immune pathways including interleukin (IL)-4, IL-5, and IL-13 along with eosinophilic airway inflammation, sometimes allergic sensitization, and responsiveness to treatment with corticosteroids. Type 2-low asthma is poorly responsive to corticosteroids and is not as well characterized as type 2-high asthma. Type 2-low asthma is limited by being defined as the absence of type 2-high inflammatory markers. Choosing a biologic for the treatment of severe asthma involves the evaluation of a panel of biomarkers such as blood eosinophils, total and specific immunoglobulin E/allergic sensitization, and fractional exhaled nitric oxide. In this review, we focus on the underlying pathobiology of adult and pediatric asthma, discuss the different phenotype-based treatment options for adult and pediatric type 2-high with or without allergic asthma and type 2-low asthma, and describe a clinical phenotyping approach to patients to guide out-patient therapy. Finally, we end with a discussion of whether pediatric asthma exacerbations necessitating admission to an ICU constitute their own high-risk phenotype and/or whether it is a part of other previously defined high-risk subgroups such as difficult-to-control asthma, exacerbation-prone asthma, and severe treatment-resistant asthma.
Collapse
Affiliation(s)
- Jocelyn R Grunwell
- Dr. Grunwell is affiliated with Division of Critical Care Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Anne M Fitzpatrick
- Dr. Fitzpatrick is affiliated with Division of Pulmonary, Allergy/Immunology, Cystic Fibrosis, and Sleep Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
29
|
Chen S, Zou L, Huang L, Li Z, Zeng H, Zeng Y, Wu J. SLC7A11 suppresses pyroptosis to alleviate rheumatoid arthritis development by modulating the IL-17 pathway. Int Immunopharmacol 2025; 147:114019. [PMID: 39798470 DOI: 10.1016/j.intimp.2025.114019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology. This study aims to explore the potential mechanisms by which solute carrier family 7 member 11 (SLC7A11) influences RA development. METHODS Collagen-induced arthritis (CIA) mice were constructed to observe disease onset and pathological scores. Pathological changes were examined using Hematoxylin-eosin and Safranin O-Fast Green staining. Levels of lactate dehydrogenase (LDH), inflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-18 and IL-1β), and oxidative stress (reactive oxygen species, malondialdehyde, and glutathione) were measured using ELISA. Western blotting was performed to detect the expression of pyroptosis- and pathway-related proteins. Fibroblast-like synoviocytes of RA (RA-FLS) were treated with TNF-α. Cell migration, invasion, and Caspase-1 levels were assessed through scratch assays, Transwell assays, and flow cytometry, respectively. The correlation between SLC7A11 and immune cell infiltration in RA was analyzed using bioinformatics. Additionally, downstream pathways of SLC7A11 in RA were screened, and the impacts of SLC7A11 on these pathways were validated in vitro. RESULTS CIA mice were successfully established, revealing significant downregulation of SLC7A11 in RA. Staining results indicated that overexpression of SLC7A11 significantly mitigated joint damage in CIA mice. In vitro experiments demonstrated that overexpression of SLC7A11 inhibited migration, invasion, and Caspase-1 expression levels in TNF-α-induced RA-FLSs. Furthermore, SLC7A11 suppressed inflammation, LDH release, and oxidative stress, while inhibiting pyroptosis. SLC7A11 expression was significantly different in multiple immune cells. The IL-17 pathway was identified as a downstream pathway of SLC7A11, and SLC7A11 inhibited the expression of IL-17 pathway proteins. Additionally, rhIL-17A, an activator of the IL-17 pathway, attenuated the inhibitory effects of SLC7A11 on inflammation, oxidative stress, and pyroptosis. CONCLUSION SLC7A11 suppresses pyroptosis to alleviate RA development by inhibiting the IL-17 pathway.
Collapse
Affiliation(s)
- Shaojian Chen
- Department of Sports Medical Ganzhou People's Hospital/The Affiliated Ganzhou Hospital Jiangxi Medical College Nanchang University Ganzhou China.
| | - Longqiang Zou
- Department of Sports Medical Ganzhou People's Hospital/The Affiliated Ganzhou Hospital Jiangxi Medical College Nanchang University Ganzhou China
| | - Liangcai Huang
- Department of Sports Medical Ganzhou People's Hospital/The Affiliated Ganzhou Hospital Jiangxi Medical College Nanchang University Ganzhou China
| | - Zhengnan Li
- Department of Sports Medical Ganzhou People's Hospital/The Affiliated Ganzhou Hospital Jiangxi Medical College Nanchang University Ganzhou China
| | - Hui Zeng
- Department of Sports Medical Ganzhou People's Hospital/The Affiliated Ganzhou Hospital Jiangxi Medical College Nanchang University Ganzhou China
| | - Yanmei Zeng
- Department of Clinical Laboatory Ganzhou People's Hospital/The Affiliated Ganzhou Hospital Jiangxi Medical College Nanchang University Ganzhou China
| | - Juan Wu
- Department of Rheumatology Ganzhou People's Hospital/The Affiliated Ganzhou Hospital Jiangxi Medical College Nanchang University Ganzhou China
| |
Collapse
|
30
|
Meng T, Nie L, Wang Y. Role of CD4 + T cell-derived cytokines in the pathogenesis of uveitis. Clin Exp Med 2025; 25:49. [PMID: 39909966 PMCID: PMC11799126 DOI: 10.1007/s10238-025-01565-7] [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: 11/22/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025]
Abstract
Uveitis refers to a diverse group of inflammatory diseases that affecting the uveal tract, comprising the iris, ciliary body, and choroid, with potential repercussions ranging from visual impairment to blindness. The role of autoimmunity in uveitis etiology is complex and still under investigation. CD4+ T cells intricately regulate immune responses in uveitis through their diverse subtypes: Th1, Th2, Th17, Treg (T regulatory), and Tfh (follicular T helper) cells. Each T cell subtype secretes specific cytokines with either pathogenic or protective implications in uveitis. Th1 cells, characterized by IFN-γ secretion and T-bet expression, drive type 1 immune responses against intracellular pathogens. Conversely, Th2 cells, which produce interleukin (IL)-4, IL-5, and IL-13 and express the transcription factor GATA3, mediate type 2 immune responses to larger extracellular threats like helminths. Th17 cells, generating IL-17 and IL-22 and controlled by RORγt, engage in type 3 immune responses against select pathogens. Tfh cells, releasing IL-21 and governed by Bcl6, aid B cell antibody production. Conversely, Tregs, identified by Foxp3, exert regulatory functions in immune homeostasis. This review delves into the roles of CD4+ T cell-derived cytokines in uveitis, emphasizing their intricate involvement in disease progression and resolution. Insight into these mechanisms might guide therapeutic approaches targeting CD4+ T cell responses in uveitis management.
Collapse
Affiliation(s)
- Tingting Meng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Lili Nie
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Ying Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130000, China.
- , Changchun, China.
| |
Collapse
|
31
|
Zhang HX, Hamit D, Li Q, Hu X, Li SF, Xu F, Wang MY, Bao GQ, Li HY. Integrative bioinformatic approach reveals novel melatonin-related biomarkers for Alzheimer's disease. Sci Rep 2025; 15:4193. [PMID: 39905093 PMCID: PMC11794634 DOI: 10.1038/s41598-024-80755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/21/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Melatonin (MLT) can improve mitophagy, thereby ameliorating cognitive deficits in Alzheimer's disease (AD) patients. Hence, our research focused on the potential value of MLT-related genes (MRGs) in AD through bioinformatic analysis. METHODS First, the key cells in the single-cell dataset GSE138852 were screened out based on the proportion of annotated cells and Fisher's test between the AD and control groups. The differentially expressed genes (DEGs) in the key cell and GSE5281 datasets were identified, and the MRGs in GSE5281 were selected via weighted gene coexpression network analysis. After intersecting two sets of DEGs and MRGs, we performed Mendelian randomization analysis to identify the MRGs causally related to AD. Biomarkers were further ascertained through receiver operating characteristic curve (ROC) and expression analysis in GSE5281 and GSE48350. Furthermore, gene set enrichment analysis, immune infiltration analysis and correlation analysis with metabolic pathways were conducted, as well as construction of a regulator network and molecular docking. RESULTS According to the Fisher test, oligodendrocytes were regarded as key cells due to their excellent abundance in the GSE138852 dataset, in which there were 281 DEGs between the AD and control groups. After overlapping with 3,490 DEGs and 550 MRGs in GSE5281, four genes were found to be causally related to AD, namely, G protein-coupled receptor, family C, group 5, member B (GPRC5B), Methyltransferase-like protein 7 A (METTL7A), NF-κB inhibitor alpha (NFKBIA) and RAS association domain family 4(RASSF4). Moreover, GPRC5B, NFKBIA and RASSF4 were deemed biomarkers, except for METTL7A, because of their indistinctive expression between the AD and control groups. Biomarkers might be involved in oxidative phosphorylation, adipogenesis and heme metabolism. Moreover, T helper type 17 cells, natural killer cells and CD56dim natural killer cells were significantly correlated with biomarkers. Transcription factors (GATA2, POU2F2, NFKB1, etc.) can regulate the expression of biomarkers. Finally, we discovered that all biomarkers could bind to MLT with a strong binding energy. CONCLUSION Our study identified three novel biomarkers related to MLT for AD, namely, GPRC5B, NFKBIA and RASSF4, providing a novel approach for the investigation and treatment of AD patients.
Collapse
Affiliation(s)
- Hua-Xiong Zhang
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Dilmurat Hamit
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Qing Li
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xiao Hu
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - San-Feng Li
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Fu Xu
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Ming-Yuan Wang
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Guo-Qing Bao
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Hong-Yan Li
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China.
- Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang Uygur Autonomous Region People's Hospital, No 91, Tianchi Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
32
|
Zhang G, Yu Q, Chen X, Zhao X, Xu Y, Yang X. Unraveling the complexities of immunotherapy for thymic epithelial tumors via bioinformatics and experimental analyses. Comput Biol Med 2025; 185:109488. [PMID: 39631109 DOI: 10.1016/j.compbiomed.2024.109488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Thymic epithelial tumors (TETs) are rare neoplasms typically located in the anterior mediastinum. While immune checkpoint inhibitors (ICIs) show promise for advanced or refractory TETs, their clinical application is hindered by heterogeneous responses across TET subtypes, lack of reliable predictive markers, and the risk of immune-related adverse events (irAEs). METHODS We analyzed TCGA, GEO, and GTEx databases to identify differentially expressed genes (DEGs) among three TET subtypes. Comprehensive enrichment analysis determined gene functions and pathways. CIBERSORT analysis revealed subtype-specific immune infiltration profiles. We assessed immune-related genes using immune/stromal scores, TIDE scores, and immune checkpoint gene correlation analysis. Immunohistochemistry was performed to evaluate FGF17 and PD-L1 protein expression levels and their correlation in TET samples. RESULTS Our findings revealed distinctive molecular and immune infiltration patterns across TET subtypes. Pathway analysis showed upregulation of immune-related pathways in type C. CIBERSORT analysis revealed higher fractions of plasma cells and activated CD4 T cells in type C and increased resting dendritic cells in type A or B3. Furthermore, we identified 1,100 DEGs between responders and non-responders to pembrolizumab. FGF17 emerged as a potential predictive marker for immunotherapy response, showing significantly lower expression in type C and a strong negative correlation with PD-L1 expression (P < 0.001). We identified 115 genes potentially linked to irAEs, with CXCL8, IL17A, and CD40LG among the top hub genes in the protein-protein interaction network. CONCLUSIONS This study provides insights into subtype-specific molecular and immune characteristics of TETs, identifies FGF17 as a potential negative biomarker for immunotherapy response (with lower expression potentially indicating better response), and elucidates mechanisms of irAEs. These findings contribute to the development of targeted immunotherapeutic approaches for managing TETs, particularly in predicting response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Gaowen Zhang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China; Department of Thoracic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266000, China.
| | - Qian Yu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Xiaotong Chen
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, 110001, China.
| | - Xitong Zhao
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Yang Xu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Xueying Yang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| |
Collapse
|
33
|
He Y, Shen X, Zhai K, Nian S. Advances in understanding the role of interleukins in pulmonary fibrosis (Review). Exp Ther Med 2025; 29:25. [PMID: 39650776 PMCID: PMC11619568 DOI: 10.3892/etm.2024.12775] [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: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive, irreversible disease characterized by heterogeneous interstitial lung tissue damage. It originates from persistent or repeated lung epithelial injury and leads to the activation and differentiation of fibroblasts into myofibroblasts. Interleukins (ILs) are a group of lymphokines crucial for immunomodulation that are implicated in the pathogenesis of PF. However, different types of ILs exert disparate effects on PF. In the present review, based on the effect on PF, ILs are classified into three categories: i) Promotors of PF; ii) inhibitors of PF; and iii) those that exert dual effects on PF. Several types of ILs can promote PF by provoking inflammation, initiating proliferation and transdifferentiation of epithelial cells, exacerbating lung injury, while other ILs can inhibit PF through suppressing expression of inflammatory factors, modulating the Th1/Th2 balance and autophagy. The present review summarizes the association of ILs and PF, focusing on the roles and mechanisms of ILs underlying PF.
Collapse
Affiliation(s)
- Yuqing He
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xuebin Shen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Kefeng Zhai
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, P.R. China
| | - Sihui Nian
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
34
|
Zhou W, Yuan S, Kang W, Deng X, Zhou H, Ruan J, Feng X, Qi M, Chen B. Replication Study and Meta-Analysis of the Contribution of Seven Genetic Polymorphisms in Immune-Related Genes to the Risk of Gastric and Colorectal Cancers. Int J Immunogenet 2025; 52:39-55. [PMID: 39800859 DOI: 10.1111/iji.12705] [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: 09/27/2024] [Revised: 12/27/2024] [Accepted: 01/05/2025] [Indexed: 01/30/2025]
Abstract
Recently, it has been realized that immune processes participate in the pathogenesis of human cancers. A large number of genetic polymorphisms in immune-related genes have been extensively examined for their roles in the susceptibility of gastric cancer (GC) and colorectal cancer (CRC), including IL4 gene rs2070874, IL4RA gene rs1801275, IL18 gene rs187238, IL18RAP gene rs917997, IL17A gene rs8193036, IL23R gene rs1884444 and IL23R gene rs10889677. However, there is no consistent conclusion, which calls for further research. In this case-control study, these 7 genetic polymorphisms were genotyped by Sanger sequencing in a total of 1247 patients with cancer (GC/CRC: 460/787) and 800 healthy individuals. A total of 31 previous studies and our present study were included in this meta-analysis. The case-control study revealed that in Hubei Chinese population, rs2070874, rs187238 and rs10889677 were significantly associated with CRC risk, whereas only rs917997 was significantly associated with GC risk. The meta-analysis showed that rs2070874, rs917997, rs8193036 and rs1884444 were significantly associated with GC risk in Chinese population, whereas rs2070874 in total population, rs1801275 in Asian population and rs187238 in Chinese population were significantly associated with CRC risk. IL4 gene rs2070874, IL18RAP gene rs917997, IL17A gene rs8193036 and IL23R gene rs1884444 may serve as the susceptible factors for GC carcinogenesis in Chinese population. IL4 gene rs2070874 in total population, IL4RA gene rs1801275 in Asian population and IL18 gene rs187238 in Chinese population may be genetic biomarkers for CRC susceptibility.
Collapse
Affiliation(s)
- Weiguang Zhou
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Siqi Yuan
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Wenqiang Kang
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xiangyuan Deng
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| | - Hang Zhou
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jiangyi Ruan
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xianhong Feng
- Department of Clinical Laboratory, Wuhan Xinzhou District People's Hospital, Wuhan, Hubei, China
| | - Meifang Qi
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Bifeng Chen
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| |
Collapse
|
35
|
Giakomidi D, Ishola A, Nus M. Targeting gut microbiota to regulate the adaptive immune response in atherosclerosis. Front Cardiovasc Med 2025; 12:1502124. [PMID: 39957996 PMCID: PMC11825770 DOI: 10.3389/fcvm.2025.1502124] [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: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Atherosclerosis, the leading cause of death worldwide, is a chronic inflammatory disease leading to the accumulation of lipid-rich plaques in the intima of large and medium-sized arteries. Accumulating evidence indicates the important regulatory role of the adaptive immune system in atherosclerosis during all stages of the disease. The gut microbiome has also become a key regulator of atherosclerosis and immunomodulation. Whilst existing research extensively explores the impact of the microbiome on the innate immune system, only a handful of studies have explored the regulatory capacity of the microbiome on the adaptive immune system to modulate atherogenesis. Building on these concepts and the pitfalls on the gut microbiota and adaptive immune response interaction, this review explores potential strategies to therapeutically target the microbiome, including the use of prebiotics and vaccinations, which could influence the adaptive immune response and consequently plaque composition and development.
Collapse
Affiliation(s)
- Despina Giakomidi
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Ayoola Ishola
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
36
|
Han L, Li X, Ke G, Dong K, Wang G, Sheng Y, Tao L. Dexamethasone-loaded fibroin nanoparticles promote retinal reattachment in rats by regulating the Th17/Treg balance. NANOTECHNOLOGY 2025; 36:115101. [PMID: 39667017 DOI: 10.1088/1361-6528/ad9df2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 12/12/2024] [Indexed: 12/14/2024]
Abstract
Retinal detachment (RD) is a common acute blinding eye disease, and dexamethasone (DEX), an adrenocorticosteroid, shows protective effects against RD. However, its poor water solubility and low bioavailability limit its effectiveness. To address this, we developed SF@DEX nanomaterials and investigated their therapeutic potential and mechanisms in RD. The nanomaterials were successfully synthesized and characterized, achieving 90% encapsulation efficiency and releasing 60% of DEX within 12 h.In vitro, phagocytosis was measured by flow cytometry, and enzyme-linked immunosorbent assay determined interleukin-17 (IL-17) and interleukin-10 (IL-10) levels. A rat RD model was established surgically, followed by oral administration of silk fibroin (SF), SF@DEX, and DEX. Polymerase chain reaction (PCR) assessed IL-17A and forkhead box P3 (FOXP3) expression, while Western blot analysed transforming growth factor-β1 (TGF-β1), IL-10, IL-17A, and FOXP3 levels. Apoptosis of retinal ganglion cells was evaluated using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, and confocal microscopy detected colocalization of IL-17A and FOXP3. SF@DEX treatment significantly reduced Th17 cells and IL-17A while increasing Tregs, FOXP3, TGF-β1, and IL-10 levels. The severity of RD in rats was notably alleviated by SF@DEX, demonstrating its anti-inflammatory effects through modulation of the Th17/Treg immune balance. These results highlight SF@DEX as a promising nano-based therapy for RD.
Collapse
Affiliation(s)
- Linfeng Han
- Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, People's Republic of China
- Ophthalmology, Wuhu Eye Hospital, Wuhu 241002, People's Republic of China
| | - Xiaomeng Li
- Ophthalmology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Hefei 230036, People's Republic of China
| | - Genjie Ke
- Ophthalmology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Hefei 230036, People's Republic of China
| | - Kai Dong
- Ophthalmology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Hefei 230036, People's Republic of China
| | - Guoping Wang
- Ophthalmology, Wuhu Eye Hospital, Wuhu 241002, People's Republic of China
| | - Yonghong Sheng
- Ophthalmology, Wuhu Eye Hospital, Wuhu 241002, People's Republic of China
| | - Liming Tao
- Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, People's Republic of China
| |
Collapse
|
37
|
Pramanik R, Chattopadhyay S, Bishayi B. Dual neutralization of TGF-β and IL-21 regulates Th17/Treg balance by suppressing inflammatory signalling in the splenic lymphocytes of Staphylococcus aureus infection-induced septic arthritic mice. Immunol Res 2025; 73:38. [PMID: 39831928 DOI: 10.1007/s12026-024-09586-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025]
Abstract
Septic arthritis (SA) caused by Staphylococcus aureus is a severe inflammatory joint disease, characterized by synovitis accompanied with cartilage destruction and bone erosion. The available antibiotic treatment alone is insufficient to resolve the inflammation that leads to high rates of morbidity and mortality. Among the CD4+ T helper lymphocytes, the Th17 and Tregs are key regulators of immune homeostasis. A high Th17 could lead to autoimmunity, whereas an increase in Tregs indicates immunosuppression. Depending on the external cytokine milieu, naïve CD4+ T cells transform into either Th17 or Treg cell lineage. TGF-β in the presence of IL-21 produces Th17 cells and drives the inflammatory cascade of reactions. We studied the effects of in vivo neutralization of TGF-β and IL-21 in septic arthritic mice to control arthritic inflammation, which has not been studied before. The arthritic index showed maximum severity in the SA group which substantially reduced in the Ab-treated groups. Flow cytometric analyses of peripheral blood collected from mice at 9DPI revealed the highest Th17/Treg ratio in the SA group but least in the combined-antibody-treated group. TGF-β1 and IL-21 cytokine production from serum, spleen, and synovial tissue homogenates was significantly reduced in the dual Ab-treated group than in the untreated SA group. From the Western blot analyses obtained from splenic lymphocytes at 9 DPI, we elucidated the possible underlying mechanism of interplay in downstream signalling involving the interaction between different STAT proteins and SOCS, NF-κB, RANKL, mTOR, iNOS, and COX-2 in regulating inflammation and osteoclastogenesis. On endogenous blockade with TGF-β and IL-21, the Th17/Treg ratio and resultant arthritic inflammation in SA were found to be reduced. Therefore, maintaining the Th17/Treg balance is critical to eradicate infection as well as suppress excessive inflammation and neutralization of TGF-β and IL-21 could provide a novel therapeutic strategy to treat staphylococcal SA.
Collapse
Affiliation(s)
- Rochana Pramanik
- Immunology Laboratory, Department of Physiology, University Colleges of Science and Technology, University of Calcutta, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Sreya Chattopadhyay
- Immunology Laboratory, Department of Physiology, University Colleges of Science and Technology, University of Calcutta, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Biswadev Bishayi
- Immunology Laboratory, Department of Physiology, University Colleges of Science and Technology, University of Calcutta, 92 APC Road, Calcutta, 700009, West Bengal, India.
| |
Collapse
|
38
|
Mapuskar KA, London B, Zacharias ZR, Houtman JC, Allen BG. Immunometabolism in the Aging Heart. J Am Heart Assoc 2025; 14:e039216. [PMID: 39719411 PMCID: PMC12054428 DOI: 10.1161/jaha.124.039216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
Structural, functional, and molecular-level changes in the aging heart are influenced by a dynamic interplay between immune signaling and cellular metabolism that is referred to as immunometabolism. This review explores the crosstalk between cellular metabolic pathways including glycolysis, oxidative phosphorylation, fatty acid metabolism, and the immune processes that govern cardiac aging. With a rapidly aging population that coincides with increased cardiovascular risk and cancer incidence rates, understanding the immunometabolic underpinnings of cardiac aging provides a foundation for identifying therapeutic targets to mitigate cardiac dysfunction. Aging alters the immune environment of the heart by concomitantly driving the changes in immune cell metabolism, mitochondrial dysfunction, and redox signaling. Shifts in these metabolic pathways exacerbate inflammation and impair tissue repair, creating a vicious cycle that accelerates cardiac functional decline. Treatment with cancer therapy further complicates this landscape, as aging-associated immunometabolic disruptions augment the susceptibility to cardiotoxicity. The current review highlights therapeutic strategies that target the immunometabolic axis to alleviate cardiac aging pathologies. Interventions include modulating metabolic intermediates, improving mitochondrial function, and leveraging immune signaling pathways to restore cardiac health. Advances in immunometabolism thus hold significant potential for translating preclinical findings into therapies that improve the quality of life for the aging population and underscore the need for approaches that address the immunometabolic mechanisms of cardiac aging, providing a framework for future research.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Barry London
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Internal MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Zeb R. Zacharias
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Jon C.D. Houtman
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Microbiology and ImmunologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Bryan G. Allen
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| |
Collapse
|
39
|
Wei Y, Zhang S, Shao F, Sun Y. Ankylosing spondylitis: From pathogenesis to therapy. Int Immunopharmacol 2025; 145:113709. [PMID: 39644789 DOI: 10.1016/j.intimp.2024.113709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Ankylosing spondylitis (AS) is an autoimmune rheumatic disease that primarily affects the axial joints, with its etiology complex and still not fully understood. The unknown pathogenesis of AS limits the development of treatment strategies, so keeping up-to-date with the current research on AS can help in searching for potential therapeutic targets. In addition to the classic HLA-B27 genetic susceptibility and Th17-related inflammatory signals, increasing research is focusing on the influence of autoantigen-centered autoimmune responses and bone stromal cells on the onset of AS. Autoantigens derived from gut microbiota and preferential TCR both exacerbate the autoimmune response in patients with AS. Furthermore, dysregulated bone metabolism also promotes pathological new bone formation in AS. Current treatments approved for AS almost focus on the management of inflammation with inconsistent treatment results due to the heterogeneity of patients. In this review, we systematically summarized various pathogenesis and management of AS, meanwhile discussed the underlying risk factors and potential therapeutic targets.
Collapse
Affiliation(s)
- Yuxiao Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| | - Shuqiong Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| | - Fenli Shao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
40
|
Cai J, Wang F, Shi S. Expression of serum miR-135a in patients with allergic rhinitis and its relationship with Treg/Th17 balance. Kaohsiung J Med Sci 2025; 41:e12918. [PMID: 39661490 PMCID: PMC11724171 DOI: 10.1002/kjm2.12918] [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: 07/15/2024] [Revised: 10/31/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024] Open
Abstract
The dysregulation of microRNA (miRNA) expression contributes to the development of allergic rhinitis (AR). This study investigates serum miR-135a levels and their association with regulatory T cell (Treg) and T helper cell 17 (Th17) balance in AR patients. A total of 93 AR patients and 76 healthy controls were retrospectively recruited. Levels of serum miR-135a, peripheral blood Th17 and Treg cells, and Treg/Th17-related cytokines were measured. We assessed the diagnostic value of serum miR-135a for AR and its relationship with Treg/Th17 balance. AR patients showed significantly elevated immunoglobulin E (IgE), peripheral blood Th17 cells, and IL-17 and IL-6 levels, alongside reduced serum miR-135a, Treg cells, IL-10, TGF-β1, and Treg/Th17 ratios. A serum miR-135a of ≤0.536 demonstrated diagnostic potential for AR. Patients with higher serum miR-135a levels displayed increased Treg cell level and Treg/Th17 ratios, reduced Th17 cell, and lower total nasal symptom score (TNSS). Serum miR-135a levels in AR patients negatively correlated with TNSS, IL-17, IL-6, and Th17 cell percentages, and positively correlated with IL-10, TGF-β1, Treg cell percentages, and Treg/Th17 ratios. Collectively, decreased serum miR-135a levels in AR patients are associated with Treg/Th17 balance, supporting miR-135a as a potential biomarker for AR diagnosis.
Collapse
Affiliation(s)
- Jing Cai
- Department of OctolaryngologyFirst Affiliated Hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Fang Wang
- Department of OctolaryngologyFirst Affiliated Hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Sheng‐Liu Shi
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| |
Collapse
|
41
|
Yang K, Li Q, Zhuang X, Ma H, Chen B, Yu K, Chen Y. A retrospective analysis of autoimmune hemolytic anemia after allogeneic hematopoietic stem cell transplantation for transfusion-dependent β-thalassemia: focus on T and B lymphocyte reconstitution. Ann Hematol 2025; 104:721-728. [PMID: 39751849 DOI: 10.1007/s00277-024-06157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Autoimmune hemolytic anemia (AIHA) following allogeneic hematopoietic stem cell transplantation (allo-HSCT) is often refractory and relapsing, leading to increased mortality post-HSCT. METHODS We retrospectively analyzed the cases of patients with transfusion-dependent β-thalassemia (TDT) who underwent allo-HSCT to study their clinical features, the occurrence of AIHA post-HSCT, and treatment response and to explore the possible pathogenesis of AIHA. RESULT A total of 113 patients were registered in the study, out of whom 14 developed AIHA following allo-HSCT, resulting in a cumulative incidence of 12.4%. The median age at HSCT was 5 (range: 2-14) years, and the median time of occurrence was 8 (range: 4-17) months after HSCT. Patients who are less than 4 years old at the time of HSCT (P = 0.032) exhibit a higher incidence of AIHA. Compared to patients without AIHA, AIHA patients demonstrate a lower percentage of B lymphocytes at the first 100 days (day + 100) post-HSCT(P = 0.002). There were no statistically significant differences in gender, unrelated donors, HLA incomplete mismatch, iron overload, ABO incompatibility, cytomegalovirus (CMV) reactivation, Epstein Barr virus (EBV) reactivation, acute and chronic graft-versus-host disease (GvHD). When AIHA occurred, the absolute value of regulatory T cells decreased without a clear reduction in the proportion of CD4 + cells, and there was no significant elevation of interleukin-17. Eventually, 78.6% (11/14) of patients achieved complete remission with corticosteroids and rituximab, and patients who failed were efficacious with the bortezomib in combination with corticosteroids. Four patients experienced relapse, with one of them relapsing twice. Two patients relapsed after bortezomib and subsequently achieved remission with retreatment using a combination of corticosteroids and rituximab. All AIHA patients were alive and without relapse at the follow-up cutoff. CONCLUSIONS Patients suffering from TDT are more prone to developing AIHA following allo-HSCT, potentially due to a disruption in the reconstitution balance of T and B lymphocytes. Despite the high incidence, the response to treatment was excellent. For relapsed/refractory patients, alternate therapy with bortezomib and rituximab may be considered.
Collapse
MESH Headings
- Humans
- beta-Thalassemia/therapy
- beta-Thalassemia/immunology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Anemia, Hemolytic, Autoimmune/etiology
- Anemia, Hemolytic, Autoimmune/epidemiology
- Anemia, Hemolytic, Autoimmune/immunology
- Anemia, Hemolytic, Autoimmune/drug therapy
- Retrospective Studies
- Male
- Female
- Child, Preschool
- Adolescent
- Child
- B-Lymphocytes/immunology
- T-Lymphocytes/immunology
- Transplantation, Homologous/adverse effects
- Rituximab
- Immune Reconstitution
- Blood Transfusion
Collapse
Affiliation(s)
- Kaiqian Yang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Qianping Li
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xin Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Haoyuan Ma
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Binbin Chen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Kang Yu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Yi Chen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
42
|
Geng Z, Cao Y, Zhao L, Wang L, Dong Y, Bi Y, Liu G. Function and Regulation of Age-Associated B Cells in Diseases. J Cell Physiol 2025; 240:e31522. [PMID: 39749652 DOI: 10.1002/jcp.31522] [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: 10/10/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/04/2025]
Abstract
The aging process often leads to immune-related diseases, including infections, tumors, and autoimmune disorders. Recently, researchers identified a special subpopulation of B cells in elderly female mice that increases with age and accumulates prematurely in mouse models of autoimmune diseases or viral infections; these B cells are known as age-related B cells (ABCs). These cells possess distinctive cell surface phenotypes and transcriptional characteristics, and the cell population is widely recognized as CD11c+CD11b+T-bet+CD21-CD23- cells. Research has shown that ABCs are a heterogeneous group of B cells that originate independently of the germinal center and are insensitive to B-cell receptor (BCR) and CD40 stimulation, differentiating and proliferating in response to toll-like receptor 7 (TLR7) and IL-21 stimulation. Additionally, they secrete self-antibodies and cytokines to regulate the immune response. These issues have aroused widespread interest among researchers in this field. This review summarizes recent research progress on ABCs, including the functions and regulation of ABCs in aging, viral infection, autoimmune diseases, and organ transplantation.
Collapse
Affiliation(s)
- Zi Geng
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Longhao Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
43
|
Yao M, Qu Y, Zheng Y, Guo H. The effect of exercise on depression and gut microbiota: Possible mechanisms. Brain Res Bull 2025; 220:111130. [PMID: 39557221 DOI: 10.1016/j.brainresbull.2024.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Exercise can effectively prevent and treat depression and anxiety, with gut microbiota playing a crucial role in this process. Studies have shown that exercise can influence the diversity and composition of gut microbiota, which in turn affects depression through immune, endocrine, and neural pathways in the gut-brain axis. The effectiveness of exercise varies based on its type, intensity, and duration, largely due to the different changes in gut microbiota. This article summarizes the possible mechanisms by which exercise affects gut microbiota and how gut microbiota influences depression. Additionally, we reviewed literature on the effects of exercise on depression at different intensities, types, and durations to provide a reference for future exercise-based therapies for depression.
Collapse
Affiliation(s)
- Mingchen Yao
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yaqi Qu
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yalin Zheng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Hao Guo
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
44
|
Chen M, Qiao Y, Yu L, Wang W, Wang W, Sun H, Xu Y, Bai J, Zhou J, Geng D. A microenvironment responsive polyetheretherketone implant with antibacterial and osteoimmunomodulatory properties facilitates osseointegration. Bioact Mater 2025; 43:273-291. [PMID: 39399839 PMCID: PMC11470486 DOI: 10.1016/j.bioactmat.2024.09.017] [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: 06/17/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Failure of intraosseous prostheses is primarily attributed to implant loosening and infections. Current primary therapeutic modalities, such as antibiotics and local debridement, not only face challenges in thoroughly eliminating obstinate adhered bacteria but also encounter difficulties in ameliorating undue inflammatory reactions and regenerating impaired peri-implant bone tissues. Polyetheretherketone (PEEK) has excellent mechanical and physicochemical characteristics and has been used extensively as a medical biomaterial. However, the limited bactericidal and osseointegrative activities of bioinert PEEK restrict its clinical application. Herein, a microenvironment responsive coating with immobilised immunomodulatory magnesium ions (Mg2+) and disinfectant cerium oxide nanoparticles (CNPs) is designed via ion coordination mediated by polydopamine (PDA) and electrospinning based on collagen structure-bionic silk fibroin (SF). By utilising the pH responsiveness of SF, CNPs exhibit potent antibacterial effects in an acidic environment (pH 5.0) caused by local bacterial infection. Due to the chelation interaction with PDA and the constraint of SF, Mg2+ is slowly released, ameliorating the local immune microenvironment and boosting osteogenesis by upregulating M2 phenotype macrophages. Bioinformatics analysis indicates that the inflammation is suppressed via the NF-κB signaling pathway. Overall, this SF-based coating maximizes the synergistic effect of CNPs and Mg2+, offering enhanced antibacterial and osteoimmunomodulatory bioactivity for successful implantation.
Collapse
Affiliation(s)
- Miao Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yusen Qiao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Haifu Sun
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Jun Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
45
|
You K, Yang L, Su Z, Shen J, Fan X, Guo Y, Yuan Z, Lu H. Butyric Acid Modulates Gut Microbiota to Alleviate Inflammation and Secondary Bone Loss in Ankylosing Spondylitis. Biomedicines 2024; 13:9. [PMID: 39857593 PMCID: PMC11762490 DOI: 10.3390/biomedicines13010009] [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: 10/28/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Ankylosing spondylitis (AS) is a chronic inflammatory and autoimmune disease that primarily affects the sacroiliac joints and axial skeleton. While the exact pathogenetic mechanism of AS remains unclear, previous reports have highlighted the involvement of genetic factors, immune responses, and gut microbiota dysregulation in the development of this condition. Short-chain fatty acids (SCFAs), which are microbial fermentation products derived from sugar, protein, and dietary fibers, play a role in maintaining the intestinal barrier function and reducing inflammatory responses. The aim of this study was to investigate the therapeutic potential of butyric acid (BA), an important SCFA, in the treatment of AS. Methods: To evaluate the anti-inflammatory and anti-bone loss effects of BA, a murine AS model was established using proteoglycan and dimethyl dioctadecyl ammonium (DDA) adjuvants. Various techniques, including an enzyme-linked immunosorbent assay (ELISA), magnetic resonance imaging (MRI), micro-CT, histology, quantitative PCR (qPCR) for intestinal tight junction protein expression, and 16S rDNA sequencing to analyze gut microbiota abundance, were employed to assess the inflammation and bone health in the target tissues. Results: The results indicated that BA demonstrated potential in alleviating the inflammatory response in the peripheral joints and the axial spine affected by AS, as evidenced by the reductions in inflammatory infiltration, synovial hyperplasia, and endplate erosion. Furthermore, BA was found to impact the intestinal barrier function positively. Notably, BA was associated with the downregulation of harmful inflammatory factors and the reversal of bone loss, suggesting its protective effects against AS. Conclusions: These beneficial effects were attributed to the modulation of gut microbiota, anti-inflammatory properties, and the maintenance of skeletal metabolic homeostasis. This study contributes new evidence supporting the relationship between gut microbiota and bone health.
Collapse
Affiliation(s)
- Ke You
- Department of Spine Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519082, China; (K.Y.); (L.Y.); (Z.S.); (Y.G.)
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Lianjun Yang
- Department of Spine Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519082, China; (K.Y.); (L.Y.); (Z.S.); (Y.G.)
| | - Zhihai Su
- Department of Spine Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519082, China; (K.Y.); (L.Y.); (Z.S.); (Y.G.)
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Jun Shen
- Department of Spine Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China;
| | - Xinyang Fan
- Centre of Education Development, South China Normal University, Guangzhou 510006, China;
| | - Yuanqing Guo
- Department of Spine Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519082, China; (K.Y.); (L.Y.); (Z.S.); (Y.G.)
| | - Zhen Yuan
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Hai Lu
- Department of Spine Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519082, China; (K.Y.); (L.Y.); (Z.S.); (Y.G.)
| |
Collapse
|
46
|
Gong Y, Jia H, Dang W, Zhou T, He P, Wang X, Zhu B. Enhancing cell-mediated immunity through dendritic cell activation: the role of Tri-GalNAc-modified PLGA-PEG nanoparticles encapsulating SR717. Front Immunol 2024; 15:1490003. [PMID: 39763671 PMCID: PMC11701044 DOI: 10.3389/fimmu.2024.1490003] [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: 09/02/2024] [Accepted: 11/25/2024] [Indexed: 05/16/2025] Open
Abstract
Introduction Vaccines against intracellular pathogens like Mycobacterium tuberculosis (M. tuberculosis) require the induction of effective cell-mediated immunity. Adjuvants primarily enhance antigen-induced adaptive immunity by promoting the activation of antigen-presenting cells (APCs).This study is to develop an adjuvant targeted to dendritic cells (DCs), one of the main APCs, so as to assist in inducing a long-term cellular immune response to M. tuberculosis protein antigens. Methods Polylactic-co-glycolic acid-polyethylene glycol (PLGA-PEG) nanoparticles (NPs) modified with Triantennary N-Acetylgalactosamine (Tri-GalNAc) were prepared to target DCs. Additionally, the stimulator of interferon genes (STING) agonist SR717 was encapsulated within PLGA-PEG NPs to activate DCs. Meanwhile, M. tuberculosis fusion protein (TP) was encapsulated in PLGA-PEG NPs to construct vaccine candidates: TP/Tri-GalNAc-PLGA-PEG-SR717 (TP/GPS in short) and TP/ Tri-GalNAc-PLGA-PEG (TP/GP in short). The targeting and activation effects of these NPs were assessed in vitro and in vivo, and their immunogenicity were evaluated in mice. Results Tri-GalNAc modification significantly enhanced the targeting of NPs to DCs, and encapsulated SR717 effectively promoted the maturation and activation of DCs. TP/GPS elicited a potent antigen-specific T cell immune response and successfully induced long-term immune memory in mice. Moreover, after the mice were infected with H37Ra via nasal instillation, TP/GPS significantly reduced the bacterial load in their lungs. Discussion Tri-GalNAc-modified PLGA-PEG NPs in combination with SR717 targeted and activated DCs, effectively assisting M. tuberculosis antigen in inducing long-term T cell-mediated immunity. This approach offers an innovative and effective adjuvant strategy for the development of subunit vaccine against intracellular pathogen.
Collapse
Affiliation(s)
- Yang Gong
- State Key Laboratory for Animal Disease Control and Prevention & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hongbin Jia
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Wenrui Dang
- State Key Laboratory for Animal Disease Control and Prevention & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Ting Zhou
- State Key Laboratory for Animal Disease Control and Prevention & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Pu He
- State Key Laboratory for Animal Disease Control and Prevention & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaolei Wang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
- College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- State Key Laboratory for Animal Disease Control and Prevention & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
47
|
Luo M, Huang H, Nie H, Liu Y, Chen Y, Zheng F, Xi L, Liu H. Recurrent Enteritis and Intestinal Obstruction in a Patient with Chronic Mucocutaneous Candidiasis due to STAT1 Gain-of-Function Mutation. Mycopathologia 2024; 190:3. [PMID: 39707011 DOI: 10.1007/s11046-024-00912-8] [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: 08/20/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
We presented a case of chronic mucocutaneous candidiasis (CMC) due to STAT1 GOF mutation with recurrent enteritis and intestinal obstruction. A 33-year-old woman complained of recurrent oral erosion and finger (toe) nails damage for over 30 years. Candida albicans were cultured from the oral mucosa and nails. Sanger sequencing revealed a gain-of-function mutation in STAT1 (c.A1159 G, p.T387A). Since the age of 37, she developed recurrent enteritis and intestinal obstruction. Laboratory examinations revealed an increased pSTAT1 protein expression and a decreased proportion of Th17 cells in peripheral blood lymphocyte (PBMC), with a high expression of pSTAT1 and scarce expression of IL17A observed in intestinal immunohistochemistry. Intestinal obstruction had not previously been reported as the main clinical manifestation in STAT1 GOF patients. We speculated that the low levels of IL17A impaired the intestinal barrier, which might lead to gastrointestinal disorders in this patient. This case expanded the clinical phenotype of heterozygous STAT1 GOF patients.
Collapse
Affiliation(s)
- Mingfen Luo
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Dermatology Department, The Third People's Hospital of Huizhou, Huizhou, China
| | - Huan Huang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Dermatology, The First People Hospital of Foshan, Foshan, China
| | - Hanhui Nie
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yinghui Liu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yangxia Chen
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Fuying Zheng
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Liyan Xi
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Hongfang Liu
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
48
|
Lv L, Chen B, Gao Y, Sun N, Li W, Fu W. Discovery of Novel N-Sulfonamide-tetrahydroquinolines as Potent Retinoic Acid Receptor-Related Orphan Receptor γt (RORγt) Inverse Agonists for the Treatment of Psoriasis. J Med Chem 2024; 67:21400-21420. [PMID: 39611350 DOI: 10.1021/acs.jmedchem.4c02318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Guided by the mode of action of 13, our previously discovered RORγt inverse agonist, we conducted five rounds of design syntheses and structure-activity relationship (SAR) studies, ultimately identifying RORγt inverse agonist 5a, which exhibited superior in vitro activity compared to 13. Besides, 5a showed promising therapeutic effects in alleviating psoriasis in mice by intraperitoneal injection. Due to the high lipophilicity and in vitro pharmacokinetic properties of 5a, it was formulated into an ointment, which enabled effective skin retention and mitigated systemic side effects. The ointment of 5a was assessed in the mouse model, where it demonstrated significant antipsoriatic effects, superior to 13 and comparable to the positive control GSK2981278, without obvious toxicity. Furthermore, we elucidated molecular mechanism of action for inverse agonist 5a and agonist 1e by means of molecular dynamics (MD) simulation. In summary, 5a holds great promise as a novel antipsoriatic drug candidate.
Collapse
Affiliation(s)
- Lunan Lv
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Baiyu Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yang Gao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Nannan Sun
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
49
|
Trotta MC, Esposito D, Carotenuto R, di Fraia R, Selvaggio LD, Allosso F, Russo M, Accardo G, Alfano R, D'Amico M, Pasquali D. Thyroid dysfunction in Hashimoto's thyroiditis: a pilot study on the putative role of miR-29a and TGFβ1. Endocrine 2024; 86:1090-1096. [PMID: 39023839 PMCID: PMC11554689 DOI: 10.1007/s12020-024-03965-3] [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] [Received: 05/21/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024]
Abstract
PURPOSE Hashimoto's thyroiditis (HT) is one of the most common causes of thyroid dysfunction in iodine sufficient worldwide areas, but its molecular mechanisms are not completely understood. To this regard, this study aimed to assess serum levels of miRNA-29a (miR-29a) and transforming growth factor beta 1 (TGFβ1) in HT patients with different patterns of thyroid function. METHODS A total of 29 HT patients, with a median age of 52 years (21-68) were included. Of these, 13 had normal thyroid function (Eu-HT); 8 had non-treated hypothyroidism (Hypo-HT); 8 had hypothyroidism on replacement therapy with LT4 (subst-HT). All patients had serum miR-29a assayed through qRT-PCR and serum TGFβ1 assayed by ELISA. RESULTS Serum miR-29a levels were significantly down-regulated in patients with Hypo-HT compared to Eu-HT patients (P < 0.01) and subst-HT patients (P < 0.05). A significant negative correlation was detected between serum miR-29a levels and TSH levels (r = -0.60, P < 0.01). Serum TGFβ1 levels were significantly higher in Hypo-HT than both Eu-HT (P < 0.01) and subst-HT patients (P < 0.05). A negative correlation was observed between serum miR-29a and TGFβ1 (r = -0.75, P < 0.01). CONCLUSIONS In conclusion, Hypo-HT patients had lower levels of serum miR-29a and higher levels of TGFβ1 in comparison with Eu-HT patients. Worthy of note, subst-HT patients showed restored serum miR-29a levels compared with Hypo-HT group, associated with lower serum TGFβ1. These novel findings may suggest a possible impact of replacement therapy with levothyroxine on serum miR-29a levels in HT.
Collapse
Affiliation(s)
- Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Daniela Esposito
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Endocrinology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Raffaela Carotenuto
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy
| | - Rosa di Fraia
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy
| | - Lucia Digitale Selvaggio
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy
| | - Francesca Allosso
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy
| | - Marina Russo
- PhD Course in National Interest in Public Administration and Innovation for Disability and Social Inclusion, Department of Mental, Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- School of Pharmacology and Clinical Toxicology, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | | | - Roberto Alfano
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Daniela Pasquali
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
50
|
Giacomini F, Rho HS, Eischen‐Loges M, Tahmasebi Birgani Z, van Blitterswijk C, van Griensven M, Giselbrecht S, Habibović P, Truckenmüller R. Enthesitis on Chip - A Model for Studying Acute and Chronic Inflammation of the Enthesis and its Pharmacological Treatment. Adv Healthc Mater 2024; 13:e2401815. [PMID: 39188199 PMCID: PMC11650547 DOI: 10.1002/adhm.202401815] [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: 06/12/2024] [Revised: 08/14/2024] [Indexed: 08/28/2024]
Abstract
Enthesitis, the inflammation of the enthesis, which is the point of attachment of tendons and ligaments to bones, is a common musculoskeletal disease. The inflammation often originates from the fibrocartilage region of the enthesis as a consequence of mechanical overuse or -load and consequently tissue damage. During enthesitis, waves of inflammatory cytokines propagate in(to) the fibrocartilage, resulting in detrimental, heterotopic bone formation. Understanding of human enthesitis and its treatment options is limited, also because of lacking in vitro model systems that can closely mimic the pathophysiology of the enthesis and can be used to develop therapies. In this study, an enthes(it)is-on-chip model is developed. On opposite sides of a porous culture membrane separating the chip's two microfluidic compartments, human mesenchymal stromal cells are selectively differentiated into tenocytes and fibrochondrocytes. By introducing an inflammatory cytokine cocktail into the fibrochondrocyte compartment, key aspects of acute and chronic enthesitis, measured as increased expression of inflammatory markers, can be recapitulated. Upon inducing chronic inflammatory conditions, hydroxyapatite deposition, enhanced osteogenic marker expression and reduced secretion of tissue-related extracellular matrix components are observed. Adding the anti-inflammatory drug celecoxib to the fibrochondrocyte compartment mitigates the inflammatory state, demonstrating the potential of the enthesitis-on-chip model for drug testing.
Collapse
Affiliation(s)
- Francesca Giacomini
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Hoon Suk Rho
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Maria Eischen‐Loges
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Clemens van Blitterswijk
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Martijn van Griensven
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| |
Collapse
|