1
|
Liu XR, Li M, Hao QQ, Yu YJ, Liao C, Yu R, Kong DL, Wang Y. Unraveling cysteinyl leukotrienes and their receptors in inflammation through the brain-gut-lung axis. Virulence 2025; 16:2502555. [PMID: 40351036 PMCID: PMC12077450 DOI: 10.1080/21505594.2025.2502555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025] Open
Abstract
Cysteinyl leukotrienes (CysLTs), as potent lipid inflammatory mediators, play a pivotal role in systemic multi-organ inflammation and inter-organ communication through interactions with their receptors (CysLTRs). However, However, the function of CysLT3R is unclear and lacks a network of cross-organ metabolite interactions, and the clinical use of leukotriene receptor antagonists (LTRAs) has certain limitations. This review systematically synthesizes existing evidence and proposes future directions by clarifying receptor subtype specificity, optimizing targeted therapies, exploring CysLTs' applications in neuroimmunology, and elucidating the dual roles of CysLTs in chronic inflammation. It is indicated that CysLTs activate eosinophils, mast cells, and airway tuft cells, driving type 2 immune responses and mucus secretion in the lungs, thereby exacerbating respiratory diseases such as asthma. In the nervous system, CysLTs aggravate neurodegenerative disorders like cerebral ischemia and Alzheimer's disease by disrupting the blood-brain barrier, promoting glial activation, and inducing neuronal damage. In the gut, CysLTs regulate anti-helminth immunity via the tuft cell-ILC2 pathway and collaborate with prostaglandin D2 (PGD2) to modulate bile excretion and mucosal protection. Furthermore, CysLTs mediate communication through the gut-lung and gut-brain axes via metabolites such as succinate, contributing to cross-organ inflammatory regulation. In conclusion, this review highlights the complex roles of CysLTs in chronic inflammation, providing a theoretical foundation for precise intervention in multi-organ inflammatory diseases, which provides a theoretical framework for precision interventions in multi-organ inflammatory diseases and inspires interdisciplinary breakthroughs.
Collapse
Affiliation(s)
- Xiao-Ru Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qian-Qian Hao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Ya-Jie Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Cai Liao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Rui Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - De-Lei Kong
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Guimarães JG, de Campos GY, Machado MP, Oliveira Brito PKM, dos Reis TF, Goldman GH, Bonini Palma PV, de Campos Fraga-Silva TF, Cavallin DCU, Venturini J, da Silva TA. A novel mannan-specific chimeric antigen receptor M-CAR redirects T cells to interact with Candida spp. hyphae and Rhizopus oryzae spores. Bioengineered 2025; 16:2458786. [PMID: 39891522 PMCID: PMC11792852 DOI: 10.1080/21655979.2025.2458786] [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: 04/05/2024] [Revised: 07/24/2024] [Accepted: 08/28/2024] [Indexed: 02/03/2025] Open
Abstract
Invasive fungal infections (IFIs) are responsible for elevated rates of morbidity and mortality, causing around of 1.5 million deaths annually worldwide. One of the main causative agents of IFIs is Candida albicans, and non-albicans Candida species have emerged as a spreading global public health concernment. Furthermore, COVID-19 has contributed to a boost in the incidence of IFIs, such as mucormycosis, in which Rhizopus oryzae is the most prevalent causative agent. The effector host immune response against IFIs depends on the activity of T cells, which are susceptible to the regulatory effects triggered by fungal virulence factors. The fungal cell wall plays a crucial role as a virulence factor, and its remodeling compromises the development of a specific T-cell response. The redirection of Jurkat T cells to target Candida spp. by recognizing targets expressed on the fungal cell wall can be facilitated using chimeric antigen receptor (CAR) technology. This study generated an M-CAR that contains an scFv with specificity to α-1,6 mannose backbone of fungal mannan, and the expression of M-CAR on the surface of modified Jurkat cells triggered a strong activation against Candida albicans (hyphae form), Candida tropicalis (hyphae form), Candida parapsilosis (pseudohyphal form), and Candida glabrata (yeast form). Moreover, M-CAR Jurkat cells recognized Rhizopus oryzae spores, which induced high expression of cell activation markers. Thus, a novel Mannan-specific CAR enabled strong signal transduction in modified Jurkat cells in the presence of Candida spp. or R. oryzae.
Collapse
Affiliation(s)
- Júlia Garcia Guimarães
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara, Sao Paulo State University, São Paulo, Brazil
| | - Gabriela Yamazaki de Campos
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Michele Procópio Machado
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Patricia Vianna Bonini Palma
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | | | | | - James Venturini
- Faculty of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Thiago Aparecido da Silva
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara, Sao Paulo State University, São Paulo, Brazil
| |
Collapse
|
3
|
Li X, Zhu L. Comprehensive profiling of cell type-specific expression and distribution of complement genes in mouse and human kidneys: insights into normal physiology and response to kidney transplantations. Ren Fail 2025; 47:2471568. [PMID: 40015727 PMCID: PMC11869339 DOI: 10.1080/0886022x.2025.2471568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Recent studies innovatively revealed the localized expression of complement genes in kidneys and shed light on the vital roles of the intracellular complement system in the physiologic function and pathological conditions. However, a comprehensive analysis of the expression of complement genes in the context of the evolving cellular landscape of the kidney is not available. METHODS We analyzed single-cell RNA sequencing data from healthy human subjects, C57BL/6 mice, and kidney transplant-rejected mice. The data were sourced from the NCBI Gene Expression Omnibus and processed using quality control measures and unsupervised clustering. Differential gene analyses were based on expression levels. RESULTS In total, 50 complement genes were categorized into pattern recognition molecules, proteases, complement components, receptors, and regulators. In normal mice kidneys, complement genes were expressed at relatively low levels. Among different complement gene categories, receptor genes were most widely expressed in kidney cells. Comparatively, macrophages and mesangial cells are the most abundant immune and nonimmune cell types for complement gene expression. A comparison of human and mouse data showed similar expression patterns, but human kidney complement gene expression was more abundant. Comparative analysis between mouse transplant-rejected and normal kidneys demonstrated stronger complement gene expression in transplant-rejected kidneys. CONCLUSIONS This study illustrated significant similarities in complement gene expression between murine and human kidneys and highlighted the responsive nature of complement genes to kidney injury, underscoring the dynamic nature of local complement regulation. These findings enhance our understanding of the complex regulation of the complement system within the kidney, offering insights into its role in renal disease pathogenesis.
Collapse
Affiliation(s)
- Xianzhi Li
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Li Zhu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
4
|
Li X, Lou C, Ren H, Cui L, Chen K. Fundamental knowledge and research regarding the role of immunity in triple-negative breast cancer from 2014-2024: A bibliometric analysis. Hum Vaccin Immunother 2025; 21:2483022. [PMID: 40135819 PMCID: PMC11951696 DOI: 10.1080/21645515.2025.2483022] [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: 09/13/2024] [Revised: 02/27/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Immunity has vital research value and promising applications in triple-negative breast cancer (TNBC). Nevertheless, few bibliometric analyses have systematically investigated this area. This study aimed to comprehensively review the collaboration and impact of countries, institutions, authors, and journals on the role of immunity in TNBC from a bibliometric perspective, evaluate the keyword co-occurrence of the knowledge structure, and identify hot trends and emerging topics. Articles and reviews related to immunity in TNBC were retrieved from the Web of Science core collection using subject search. A bibliometric study was conducted primarily using CiteSpace and VOSviewer. A total of 3,104 articles and reviews were included from January 1, 2014, through December 31, 2024. The number of articles on immunization in TNBC is rising. These publications are mainly from 415 institutions in 82 countries, led by China and the USA. Among these publications, Lajos Pusztai published the most papers, while Peter Schmid was co-cited the most. The most productive journals focused on molecular biology, biological immunology, and clinical medicine. Furthermore, co-citation analysis revealed that tumor microenvironment, biomarkers, and immune checkpoint inhibitors are current and developing research areas. The keywords "immunotherapy" and "nanoparticles" are also likely to be new trends and focal points for future research. This study adopted bibliometric and visualization methods to provide a comprehensive review of the research on immunization in TNBC. This article will help researchers better understand the dynamic evolution of the role of immunity in TNBC and identify areas for future research.
Collapse
Affiliation(s)
- Xudong Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chun Lou
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - He Ren
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lina Cui
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kexin Chen
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
5
|
Thomas S, Smatti MK, Mohammad AlKhatib HA, Tayyar Y, Nizar M, Zedan HT, Ouhtit A, Althani AA, Nasrallah GK, Yassine HM. Antibody-dependent enhancement of SARS-CoV-2, the impact of variants and vaccination. Hum Vaccin Immunother 2025; 21:2505356. [PMID: 40411306 PMCID: PMC12118418 DOI: 10.1080/21645515.2025.2505356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/24/2025] [Accepted: 05/09/2025] [Indexed: 05/26/2025] Open
Abstract
This study characterized antibody-dependent enhancement (ADE) in serum samples from individuals exposed to SARS-CoV-2 via infection or vaccination and evaluated its association with SARS-CoV-2 variants (Wuhan and Omicron), MERS-CoV, and NL63. ADE assays were performed on sera from SARS-CoV-2-infected patients (n = 210) with varying disease severity and vaccinated individuals (n = 225) who received adenovirus vector, inactivated virus or mRNA vaccines. ADE was assessed using pseudoviruses (PVs) in BHK cells expressing FcgRIIa. Neutralizing antibody levels, total IgG, IgG subclasses, and complement activation were analyzed using ELISA and neutralization assays. ADE was observed in 6.2% of infection samples (primarily severe cases) and 5.3% of vaccinated samples (adenovirus-vector and inactivated virus groups). ADE-positive samples showed reduced neutralizing activity, while total IgG and IgG subclasses did not differ significantly between ADE-positive and negative samples. Complement activation was elevated in severe cases but did not correlate clearly with ADE. Notably, MERS-CoV PV induced ADE in a subset of infected samples, but no ADE was detected for NL63. ADE was observed in SARS-CoV-2-infected individuals, particularly in severe cases, and in those vaccinated with adenovirus-vector and inactivated virus vaccines, but not with mRNA vaccines. Cross-reactivity leading to ADE was detected for MERS-CoV but not for NL63. ADE was associated with reduced neutralizing antibody activity and elevated complement activation in severe infections, though the specific role of complement in ADE remains unclear. These findings highlight the need to investigate the mechanisms underlying ADE and its implications for vaccine design and post-infection immunity against respiratory viruses.
Collapse
Affiliation(s)
- Swapna Thomas
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Maria K. Smatti
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
| | | | - Yaman Tayyar
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- Institute of Biomedicine and Glycomics, Griffith University, Brisbane, Australia
| | - Muna Nizar
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
| | - Hadeel T. Zedan
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- Department of Biomedical Science
| | - Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Asmaa A. Althani
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- QU Health, Qatar University, Doha, Qatar
| | - Gheyath K. Nasrallah
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- Department of Biomedical Science
| | - Hadi M. Yassine
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- Department of Biomedical Science
| |
Collapse
|
6
|
Lee AH, Rodriguez Jimenez DM, Meisel M. Limosilactobacillus reuteri - a probiotic gut commensal with contextual impact on immunity. Gut Microbes 2025; 17:2451088. [PMID: 39825615 DOI: 10.1080/19490976.2025.2451088] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
The gut microbiome plays a key role in human health, influencing various biological processes and disease outcomes. The historical roots of probiotics are traced back to Nobel Laureate Élie Metchnikoff, who linked the longevity of Bulgarian villagers to their consumption of sour milk fermented by Lactobacilli. His pioneering work led to the global recognition of probiotics as beneficial supplements, now a multibillion-dollar industry. Modern probiotics have been extensively studied for their immunomodulatory effects. Limosilactobacillus reuteri (L. reuteri), a widely used probiotic, has garnered significant attention for its systemic immune-regulatory properties, particularly in relation to autoimmunity and cancer. This review delves into the role of L. reuteri in modulating immune responses, with a focus on its impact on systemic diseases.
Collapse
Affiliation(s)
- Amanda H Lee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Marlies Meisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
7
|
Sun R, Lee KY, Mei Y, Nickles E, Le Lin J, Xia R, Liu H, Schwarz H. Induction of cell death in malignant cells and regulatory T cells in the tumor microenvironment by targeting CD137. Oncoimmunology 2025; 14:2443265. [PMID: 39716931 DOI: 10.1080/2162402x.2024.2443265] [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: 10/21/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
Regulatory T cells (Tregs) contribute significantly to the immunosuppressive nature of the tumor microenvironment which is a main barrier for immunotherapies of solid cancers. Reducing Treg numbers enhances anti-tumor immune responses but current depletion strategies also impair effector T cells (Teffs), potentially leading to reduced anti-tumor immunity and/or autoimmune diseases. CD137 has been identified as the most differentially expressed gene between peripheral Tregs and intratumoral Tregs in virtually all solid cancers. Further, CD137 is expressed by malignant cells of certain cancers, making it a potential target for tumor immunotherapy. Here, we report the development of a fully human anti-human CD137 antibody of the IgG1 isotype, clone P1A1, that induces antibody-dependent cell-mediated cytotoxicity (ADCC) in CD137+ Tregs and cancer cells. P1A1 cross-reacts with murine CD137 which allowed testing murine chimeric P1A1 in syngeneic murine tumor models where P1A1 significantly reduced the number of CD137+ Tregs and inhibited tumor growth in a murine hepatocellular carcinoma (HCC) and a melanoma lung metastasis model. P1A1 can also be internalized thus enabling it as a carrier for drugs to target CD137+ Tregs and cancer cells. These anti-cancer properties suggest a translation of P1A1 to human immunotherapy.
Collapse
Affiliation(s)
- Rui Sun
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore, Singapore
| | - Kang Yi Lee
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Mei
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Emily Nickles
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Jia Le Lin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Runze Xia
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore, Singapore
| |
Collapse
|
8
|
Li P, Huang Z, Qin Y, Liao W, Xiang T. Diagnosing pulmonary MALT lymphoma: a case of unilateral cystic lesions. Future Sci OA 2025; 11:2497214. [PMID: 40293107 PMCID: PMC12039412 DOI: 10.1080/20565623.2025.2497214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
We present an atypical case of a 62-year-old female diagnosed with pulmonary mucosa-associated lymphoid tissue (p-MALT) lymphoma, which uniquely manifested as a singular cystic lesion in the lung. Diagnostic evaluations, including comprehensive imaging, bronchoscopy, and CT-guided lung biopsy, revealed this uncommon radiological presentation. Detailed histopathological and immunohistochemical assessments further supported the diagnosis. To determine the extent of the disease, systemic evaluations, such as whole-body PET-CT, gastroscopy, colonoscopy, and bone marrow biopsy, were conducted, confirming its localized nature. Following the definitive diagnosis, the patient underwent a rituximab-centric therapeutic regimen, which yielded significant clinical improvement. This case highlights the importance of recognizing distinctive cystic lung features in p-MALT lymphoma and the indispensable role of holistic diagnostic approaches in guiding precise therapeutic and prognostic decisions.
Collapse
Affiliation(s)
- Ping Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, P.R. China
| | - Zhisheng Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Yan Qin
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Wenjian Liao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Tianxin Xiang
- Jiang Xi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, P.R. China
- Jiangxi Medical Center for Critical Public Health Events, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
9
|
Danev N, Harman RM, Sipka AS, Oliveira L, Huntimer L, Van de Walle GR. The secretomes of bovine mammary epithelial cell subpopulations differentially modulate macrophage function. Vet Q 2025; 45:1-14. [PMID: 39921381 PMCID: PMC11809179 DOI: 10.1080/01652176.2025.2463338] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Bovine mammosphere-derived epithelial cell (MDEC) cultures are heterogeneous and enriched for stem and progenitor cells. We previously reported that the bovine MDEC secretome, comprised of all bioactive factors secreted by the cells, displays regenerative properties, exerts antimicrobial effects, and modulates neutrophil activity, positioning it as a promising non-antibiotic biologic therapy for infectious diseases important to the dairy industry, like mastitis. Mastitis is defined as inflammation of the udder, and it is typically caused by bacterial infection. The effect of the MDEC secretome on macrophages, a first line of defense against bacterial infections in the udder, is unknown and could impact the utility of the secretome as a therapy for mastitis. To address this, we isolated bovine monocytes from peripheral blood and maintained them as an unpolarized (M0) population or polarized them into M1 or M2 phenotypes. Macrophages cultured with the secretome of bovine MDECs were assessed for their ability to phagocytose labeled bacterial particles and accumulate reactive oxygen species (ROS). We used single-cell RNA sequencing (scRNA-seq) and fluorescence-activated cell sorting (FACS) to isolate a subpopulation of MDECs that exert enhanced effects on macrophages. We found that the secretome of MDECs that do not express cluster of differentiation (CD) 73, a cell surface enzyme used as a marker for mesenchymal stromal cells, most strongly increased macrophage phagocytosis and ROS accumulation. These findings will help optimize the generation of the bovine MDEC secretome as a suitable treatment option for mastitis.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Rebecca M. Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Anja S. Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- The Royal (Dick) School of Veterinary Studies and the Roslin Institute, University of Edinburgh, Midlothian, Scotland
| |
Collapse
|
10
|
Laurent PA, André F, Bobard A, Deandreis D, Demaria S, Depil S, Eichmüller SB, Fernandez-Palomo C, Foijer F, Galluzzi L, Galon J, Guckenberger M, Harrington KJ, Herrera FG, Huber PE, Italiano A, Karam SD, Kroemer G, Lambin P, Leuschner C, Mantovani A, Meylan E, Mondini M, Pittet MJ, Pouget JP, Remon J, Sørensen CS, Sotiriou C, Vanpouille-Box C, Weichselbaum RR, Welsh JW, Zitvogel L, Formenti SC, Deutsch E. Pushing the boundaries of radiotherapy-immunotherapy combinations: highlights from the 7 th immunorad conference. Oncoimmunology 2025; 14:2432726. [PMID: 39696783 DOI: 10.1080/2162402x.2024.2432726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Over the last decade, the annual Immunorad Conference, held under the joint auspicies of Gustave Roussy (Villejuif, France) and the Weill Cornell Medical College (New-York, USA) has aimed at exploring the latest advancements in the fields of tumor immunology and radiotherapy-immunotherapy combinations for the treatment of cancer. Gathering medical oncologists, radiation oncologists, physicians and researchers with esteemed expertise in these fields, the Immunorad Conference bridges the gap between preclinical outcomes and clinical opportunities. Thus, it paves a promising way toward optimizing radiotherapy-immunotherapy combinations and, from a broader perspective, improving therapeutic strategies for patients with cancer. Herein, we report on the topics developed by key-opinion leaders during the 7th Immunorad Conference held in Paris-Les Cordeliers (France) from September 27th to 29th 2023, and set the stage for the 8th edition of Immunorad which will be held at Weill Cornell Medical College (New-York, USA) in October 2024.
Collapse
Affiliation(s)
- Pierre-Antoine Laurent
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM, U1030 "Molecular Radiotherapy and Therapeutic Innovations", Gustave Roussy, Villejuif, France
| | - Fabrice André
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
- INSERM U981 "Molecular predictors and new targets in oncology", Gustave Roussy, Villejuif, France
- IHU PRISM Precision Medicine Cancer Center, Gustave Roussy, Villejuif, France
| | | | | | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Stephane Depil
- Cancer Research Center of Lyon, Centre Léon Bérard, Université Claude Bernard, Lyon, France
- ErVimmune, Lyon, France
| | - Stefan B Eichmüller
- Research Group GMP & T-cell therapy, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | | | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology; Sorbonne Université; Sorbonne Paris Cité, Université de Paris, Paris, France
- Centre de Recherche des Cordeliers, Paris, France
| | | | - Kevin J Harrington
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, National Institute of Health Research Biomedical Research Centre, London, UK
| | - Fernanda G Herrera
- Radiation Oncology Service, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
- Immuno-oncology Service, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Peter E Huber
- Department of Radio-oncology and Radiotherapy, University Hospital Heidelberg; Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antoine Italiano
- Department of therapeutic innovations (DITEP), Gustave Roussy, Villejuif, France
- Department of Medicine, Institut Bergonié, Bordeaux, France
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Department of Biology, Hôpital Européen Georges Pompidou AP-HP, Paris, France
- Institut du Cancer Paris CARPEM, Paris, France
| | - Philippe Lambin
- Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Carola Leuschner
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
- William Harvey Research Institute, Queen Mary University, London, UK
| | - Etienne Meylan
- Laboratory of Immunobiology, Department of Molecular Biology, Faculty of Sciences, Université Libre de Bruxelles, Bruxelles, Belgium
- Lung Cancer and Immuno-Oncology laboratory, Bordet Cancer Research Laboratories, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Faculty of Medicine, Université libre de Bruxelles, Bruxelles, Belgium
- ULB Cancer Research Center (U-CRC) and ULB Center for Research in Immunology (U-CRI), Bruxelles, Belgium
| | - Michele Mondini
- INSERM, U1030 "Molecular Radiotherapy and Therapeutic Innovations", Gustave Roussy, Villejuif, France
| | - Mikael J Pittet
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland. Swiss Cancer Center Leman, Lausanne, Switzerland
- Translational Research Center in Onco-Haematology (CRTOH), University of Geneva, Geneva, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM)INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Jordi Remon
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Claus S Sørensen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research; University of Chicago, Chicago, IL, USA
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurence Zitvogel
- ClinicObiome, Gustave Roussy, Villejuif, France
- INSERM U1015 "Tumor Immunology and Anti-Cancer Immunotherapy Unit", Gustave Roussy, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France
- Division of Medicine, Paris-Saclay University, Ile-de-France, France
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New-York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM, U1030 "Molecular Radiotherapy and Therapeutic Innovations", Gustave Roussy, Villejuif, France
- Division of Medicine, Paris-Saclay University, Ile-de-France, France
- RHU LySAIRI "Lymphocyte-Sparing Artificial Intelligence-guided Radio-Immunotherapy", Gustave Roussy, Villejuif, France
| |
Collapse
|
11
|
Thulasinathan B, Suvilesh KN, Maram S, Grossmann E, Ghouri Y, Teixeiro EP, Chan J, Kaif JT, Rachagani S. The impact of gut microbial short-chain fatty acids on colorectal cancer development and prevention. Gut Microbes 2025; 17:2483780. [PMID: 40189834 PMCID: PMC11980463 DOI: 10.1080/19490976.2025.2483780] [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: 12/18/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer is a long-term illness that involves an imbalance in cellular and immune functions. It can be caused by a range of factors, including exposure to environmental carcinogens, poor diet, infections, and genetic alterations. Maintaining a healthy gut microbiome is crucial for overall health, and short-chain fatty acids (SCFAs) produced by gut microbiota play a vital role in this process. Recent research has established that alterations in the gut microbiome led to decreased production of SCFA's in lumen of the colon, which associated with changes in the intestinal epithelial barrier function, and immunity, are closely linked to colorectal cancer (CRC) development and its progression. SCFAs influence cancer progression by modifying epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNA functions thereby affecting tumor initiation and metastasis. This suggests that restoring SCFA levels in colon through microbiota modulation could serve as an innovative strategy for CRC prevention and treatment. This review highlights the critical relationship between gut microbiota and CRC, emphasizing the potential of targeting SCFAs to enhance gut health and reduce CRC risk.
Collapse
Affiliation(s)
- Boobalan Thulasinathan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Kanve N. Suvilesh
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Sumanas Maram
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Erik Grossmann
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Yezaz Ghouri
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Emma Pernas Teixeiro
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Joshua Chan
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | - Jussuf T. Kaif
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| |
Collapse
|
12
|
Zhou JY, Shen QH, Xiong YW, Chen JW, Hui Q, Zhou R, Luo YF, Cheng B, Tan CP, Wu T. Zinc-mediated metalloimmunotherapy with dual elimination of tumor and intratumoral bacteria in oral squamous cell carcinoma. Biomaterials 2025; 323:123439. [PMID: 40450766 DOI: 10.1016/j.biomaterials.2025.123439] [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: 04/15/2025] [Accepted: 05/25/2025] [Indexed: 06/11/2025]
Abstract
Metal dyshomeostasis has been implicated in the immune evasion, host-microbiota interaction and tumor progression during carcinogenesis. Specifically, zinc dyshomeostasis has been observed in oral squamous cell carcinoma (OSCC), and strongly associated with Porphyromonas gingivalis (P. gingivalis) infection. Therefore, developing a zinc-mediated metalloimmunotherapy with synergistic antitumor, antibacteria and immunoactivation effects could be an efficient strategy to combat OSCC. Herein, PYT@ZIF8@siRNA nanoparticles (NPs) were constructed from zeolite imidazolate framework-8 (ZIF8) NPs loaded with pyrithione (PYT, a zinc ionophore) and small interfering RNA (siRNA) for SLC30A1 (zinc transporter 1). Zinc overload caused by PYT@ZIF8@siRNA NPs effectively eradicated OSCC cells by causing mitochondrial dysfunction and pro-death mitophagy, and simultaneously eliminated P. gingivalis by destroying the membrane structure and elevating the reactive oxygen species. Concurrently, PYT@ZIF8@siRNA NPs evoked robust antitumor immunity by eliciting immunogenic cell death and attenuating immunosuppression mediated by P. gingivalis. Moreover, PYT@ZIF8@siRNA significantly interrupted epithelial malignant transformation in experimental oral carcinogenesis model, and synergized with programmed death-1 blockade to establish a collaborative tumoricidal network by reprogramming the immunosuppressive tumor microenvironment in P. gingivalis-infected tumor model. In all, our work proposes a strategy for the simultaneous elimination of tumor and intratumoral P. gingivalis, providing a new avenue for precision metalloimmunotherapy in OSCC.
Collapse
Affiliation(s)
- Jia-Ying Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China
| | - Qing-Hua Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yu-Wen Xiong
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China
| | - Jia-Wen Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China
| | - Qi Hui
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China
| | - Rui Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China
| | - Yi-Fei Luo
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China.
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | - Tong Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, PR China.
| |
Collapse
|
13
|
Zhong X, Tai W, Liu ML, Ma S, Shen T, Zou Y, Zhang CL. The Citron homology domain of MAP4Ks improves outcomes of traumatic brain injury. Neural Regen Res 2025; 20:3233-3244. [PMID: 39314140 PMCID: PMC11881717 DOI: 10.4103/nrr.nrr-d-24-00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/19/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00027/figure1/v/2024-12-20T164640Z/r/image-tiff The mitogen-activated protein kinase kinase kinase kinases (MAP4Ks) signaling pathway plays a pivotal role in axonal regrowth and neuronal degeneration following insults. Whether targeting this pathway is beneficial to brain injury remains unclear. In this study, we showed that adeno-associated virus-delivery of the Citron homology domain of MAP4Ks effectively reduces traumatic brain injury-induced reactive gliosis, tauopathy, lesion size, and behavioral deficits. Pharmacological inhibition of MAP4Ks replicated the ameliorative effects observed with expression of the Citron homology domain. Mechanistically, the Citron homology domain acted as a dominant-negative mutant, impeding MAP4K-mediated phosphorylation of the dishevelled proteins and thereby controlling the Wnt/β-catenin pathway. These findings implicate a therapeutic potential of targeting MAP4Ks to alleviate the detrimental effects of traumatic brain injury.
Collapse
Affiliation(s)
- Xiaoling Zhong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meng-Lu Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shuaipeng Ma
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tianjin Shen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
14
|
Griffin S, de Oliveira Mallia J, Psakis G, Attard J, Caruana M, Gatt R. Comparative analysis of N/TERT-1 and N/TERT-2G keratinocyte responses to oxidative stress and immune challenges. Cell Signal 2025; 132:111861. [PMID: 40355015 DOI: 10.1016/j.cellsig.2025.111861] [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: 12/16/2024] [Revised: 04/05/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
The responses of N/TERT-1 and N/TERT-2G keratinocyte cell lines to oxidative stress and immune challenges were investigated to assess their suitability for dermatological testing. The cell lines were exposed to various stimuli, including PAMPs, DAMPs, H₂O₂, and menadione, to assess cytokine production, oxidative stress markers, cell viability, apoptosis, and membrane integrity. IL-1α, IL-6, IL-8, TNF-α, and TGF-β levels significantly increased in N/TERT-1 cells following exposure to LPS, while N/TERT-2G cells remained unaffected. Both cell lines showed increased production of IL-1α, IL-1β, TNF-α, IL-6, and IL-8 in response to dsDNA and LMW and HMW Poly I:C, although TGF-β significantly decreased only in N/TERT-1 cells. In response to H₂O₂, a dose-dependent increase in cytokine levels was observed in N/TERT-2G, whereas N/TERT-1 did not exhibit a clear dose-dependent response. Markers of oxidative stress, including SOD and GSH, displayed similar patterns in both cell lines, with N/TERT-2G showing slightly higher sensitivity. Lipid peroxidation and mitochondrial membrane potential fluctuations were more pronounced in N/TERT-2G, suggesting greater oxidative stress sensitivity. The baseline GSH levels were higher in N/TERT-1 cells, which may contribute towards the enhanced resilience to oxidative stress. Despite decreased viability in MTT assays following H₂O₂ exposure, the lack of significant changes in cleaved Caspase-3 levels indicated that apoptosis was not the primary mechanism of cell death. These findings highlight the distinct characteristics of N/TERT-1 and N/TERT-2G cells, with N/TERT-1 showing higher baseline resilience to oxidative stress and N/TERT-2G displaying greater sensitivity, particularly to H₂O₂. The study underscores the importance of selecting the appropriate cell line for specific research applications in skin biology and disease modelling, considering the differences in their responses to oxidative and immune challenges.
Collapse
Affiliation(s)
- Sholeem Griffin
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta.
| | - Jefferson de Oliveira Mallia
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| | - Georgios Psakis
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| | - Juan Attard
- Department of Food Sciences and Nutrition, Faculty of Health Sciences, University of Malta, Msida MSD2080, Malta
| | - Matthias Caruana
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta
| | - Ruben Gatt
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| |
Collapse
|
15
|
Ahn JS, Lee YB, Han EJ, Choi YJ, Kim DH, Kwok SK, Choi HK, Chung HJ. Identification of specific gut microbes and their therapeutic potential in ameliorating systemic lupus erythematosus in a mouse model. Life Sci 2025; 374:123684. [PMID: 40320135 DOI: 10.1016/j.lfs.2025.123684] [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: 02/13/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025]
Abstract
AIMS The gut microbiome significantly influences autoimmune diseases, including systemic lupus erythematosus (SLE). This study aimed to characterize the gut microbiome and metabolome in SLE and evaluate the therapeutic potential of specific microbial supplementation in MRL/lpr mice. MATERIALS AND METHODS MRL/lpr mice, a well-established model for SLE, were used to analyze gut microbiome changes before and after SLE symptom onset. 16S rRNA sequencing and GC-MS-based metabolic profiling were performed to identify key microbial species and associated metabolites. Selected microbes were supplemented in MRL/lpr mice for 10 weeks, and their effects on SLE symptoms and Th17/Treg balance were evaluated. KEY FINDINGS Eisenbergiella massiliensis, Lacrimispora saccharolytica, and Hungatella xylanolytica were significantly decreased in MRL/lpr mice following the onset of SLE symptoms. These microbes were strongly correlated with specific metabolites, including 5-cholestanol, cholesterol, p-cresol, and indole. Supplementation with these microbes alleviated SLE symptoms and modulated the Th17/Treg balance. SIGNIFICANCE This study highlights the critical role of gut microbiota in immune regulation and SLE symptom relief. Targeted microbial supplementation may serve as a novel therapeutic strategy for managing SLE.
Collapse
Affiliation(s)
- Ji-Seon Ahn
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Ye-Been Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Eui-Jeong Han
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Yu-Jin Choi
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea; Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Da-Hye Kim
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea; Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54907, Republic of Korea
| | - Seung Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Hea-Jong Chung
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; Department of Bio-Analysis Science, University of Science & Technology, Daejeon 34413, Republic of Korea.
| |
Collapse
|
16
|
Nugteren S, Wang H, van Kooten C, Gelderman KA, Trouw LA. Autoantibodies and therapeutic antibodies against complement factor H. Immunol Lett 2025; 274:107002. [PMID: 40118156 DOI: 10.1016/j.imlet.2025.107002] [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: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
The complement system is a crucial part of our immune defense as, upon recognition, it can kill pathogens fast and effectively. However, misguided complement activation could cause damage to host tissues. Therefore, a well-controlled regulation of the complement system is a necessity to prevent collateral damage. Regulation is achieved by several complement inhibitory proteins, acting at different levels of the complement system. One of these complement regulators is factor H, the main regulator of the alternative complement activation pathway. Factor H can regulate the complement system both in fluid-phase and on the host cell surface by, for example, acting as co-factor for factor I, inactivating C3b. The functional properties of factor H are located within different regions of the protein. Functional impairment of factor H, either because of genetic variants, competing proteins such as the factor H-related proteins and proteins from certain pathogens, but also the presence of autoantibodies will impact on complement activation. However, exact consequences are dependent on the region within factor H that is affected. Autoantibodies binding to factor H have been shown to inhibit several regulatory functions of factor H, which is observed in diseases such as membranoproliferative glomerulonephritis and atypical hemolytic uremic syndrome. As more recently the presence of anti-factor H autoantibodies has also been discovered in several other diseases, ranging from autoimmune diseases to cancer, this review provides an overview of the presence of factor H autoantibodies described in these diseases. Factor H autoantibodies are reported to have inhibitory, or enhancing, effects on factor H, depending on the epitopes that are recognized. Formal conclusions about the pathogenicity of the factor H autoantibodies in some of these diseases cannot be drawn yet. Importantly, understanding the binding and functional impact of anti-factor H (auto)antibodies will allow targeted interventions to diminish pathological consequences of anti-factor H autoantibodies but may also open up additional avenues for the use of anti-factor H antibodies as therapeutic agents.
Collapse
Affiliation(s)
- Saskia Nugteren
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Haiyu Wang
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Cees van Kooten
- Department of Internal Medicine section Nephrology, Center of Expertise for Lupus, Vasculitis and Complement- mediated Systemic Autoimmune Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Kyra A Gelderman
- Erasmus Medical Center, Department of Immunology, Laboratory Medical Immunology, Rotterdam, the Netherlands
| | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
17
|
Nie W, Zhao X, Zhang Y, Zeng C, Yang H, Liu B. Chlorogenic acid alleviates DNCB-induced atopic dermatitis by inhibiting the Akt1/NF-κB signaling pathway. Eur J Pharmacol 2025; 998:177534. [PMID: 40118327 DOI: 10.1016/j.ejphar.2025.177534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE Atopic dermatitis (AD) is a prevalent chronic inflammatory skin disease that significantly impacts patients' quality of life. Chlorogenic acid (CGA), a polyphenol present in various dietary sources and plants, has been shown to reduce skin inflammation. However, its efficacy and mechanisms of action in AD have not been thoroughly investigated. This study aimed to evaluate the therapeutic effect of CGA on AD in mice and explored its mechanism. METHODS To establish a BALB/c mouse model of AD induced by 2,4-dinitrochlorobenzene (DNCB) to evaluate the therapeutic potential of CGA. The anti-inflammatory effects of CGA were assessed by measuring IL-1β and IL-6 levels in TNF-α-stimulated HaCaT cells. The phosphorylation levels of PI3K, Akt, Akt1, NF-κB, and IκB-α were analyzed using Western blotting. Molecular docking was conducted to evaluate the binding affinity of CGA to Akt1. RESULTS Topical application of CGA significantly reduced dermatitis scores, spleen index, epidermal thickness, mast cell infiltration, and skin fibrosis. CGA reversed DNCB-induced increases in IgE, histamine, TNF-α, IL-1β, IL-6, and IL-8 levels. Western blot analysis showed that CGA inhibited the PI3K/Akt and NF-κB signaling pathways. In vitro, CGA exerts its anti-inflammatory effects by inhibiting the Akt1/NF-κB pathway, and the Akt activator (SC79) can counteract this effect. Molecular docking and dynamics simulations suggest that CGA may inhibit Akt1 activity by interacting with specific residues (ALA-50, GLY-37, TYR-326, ASP-323). CONCLUSIONS CGA improves AD by inhibiting the Akt1/NF-κB pathway, suggesting its potential as a natural treatment for AD.
Collapse
Affiliation(s)
- Wenkai Nie
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xuan Zhao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Cheng Zeng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Huiwen Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Bing Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, China.
| |
Collapse
|
18
|
Abós B, Morel E, Ama LFD, Ordás MC, Vicente-Gil S, Carrasco JC, Koppang E, Tafalla C, Herranz-Jusdado JG. Immunological characterization of the rainbow trout bursa. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110345. [PMID: 40246038 DOI: 10.1016/j.fsi.2025.110345] [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: 01/30/2025] [Revised: 03/19/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
The bursa of Fabricius is an immune organ, located in the caudo-dorsal surface of the cloaca, responsible for the development and maturation of avian B cells. A few years ago, a lymphoepithelial tissue placed caudal to the urogenital papilla of the cloaca analogous to the bursa was identified for the first time in Atlantic salmon (Salmo salar). The salmon bursa was demonstrated to involute around sexual maturation, as in birds. However, no primary lymphoid functions were identified in this tissue. In the current study, we have identified a homologous immune organ in rainbow trout (Oncorhynchus mykiss), a different salmonid species. This lymphoepithelium covering a blind sac, caudal to the anus, was identified in rainbow trout at different stages of development and it also experienced regression in an age-dependent way. It contained abundant IgM+ B cells and CD3+ cells and especially numerous was the number of MHC II-expressing cells. In contrast to Atlantic salmon, in rainbow trout, the bursa epithelium contained quite a few IgT+ B cells but very few IgD+ B cells. Thus, by flow cytometry, we could determine that the IgM+ B cells identified in the trout bursa had lost surface IgD expression. Interestingly, although an immunization of rainbow trout by bath barely had effects on the bursa at a transcriptional level, when fish were immunized anally with a model antigen, there were significant changes in the levels of transcription of immune genes in this tissue. These included secreted igm, secreted and membrane igd, bcma and prdm1-a2. Altogether these results evidence the existence of a bursa-like immune structure in another teleost species and provide novel information to understand the immune role of this tissue in fish, pointing to a relation to gut immune responses.
Collapse
Affiliation(s)
- Beatriz Abós
- Biotechnology Department, National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Esther Morel
- Biotechnology Department, National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Laura Fernández-Del Ama
- Animal Health Research Center (CISA), National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Valdeolmos, Madrid, Spain
| | - M Camino Ordás
- Biology, Geology, Physics and Chemistry Department, Rey Juan Carlos University, Móstoles, Madrid, Spain
| | - Samuel Vicente-Gil
- Biotechnology Department, National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Juan Carlos Carrasco
- Biotechnology Department, National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Erling Koppang
- Unit of Anatomy, Veterinary Faculty, Norwegian University of Life Sciences, Ås, Norway
| | - Carolina Tafalla
- Biotechnology Department, National Institute for Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain.
| | | |
Collapse
|
19
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
20
|
Fang F, Gong Z, Guo C, Wang C, Ding L, Zhou B, Chen S. Establishment of an ovarian cell line from tomato grouper (Cephalopholis sonnerati) and its transcriptome response to ISKNV infection. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110304. [PMID: 40185294 DOI: 10.1016/j.fsi.2025.110304] [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: 01/22/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Tomato grouper (Cephalopholis sonnerati) is an economically efficient and nutritious species, whose expansion through factory farming in recent years has been hindered by the frequent occurrence of diseases, limiting the development of its aquaculture industry. The establishment of reliable cell lines is fundamental for conducting comprehensive immunological and virological research on the tomato grouper. In this study, we established an ovarian cell line from tomato grouper, designated TGGO. The TGGO cells were passaged for over 70 passages and cultured in L-15 medium supplemented with 15 % FBS at 27 °C, exhibiting a fibroblast-like morphology. It was determined that the TGGO cells were derived from the tomato grouper through mitochondrial coI gene sequencing. Karyotype analysis determined a chromosome number of 2n = 48. The survival rate of cells cryopreserved in liquid nitrogen for 5 months exceeded 70 % upon thawing. The cells were transfected with the EGFP-N3 plasmid and Cy3-labeled scrambled siRNA, and clear green and red fluorescence were observed. Additionally, the cells exhibited sensitivity to ISKNV, displaying a clear cytopathic effect (CPE) at 24 h post-infection, with viral particles observed under transmission electron microscopy. Transcriptomic analysis of ISKNV-infected TGGO cells showed significant enrichment of differentially expressed genes in pathways related to viral infection, nucleic acid replication, and immune response. Notable pathways include ECM-receptor interaction, PI3K-Akt signaling, viral protein interaction with cytokines and cytokine receptors, ribosome biogenesis, and DNA replication. These findings suggest that the TGGO cell line is susceptible to ISKNV infection and can be used to study this virus. Therefore, the TGGO cell line is anticipated to become a valuable resource for in vitro research on virology and other biological processes in tomato grouper.
Collapse
Affiliation(s)
- Fei Fang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China; State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China
| | - Zhihong Gong
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China
| | - Chenfei Guo
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China
| | - Chongwei Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China
| | - Lanqing Ding
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China
| | - Bo Zhou
- Wanning Linlan Aquaculture Co., Ltd, Wanning, Hainan, 571528, China
| | - Songlin Chen
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China; Yazhoubay Agriculture and Aquaculture Development Co., Ltd, Sanya, Hainan, 572025, China.
| |
Collapse
|
21
|
Yi Z, Wang J, Tan S, Li Z, Wang X, Yu J, Yue R, Xiao J, Wu H, Feng H. ZFYVE1 suppresses IRF3/7-mediated antiviral innate immunity in black carp. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110341. [PMID: 40250505 DOI: 10.1016/j.fsi.2025.110341] [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: 01/22/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Zinc finger FYVE-type containing 1 (ZFYVE1), a FYVE-containing protein, exerts a significant function during autophagosome formation and lipid droplet metabolism. Nevertheless, it is currently unclear whether teleost ZFYVE1 influences antiviral innate immunity. In this paper, the black carp (Mylopharyngodon piceus) homolog of ZFYVE1 (bcZFYVE1) was cloned to explore its regulatory function within the interferon (IFN) signaling cascade of teleost fish. The coding region of bcZFYVE1 is composed of 2337 nucleotides and encodes 778 amino acids, including two zinc-binding FYVE domains and a guanylate-binding protein (GBP) domain. bcZFYVE1 transcription in host cells varied upon stimulation with LPS, grass carp reovirus (GCRV) and spring viremia of carp virus (SVCV), respectively. bcZFYVE1 migrated at approximately 86 kDa in immunoblot assay and was primarily detected in the cytoplasm in immunofluorescence staining assay. Upon the co-expression of bcZFYVE1 and bcIRF3/7 in EPC cells, bcZFYVE1 markedly reduced the transcription of the IFN promoter activated by bcIRF3/7. Correspondingly, the mRNA levels of interferon-stimulated genes (ISGs) downstream were reduced. And, overexpressed bcZFYVE1 markedly inhibited the antiviral activity mediated by bcIRF3/7. Additionally, knockdown of bcZFYVE1 attenuated SVCV replication and improved the antiviral capability of host cells. The co-immunoprecipitation assays verified the interaction between bcZFYVE1 and bcIRF3/7. Furthermore, we found that co-expression of bcZFYVE1 with bcIRF3/7 decreased the protein levels of bcIRF3/7 and both MG132 or chloroquine treatment could restore the protein levels. In summary, our data conclude that ZFYVE1 suppresses IRF3/7-mediated antiviral innate immune response in black carp, which expands the understanding of the function of ZFYVE1.
Collapse
Affiliation(s)
- Zewen Yi
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jiaxin Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Shasha Tan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zhan'ao Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xinyu Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jiajie Yu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Ruimin Yue
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Hui Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
22
|
Chen Y, Wang X, Chen S, Zhang M, Cheng Z, Zhang W, Liu D, Shan Y, Du G, Li W, Yang L, Wang J, Chu Y, Liu M. Evaluation of immune effect to recombinant potential protective antigens of Mycoplasma ovipneumoniae in mice. Microb Pathog 2025; 204:107555. [PMID: 40203960 DOI: 10.1016/j.micpath.2025.107555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Mycoplasma ovipneumoniae is a primary causative agent of pneumonia in ruminants, causing chronic non-progressive pneumonia in domestic sheep and goats, but leading to higher morbidity and mortality in bighorn sheep and wild small ruminants. This disease has become a widespread epidemic, resulting in significant losses to the sheep industry. In this study, we evaluated the immunogenicity and initial protective effects of four antigenic proteins of M. ovipneumoniae, namely Eno, EF-Tu, Ulad, and T4SS. These proteins were used to immunize BALB/c mice either individually or in a combination (rProteins group). The mice were intranasally infected with 109 CCU50/mL M. ovipneumoniae strain NJ01 twice, on days 28 and 30 after immunization. Among the four recombinant proteins, rEno demonstrated the most promising results in terms of inducing specific humoral and cellular immune responses. It also resulted in the lowest lung lesion scores and the lowest M. ovipneumoniae loads in the lungs and bronchoalveolar lavage fluid (BALF). Compared to the other three proteins, rEno provided superior protection. Furthermore, the rEno vaccine significantly reduced the inflammatory response in the lungs of mice, as evidenced by the evaluation of pro-inflammatory cytokines. The expression of IL-1β and NF-κB was significantly reduced, while the expression of IL-4 was significantly increased. In conclusion, the rEno vaccine elicited a favorable immunological response and conferred protection against M. ovipneumoniae. This finding presents a novel approach to controlling the global spread of this pathogen.
Collapse
MESH Headings
- Animals
- Mice, Inbred BALB C
- Mycoplasma ovipneumoniae/immunology
- Mycoplasma ovipneumoniae/genetics
- Pneumonia, Mycoplasma/prevention & control
- Pneumonia, Mycoplasma/immunology
- Pneumonia, Mycoplasma/microbiology
- Pneumonia, Mycoplasma/pathology
- Mice
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Cytokines/metabolism
- Lung/pathology
- Lung/microbiology
- Lung/immunology
- Recombinant Proteins/immunology
- Recombinant Proteins/genetics
- Antibodies, Bacterial/blood
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Bronchoalveolar Lavage Fluid/microbiology
- Female
- Immunity, Cellular
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Disease Models, Animal
- NF-kappa B/metabolism
- Interleukin-1beta/metabolism
- Immunity, Humoral
- Sheep
- Immunization
Collapse
Affiliation(s)
- Yi Chen
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Xiaonan Wang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Siyu Chen
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Mengjie Zhang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Zilong Cheng
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Wenwen Zhang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Diyue Liu
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China
| | - Yiyi Shan
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Gaimei Du
- College of Animal Science and Food Engineering, Jinling Institute of Technology, Nanjing, 211169, PR China
| | - Wenliang Li
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Leilei Yang
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, PR China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, PR China
| | - Maojun Liu
- Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture and Rural Affairs, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, PR China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China; Guotai Technology Innovation Center for Veterinary Bioproducts (Taizhou), Taizhou, 225300, PR China.
| |
Collapse
|
23
|
Yin Y, Wang C, Liang H, Ji Y, Zhao D, Wang L, Shi W, Wang X, Gao Y, Liu B. Design, synthesis, and biological evaluation of 2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-1-one derivatives as potent hematopoietic progenitor kinase 1 (HPK1) inhibitors. Bioorg Chem 2025; 161:108554. [PMID: 40347769 DOI: 10.1016/j.bioorg.2025.108554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Yuan Yin
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Chengqi Wang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Huan Liang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Yanpeng Ji
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Damin Zhao
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Lilong Wang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Wei Shi
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Xiaojin Wang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Yong Gao
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China.
| | - Baomin Liu
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China.
| |
Collapse
|
24
|
Randon PM, Gudjonsson JE, Ward NL. What are mice teaching us about psoriatic arthritis? Curr Opin Rheumatol 2025; 37:243-253. [PMID: 40265275 PMCID: PMC12119223 DOI: 10.1097/bor.0000000000001093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW This review summarizes important mouse models of psoriatic arthritis (PsA), shedding light on their advantages and disadvantages in modeling human disease. RECENT FINDINGS Two newly created mouse models of PsA validate NF-κB signaling as disease-causing and identify pathogenic roles for CD8 + and CD4 + FoxP3 + T cells in the development of specific PsA phenotypes. The IkbkbGoF/GoF model demonstrates that homozygosity for a gain-of-function mutation in Ikbkb results in expansion of FoxP3 + CD25 + IL-17A + Tregs that lead to the development of dactylitis, spondylitis and PsA-like changes to the nails and skin, and when transferred to wildtype mice, reproduce these outcomes. The humanized mouse PsA model (Hu-PsA) establishes that introduction of PsA patient sera and PBMCs into NSG-SGM3 mice has the capacity to elicit distinct subtypes of PsA and identifies a critical role for CD8 + IL-32 + CXCL14 + T cells and immunoglobulins in disease development. SUMMARY Mouse models of PsA are powerful research tools for elucidating pathogenesis of disease, biomarker identification and may assist in the discovery of a cure.
Collapse
Affiliation(s)
- Peggy M. Randon
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Nicole L. Ward
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4) and Vanderbilt Center for Immunobiology (VCI), Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
25
|
Lu G, Wang Y, Huang H, Hua HL, Xi Y, Chen SM, Kong YG, Xu Y, Deng YQ, Tao ZZ. Association Between Diabetes Mellitus and Allergic Diseases Sensitized by Different Allergens and the Potential Mechanism of Diabetes Mellitus Affecting Ovalbumin-Induced Allergic Rhinitis. Am J Rhinol Allergy 2025; 39:271-283. [PMID: 40289517 DOI: 10.1177/19458924251332786] [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: 04/30/2025]
Abstract
ObjectiveTo investigate the association between diabetes mellitus (DM) and allergic diseases caused by different allergens and explore the mechanism of DM in allergic rhinitis (AR) induced by ovalbumin (OVA).MethodsWe established linear regression models to examine the correlation of specific immunoglobulin E (sIgE) and total immunoglobulin E (TIgE) with glycohemoglobin (A1c) and fasting plasma glucose (FPG) in individuals allergic to different allergens by retrospectively analyzing the 2005-2006 National Health and Nutrition Examination Survey database. Participants were classified into Allergy + DM and Allergy groups based on whether they had allergic disease and DM or only allergic disease. An AR mouse model was established using OVA. Mice were randomly assigned to the control, AR, diabetic (db), or db & AR group, with 6 mice in each group. The expression of GATA3, T-bet, and Foxp3 was detected using immunofluorescence and western blotting. Cytokines, FPG, A1c, and immunoglobulin in serum were detected using enzyme-linked immunosorbent assays.ResultsBased on the database, the TIgE and SIgE levels in participants with perennial allergies were lower in the Allergy + DM group than in the Allergy group, whereas those in participants with food allergies were higher in the Allergy + DM group than in the Allergy group. TIgE was negatively correlated with FPG or A1c in participants with perennial allergies and positively correlated with FPG and A1c in participants with egg allergy (among food allergens). In mice, AR symptoms, eosinophilic infiltration, and OVA-induced IgE levels were more serious in the db & AR group than in the AR group. TIgE was positively correlated with FPG, and interleukin-2 (IL-2) was negatively correlated with FPG; this was most pronounced in db & AR mice. The expression of T-bet and Foxp3 in mice was negatively associated with A1c.ConclusionsThe effect of DM on allergic diseases is related to the type of allergen. Decreased IL-2, T-bet, and Foxp3 levels resulting from elevated FPG and A1c levels may be involved in the association between OVA-induced AR and DM.
Collapse
Affiliation(s)
- Gan Lu
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Yan Wang
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Huan Huang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Hong-Li Hua
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Yang Xi
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Shi-Ming Chen
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Yong-Gang Kong
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Yu Xu
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Yu-Qin Deng
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Ze-Zhang Tao
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, P.R. China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| |
Collapse
|
26
|
Ahn W, Han J, Kim N, Hwang YH, Kim W, Lee Y, Lee DY, Cheong IW, Han K, Nam GH, Kim IS, Lee EJ. Hierarchical protein nano-crystalline hydrogel with extracellular vesicles for ectopic lymphoid structure formation. Biomaterials 2025; 318:123166. [PMID: 39933315 DOI: 10.1016/j.biomaterials.2025.123166] [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: 10/17/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Among cancer therapies, immune checkpoint blockade (ICB) has emerged as a prominent approach, substantially enhancing anti-tumor immune responses. However, the efficacy of ICB is often limited in the absence of a pre-existing immune response within the tumor microenvironment. Here, we introduce a novel hierarchical protein hydrogel platform designed to facilitate the formation of artificial tertiary lymphoid structures (aTLS), thereby improving ICB efficacy. Through the integration of self-assembling ferritin protein nanocages, rec1-resilin protein, and CP05 peptide, our hierarchical hydrogels provide a structurally supportive and functionally adaptive scaffold capable of on-demand self-repair in response to mild thermal treatments. The effective encapsulation of extracellular vesicles (EVs) via the CP05 peptide ensures the formation of aTLS with germinal center-like structures within the hierarchical hydrogel. We demonstrate that, combined with ICB therapy, EV-loaded hierarchical hydrogels also induce the TLS within the tumor, markedly promoting immune responses against ICB-resistant tumor. This bioactive hydrogel platform offers a versatile tool for enhancing a broad range of immunotherapies, with potential applications extending beyond TLS to other frameworks that support complex tissue architectures.
Collapse
Affiliation(s)
- Wonkyung Ahn
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea; Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jihoon Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Nayeon Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeong Ha Hwang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Wonjun Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeram Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dong Yun Lee
- Department of Polymer Science and Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - In Woo Cheong
- Department of Applied Chemistry, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Koohee Han
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Gi-Hoon Nam
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
27
|
Goloudina A, Le Chevalier F, Authié P, Charneau P, Majlessi L. Shared neoantigens for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200978. [PMID: 40256120 PMCID: PMC12008704 DOI: 10.1016/j.omton.2025.200978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Exploration of neoantigens holds the potential to be productive in immuno-oncotherapy. Among tumor-specific antigens, neoantigens result from genetic instability that gives rise to non-synonymous somatic mutations, highly specific to tumor cells. In addition to point mutations, gene rearrangements, indels leading to frameshifts, chromosomal translocations or inversions that may lead to fusion proteins, alternative mRNA splicing, and integration of genetic material of oncogenic viruses into the host genome provide consistent sources of neoantigens that are absent in healthy tissues. Out of these alterations, 2%-3% may generate T cell neoepitopes, possibly detectable by TCRs. Neoantigens are absent in healthy tissues and are thus at low risk of triggering autoimmunity. In addition, the host lymphocytes have not been rendered tolerant toward them and it is possible to induce immune responses against them. Here, we overview the two categories of neoantigens, i.e., private and shared, and their use in immuno-oncotherapy in selected pre-clinical and clinical studies. The vast majority of commonly occurring tumor-specific mutations are cancer causing and are permanently expressed by all malignant tumor cells, preventing the latter from escaping vaccine-induced anti-neoantigen immunity. The use of public neoantigens combined with efficient vaccine platforms can provide non-personalized "off-the-shelf" therapeutic vaccine candidates for broad-spectrum immunotherapy purposes.
Collapse
Affiliation(s)
- Anastasia Goloudina
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
28
|
Lu H, Zhang K, Zhang W, Zhang W, Wu J, Ming X, Huang Y, Xu F, Wang B. Inflammation-targeting and self-limited neutrophilic membrane-encapsulated teicoplanin for the treatment of infectious pneumonia. Trends Biotechnol 2025:S0167-7799(25)00175-1. [PMID: 40517114 DOI: 10.1016/j.tibtech.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 06/16/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) pneumonia has a high clinical incidence and is associated with a significant mortality risk. The existence of intracellular pathogens and the infection-induced swarming of neutrophils exacerbate the challenges in treating pneumonia. Here, we addressed these issues by developing a platform based on neutrophilic membrane-camouflaged teicoplanin (Teic@NEV) to kill bacteria in tissue and prevent inflammatory lung injury associated with MRSA pneumonia. Teic@NEV improved the efficiency of drug entry into cells, meaningfully increasing the intracellular drug concentration in infected cells and eliminating intracellular MRSA. Moreover, Teic@NEV enhanced the penetration of teicoplanin into the biofilm and improved antimicrobial and antibiofilm activities in vitro. Surprisingly, Teic@NEV delivered teicoplanin specifically to sites of inflammation and reduced lung injury by hindering neutrophil swarming in vivo. Thus, this platform represents an effective strategy to limit neutrophil swarming and kill intracellular pathogens in patients with MRSA pneumonia, demonstrating its significant potential for use in clinical practice.
Collapse
Affiliation(s)
- Huidan Lu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Kaixin Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Wanying Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wenting Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jicheng Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xinliang Ming
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yuqiao Huang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Institute of Translational Medicine, Zhejiang University; Hangzhou 310029, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Ben Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Institute of Translational Medicine, Zhejiang University; Hangzhou 310029, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou 310029, China; Cancer Center, Zhejiang University, Hangzhou 310029, China.
| |
Collapse
|
29
|
Simeone PG, Costantino S, Liani R, Tripaldi R, Di Castelnuovo A, Tartaro A, Mengozzi A, Cosentino F, Cipollone F, Consoli A, Paneni F, Santilli F. Interleukin-1β in circulating mononuclear cells predicts steatotic liver disease improvement after weight loss in subjects with obesity and prediabetes or type 2 diabetes. Cardiovasc Diabetol 2025; 24:247. [PMID: 40514652 DOI: 10.1186/s12933-025-02706-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/24/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major cardiovascular risk (CV) factor. Interleukin-1β (IL-1β), a cytokine involved in the pathogenesis of obesity-associated inflammation and type 2 diabetes (T2D), promotes hepatic steatosis. The Canakinumab Anti-inflammatory Thrombosis Outcome (CANTOS) trial showed that the inhibition of the IL-1β pathway was associated with a reduction of CV events in high-risk patients. The present study was designed to determine: (i) whether an equal degree of weight loss by liraglutide or lifestyle changes has a different impact on MASLD extent and IL-1β expression in peripheral blood mononuclear cells from obese subjects with prediabetes or early T2D; (ii) whether baseline IL-1β levels may predict the extent of weight loss and related metabolic changes. METHODS Thirty-two obese subjects with prediabetes (n = 16) or newly diagnosed T2D (n = 16), were randomized to the glucagon-like peptide receptor agonist (GLP1-RA) liraglutide or lifestyle counselling until achieving a comparable weight loss. Visceral adipose tissue (VAT) and gene expression of IL-1β in peripheral blood mononuclear cells were assessed by magnetic resonance and real time PCR, respectively. RESULTS At baseline, IL-1β was positively correlated to body mass index (BMI), fasting plasma glucose, HbA1c, VAT, MASLD extent, platelet count, chemerin and interleukin-1 receptor antagonist (IL1-RA). After achievement of the weight loss target in the two groups, a significant but comparable reduction of IL-1β (p for difference = 0.56) was observed in both arms, in parallel with a comparable improvement in glycaemic control, C reactive protein (CRP), BMI and MASLD. Furthermore, basal IL-1β levels independently predicted the extent of MASLD decrease (p = 0.030); subjects in the highest tertile showed a median decrease of - 8.0 (95% CI - 12.3 to - 4.8) compared with - 23.0 (95% CI - 39.5 to - 16.3) in the lowest tertile. CONCLUSION In patients with obesity with initial impairment of glucose metabolism successful weight loss is associated with a reduction of both IL-1β levels and MASLD degree. Of interest, basal levels of IL-1β predict the extent of MASLD improvement, regardless of the intervention. Our results may set the stage for ad-hoc studies investigating the usefulness of baseline IL-1β a level as a drug-response biomarker.
Collapse
Affiliation(s)
- Paola Giustina Simeone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Rossella Liani
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Romina Tripaldi
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | | | - Armando Tartaro
- Department of Neuroscience & Imaging, University of Chieti, Chieti, Italy
| | - Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Agostino Consoli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Francesca Santilli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy.
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
30
|
Pan J, Gan L, Chen Y, Zhong L, Deng H, Huang L, Liang H. Agmatine ameliorates sepsis-related intestinal injury via the AhR-STAT3-IL-10 pathway. Mol Immunol 2025; 184:76-88. [PMID: 40516501 DOI: 10.1016/j.molimm.2025.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 05/01/2025] [Accepted: 06/07/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND Sepsis-related intestinal injury is essential in multi-organ dysfunction induced by the systemic inflammatory response. Agmatine (AGM) has anti-inflammatory, antioxidative, and immunomodulatory properties. However, no reports of AGM alleviating sepsis-related intestinal injury have been found. Therefore, this study intends to investigate the protective effects of AGM on sepsis-related intestinal injury and its potential mechanism. METHODS We first established a sepsis model by lipopolysaccharide (LPS) or cecum ligation perforation (CLP), and then used H&E staining, transmission electron microscopy (TEM), and TUNEL to examine the pathological changes in rat intestines after AGM treatment. ELISA was used to detect expression levels of inflammatory factors in ileal tissues and biochemical indexes of infectionin in serum. TNF-α induced Caco-2 cell inflammation model was established in vitro, and cell activity and proliferative capacity were detected by CCK8 and EdU after AGM treatment. The Caco-2 cell inflammation model with high or low expression of Aryl hydrocarbon receptor (AhR) was established, and whether AGM exerts anti-inflammatory effects via AhR/STAT3/IL-10 pathway was investigated using molecular docking, Co-IP, and Western Blot. RESULTS The histopathological staining demonstrated that AGM significantly reduced intestinal injury and the content of inflammatory factors IL-1β, IL-6, and TNF-α, improved systemic infection and the survival rate of septic mice, and restored intestinal barrier function in CLP rats. In vitro, AGM improved cell viability and proliferative capacity of the Caco-2 cell inflammation model. AGM inhibits inflammatory factor expression by activating AhR, decreasing HIF-1 protein expression, and activating the STAT3/IL-10 signaling pathway. DISCUSSION AND CONCLUSION AGM has the potential to ameliorate sepsis-related intestinal injury via the AhR-STAT3-IL-10 pathway, thereby offering theoretical basis for clinical treatment of sepsis.
Collapse
Affiliation(s)
- Junhua Pan
- College of Pharmacy, Hainan Medical University, Haikou, China; School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China; Guangdong Zhaoqing Aviation Vocational College, Zhaoqing, China
| | - Lianfang Gan
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Yaying Chen
- College of Pharmacy, Hainan Medical University, Haikou, China; School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Lifan Zhong
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Huiming Deng
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Ling Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China; Hainan Center for Drug and Medical Device Evaluation and Service, Hainan Medical Products Administration, Haikou, China.
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
31
|
Kumar R, Vats P, Panchal P, Kumar S. Phospholipases: Paving the Way for a New Life. Reprod Sci 2025:10.1007/s43032-025-01900-z. [PMID: 40514594 DOI: 10.1007/s43032-025-01900-z] [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: 07/17/2024] [Accepted: 06/02/2025] [Indexed: 06/16/2025]
Abstract
Phospholipases are a large group of enzymes with ramifications on diverse physiological processes. These enzymes are involved in releasing phospholipids, which are subsequently utilized in the production of prostaglandin and calcium signaling. Calcium signaling is central to the function of phospholipases and calcium oscillations have an impact on the variety of events of embryo development. Additionally, the diverse isoforms of the enzymes can activate gene expression and regulatory factors in a stage-specific manner. Any alterations in the level of these enzymes or the pathways regulating these can have a detrimental effect on the development of the embryo and can also cause early embryonic mortality. In addition to contraction, the enzyme is also critical for the maintenance of quiescence in the uterus during pregnancy. This review discusses the crucial roles of the enzymes, starting from sperm capacitation to the parturition and the putative signaling pathways that are involved in these events. The phospholipases has a significant role in many events in the developmental biology of an animal. Any pathology associated with this enzyme group will have significant consequences on the progression of pregnancy.
Collapse
Affiliation(s)
- Rohit Kumar
- School of Nutrition and Public Health, College of Health, Oregon State University, Corvallis, OR, 97331, USA.
| | - Preeti Vats
- Cell Biology and Proteomics Lab, Animal Biotechnology Centre (ABTC), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Priti Panchal
- Cell Biology and Proteomics Lab, Animal Biotechnology Centre (ABTC), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Sudarshan Kumar
- Cell Biology and Proteomics Lab, Animal Biotechnology Centre (ABTC), ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
32
|
Li S, Zhou X, Yu S, Liu Z, Sun M, Si Z, Zhu W. Macrophage heterogeneity in autoimmune diseases. Cell Immunol 2025; 414:104993. [PMID: 40516246 DOI: 10.1016/j.cellimm.2025.104993] [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: 04/27/2025] [Revised: 06/10/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
The pathogenesis of autoimmune diseases (AIDs) is complex and their etiology remains unclear, with multiple cell types involved in the disease progression. Macrophages, as a crucial immune cell population in AIDs, play a pivotal role in maintaining immune homeostasis. In traditional research, macrophages are frequently oversimplified into the M1 and M2 polarized subtypes. The advent of single-cell RNA sequencing (scRNA-seq) technology has significantly advanced high-throughput research in the life sciences, enabling in-depth investigations at the cellular and molecular levels. This technology has revealed the significant heterogeneity of macrophages, further enhancing our understanding of their development, phenotypic diversity, and functional plasticity. Additionally, it provides a novel perspective for exploring the molecular mechanisms underlying various diseases. In this review, we comprehensively explore the heterogeneity of macrophages across different AIDs, and summarize potential therapeutic targets for macrophage-directed interventions, aiming to provide valuable theoretical insights and novel research directions to advance precision therapy and related studies in AIDs.
Collapse
Affiliation(s)
- Shuaiyi Li
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Xiaohui Zhou
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Shidi Yu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zenghui Liu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Mingshuang Sun
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zihou Si
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Wei Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China.
| |
Collapse
|
33
|
Sahu A, Ruhal R. Immune system dynamics in response to Pseudomonas aeruginosa biofilms. NPJ Biofilms Microbiomes 2025; 11:104. [PMID: 40506442 PMCID: PMC12162861 DOI: 10.1038/s41522-025-00738-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 05/24/2025] [Indexed: 06/16/2025] Open
Abstract
Pseudomonas aeruginosa biofilms contribute to chronic infections by resisting immune attacks and antibiotics. This review explores how innate immunity, including neutrophils, macrophages, and dendritic cells, responds to biofilms and how adaptive mechanisms involving T cells, B cells, and immunoglobulins contribute to infection persistence. Additionally, it highlights immune evasion strategies and discusses emerging therapies such as immunotherapy, monoclonal antibodies, and vaccines, offering insights into enhancing biofilm clearance and improving treatment outcomes.
Collapse
Affiliation(s)
- Abhijeet Sahu
- School of Bio Science and Technology, VIT Vellore, Vellore, Tamil Nadu, India
| | - Rohit Ruhal
- School of Bio Science and Technology, VIT Vellore, Vellore, Tamil Nadu, India.
| |
Collapse
|
34
|
Wang Q, Yu Y, Zhuang J, Liu R, Sun C. Demystifying the cGAS-STING pathway: precision regulation in the tumor immune microenvironment. Mol Cancer 2025; 24:178. [PMID: 40506729 PMCID: PMC12160120 DOI: 10.1186/s12943-025-02380-0] [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: 04/09/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway serves as an immune sentinel for cytosolic DNA, recognizing double-stranded DNA (dsDNA) derived from abnormally localized nuclear DNA or mitochondrial DNA (mtDNA), and plays a pivotal role in innate immune responses and tumor immune surveillance. Conventional antitumor therapies induce genomic instability and mitochondrial stress, leading to the release of nuclear DNA and mtDNA into the cytosol, thereby activating the cGAS-STING pathway. This activation triggers the production of type I interferons (IFN-I) and pro-inflammatory cytokines, which reshape the tumor immune microenvironment (TIME). However, the complexity of TIME reveals a "double-edged sword" effect of cGAS-STING signaling: while it activates antitumor immune responses, it also promotes immune escape and metastasis through the regulation of immunosuppressive cells and stromal components. This review comprehensively delineates the differential regulatory mechanisms of the pathway within TIME constituents, highlighting its multifaceted roles in tumor immunity. Furthermore, it reviews recent advances and challenges in targeting the cGAS-STING pathway for cancer immunotherapy, with the aim of advancing cGAS-STING signaling modulation as a key therapeutic strategy to reprogram TIME and overcome immunosuppression in antitumor treatment.
Collapse
Affiliation(s)
- Qingyang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Yu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China.
| | - Ruijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
35
|
Wang Z, Kulkarni S, Nong J, Zamora M, Ebrahimimojarad A, Hood E, Shuvaeva T, Zaleski M, Gullipalli D, Wolfe E, Espy C, Arguiri E, Wu J, Wang Y, Marcos-Contreras OA, Song W, Muzykantov VR, Fu J, Radhakrishnan R, Myerson JW, Brenner JS. A percolation phase transition controls complement protein coating of surfaces. Cell 2025:S0092-8674(25)00576-8. [PMID: 40516526 DOI: 10.1016/j.cell.2025.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 12/06/2024] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
When a material enters the body, it is immediately attacked by hundreds of proteins, organized into complex networks of binding interactions and reactions. How do such complex systems interact with a material, "deciding" whether to attack? We focus on the complement system of ∼40 blood proteins that bind microbes, nanoparticles, and medical devices, initiating inflammation. We show a sharp threshold for complement activation upon varying a fundamental material parameter, the surface density of potential complement attachment points. This sharp threshold manifests at scales spanning single nanoparticles to macroscale pathologies, shown here for diverse engineered and living materials. Computational models show these behaviors arise from a minimal subnetwork of complement, manifesting percolation-type critical transitions in the complement response. This criticality switch explains the "decision" of a complex signaling network to interact with a material.
Collapse
Affiliation(s)
- Zhicheng Wang
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sahil Kulkarni
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marco Zamora
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alireza Ebrahimimojarad
- Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, NJ 08103, USA
| | - Elizabeth Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Zaleski
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Wolfe
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Carolann Espy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evguenia Arguiri
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jichuan Wu
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yufei Wang
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenchao Song
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jinglin Fu
- Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, NJ 08103, USA
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jacob S Brenner
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Huang Y, Wang X, Li J, Li T, Liu X, Liu J, Xiao J, Wu H, Zhang Y, Feng H. Black carp STAT2 enhances IRF3-mediated antiviral signaling by regulating its ubiquitination and improving its nuclear translocation. FISH & SHELLFISH IMMUNOLOGY 2025; 165:110493. [PMID: 40513786 DOI: 10.1016/j.fsi.2025.110493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/28/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
Interferon (IFN) regulatory factor 3 (IRF3) is a critical transcription factor involved in inducing IFN production. However, the regulatory mechanisms of piscine IRF3 remain inadequately explored. Here, we report that the signal transducer and activator of transcription 2 (STAT2) of black carp (Mylopharyngodon piceus) functions as a positive regulator in the black carp IRF3 (bcIRF3)-mediated IFN signaling pathway. Knockdown of bcSTAT2 in vivo facilitates viral replications, and the mRNA levels of bcSTAT2 gene increase after IFN stimulation in host cells. Additionally, overexpression of bcSTAT2 promotes the activation of interferon stimulated response element (ISRE) ex vivo. Co-immunoprecipitation assays identify the interaction between bcSTAT2 and bcIRF3, and both molecules exhibit similar subcellular distributions in immunofluorescent staining assay. When co-expressed, bcSTAT2 improves the protein level of bcIRF3, and enhances the bcIRF3-mediated IFN production and antiviral capabilities. Mechanistically, we demonstrate that bcSTAT2 significantly enhances the nuclear translocation of bcIRF3. Besides, bcSTAT2 enhances K29-linked ubiquitination, while reduces K33- and K48-linked ubiquitination of bcIRF3. Taken together, our data concludes that bcSTAT2 positively regulates bcIRF3-mediated antiviral activity by regulating its ubiquitination and facilitating its nuclear translocation, which has shed a light on the regulation of IFN signaling.
Collapse
Affiliation(s)
- Yiru Huang
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xinyu Wang
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jinyi Li
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Ting Li
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xiaoyu Liu
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Ji Liu
- College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Xiao
- College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Hui Wu
- College of Life Science, Hunan Normal University, Changsha, 410081, China.
| | - Yong'an Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Hao Feng
- College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
37
|
Hu M, Wang Y, Yin XB. STING-activable immunomodulatory bio-glue for multiple postsurgical management. J Control Release 2025; 382:113714. [PMID: 40210124 DOI: 10.1016/j.jconrel.2025.113714] [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: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
In addition to the robust adhesive properties, there is a pressing demand for ideal adhesives in tumor surgery that possess anti-tumor therapeutic effects. In this study, we introduce BSA-MnO2-GP@Ca-Y (BMGY) bio-glue by integrating bovine serum albumin (BSA)-MnO2, genipin (GP), and Ca-Y zeolite. Ca-Y zeolite exhibits the thrombin activity for hemostasis, while the cross-linking of BSA, GP, and skin tissue induces wound adherence upon laser irradiation for normalized skin structure within nine days. The heat generated during the "photothermal suture" process ablates residual tumor cells and produces antigen fragments, which are internalized by antigen presenting cells. The released Mn ions subsequently activate the cGAS-STING pathway, enhancing immunogenicity. Consequently, tumor-infiltrating p-TBK1 and interferon-β levels are significantly increased, ensuring robust anti-tumor immunity following BMGY treatment. Thus, BMGY bio-glue achieves hemostasis, wound bonding, ablation of residual tumor cells, and tumor recurrence inhibition, simultaneously. Beyond Ca-Y zeolite, BSA-MnO2-GP serves as a versatile platform for loading other drugs or active species to boost therapeutic efficacy. Therefore, we present a successful bio-glue paradigm with significant translational potential for various postsurgical management applications.
Collapse
Affiliation(s)
- Mengzi Hu
- Institute for Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Yaqiong Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Xue-Bo Yin
- Institute for Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|
38
|
Yun JI, Yun SI, Kim JH, Kim DG, Lee DW. Mediation of Osseointegration, Osteoimmunology, and Osteoimmunologic Integration by Tregs and Macrophages: A Narrative Review. Int J Mol Sci 2025; 26:5421. [PMID: 40508228 PMCID: PMC12154945 DOI: 10.3390/ijms26115421] [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: 04/09/2025] [Revised: 05/15/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025] Open
Abstract
Osseointegration is the direct contact between living bone and a dental implant, with supporting evidence confirming the direct connection between bone and titanium, found using an electron microscope. However, the fundamental mechanisms and interconnections between the bone and titanium are not clearly understood. At present, osteoimmunology explores the interaction between bone and immune cells not only in the medical field but also in dentistry. Immunology in bone cell formation has long been a research topic; however, interest in these effects has recently surged. Through subsequent studies, osteoimmune reaction occurs in response to dental implant insertion into the bone and this mechanism portrays more accurate tissue response compared to the traditional term osseointegration. Additionally, osseointegration is a foreign body defense mechanism to protect the implant when bone forms at the contact surface between the dental implant and the alveolar bone. The term "osteoimmunology" refers to the relationship between the immune system and bone tissues. Understanding osteoimmunologic concepts may enable the development of immunomodulatory strategies to improve, maintain, and ultimately restore osseointegration. In order for biocompatible materials such as dental implants to settle and be maintained in the body, it is necessary to understand the complex interrelationships of the bone immune environment, which will enable the development of biomaterials that are more favorable to osteoimmune environments. Therefore, this review presents previous insights into cellular and molecular interactions between bone and the immune system, specifies the roles of T-regulatory cells (Tregs) and macrophages, and demonstrates their potential for translational applications worldwide.
Collapse
Affiliation(s)
- Jong Il Yun
- Dental R&D Center, Zerone Cellvane Inc., Seoul 04363, Republic of Korea; (J.I.Y.); (J.H.K.)
| | - Su In Yun
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Jae Hong Kim
- Dental R&D Center, Zerone Cellvane Inc., Seoul 04363, Republic of Korea; (J.I.Y.); (J.H.K.)
| | - Duk Gyu Kim
- Zerone Cellvane Inc., Cheonan 31116, Republic of Korea;
| | | |
Collapse
|
39
|
Liu M, Li L, Tong H, Kong X, Wang L, Jia X, Wang T, Yu D, Li Y, Wang S. Puerarin exerts an inhibitory effect on inflammatory infiltration in the cardiac tissue of EAM mice by regulation of the TNF-α/CCL2/CCR2 signal pathway. Int Immunopharmacol 2025; 157:114721. [PMID: 40300354 DOI: 10.1016/j.intimp.2025.114721] [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: 10/23/2024] [Revised: 04/09/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
Autoimmune myocarditis can result in dilated cardiomyopathy and heart failure, but effective drugs and clear therapeutic targets are still lacking. Experimental autoimmune myocarditis (EAM) serves as the primary animal model utilized for investigating human myocarditis. Puerarin (PUE), a compound derived from the root of Pueraria lobata, exhibits a broad spectrum of antioxidant and anti-inflammatory effects; nevertheless, its underlying mechanism remains elusive. The findings of this study suggested that PUE may attenuate the infiltration of inflammatory cells into the cardiac tissue by suppressing the secretion of chemokine CCL2 from endothelial cells and macrophages at the site of injury, as well as inhibiting the interaction between CCR2 and CCL2 in recruited inflammatory cells such as macrophages and Th1 cells. In this study, the focus was on investigating the impact of PUE on the chemotactic signal axis TNF-α/CCL2/CCR2. Through the utilization of Small Animal Ultrasound, Real-Time quantitative PCR, Co-Immunoprecipitation (Co-IP), and Immunofluorescence techniques on both cellular and animal models, it has been demonstrated that PUE effectively inhibits the production of CCL2 by disrupting the TNF-α/TNFR signaling pathway in macrophages and endothelial cells through its binding affinity with TNF-α. Additionally, PUE disrupts the transmission of chemotactic signals mediated by CCL2/CCR2 interaction through its binding to CCR2.This ultimately leads to a reduction in the infiltration of inflammatory cells into the heart. Moreover, the study highlights that PUE can effectively inhibit the transduction of the TNF-α/CCL2/CCR2 chemotactic signal, resulting in decreased infiltration of macrophages and Th1 cells in the heart and subsequently reducing inflammatory damage to myocardial tissue in EAM mice.
Collapse
Affiliation(s)
- Meng Liu
- Department of Biochemistry, School of Basic Medical, Qingdao University, Qingdao, China
| | - Ling Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical, Qingdao University, Qingdao, China
| | - Huimin Tong
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical, Qingdao University, Qingdao, China
| | - Xiaowen Kong
- School of Stomatology, Qingdao University, Qingdao 266071, China
| | - Li Wang
- Department of small infants, Qingdao Women and Children's Hospital, Qingdao, China
| | - Xihui Jia
- Department of Biochemistry, School of Basic Medical, Qingdao University, Qingdao, China
| | - Tiantian Wang
- School of Medicine, Qing dao Binhai University, Qingdao, China
| | - Dongyu Yu
- Department of Biochemistry, School of Basic Medical, Qingdao University, Qingdao, China
| | - Yuanyuan Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical, Qingdao University, Qingdao, China
| | - Shuang Wang
- Department of Biochemistry, School of Basic Medical, Qingdao University, Qingdao, China.
| |
Collapse
|
40
|
Islam T, Xu B, Bian Z. Anti-inflammatory and gut microbiota regulatory effects of ultrasonic degraded polysaccharides from Auricularia auricula-judae in DSS-induced colitis mice. ULTRASONICS SONOCHEMISTRY 2025; 117:107339. [PMID: 40215791 PMCID: PMC12008650 DOI: 10.1016/j.ultsonch.2025.107339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/22/2025]
Abstract
Auricularia auricula-judae is a widely cultivated mushroom species known for its edible and medicinal properties. Polysaccharides have been the focus of research because of their potential bioactivities; nonetheless, the structural complexity and molecular weight have hindered a complete understanding of their bioactivities. In this study, AP-1 polysaccharide was isolated from A. auricula-judae and subjected to ultrasonic degradation at different time points to improve their anti-inflammatory effects. The results showed that when AP-1 was degraded for 9 min (AP-2) and 20 min (AP-3), the NO inhibition rate was significantly increased in LPS-stimulated RAW 264.7 cells. The structural and physiochemical properties of native and degraded polysaccharides were analyzed, and it was found that the degradation process significantly reduced molecular weight and altered the particle size, viscosity, crystallinity, and helical structure. Furthermore, native and degraded polysaccharides (AP-1, AP-2, and AP-3) anti-inflammatory effects were investigated in the DSS-induced colitis mouse model. Degraded polysaccharides resulted in significant improvements, including recovery from weight loss, reduced disease activity, shortened colon length, and decreased inflammation, while AP-3 showed the most promising effects. Gut microbiota 16S rRNA sequencing revealed that AP-3 potentially increases healthy gut microbiota and inhibits unhealthy gut microbiota. Overall, this study demonstrates that ultrasonic degradation could be a great technique to modify polysaccharides' MW and physiochemical properties to improve anti-inflammatory and gut microbiota regulatory effects.
Collapse
Affiliation(s)
- Tahidul Islam
- Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, Beijing Normal-Hong Kong Baptist University, China.
| | - Zhaoxiang Bian
- Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
41
|
Feng H, Jin Y, Wu B. Strategies for neoantigen screening and immunogenicity validation in cancer immunotherapy (Review). Int J Oncol 2025; 66:43. [PMID: 40342048 PMCID: PMC12101193 DOI: 10.3892/ijo.2025.5749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer immunotherapy stimulates and enhances antitumor immune responses to eliminate cancer cells. Neoantigens, which originate from specific mutations within tumor cells, are key targets in cancer immunotherapy. Neoantigens manifest as abnormal peptide fragments or protein segments that are uniquely expressed in tumor cells, making them highly immunogenic. As a result, they activate the immune system, particularly T cell‑mediated immune responses, effectively identifying and eliminating tumor cells. Certain tumor‑associated antigens that are abnormally expressed in normal host proteins in cancer cells are promising targets for immunotherapy. Neoantigens derived from mutated proteins in cancer cells offer true cancer specificity and are often highly immunogenic. Furthermore, most neoantigens are unique to each patient, highlighting the need for personalized treatment strategies. The precise identification and screening of neoantigens are key for improving treatment efficacy and developing individualized therapeutic plans. The neoantigen prediction process involves somatic mutation identification, human leukocyte antigen (HLA) typing, peptide processing and peptide‑HLA binding prediction. The present review summarizes the major current methods used for neoantigen screening, available computational tools and the advantages and limitations of various techniques. Additionally, the present review aimed to summarize experimental strategies for validating the immunogenicity of the predicted neoantigens, which will determine whether these neoantigens can effectively trigger immune responses, as well as challenges encountered during neoantigen screening, providing relevant recommendations for the optimization of neoantigen‑based immunotherapy.
Collapse
Affiliation(s)
- Hua Feng
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yuanting Jin
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, P.R. China
| | - Bin Wu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
42
|
Mieda K, Nakanishi T, Kuramoto H, Hosokawa Y, Hosokawa I, Takegawa D, Hosaka K. Sudachitin Reduces Inflammatory Mediator Expression in Toll-Like Receptor 2 Ligand-Stimulated Human Dental Pulp Cells. Cell Biochem Biophys 2025; 83:2431-2439. [PMID: 39739289 DOI: 10.1007/s12013-024-01652-8] [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] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Sudachitin, which is a polymethoxy flavonoid derived from the peer of Citrus sudachi, has several biological properties. However, the effect of sudachitin on human dental pulp cells (HDPCs) remains unclear. The aim of this study was to investigate whether sudachitin could decrease the expression of inflammatory mediators such as cytokines and prostaglandin in HDPCs stimulated with Pam3CSK4, a ligand for toll-like receptor (TLR) 2. HDPCs were pre-incubated with different concentrations of sudachitin (6.25, 12.5, 25, or 50 μM) and stimulated with Pam3CSK4 (100 ng/mL). The quantification of inflammatory cytokines (interleukin (IL)-6, IL-8, and C-X-C motif chemokine ligand (CXCL) 10) and prostaglandin E2 (PGE2) were performed by enzyme-linked immunosorbent assay (ELISA). The expression of cyclooxygenase (COX)-2, a key enzyme for PGE2 formation, was analyzed by western blot. Moreover, the activations of cell signal pathways were examined by western blot analysis. Sudachitin suppressed IL-6, IL-8, CXCL10, and PGE2 production and COX-2 protein expression in Pam3CSK4-stimulated HDPCs. In addition, we revealed that nuclear factor-kappa B (NF-κB) and protein kinase B (Akt) pathways in the Pam3CSK4-stimulated HDPCs were inhibited by sudachitin treatment. These findings suggest that sudachitin can reduce inflammatory mediator production in HDPCs stimulated with TLR2 ligand by inhibiting NF-κB and Akt activations.
Collapse
Affiliation(s)
- Katsuhiro Mieda
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tadashi Nakanishi
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.
| | - Hitomi Kuramoto
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Department of Pediatric Dentistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoshitaka Hosokawa
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ikuko Hosokawa
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Daisuke Takegawa
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keiichi Hosaka
- Department of Regenerative Dental Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
43
|
Palm CL, de Wit S, Gorter TM, Rienstra M, Vos MJ, Kema IP, van der Ley CP, Bakker SJL, Bakker BM, de Boer RA, van Veldhuisen DJ, Meijers WC, Westenbrink BD. Beyond the gut: Systemic levels of short-chain fatty acids are altered in patients with heart failure. Int J Cardiol 2025; 428:133124. [PMID: 40068788 DOI: 10.1016/j.ijcard.2025.133124] [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: 02/06/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND & AIM The gut microbiome produces short-chain fatty acids (SCFAs), which serve as a substantial energy source and provide a link between the microbiome and (cardiac) metabolism. It has been demonstrated that the composition of the microbiome is altered in patients with heart failure (HF), but whether circulating levels of SCFAs are altered in HF is unknown. METHODS & RESULTS Serum concentrations of the SCFAs acetate, propionate, and butyrate were measured in 205 patients with HF and in 54 healthy controls, using isotope dilution liquid chromatography-tandem mass spectrometry. Of the patients with HF, 99 had HF with a reduced ejection fraction (HFrEF) and 106 had HF with mildly-reduced or preserved ejection fraction (HFmrEF/HFpEF). Healthy controls were age and sex matched to the HFrEF patients. Serum concentrations of acetate and propionate were significantly lower in patients with HF than in healthy controls, whereas butyrate levels were higher in patients with HF. Analyses by HF type revealed that acetate and propionate levels were lower in both HFrEF and HFpEF/HFmrEF patients in comparison to healthy controls. However, butyrate levels were observed to be lower in patients with HFmrEF/HFpEF in comparison to healthy controls, while they were higher in patients with HFrEF. CONCLUSIONS In patients with HF, serum levels of acetate and propionate are lower across the HF spectrum, whereas serum butyrate levels are elevated in HFrEF, but lower in HFmrEF/HFpEF. These alterations in SCFA profiles suggest a microbiome-driven metabolic dysregulation, which appears to differ between HF subtypes.
Collapse
Affiliation(s)
- C L Palm
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Institute of Clinical Chemistry and Laboratory Medicine, Oldenburg Clinic, University of Oldenburg, Germany
| | - S de Wit
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - T M Gorter
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M Rienstra
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M J Vos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - I P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - C P van der Ley
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - S J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - B M Bakker
- Laboratory of Pediatrics, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - R A de Boer
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - D J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - W C Meijers
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - B D Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
44
|
Kumar KP, Madhusoodanan M, Pangath M, Menon D. Innovative landscapes in intraperitoneal therapy of ovarian cancer. Drug Deliv Transl Res 2025; 15:1877-1906. [PMID: 39888579 DOI: 10.1007/s13346-024-01765-w] [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: 12/09/2024] [Indexed: 02/01/2025]
Abstract
Epithelial ovarian cancer is the most prevalent gynecological malignancy, characterized by high mortality rates due to its late-stage diagnosis and frequent recurrence. The current standard of care for ovarian cancer is a combination of debulking surgery followed by the conventional mode of chemotherapy. Despite significant advances in therapeutic modalities, the overall survival rate of EOC continues to be poor, mainly because low concentrations of the chemotherapeutics reach the peritoneum, which is the primary site of ovarian cancer, leading to disease relapse. Here, intraperitoneal chemotherapy gains advantage due to its ability to deliver the drug molecules directly to the peritoneal cavity and provide localized and sustained effects. This is facilitated by the use of diverse kinds of nano or micron sized delivery systems, which help in transporting drugs, vaccines, antibodies and genes appropriately to the peritoneum for its desired function. This review article delves on how intraperitoneal delivery impacts the therapy of epithelial ovarian cancer spanning the conventional therapeutic modes to the recent nanoinnovations in chemotherapy, immunotherapy and gene therapy.
Collapse
Affiliation(s)
- Krishna Pradeep Kumar
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Maneesha Madhusoodanan
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Meghna Pangath
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Deepthy Menon
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
45
|
Melkonian AL, Cheung MD, Erman EN, Moore KH, Lever JMP, Jiang Y, Yang Z, Lasseigne BN, Agarwal A, George JF. Single-cell RNA sequencing and spatial transcriptomics reveal unique subpopulations of infiltrating macrophages and dendritic cells following AKI. Am J Physiol Renal Physiol 2025; 328:F907-F920. [PMID: 40331777 DOI: 10.1152/ajprenal.00059.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Kidney infiltrating macrophages (KIMs) and kidney dendritic cells (KDCs) are strongly associated with inflammation and fibrosis in acute kidney injury (AKI) and chronic kidney disease (CKD). Contrary to kidney resident macrophages (KRMs), which are self-renewing and present in the kidney prior to injury, KIMs are bone-marrow derived F4/80int, CD11bhigh macrophages that infiltrate the kidney during AKI. Here, we combined single-cell RNA sequencing (scRNAseq), spatial transcriptomics, and cellular indexing of transcriptomes and epitopes (CITE)-sequencing to elucidate temporal, spatial, and transcriptional characteristics of unique subpopulations of KIMs and KDCs in ischemia-induced AKI. scRNAseq revealed three KIM, two KDC, and one proliferative macrophage subpopulation. All six clusters were localized in unique, spatially constrained microenvironments and their locations were dynamically regulated following bilateral ischemia reperfusion injury. We showed that a specific Arginase 1-expressing KIM cluster infiltrates the kidney cortex at day 1 after ischemia. We also identified a macrophage subpopulation that expresses genes specific to cell proliferation that resides in the cortex in uninjured states and in the medulla at day 6 during the reparative phase of AKI. Gene ontology analysis revealed functional characteristics that distinguish each KIM and KDC population. By day 28 after ischemia, the transcriptional profiles of KIMs upregulate C1q, Cd81, and Cd74, markers normally limited to KRMs in quiescence and early AKI. Since KIMs and KDCs are profoundly involved in AKI, it is paramount that we understand their dynamics-temporally and spatially-and identify their key genes and surface protein markers to develop macrophage-specific therapeutics aimed toward targeting kidney disease.NEW & NOTEWORTHY In this work, we fully characterized both single cell and spatial transcriptomes of kidney infiltrating macrophages (KIMs) and kidney dendritic cells (KDCs) following bilateral ischemia reperfusion injury. We also discovered distinct markers that differentiate KIMs from one another and kidney resident macrophages (KRMs). Finally, we show evidence suggesting that KIMs may reprogram and express genes previously limited to KRMs by day 28 following injury resolution.
Collapse
Affiliation(s)
- Arin L Melkonian
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Matthew D Cheung
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Elise N Erman
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kyle H Moore
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jeremie M P Lever
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Yanlin Jiang
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Zhengqin Yang
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Brittany N Lasseigne
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - James F George
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
46
|
Liu X, Cui L, Dong J, Ren J, Xu D, Han Y, Zhang J. Potential of bone marrow mesenchymal stem cells in iodine-induced autoimmune thyroiditis therapy. Eur Thyroid J 2025; 14:e240137. [PMID: 40435196 PMCID: PMC12164285 DOI: 10.1530/etj-24-0137] [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: 05/05/2024] [Revised: 04/25/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Objective Hashimoto's thyroiditis (HT) is a prevalent autoimmune disease without a cure. Mesenchymal stem cells (MSCs) may offer the opportunity to improve autoimmune thyroiditis. Methods We replicated the pathogenic factors of HT and established a stable autoimmune thyroiditis model in NOD.H-2h4 mice by administering iodine for 12 weeks. We used orthotopic injection to transplant bone MSCs (BMSCs) into the thyroid. Immunohistochemistry, enzyme-linked immunosorbent assay, flow cytometry, and hematoxylin and eosin and immunofluorescence staining were used to evaluate the effects of cell transplantation. Results Orthotopic BMSC transplantation decreased serum thyroglobulin antibody and caspase 3 levels; increased proliferating cell nuclear antigen levels; decreased CD4+/CD3+ T cells, Th1/Th2, and Th17/Treg ratios; decreased TNF-alpha (a proinflammatory cytokine) and interferon-gamma levels; and increased transforming growth factor-beta and interleukin-10 levels. In addition, it increased CD90/S100A4 and CD90/TTF-1 co-expression. Conclusion Orthotopic BMSC transplantation improved the inflammatory environment by regulating the secretion of anti-inflammatory cytokines, promoting regeneration, and reducing apoptosis in the thyroid tissue. Bone marrow-derived stem cells inhibited T cell activation, maintained a balance between T cell subpopulation ratios, and halted thyroiditis progression. Finally, transplanted BMSCs could transform into fibroblasts and thyroid cells. This study elucidated the pathogenesis of HT and provided evidence supporting the potential of MSCs in HT treatments.
Collapse
Affiliation(s)
- Xun Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Likun Cui
- Department of Ultrasound, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianwei Dong
- Department of Vascular & Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jiancong Ren
- Department of Vascular & Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Dongdong Xu
- Department of Vascular & Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yanshuo Han
- Department of Vascular Surgery, Central Hospital of Dalian University of Technology, Dalian, China
- School of Chemical Engineering, Ocean Technology and Life Science, Dalian University of Technology, Panjin, China
| | - Jian Zhang
- Department of Vascular & Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
47
|
Dorscheid D, Gauvreau GM, Georas SN, Hiemstra PS, Varricchi G, Lambrecht BN, Marone G. Airway epithelial cells as drivers of severe asthma pathogenesis. Mucosal Immunol 2025; 18:524-536. [PMID: 40154790 DOI: 10.1016/j.mucimm.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/31/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Our understanding of the airway epithelium's role in driving asthma pathogenesis has evolved over time. From being regarded primarily as a physical barrier that could be damaged via inflammation, the epithelium is now known to actively contribute to asthma development through interactions with the immune system. The airway epithelium contains multiple cell types with specialized functions spanning barrier action, mucociliary clearance, immune cell recruitment, and maintenance of tissue homeostasis. Environmental insults may cause direct or indirect injury to the epithelium leading to impaired barrier function, epithelial remodelling, and increased release of inflammatory mediators. In severe asthma, the epithelial barrier repair process is inhibited and the response to insults is exaggerated, driving downstream inflammation. Genetic and epigenetic mechanisms also maintain dysregulation of the epithelial barrier, adding to disease chronicity. Here, we review the role of the airway epithelium in severe asthma and how targeting the epithelium can contribute to asthma treatment.
Collapse
Affiliation(s)
- Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gail M Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| | - Bart N Lambrecht
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium.
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy.
| |
Collapse
|
48
|
Hu Y, Tuo B. The function of chloride channels in digestive system disease (Review). Int J Mol Med 2025; 55:99. [PMID: 40314091 PMCID: PMC12045473 DOI: 10.3892/ijmm.2025.5540] [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: 12/16/2024] [Accepted: 03/11/2025] [Indexed: 05/03/2025] Open
Abstract
Cation channels have been extensively studied in the context of digestive disorders, but comparatively little attention has been given to anions and their associated channels. Chloride ions, the most abundant anions in the human body, act as signaling molecules, modulating cellular behavior and playing a key role in regulating multiorgan physiological and pathophysiological mechanisms. The intra‑ and extracellular distributions of chloride ions are primarily controlled by various chloride channels and transporters. Currently, these chloride channels are classified into several groups: The chloride channels family, cystic fibrosis transmembrane conductance regulator, calcium‑activated chloride channels, volume‑regulated anion channels, proton‑activated chloride channels and ligand‑gated anion channels. This review aims to summarize the roles of chloride ion channels and transporter proteins in digestive system diseases, providing a theoretical basis for future research and offering potential new strategies for disease treatment.
Collapse
Affiliation(s)
- Yanxia Hu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
49
|
Yu H, Resch W, Moss B. Poxvirus structural biology for application to vaccine design. Trends Immunol 2025; 46:455-470. [PMID: 40340168 DOI: 10.1016/j.it.2025.04.002] [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: 02/17/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 05/10/2025]
Abstract
The upsurge of mpox (formerly known as monkeypox) in Africa and its global spread highlight the need for improved vaccines. The development of new recombinant vaccines, including mRNA and protein nanoparticles, depends on understanding the biology of poxviruses and selecting the most protective immunogens. Animal studies demonstrate that vaccines need to target the antigens of both infectious forms - the mature virion and the enveloped virion - which display surface proteins responsible for cell entry and cell-to-cell spread, respectively. Although some of these proteins have been shown to induce protective antibodies, others including most of those that are essential for membrane fusion remain to be tested. We review the structures of orthopoxvirus surface proteins as a guide to the selection of optimal antigens for recombinant vaccines.
Collapse
Affiliation(s)
- Huibin Yu
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Wolfgang Resch
- Center for Information Technology, NIH, Bethesda, MD, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
50
|
Pereda J, Espinosa R, García-Solís B, Guerra-Galán T, Van-Den-Rym A, Kars M, Mena R, Galán V, de Andrés-Martín A, Rodríguez-Gallego C, López-Lera A, Corvillo F, Pérez-Martínez A, López-Collazo E, Sánchez-Ramón S, Martínez-Barricarte R, Quintana-Murci L, Lorenzo-Salazar J, Itan Y, Flores C, Pérez-de-Diego R. IEIVariantFilter: a bioinformatics tool to speed up genetic diagnosis of inborn errors of immunity patients. NAR Genom Bioinform 2025; 7:lqaf069. [PMID: 40438610 PMCID: PMC12117399 DOI: 10.1093/nargab/lqaf069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/25/2025] [Accepted: 05/14/2025] [Indexed: 06/01/2025] Open
Abstract
Severe infectious diseases remain the leading cause of death in children and young adults worldwide. Monogenic inborn errors of immunity (IEIs) are traditionally defined as a heterogeneous group of rare inborn genetic diseases affecting the functioning of the immune system. Greater awareness has led to the clinical definition of 485 monogenic IEIs and whole exome sequencing (WES) is becoming increasingly relevant for IEI genetic diagnosis. The current protocol for IEI genetic studies includes manual filtering of the list of genes obtained as a WES read-out providing a short list of candidate genes. This procedure is time-consuming and can produce mistakes due to human error in manual filtering. IEIVariantFilter is a new web-based bioinformatics tool to speed up and refine the genetic diagnosis of IEI patients oriented for users in the biomedical field without needing bioinformatics expertise. IEIVariantFilter prioritizes genetic variants based on ranges of zygosity, the quality of reads, the predicted variant effect, and genes related to immunity, considering a consanguineous hypothesis whenever necessary. IEIVariantFilter facilitates gene and variant list prioritization, speeding up the identification of candidate disease-causing variants for validation by experimental studies. The software improves the genetic diagnosis of patients, thereby facilitating precision medicine and fast and proper treatment.
Collapse
Affiliation(s)
| | | | - Blanca García-Solís
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Teresa Guerra-Galán
- Clinical Immunology Department, San Carlos Clinical Hospital, Madrid 28040, Spain
| | - Ana Van-Den-Rym
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Meltem Ece Kars
- The Charles Bronfman Institute for Personalized Medicine and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Rocío Mena
- Institute of Medical & Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid 28046, Spain
| | - Victor Galán
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
| | | | - Carlos Rodríguez-Gallego
- Department of Immunology, University Hospital of Gran Canaria Dr. Negrin, Las Palmas de Gran Canaria 35010, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical and Surgical Sciences, School of Medicine, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria 35016, Spain
- Department of Clinical Sciences, University Fernando Pessoa Canarias, Las Palmas de Gran Canaria 35450, Spain
| | - Alberto López-Lera
- IdiPAZ Institute for Health Research, La Paz University Hospital, CIBERER U-754, Madrid 28046, Spain
| | - Fernando Corvillo
- IdiPAZ Institute for Health Research, La Paz University Hospital, CIBERER U-754, Madrid 28046, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
| | - Eduardo López-Collazo
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
| | - Silvia Sánchez-Ramón
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
- Clinical Immunology Department, San Carlos Clinical Hospital, Madrid 28040, Spain
| | - Rubén Martínez-Barricarte
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Lluis Quintana-Murci
- Unit of Human Evolutionary Genetics, Institut Pasteur and CNRS URA3012, Paris 75015, France
| | - José Miguel Lorenzo-Salazar
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), Santa Cruz de Tenerife 38600, Spain
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Carlos Flores
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Department of Clinical Sciences, University Fernando Pessoa Canarias, Las Palmas de Gran Canaria 35450, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), Santa Cruz de Tenerife 38600, Spain
- Research Unit, Hospital Universitario Ntra. Sra. de Candelaria, Instituto de Investigación Sanitaria de Canarias (IISC), Santa Cruz de Tenerife 38010, Spain
| | - Rebeca Pérez-de-Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, Madrid 28046, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|