1
|
Chuang CC, Chen CC, Chen XA, Lee CY, Chang YH, Tsai MH, Young JJ, Chuang CC. Enhancing the in vivo efficacy of anthrax vaccine using trimethylchitosan covalently coated chitosomes in a single-step microfluidic synthesis. Int J Biol Macromol 2025; 304:140689. [PMID: 39914530 DOI: 10.1016/j.ijbiomac.2025.140689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Liposomal vaccines have been developed extensively in recent years, and considerable progress has been made in the prevention of infectious diseases. Quaternary chitosan-coated liposomes were used in the present study. Liposomes coated with TMC (namely chitosomes) were synthesized in a single microfluidic step in which liposomes were formed and coated with trimethylchitosan (TMC) via covalent and/or hydrogen bonds in situ. The endocytosis of chitosomes by JAWS II dendritic cells was detected using flow cytometry. Chitosomes covalently coated with TMC were more efficiently internalized than uncoated liposomes, showed lower cytotoxicity than those coated via hydrogen bonds, and could significantly lower the cytotoxicity via protein mixing. In a BALB/c model, covalent modification of chitosomes improved the efficiency of anti-PA IgG induction via anthrax vaccine adsorbed (AnV), compared with those with hydrogen bonds and those with AnV only, where humoral immunity is dominant. These results suggest that covalently TMC-coated chitosomes serve as a promising delivery system for proteins and that they can be reproducibly obtained via rapid, cost-efficient, and automated methods with few scale-up barriers; meanwhile, their versatile practicability favors commercialization. STATEMENT OF SIGNIFICANCE: In this study, we proved that liposomes coated with TMC (namely chitosomes) via covalent bonds induced better uptake efficiency, stronger in vivo immune responses, and lower cytotoxicity than those coated via hydrogen bonds. The endocytosis efficiency and cytotoxicity of chitosomes were positively correlated with the concentration of the TMC coating. Additionally, this invention provides a facile single-step microfluidic process for the rapid, low-cost, and high-quality preparation of an innovative high-performance chitosomal delivery system that is convenient and easy to scale up. Based on these benefits, the current disclosure provides an alternative platform for the development of new chitosomal drugs.
Collapse
Affiliation(s)
- Chuan-Chung Chuang
- School of Dentistry and Graduate Institute of Dental Science, National Defense Medical Center, Taipei City 11490, Taiwan, ROC; Department of Dentistry, Tri-Service General Hospital, Taipei City 11490, Taiwan, ROC
| | - Cheng-Cheung Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City 11490, Taiwan, ROC
| | - Xin-An Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Chia-Ying Lee
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Yu-Hsiu Chang
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Meng-Hung Tsai
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City 11490, Taiwan, ROC
| | - Jenn-Jong Young
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC.
| | - Chuan-Chang Chuang
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC; Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei City 11490, Taiwan, ROC.
| |
Collapse
|
2
|
Lu L, Feng J, Zhang S, He H, Hu Z, Yang L, Liu Y, Zhao B, Wang T. Vitiligo associated with type 2 immune inhibitors: FAERS analysis and literature review. J Dermatol 2025. [PMID: 40087891 DOI: 10.1111/1346-8138.17698] [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: 11/02/2024] [Revised: 02/10/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
With the widespread use of type 2 immune response inhibitors (IRIs), there is growing concern about their association with the occurrence of vitiligo. This study aimed to comprehensively search for cases of vitiligo associated with type 2 IRIs in the US Food and Drug Administration Adverse Event Reporting System (FAERS). We retrieved the clinical characteristics of cases from January 2004 to September 2024 from the FAERS database. Disproportionality and Bayesian analyses were conducted to detect signals for vitiligo associated with type 2 IRIs. A total of 86 cases of vitiligo were identified in association with these inhibitors. The mean onset time was 326 days. Vitiligo associated with dupilumab was the most common (81.4%), with the highest reporting odds ratio (2.67, 95% confidence interval 2.11-3.4), proportional reporting ratio (2.67, χ2 = 70.59), information component (1.38, [IC025 = 1.09), and empirical Bayes geometric mean (2.61, EBGM05 = 2.14). The link between vitiligo and type 2 IRIs underscores the need for continued pharmacovigilance to better understand these drugs and the incidence of related conditions.
Collapse
Affiliation(s)
- Lu Lu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Jindi Feng
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Shiyu Zhang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Huimin He
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Zhonghui Hu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Lu Yang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yuehua Liu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Bin Zhao
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- Department of Pharmacy, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, SAR, China
| | - Tao Wang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| |
Collapse
|
3
|
Wang X, Sarangi V, Wickland DP, Li S, Chen D, Aubrey Thompson E, Jenkinson G, Asmann YW. Identification of Gene Regulatory Networks Associated with Breast Cancer Patient Survival Using an Interpretable Deep Neural Network Model. EXPERT SYSTEMS WITH APPLICATIONS 2025; 262:125632. [PMID: 39676894 PMCID: PMC11643596 DOI: 10.1016/j.eswa.2024.125632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Artificial neural networks have recently gained significant attention in biomedical research. However, their utility in survival analysis still faces many challenges. In addition to designing models for high accuracy, it is essential to optimize models that provide biologically meaningful insights. With these considerations in mind, we developed a deep neural network model, MaskedNet, to identify genes and pathways whose expression at the time of diagnosis is associated with overall survival. MaskedNet was trained using TCGA breast cancer transcriptome and clinical data, and the model's final output was the predicted logarithm of the hazard ratio for death. The trained model was interpreted using SHapley Additive exPlanations (SHAP), a technique grounded in robust mathematical principles that assigns importance scores to input features. Compared to traditional Cox proportional hazards regression, MaskedNet had higher accuracy, as measured by Harrell's C-index. We also found that aggregating outputs from several model runs identified multiple genes and pathways associated with overall survival, including IFNG and PIK3CA genes, along with their related pathways. To further elucidate the role of the IFNG gene, tumors were partitioned into two groups based on low and high IFNG SHAP values, respectively. Tumors with lower IFNG SHAP values exhibited higher IFNG expression and better overall survival, which were linked to more abundant presence of M1 macrophages and activated CD4+ and CD8+ T cells in the tumor microenvironment. The association of the IFNG pathway with overall survival was validated in the trastuzumab arm of the NCCTG-N9831 trial, an independent breast cancer study.
Collapse
Affiliation(s)
- Xue Wang
- Department of Quantitative Health Sciences, Mayo Clinic, 4500 San Pablo Rd. S., Jacksonville, FL, USA, 32224
| | - Vivekananda Sarangi
- Department of Quantitative Health Sciences, Mayo Clinic, 200 1st St SW, Rochester, MN, USA, 55905
| | - Daniel P. Wickland
- Department of Quantitative Health Sciences, Mayo Clinic, 4500 San Pablo Rd. S., Jacksonville, FL, USA, 32224
| | - Shaoyu Li
- Mathematics & Statistics Department, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, USA, 28223
| | - Duan Chen
- Mathematics & Statistics Department, University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte, NC, USA, 28223
| | - E. Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Rd. S., Jacksonville, FL, USA, 32224
| | - Garrett Jenkinson
- Department of Quantitative Health Sciences, Mayo Clinic, 200 1st St SW, Rochester, MN, USA, 55905
- Department of Data Science, Johnson & Johnson Innovative Medicine R&D, 1400 McKean Rd, Springhouse, PA 19477
| | - Yan W. Asmann
- Department of Quantitative Health Sciences, Mayo Clinic, 4500 San Pablo Rd. S., Jacksonville, FL, USA, 32224
| |
Collapse
|
4
|
Deng M, Zhao R, Zou H, Guan R, Wang J, Lee C, He B, Zhou J, Li S, Wei W, Cai H, Guo R. Oxaliplatin induces pyroptosis in hepatoma cells and enhances antitumor immunity against hepatocellular carcinoma. Br J Cancer 2025; 132:371-383. [PMID: 39748129 PMCID: PMC11832738 DOI: 10.1038/s41416-024-02908-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 11/03/2024] [Accepted: 11/08/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Pyroptosis is closely associated with chemotherapeutic drugs and immune response. Here, we investigated whether oxaliplatin, a key drug in FOLFOX-hepatic artery infusion chemotherapy (FOLFOX-HAIC), induces pyroptosis in hepatoma cells and enhances antitumor immunity after tumor cell death. METHODS Hepatoma cells were treated with oxaliplatin. Pyroptosis and immunoreactivity were evaluated in vitro and in vivo. RESULTS Oxaliplatin activated caspase-3-mediated gasdermin E (GSDME) cleavage and induced pyroptosis in Hep G2 and SK-Hep-1 cells in vitro. Liver cancer cells with high levels of GSDME expression are prone to pyroptosis. Bioinformatic analysis revealed that pyrolysis-related genes are closely related to immunity. In vivo experiments revealed that oxaliplatin exhibited superior antitumor efficacy in mice with normal immune function and more pronounced inhibitory effect on hepatocellular carcinoma with high GSDME levels. Higher levels of cytokines and greater CD8+ T cell infiltration were observed in tumor tissues with better efficacy. Furthermore, an in vitro coculture assay confirmed that oxaliplatin-induced pyroptosis in Hep G2 cells overexpressing GSDME and activated the p38/MAPK signaling pathway to improve the cytotoxicity of CD8+ T cells. Analysis of clinical samples of HCC suggested that the efficacy of FOLFOX-HAIC in patients with high GSDME expression was better than that in patients with low GSDME expression. CONCLUSIONS Oxaliplatin induced pyroptosis in hepatoma cells by activating caspase-3-mediated cleavage of GSDME, which enhanced the cytotoxicity of CD8+ T cells by regulating the p38/MAPK signaling pathway. These results suggest that GSDME level may be used as a marker to predict the efficacy of FOLFOX-HAIC.
Collapse
Affiliation(s)
- Min Deng
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of General Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Rongce Zhao
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hao Zou
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Renguo Guan
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiongliang Wang
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Carol Lee
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Benyi He
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shaohua Li
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wei Wei
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hao Cai
- Department of General Surgery, Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rongping Guo
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
5
|
Roy P, Sharma S, Baranwal M. Computational Insight in the Identification of Non-Synonymous Single-Nucleotide Polymorphism Affecting the Structure and Function of Interleukin-4. Proteomics Clin Appl 2025; 19:e202400070. [PMID: 39648289 DOI: 10.1002/prca.202400070] [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: 07/13/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND IL4 is a versatile cytokine essentially known for differentiation, proliferation and cell death in cells. Its dysregulation has been found to be associated with the development of inflammatory disorders. OBJECTIVE The goal of the current investigation is to identify and select non-synonymous single-nucleotide polymorphisms (nsSNPs) in the IL-4 gene by employing computational methods which may have a potential functional impact on the occurrence of disease. METHOD AND RESULT Six different nsSNPs were predicted to be deleterious based on the consensus of different algorithms: SIFT, Polyphen2 (Humdiv and HumVar), PredictSNP and SNP&GO. I-mutant and MuPro assessment revealed a decrease in the stability of these mutants except K150M. Modelling was then carried out to build the wild type along with its mutants, followed by superimposition of the wild type with mutants to evaluate the RMSD value, which lies between 0.26 and 0.34. Simulation results of mutant models, along with wild type, showed that four of the mutants (N113Y, A118G, R109W and K150M) deviated most and were unstable. A118G showed a significant deviation from the wild type, while V53A and C123R were stable. CONCLUSION The finding establishes the evidence that the identified six nsSNPs of IL-4 can be the new entrant presenting their candidature for genetic testing.
Collapse
Affiliation(s)
- Pratima Roy
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| |
Collapse
|
6
|
Wu Q, Chen K, Xue W, Wang G, Yang Y, Li S, Xia N, Chen Y. An insect cell-derived extracellular vesicle-based gB vaccine elicits robust adaptive immune responses against Epstein-Barr virus. SCIENCE CHINA. LIFE SCIENCES 2025; 68:734-745. [PMID: 39499444 DOI: 10.1007/s11427-023-2599-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/19/2024] [Indexed: 11/07/2024]
Abstract
Epstein-Barr virus (EBV), the first identified human tumor virus, is implicated in various human malignancies, infectious mononucleosis, and more recently, multiple sclerosis. Prophylactic vaccines have the potential to effectively prevent EBV infection. Glycoprotein B (gB) serves as the fusogen and plays a pivotal role in the virus entry process, making it a critical target for EBV vaccine development. Surface membrane proteins of enveloped viruses serve as native conformational antigens, making them susceptible to immune recognition. Utilizing lipid membrane-bound viral antigens is a promising strategy for effective vaccine presentation in this context. In this study, we employed a truncated design for gB proteins, observing that these truncated gB proteins prompted a substantial release of extracellular vesicles (EVs) in insect cells. We verified that EVs exhibited abundant gB proteins, displaying the typical virus particle morphology and extracellular vesicle characteristics. gB EVs demonstrated a more efficient humoral and cellular immune response compared with the gB ectodomain trimer vaccine in mice. Moreover, the antisera induced by the gB EVs vaccine exhibited robust antibody-dependent cytotoxicity. Consequently, gB EVs-based vaccines hold significant potential for preventing EBV infection and offer valuable insights for vaccine design.
Collapse
Affiliation(s)
- Qian Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Kaiyun Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Wenhui Xue
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Guosong Wang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Yanbo Yang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| | - Yixin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Life Sciences, School of Public Health, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
7
|
Ghajar-Rahimi G, Yusuf N, Xu H. Ultraviolet Radiation-Induced Tolerogenic Dendritic Cells in Skin: Insights and Mechanisms. Cells 2025; 14:308. [PMID: 39996779 PMCID: PMC11854269 DOI: 10.3390/cells14040308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
Ultraviolet (UV) radiation has profound effects on the immune system, including the induction of tolerogenic dendritic cells (DCs), which contribute to immune suppression and tolerance. This review explores the roles of conventional CD11c⁺ DCs, as well as cutaneous Langerhans cells and CD11b⁺ myeloid cells, in UV-induced immune modulation. Two key mechanisms underlying the immunosuppressive relationship between UV and DCs are discussed: the inactivation of DCs and the induction of tolerogenic DCs. DCs serve as a critical link between the innate and adaptive immune systems, serving as professional antigen-presenting cells. In this context, we explore how UV-induced DCs influence the activity of specific T cell subsets, including regulatory T lymphocytes and T helper cells, and shape immune outcomes. Finally, we highlight the implications of UV-induced tolerogenic DCs in select dermatologic pathologies, including cutaneous lupus, polymorphic light eruption, and skin cancer. Understanding the mechanisms by which UV radiation alters DC function offers insights into the complex interplay between environmental factors and immune regulation, providing potential avenues for preventive and therapeutic intervention in UV-induced skin diseases.
Collapse
Affiliation(s)
| | - Nabiha Yusuf
- Department of Dermatology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | - Hui Xu
- Department of Dermatology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| |
Collapse
|
8
|
Lantz AM, Baxter VK. Neuropathogenesis of Old World Alphaviruses: Considerations for the Development of Medical Countermeasures. Viruses 2025; 17:261. [PMID: 40007016 PMCID: PMC11860675 DOI: 10.3390/v17020261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Chikungunya virus (CHIKV) and other alphaviruses that primarily induce arthritogenic disease in humans, known as "Old World" alphaviruses, present an emerging public health concern as geographic ranges of mosquito vectors expand due to climate change. While a vaccine against CHIKV has recently been approved by several countries in North America and Europe, access to effective preventative countermeasures against disease induced by Old World alphaviruses remains elusive for the most vulnerable populations. Furthermore, treatment options continue to be limited to supportive care. Atypical neurological disease manifestations caused by Old World alphaviruses, which make up as many as 25% of the cases in some CHIKV outbreaks, present special challenges when considering strategies for developing effective countermeasures. This review focuses on Old World alphaviruses, specifically CHIKV, Ross River virus, O'nyoug-nyoug virus, and Mayaro virus, concentrating on the atypical neurological disease manifestations they may cause. Our current understanding of Old World alphavirus neuropathogenesis, gained from human cases and preclinical animal models, is discussed, including viral and host factors' roles in disease development. The current state of alphavirus preventatives and treatments, both virus-targeting and host-directed therapies, is then summarized and discussed in the context of addressing neurological disease induced by Old World alphaviruses.
Collapse
|
9
|
Panda S, Swain SK, Sahu BP, Mahapatra SR, Dey J, Sarangi R, Ranade AV, Mishra N. Designing a potent multivalent epitope vaccine candidate against Orientia tsutsugamushi via reverse vaccinology technique - bioinformatics and immunoinformatic approach. Front Immunol 2025; 16:1513245. [PMID: 40018038 PMCID: PMC11865050 DOI: 10.3389/fimmu.2025.1513245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/02/2025] [Indexed: 03/01/2025] Open
Abstract
Scrub typhus is a life-threatening, undifferentiated febrile illness caused by a gram-negative bacterium, Orientia tsutsugamushi. The bacterial strain is a global health concern that should be considered. Despite several years of effort for the development of an effective immunogenic vaccine, no successful licensed vaccine is available. The aim of the study is to construct an epitope response using a reverse vaccinology approach. The TSA56 and ScaA proteins combined can be the most promising subunit vaccine candidates against O. tsutsugamushi. B-cell, CTL, and HTL epitopes were predicted, and subsequently, all the epitopes were linked by KK, AAY, and GPGPG linkers, respectively, along with an adjuvant at the N-terminal region. Furthermore, molecular docking and MD simulations were performed that exhibited a higher affinity towards TLR-2. A total of 16 linear B-cells, 6 CTL, and 2 HTL epitopes were identified and validated. The final vaccine construct showed high antigenicity, stability, and solubility. Molecular docking and MD simulations indicated strong binding interactions with TLR-2 and a stable vaccine-receptor complex. The expression of the vaccine in pET28a (+) vector was successfully implemented via in silico cloning as well as significant results from immune simulation demonstrated the efficacy of the vaccine in the immune cell interaction during the innate and adaptive immune responses immune simulation. In conclusion, the outcome suggested that the newly developed vaccine will be a promising candidate for controlling and providing definitive preventive measures against scrub typhus if further investigation is conducted experimentally.
Collapse
Affiliation(s)
- Subhasmita Panda
- Institute of Medical Sciences and SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, India
| | - Subrat Kumar Swain
- Institute of Medical Sciences and SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, India
| | - Basanta Pravas Sahu
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Soumya Ranjan Mahapatra
- School of Biotechnology, Kalinga Institute of Information and Technology (KIIT) University, Bhubaneswar, India
| | - Jyotirmayee Dey
- School of Biotechnology, Kalinga Institute of Information and Technology (KIIT) University, Bhubaneswar, India
| | - Rachita Sarangi
- Institute of Medical Sciences and SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, India
| | - Anu Vinod Ranade
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Namrata Mishra
- School of Biotechnology, Kalinga Institute of Information and Technology (KIIT) University, Bhubaneswar, India
| |
Collapse
|
10
|
Ramirez GA, Cardamone C, Lettieri S, Fredi M, Mormile I. Clinical and Pathophysiological Tangles Between Allergy and Autoimmunity: Deconstructing an Old Dichotomic Paradigm. Clin Rev Allergy Immunol 2025; 68:13. [PMID: 39932658 DOI: 10.1007/s12016-024-09020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2024] [Indexed: 02/14/2025]
Abstract
Allergic and autoimmune disorders are characterised by dysregulation of the immune responses to otherwise inert environmental substances and autoantigens, leading to inflammation and tissue damage. Their incidence has constantly increased in the last decades, and their co-occurrence defies current standards in patient care. For years, allergy and autoimmunity have been considered opposite conditions, with IgE and Th2 lymphocytes cascade driving canonical allergic manifestations and Th1/Th17-related pathways accounting for autoimmunity. Conversely, growing evidence suggests that these conditions not only share some common inciting triggers but also are subtended by overlapping pathogenic pathways. Permissive genetic backgrounds, along with epithelial barrier damage and changes in the microbiome, are now appreciated as common risk factors for both allergy and autoimmunity. Eosinophils and mast cells, along with autoreactive IgE, are emerging players in triggering and sustaining autoimmunity, while pharmacological modulation of B cells and Th17 responses has provided novel clues to the pathophysiology of allergy. By combining clinical and therapeutic evidence with data from mechanistic studies, this review provides a state-of-the-art update on the complex interplay between allergy and autoimmunity, deconstructing old dichotomic paradigms and offering potential clues for future research.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Chiara Cardamone
- Immunorheumatology Unit, University Hospital "San Giovanni Di Dio E Ruggi d'Aragona", Largo Città d'Ippocrate, Via San Leonardo 1, 84131, Salerno, Italy.
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.
| | - Sara Lettieri
- Pulmonology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Mormile
- Division of Internal Medicine and Clinical Immunology, Department of Internal Medicine and Clinical Complexity, AOU Federico II, Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
11
|
Corripio-Miyar Y, Hayward AD, Lemon H, Bal X, Cunnea C, Kenyon F, Pilkington JG, Pemberton JM, Nussey DH, McNeilly TN. T-helper cell phenotypes are repeatable, positively correlated, and associated with helminth infection in wild Soay sheep. DISCOVERY IMMUNOLOGY 2025; 4:kyae017. [PMID: 39963298 PMCID: PMC11832277 DOI: 10.1093/discim/kyae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/04/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Background T-helper (Th) cells co-ordinate immune responses to ensure that infections with diverse parasites are controlled effectively. Helminth parasites such as gastrointestinal nematodes (GIN) are generally associated with T-helper type 2 (Th2) responses, while intracellular parasites are associated with Th1 responses. Although laboratory models have reported that Th1 and Th2 can be antagonistic, this has been challenged by studies of natural infections. Methods Between 2019 and 2022 we completed 759 captures of 538 wild Soay sheep (1-4 captures per animal) and monitored body weight, parasite egg counts, Th phenotypes, cytokines, and GIN-specific antibodies. Results While different Th cell counts, cytokines and antibody isotypes were generally positively correlated with each other, no strong positive associations were observed between these measurements. Cell counts had low repeatability (among-individual variation) across 4 years, while antibody levels were highly repeatable. The Th1 and Th2 cytokines Interferon-gamma (IFN-γ) and Interleukin-4 (IL-4) were moderately repeatable and were positively correlated at both the between- and within-individual levels independent of body condition or parasite exposure. IL-4 was negatively associated with GIN faecal egg count, while IFN-γ was negatively associated with coccidian faecal oocyst count, suggesting that these cytokines reflect resistance to these parasites. None of our immune markers were strongly associated with lamb survival. Conclusions Our results provide insights into how different aspects of immune function interact to produce effective responses to complex infections but suggest longer-term data collection is required to address the causes of these interactions and to detect fitness consequences of variation in T cell phenotypes under natural conditions.
Collapse
Affiliation(s)
| | - Adam D Hayward
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, UK
| | - Hannah Lemon
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
- Department of Biology, University of Oxford, Oxford, UK
| | - Xavier Bal
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
| | - Cameron Cunnea
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, UK
| | - Fiona Kenyon
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, UK
| | - Jill G Pilkington
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
| | - Josephine M Pemberton
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
| | - Daniel H Nussey
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
| | - Tom N McNeilly
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian, UK
| |
Collapse
|
12
|
Kar S, Verma D, Mehrotra S, Prajapati VK. Reconfiguring the immune system to target cancer: Therapies based on T cells, cytokines, and vaccines. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:77-150. [PMID: 39978976 DOI: 10.1016/bs.apcsb.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Over the years, extensive research has been dedicated to performing in-depth analysis of cancer to uncover the intricate details of its nature - including the types of cancer, causative agents, stimulators of disease progression, factors contributing to poor prognosis, and efficient therapies to restrict the metastatic aggressiveness. This chapter highlights the mechanisms through which different arms of the host immune system - namely cytokines, lymphocytes, antigen-presenting cells (APCs) -can be mobilized to eradicate cancer. Most malignant tumors are either poorly immunogenic, or are harbored in a highly immuno-suppressive microenvironment. This is why reinforcing the host's anti-tumor defenses, through infusion of pro-inflammatory cytokines, tumor antigen-loaded APCs, and anti-tumor cytotoxic cells has emerged as a viable treatment option against cancer. The chapter also highlights the ongoing preclinical and clinical studies in different malignancies and the outcome of various therapies. Although these methods are not foolproof, and antigen escape variants can still evade or develop resistance to customized therapies, they achieve disease stabilization in several cases when conventional treatments fail. In many instances, combination therapies involving cytokines, T cells, and vaccinations prove more effective than monotherapies. The limitations of the current therapies are also discussed, along with ongoing modifications aimed at improving efficacy.
Collapse
Affiliation(s)
- Sramona Kar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Divya Verma
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
13
|
Yan W, Cao Y, Xu S, Li Y, Wu T, Yuan W, Yin Q, Li Y. Personalized Multi-Epitope Nanovaccine Unlocks B Cell-Mediated Multiple Pathways of Antitumor Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411361. [PMID: 39711226 DOI: 10.1002/adma.202411361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/13/2024] [Indexed: 12/24/2024]
Abstract
B lymphocytes have emerged as an important immune-regulating target. Inoculation with tumor cell membrane-derived vaccines is a promising strategy to activate B cells, yet their efficiency is limited due to lack of costimulatory molecules. To amplify B cell responses against tumor, herein, a spatiotemporally-synchronized antigen-adjuvant integrated nanovaccine, termed as CM-CpG-aCD40, is constructed by conjugating the immune stimulative CpG oligonucleotide and the anti-CD40 antibody (aCD40) onto the membrane vesicles derived from triple negative breast cancer cells. CM-CpG-aCD40 actively accumulates in lymph nodes and is effectively captured by antigen-presenting cells via the recognition of CD40 by aCD40. Tumor antigens on CM-CpG-aCD40 bind to B cell receptors, providing the first stimulation signal for B cells. Meanwhile, the interaction between CpG/Toll like receptor and aCD40/CD40 provides superposed co-stimulation signals, improving the antibody-secreting and antigen-presenting abilities of B cells. The nanovaccine also stimulates dendritic cells to activate CD8+ T cells, and reprograms tumor associated macrophages. CM-CpG-aCD40 activating humoral, cellular, and innate antitumor immunity achieves a tumor inhibition rate of 89.3%, which is further improved to 95.4% when combined with the anti-programmed death ligand 1 (PD-L1) antibody. CM-CpG-aCD40, as a personalized multi-epitope nanovaccine, paves the way for ushering the era of B cell-based immunotherapy.
Collapse
Affiliation(s)
- Wenlu Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Shanshan Xu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ting Wu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China
| | - Wenhui Yuan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264000, China
| |
Collapse
|
14
|
Tokura Y, Yunoki M, Kondo S, Otsuka M. What is "eczema"? J Dermatol 2025; 52:192-203. [PMID: 39301836 PMCID: PMC11807370 DOI: 10.1111/1346-8138.17439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/22/2024]
Abstract
Eczema is the most common category of inflammatory skin disorders as dermatologists see many patients with eczematous diseases in daily practice. It is characterized by the three major morphological features: multiple-pinpoint condition, polymorphism, and itch. To describe polymorphism, "eczema triangle" has been used in German/Japanese dermatology. The multiple pinpoints correspond to numerous tiny foci from which individual papules/vesicles arise. The polymorphism betrays composition of erythema, papule, seropapule, vesicle, pustule, scale, and crust, which are seen in acute eczema. Meanwhile, chronic eczema is represented by lichenification and hyperpigmentation, and possibly by hypopigmentation. In acute eczema, spongiosis is associated with overproduction of hyaluronic acid, secretion of self-protective galectin-7, and decreased expression of E-cadherin. In the upper dermis, Th1/Tc1 or Th2/Tc2, and additional Th17, Th22, and/or Tc22 infiltrate, depending on each eczematous disease. Innate lymphoid cells are also involved in the formation of eczema. In chronic eczema, periostin contributes to remodeling of inflammatory skin with dermal fibrosis, and epidermal melanogenesis and dermal pigment deposition result in hyperpigmentation. Finally, eczematous diseases are potentially associated with increased risk of comorbidities, including not only other allergic diseases but also coronary heart disease and mental problems such as depression. Although the original word for eczema is derived from old Greek "ekzein," eczema remains a major target of modern science and novel therapies.
Collapse
Affiliation(s)
- Yoshiki Tokura
- Department of Dermatology and Skin OncologyChutoen General Medical CenterKakegawaJapan
- Allergic Disease Research CenterChutoen General Medical CenterKakegawaJapan
| | - Marina Yunoki
- Department of Dermatology and Skin OncologyChutoen General Medical CenterKakegawaJapan
| | - Shumpei Kondo
- Department of Dermatology and Skin OncologyChutoen General Medical CenterKakegawaJapan
| | - Masaki Otsuka
- Department of Dermatology and Skin OncologyChutoen General Medical CenterKakegawaJapan
| |
Collapse
|
15
|
Bucheli OTM, Rodrigues D, Ulbricht C, Hauser AE, Eyer K. Dynamic Activation of NADPH Oxidases in Immune Responses Modulates Differentiation, Function, and Lifespan of Plasma Cells. Eur J Immunol 2025; 55:e202350975. [PMID: 39931760 PMCID: PMC11811814 DOI: 10.1002/eji.202350975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
NADPH-oxidase (NOX)-derived reactive oxygen species (ROS) have been described to play essential roles in B-cell activation processes. However, several key questions concerning NOX activity and subsequent ROS production remain unaddressed, including fundamental processes such as differentiation, functional competence, cellular metabolism, and viability. This study investigated these questions in a murine B-cell response after secondary immunization. We combined single-cell transcriptomics and single-cell detection of NOX activity and observed that various subsets of B cells dynamically express NOX1 and NOX2. The NOX+ cellular phenotype correlated with increased activity of metabolic pathways, augmented lactate production, lower IgG secretion rates, and markers for longevity. The NOX+ cellular phenotype was also associated with increased cellular stress and apoptosis, underscoring the intricate relationship between ROS and cellular survival. Consequently, these insights advance our understanding of how long-lived humoral immunity is formed.
Collapse
Affiliation(s)
- Olivia T. M. Bucheli
- ETH Laboratory for Functional Immune Repertoire AnalysisInstitute of Pharmaceutical Sciences, D‐CHAB, ETH ZürichZürichSwitzerland
| | - Daniela Rodrigues
- ETH Laboratory for Functional Immune Repertoire AnalysisInstitute of Pharmaceutical Sciences, D‐CHAB, ETH ZürichZürichSwitzerland
| | - Carolin Ulbricht
- Department of Rheumatology and Clinical ImmunologyCharité ‐ Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Immune DynamicsDeutsches Rheuma‐Forschungszentrum (DRFZ)a Leibniz Institute, Charitéplatz 1BerlinGermany
| | - Anja E. Hauser
- Department of Rheumatology and Clinical ImmunologyCharité ‐ Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Immune DynamicsDeutsches Rheuma‐Forschungszentrum (DRFZ)a Leibniz Institute, Charitéplatz 1BerlinGermany
| | - Klaus Eyer
- ETH Laboratory for Functional Immune Repertoire AnalysisInstitute of Pharmaceutical Sciences, D‐CHAB, ETH ZürichZürichSwitzerland
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| |
Collapse
|
16
|
Balakin E, Yurku K, Ivanov M, Izotov A, Nakhod V, Pustovoyt V. Regulation of Stress-Induced Immunosuppression in the Context of Neuroendocrine, Cytokine, and Cellular Processes. BIOLOGY 2025; 14:76. [PMID: 39857306 PMCID: PMC11760489 DOI: 10.3390/biology14010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Understanding the regulatory mechanisms of stress-induced immunosuppression and developing reliable diagnostic methods are important tasks in clinical medicine. This will allow for the development of effective strategies for the prevention and treatment of conditions associated with immune system dysfunction induced by chronic stress. The purpose of this review is to conduct a comprehensive analysis and synthesis of existing data on the regulatory mechanisms of stress-induced immunosuppression. The review is aimed at identifying key neuroendocrine, cytokine, and cellular processes underlying the suppression of the immune response under stress. This study involved a search of scientific literature covering the neuroendocrine, cellular, and molecular mechanisms of stress-induced immunosuppression regulation, as well as modern methods for its diagnosis. Major international bibliographic databases covering publications in biomedicine, psychophysiology, and immunology were selected for the search. The results of the analysis identified key mechanisms regulating stress-induced immunosuppression. The reviewed publications provided detailed descriptions of the neuroendocrine and cytokine processes underlying immune response suppression under stress. A significant portion of the data confirms that the activation of the hypothalamic-pituitary-adrenal (HPA) axis and subsequent elevation of cortisol levels exert substantial immunosuppressive effects on immune cells, particularly macrophages and lymphocytes, leading to the suppression of innate and adaptive immune responses. The data also highlight the crucial role of cortisol and catecholamines (adrenaline and noradrenaline) in initiating immunosuppressive mechanisms under chronic stress.
Collapse
Affiliation(s)
- Evgenii Balakin
- Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| | - Ksenia Yurku
- Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| | - Mark Ivanov
- Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| | - Alexander Izotov
- V.N. Orekhovich Research Institute of Biomedical Chemistry, Pogodinskaya Str. 10, Bldg. 8, 119121 Moscow, Russia
| | - Valeriya Nakhod
- V.N. Orekhovich Research Institute of Biomedical Chemistry, Pogodinskaya Str. 10, Bldg. 8, 119121 Moscow, Russia
| | - Vasiliy Pustovoyt
- Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| |
Collapse
|
17
|
Li J, Long H, Chen S, Zhang Z, Li S, Liu Q, Liu J, Cai J, Luo L, Peng Y. An mRNA-Based Respiratory Syncytial Virus Vaccine Elicits Strong Neutralizing Antibody Responses and Protects Rodents Without Vaccine-Associated Enhanced Respiratory Disease. Vaccines (Basel) 2025; 13:52. [PMID: 39852831 PMCID: PMC11768429 DOI: 10.3390/vaccines13010052] [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: 11/23/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) causes the most common type of severe lower respiratory tract infection worldwide, and the fusion (F) protein is a target for neutralizing antibodies and vaccine development. This study aimed to investigate the immunogenicity and efficacy of an mRNA-based RSV vaccine with an F protein sequence. METHODS We designed an mRNA construct encoding a modified RSV F protein, which was further developed into an LNP-encapsulated mRNA vaccine (LVRNA007). LVRNA007 was administered to mice and cotton rats, followed by immunogenicity analysis and viral challenge studies. Protection of rodents from the viral infection was evaluated based on the presence of the virus in the lung and pathological examination of respiratory tissues. RESULTS LVRNA007 induced robust humoral and cellular immune responses in both mice and cotton rats, with neutralization antibody levels in the immunized animals maintained at high levels for over one year. Vaccination of LVRNA007 also protected the rodents from RSV challenge, judged by the much decreased virus titer and the pathological score in the lung tissue. In addition, no vaccine-enhanced disease (VED) phenomenon was observed with LVRNA007 vaccination. CONCLUSIONS Based on the preclinical immunogenicity and efficacy data, LVRNA007 could be a potential promising vaccine for prophylaxis of RSV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yucai Peng
- Liverna Therapeutics Inc., Zhuhai 519000, China; (J.L.); (H.L.); (S.C.); (Z.Z.); (S.L.); (Q.L.); (J.L.); (J.C.); (L.L.)
| |
Collapse
|
18
|
Zhang S, Wang X, Gao X, Chen X, Li L, Li G, Liu C, Miao Y, Wang R, Hu K. Radiopharmaceuticals and their applications in medicine. Signal Transduct Target Ther 2025; 10:1. [PMID: 39747850 PMCID: PMC11697352 DOI: 10.1038/s41392-024-02041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/28/2024] [Indexed: 01/04/2025] Open
Abstract
Radiopharmaceuticals involve the local delivery of radionuclides to targeted lesions for the diagnosis and treatment of multiple diseases. Radiopharmaceutical therapy, which directly causes systematic and irreparable damage to targeted cells, has attracted increasing attention in the treatment of refractory diseases that are not sensitive to current therapies. As the Food and Drug Administration (FDA) approvals of [177Lu]Lu-DOTA-TATE, [177Lu]Lu-PSMA-617 and their complementary diagnostic agents, namely, [68Ga]Ga-DOTA-TATE and [68Ga]Ga-PSMA-11, targeted radiopharmaceutical-based theranostics (radiotheranostics) are being increasingly implemented in clinical practice in oncology, which lead to a new era of radiopharmaceuticals. The new generation of radiopharmaceuticals utilizes a targeting vector to achieve the accurate delivery of radionuclides to lesions and avoid off-target deposition, making it possible to improve the efficiency and biosafety of tumour diagnosis and therapy. Numerous studies have focused on developing novel radiopharmaceuticals targeting a broader range of disease targets, demonstrating remarkable in vivo performance. These include high tumor uptake, prolonged retention time, and favorable pharmacokinetic properties that align with clinical standards. While radiotheranostics have been widely applied in tumor diagnosis and therapy, their applications are now expanding to neurodegenerative diseases, cardiovascular diseases, and inflammation. Furthermore, radiotheranostic-empowered precision medicine is revolutionizing the cancer treatment paradigm. Diagnostic radiopharmaceuticals play a pivotal role in patient stratification and treatment planning, leading to improved therapeutic outcomes in targeted radionuclide therapy. This review offers a comprehensive overview of the evolution of radiopharmaceuticals, including both FDA-approved and clinically investigated agents, and explores the mechanisms of cell death induced by radiopharmaceuticals. It emphasizes the significance and future prospects of theranostic-based radiopharmaceuticals in advancing precision medicine.
Collapse
Grants
- 82372002 National Natural Science Foundation of China (National Science Foundation of China)
- 0104002 Beijing Nova Program
- L248087; L234044 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Nonprofit Central Research Institute Fund of the Chinese Academy of Medical Sciences (No. 2022-RC350-04), the CAMS Innovation Fund for Medical Sciences (Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001), the National Key Research and Development Program of China (No. 2022YFE0111700),the Fundamental Research Funds for the Central Universities (Nos. 3332023044 and 3332023151), the CIRP Open Fund of Radiation Protection Laboratories (No. ZHYLYB2021005), and the China National Nuclear Corporation Young Talent Program.
- Fundamental Research Funds for the Central Universities,Nos. 3332023044
- Fundamental Research Funds for the Central Universities,Nos. 3332023151
- he Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,No. 2022-RC350-04;the CAMS Innovation Fund for Medical Sciences,Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001;the National Key Research and Development Program of China,No. 2022YFE0111700
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Linger Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Yuan Miao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 2019RU066, 730000, Lanzhou, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
19
|
Wagner M, Nishikawa H, Koyasu S. Reinventing type 2 immunity in cancer. Nature 2025; 637:296-303. [PMID: 39780006 DOI: 10.1038/s41586-024-08194-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/10/2024] [Indexed: 01/11/2025]
Abstract
Our understanding of type 2 immunity has undergone a substantial transformation in recent years, revealing previously unknown functions. Beyond its canonical role in defence against parasitic helminth infections, type 2 immunity safeguards the host through additional mechanisms, including the suppression of excessive type 1 immune responses, regulation of tissue repair and maintenance of adipose tissue homeostasis. However, unlike type 1 immune responses, type 2 immunity is perceived as a potential promoter of tumorigenesis. Emerging evidence challenges this perspective, painting a more nuanced picture in which type 2 immunity might protect against or even actively suppress tumour growth and progression. In this Review, we explore discoveries that highlight the potential of type 2 immunity in reshaping the landscape of cancer immunotherapies.
Collapse
Affiliation(s)
- Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network-PORT Polish Center for Technology Development, Wrocław, Poland.
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Cancer Immunology, Research Institute/EPOC, National Cancer Center, Tokyo, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- National Institutes for Quantum Science and Technology (QST), Chiba, Japan.
| |
Collapse
|
20
|
Koenig JFE. T follicular helper and memory B cells in IgE recall responses. Allergol Int 2025; 74:4-12. [PMID: 39562254 DOI: 10.1016/j.alit.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
IgE antibodies raised against innocuous environmental antigens cause allergic diseases like allergic rhinitis, food allergy, and allergic asthma. While some allergies are often outgrown, others (peanut, shellfish, tree nut) are lifelong in the majority of individuals. Lifelong allergies are the result of persistent production of allergen-specific IgE. However, IgE antibodies and the plasma cells that secrete them tend to be short-lived. Persistent allergen-specific IgE titres are thought to be derived from the continued renewal of IgE plasma cells from memory B cells in response to allergen encounters. The initial generation of allergen-specific IgE is driven by B cell activation by IL-4 producing Tfh cells, but the cellular and molecular mechanisms of the long-term production of IgE are poorly characterized. This review investigates the mechanisms governing IgE production and Tfh activation in the primary and recall responses, towards the objective of identifying molecular targets for therapeutic intervention that durably inactivate the IgE recall response.
Collapse
Affiliation(s)
- Joshua F E Koenig
- McMaster Immunology Research Centre, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada; Schroeder Allergy and Immunology Research Institute, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
21
|
Ohshima Y. T follicular helper cells and IgE. Allergol Int 2025; 74:2-3. [PMID: 39756839 DOI: 10.1016/j.alit.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Indexed: 01/07/2025] Open
Affiliation(s)
- Yusei Ohshima
- Department of Pediatrics, Faculty of Medical Sciences, University of Fukui, 23-3 Shimoaizuki, Matsuoka, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.
| |
Collapse
|
22
|
Portilho AI, Hermes Monteiro da Costa H, Grando Guereschi M, Prudencio CR, De Gaspari E. Hybrid response to SARS-CoV-2 and Neisseria meningitidis C after an OMV-adjuvanted immunization in mice and their offspring. Hum Vaccin Immunother 2024; 20:2346963. [PMID: 38745461 PMCID: PMC11789737 DOI: 10.1080/21645515.2024.2346963] [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: 12/15/2023] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/16/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, and meningococcal disease, caused by Neisseria meningitidis, are relevant infectious diseases, preventable through vaccination. Outer membrane vesicles (OMVs), released from Gram-negative bacteria, such as N. meningitidis, present adjuvant characteristics and may confer protection against meningococcal disease. Here, we evaluated in mice the humoral and cellular immune response to different doses of receptor binding domain (RBD) of SARS-CoV-2 adjuvanted by N. meningitidis C:2a:P1.5 OMVs and aluminum hydroxide, as a combined preparation for these pathogens. The immunization induced IgG antibodies of high avidity for RBD and OMVs, besides IgG that recognized the Omicron BA.2 variant of SARS-CoV-2 with intermediary avidity. Cellular immunity showed IFN-γ and IL-4 secretion in response to RBD and OMV stimuli, demonstrating immunologic memory and a mixed Th1/Th2 response. Offspring presented transferred IgG of similar levels and avidity as their mothers. Humoral immunity did not point to the superiority of any RBD dose, but the group immunized with a lower antigenic dose (0.5 μg) had the better cellular response. Overall, OMVs enhanced RBD immunogenicity and conferred an immune response directed to N. meningitidis too.
Collapse
MESH Headings
- Animals
- Mice
- Immunoglobulin G/blood
- Neisseria meningitidis/immunology
- Female
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- SARS-CoV-2/immunology
- Adjuvants, Immunologic/administration & dosage
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Immunity, Cellular
- Immunity, Humoral
- Mice, Inbred BALB C
- Meningococcal Infections/prevention & control
- Meningococcal Infections/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Adjuvants, Vaccine/administration & dosage
- Aluminum Hydroxide/administration & dosage
- Aluminum Hydroxide/immunology
- Immunization/methods
- Antibody Affinity
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Meningococcal Vaccines/immunology
- Meningococcal Vaccines/administration & dosage
- Immunologic Memory
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Hernan Hermes Monteiro da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | | | - Carlos Roberto Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
- Post-Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
23
|
Sun S, Chen Y, Ouyang Y, Tang Z. Regulatory Roles of SWI/SNF Chromatin Remodeling Complexes in Immune Response and Inflammatory Diseases. Clin Rev Allergy Immunol 2024; 68:2. [PMID: 39751934 DOI: 10.1007/s12016-024-09011-4] [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: 12/13/2024] [Indexed: 01/04/2025]
Abstract
The switch/sucrose non-fermentable (SWI/SNF) chromatin remodeling complexes (also referred to as BAF complexes) are composed of multiple subunits, which regulate the nucleosome translocation and chromatin accessibility. In recent years, significant advancements have been made in understanding mutated genes encoding subunits of the SWI/SNF complexes in cancer biology. Nevertheless, the role of SWI/SNF complexes in immune response and inflammatory diseases continues to attract significant attention. This review presents a summary of the significant functions of SWI/SNF complexes during the overall process from the development to the activation of innate and adaptive immune cells. In addition, the correlation between various SWI/SNF subunits and diverse inflammatory diseases is explored. Further investigations are warranted in terms of the mechanism of SWI/SNF complexes' preference for binding sites and opposite pro-/anti-inflammatory effects. In conclusion, further efforts are needed to evaluate the druggability of targeting SWI/SNF complexes in inflammatory diseases, and we hope this review will inspire the development of novel immune modulators in clinical practice.
Collapse
Affiliation(s)
- Shunan Sun
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, People's Republic of China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuzhen Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, People's Republic of China.
| |
Collapse
|
24
|
Mishra A, Kumar V, Kumar S, Singh H, Singh A. HRAMS Proteomics Insights on the Anti-Filarial Effect of Ocimum sanctum: Implications in Phytochemical-Based Drug-Targeting and Designing. Proteomes 2024; 13:2. [PMID: 39846633 PMCID: PMC11755628 DOI: 10.3390/proteomes13010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Lymphatic filariasis (LF) continues to impact 657 million individuals worldwide, resulting in lifelong and chronic impairment. The prevalent anti-filarial medications-DEC, albendazole, and ivermectin-exhibit limited adulticidal efficacy. Despite ongoing LF eradication programs, novel therapeutic strategies are essential for effective control. This study examines the mechanism of action of Ocimum sanctum on the filarial parasites Setaria cervi via a synergistic biochemical and proteomics methodology. The ethanolic extract of Ocimum sanctum (EOS) demonstrated potential anti-filarial action in the MTT reduction experiment, with an LC50 value of 197.24 µg/mL. After EOS treatment, an elevation in lipid peroxidation (51.92%), protein carbonylation (48.99%), and NADPH oxidase (88.88%) activity, along with a reduction in glutathione (GSH) (-39.23%), glutathione reductase (GR) (-60.17%), and glutathione S transferase (GST) (-50.48%) activity, was observed. The 2D gel electrophoresis identified 20 decreased and 11 increased protein spots in the EOS-treated parasites relative to the control group. Additionally, in drug docking analysis, the EOS bioactive substances ursolic acid, rutin, and rosmarinic acid show a significant binding affinity with the principal differentially expressed proteins. This paper demonstrates, for the first time, that the anti-filarial efficacy of EOS is primarily facilitated by its impact on energy metabolism, antioxidant mechanisms, and stress response systems of the parasites.
Collapse
Affiliation(s)
- Ayushi Mishra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India; (A.M.)
| | - Vipin Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India; (A.M.)
| | - Sunil Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India; (A.M.)
| | - HariOm Singh
- Department of Molecular Biology, National Aids Research Institute, Pune 411026, India
| | - Anchal Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India; (A.M.)
| |
Collapse
|
25
|
Seremet T, Di Domizio J, Girardin A, Yatim A, Jenelten R, Messina F, Saidoune F, Schlapbach C, Bogiatzi S, Minisini F, Garzorz-Stark N, Leuenberger M, Wüthrich H, Vernez M, Hohl D, Eyerich S, Eyerich K, Guenova E, Paul C, Gottardo R, Conrad C, Gilliet M. Immune modules to guide diagnosis and personalized treatment of inflammatory skin diseases. Nat Commun 2024; 15:10688. [PMID: 39695162 DOI: 10.1038/s41467-024-54559-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
Previous advances have identified immune pathways associated with inflammatory skin diseases, leading to the development of targeted therapies. However, there is a lack of molecular approaches that delineate these pathways at the individual patient level for personalized diagnostic and therapeutic guidance. Here, we conduct a cross-comparison of expression profiles from multiple inflammatory skin diseases to identify gene modules defining relevant immune pathways. Seven modules are identified, representing key immune pathways: Th17, Th2, Th1, Type I IFNs, neutrophilic, macrophagic, and eosinophilic. These modules allow the development of a molecular map with high diagnostic efficacy for inflammatory skin diseases and clinico-pathologically undetermined cases. Aligning dominant modules with treatment targets offers a rational framework for treatment selection, improving response rates in both treatment-naïve patients and non-responders to targeted therapies. Overall, our approach offers precision medicine for inflammatory skin diseases, utilizing transcriptional modules to support diagnosis and guide personalized treatment selection.
Collapse
Affiliation(s)
- Teofila Seremet
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Jeremy Di Domizio
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Antoine Girardin
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Ahmad Yatim
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Raphael Jenelten
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Francesco Messina
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Fanny Saidoune
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sofia Bogiatzi
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Frederic Minisini
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Natalie Garzorz-Stark
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Matthieu Leuenberger
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Héloise Wüthrich
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Maxime Vernez
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Daniel Hohl
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Stefanie Eyerich
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Emmanuella Guenova
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Carle Paul
- Department of Dermatology and Venereology, CHU Toulouse, Toulouse, France
| | - Raphael Gottardo
- Biomedical Data Science Center, CHUV, UNIL, and SIB, Lausanne, Switzerland
| | - Curdin Conrad
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Michel Gilliet
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
26
|
Mukhtarova N, Babu A, Coe CL, Kling PJ. Influence of Biological Sex and Congenital Iron Deficiency on Neonatal Cytokine Responses. Nutrients 2024; 16:4203. [PMID: 39683596 DOI: 10.3390/nu16234203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Stimulated cord blood mononuclear cell (CBMC) cytokine responses were previously shown to predict the risk of childhood atopic disease. Iron deficiency (ID) at birth may also program atopic disease. Males are at a higher risk of pediatric atopic disease, but it is not known whether congenital ID impacts CBMC immune responses differentially by sex. METHODS Cord blood (CB) samples were collected from healthy term or near-term neonates after elective cesarean deliveries. A transferrin saturation ≤ 25% defined congenital ID. CBMCs were stimulated with either phytohemagglutinin (PHA) or PHA plus an iron chelator. RESULTS Of the 85 neonates, the 26 neonates with congenital ID exhibited lower plasma tumor necrosis factor-α (TNF-α), as well as higher CBMC TNF-α and IL-8 responses than iron-sufficient neonates (p = 0.017, p = 0.013, and p = 0.007, respectively). Higher CBMC TNF-α responses were seen in both males and females with congenital ID. However, females with congenital ID also had lower plasma IL-6, lower plasma TNF-α, and higher CBMC interleukin (IL)-8 responses. Additionally, iron chelation during culture influenced stimulated CBMC IFN-γ and CBMC TNF-α responses. DISCUSSION Congenital ID may influence stimulated CBMC cytokine responses, but results point to a sex-specific regulation of immune balance at birth. Because males are more prone to infantile ID and more likely to develop early childhood asthma, future studies should further investigate how fetal sex and congenital iron status impacts childhood immune responsiveness to infections and antigenic stimulation from the rearing environment.
Collapse
Affiliation(s)
- Narmin Mukhtarova
- Department of Pediatrics, University of Wisconsin Hospitals and Clinics, Madison, WI 53792, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Anthony Babu
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Christopher L Coe
- Department of Psychology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Pamela J Kling
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA
| |
Collapse
|
27
|
Lai R, Deng X, Lv X, Zhong Y. Causal relationship between inflammatory proteins, immune cells, and gout: a Mendelian randomization study. Sci Rep 2024; 14:30070. [PMID: 39627303 PMCID: PMC11615377 DOI: 10.1038/s41598-024-80138-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024] Open
Abstract
Prior research has documented the association between certain circulating inflammatory proteins/immune cells and gout. However, the reliability of these associations remains contentious due to the constraints of conventional observational methodologies. This investigation seeks to reassess the causative link between circulating inflammatory proteins/immune cells and gout through the application of Mendelian randomization (MR). The study included 3576 individuals of European ancestry with gout, immune cell data from the GWAS summary of 3757 Sardinians, and circulating inflammatory protein data from 14,824 European ancestry participants for MR analysis. The principal approach employed was inverse variance weighted analysis to investigate the causal relationship between exposure and outcomes. The results indicate that CD28 on CD39+ CD4+ T cells may be associated with a reduced risk of gout. Additionally, CD45RA+ CD28- CD8bright T cells may also be associated with a reduced risk of gout. In contrast, DN (CD4-CD8-) T cells and IL-12β may increase the risk of gout. Some inflammatory proteins and immune cells show potential causal associations with gout. Nevertheless, additional experimental verification is warranted to assess the underlying mechanisms and confirm the causative role of these immune factors in gout pathogenesis.
Collapse
Affiliation(s)
- Rui Lai
- Chengdu Integrated TCM & Western Medicine Hospital/Chengdu First People's Hospital, Chengdu, China
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinmin Deng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofeng Lv
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumei Zhong
- Chengdu Integrated TCM & Western Medicine Hospital/Chengdu First People's Hospital, Chengdu, China.
| |
Collapse
|
28
|
Wang Z, Pu N, Zhao W, Chen X, Zhang Y, Sun Y, Bo X. RNA sequencing reveals dynamic expression of genes related to innate immune responses in canine small intestinal epithelial cells induced by Echinococcus granulosus protoscoleces. Front Vet Sci 2024; 11:1503995. [PMID: 39679172 PMCID: PMC11638162 DOI: 10.3389/fvets.2024.1503995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Background Dogs are definitive hosts of Echinococcus granulosus, with the small intestine being the only site of parasitic infections. However, the immunomodulatory processes that occur during interactions between E. granulosus and its definitive host remain unclear. Therefore, this study aimed to evaluate gene transcription patterns in canine small intestinal epithelial cells (CIECs) following stimulation by E. granulosus protoscoleces (PSCs). Particularly, this study investigated the roles of pattern recognition receptors (PRRs), involved in recognizing pathogen-associated molecular patterns (PAMPs) and mediating the host innate immune response to the tapeworm E. granulosus. Methods RNA sequencing (RNA-seq) was used to examine gene transcription patterns in CIECs following stimulation with PSCs for 12 and 24 h. The potential roles of differentially expressed (DE) genes were inferred through Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Results RNA-seq analysis identified 78,206,492-90,548,214 clean reads in 12 RNA samples. This included six samples stimulated with PSCs for 12 h (PSC1_12h-PSC3_12h) and 24 h (PSC1_24h-PSC3_24h) and six corresponding control samples (PBS1_12h-PBS3_12h and PBS1_24h-PBS3_24h). In the PSC_12h vs. PBS_12h and PSC_24h vs. PBS_24h groups, 3,520 (2,359 upregulated and 1,161 downregulated) and 3,287 (1765 upregulated and 1,522 downregulated) DEgenes were identified, respectively. The expression of 45 PRRs genes was upregulated in the PSC_12h and PSC_24h groups compared to those in the control groups, including 4 Toll-like receptors (TLRs), 4C-type lectin receptors (CLRs), 3 NOD-like receptors (NLRs), 17 G protein-coupled receptors (GPCRs), 4 scavenger receptors (SRs), and 13 leucine-rich repeat-containing proteins (LRRCs). GO enrichment and KEGG analyses revealed that these DEgenes were mainly involved in the regulation of host immune response processes and molecules. These included antigen processing and presentation, Th17, PI3K-Akt, Th1, and Th2 cell differentiation, neutrophil extracellular trap formation, NOD- and Toll-like receptors, TNF, intestinal immune network for IgA production and IL-17 signaling pathway. Furthermore, the identified DEgenes were involved in the regulation of signaling molecules and interaction (e.g., cell adhesion molecules and ECM-receptor interaction). Conclusion These preliminary findings provide novel perspectives on the host innate immune response to E. granulosus PSC stimulation, with a focus on the involvement of E. granulosus-specific PRRs in host defense mechanisms against infection.
Collapse
Affiliation(s)
- Zhengrong Wang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
| | - Na Pu
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Wenqing Zhao
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- College of Animal Science and Technology, Tarim University, Alar, China
| | - Xuke Chen
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yanyan Zhang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
| | - Yan Sun
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
| | - Xinwen Bo
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
29
|
Okamoto M, Kuratani A, Okuzaki D, Kamiyama N, Kobayashi T, Sasai M, Yamamoto M. IFN-γ-induced Th1-Treg polarization in inflamed brains limits exacerbation of experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 2024; 121:e2401692121. [PMID: 39560646 PMCID: PMC11621829 DOI: 10.1073/pnas.2401692121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 10/02/2024] [Indexed: 11/20/2024] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most widely used rodent model for multiple sclerosis. Interferon-γ (IFN-γ) and regulatory T cells (Tregs) are individually well known to play beneficial roles in amelioration of EAE. However, little is known about the relationship between IFN-γ and Tregs during the disease. Here, we show that IFN-γ polarizes Tregs into T helper 1 (Th1)-type Tregs (Th1-Tregs) to recover from EAE. Single-cell RNA sequencing analysis revealed that brain Tregs showed signs of IFN-γ stimulation during EAE. Loss of IFN-γ signaling in Tregs and of T cell-derived IFN-γ impaired the Th1-Treg polarization and worsened the disease. Moreover, selective ablation of Th1-Tregs using an intersectional genetic method promoted proinflammatory features of macrophages in the inflamed brains and exacerbated the EAE. Taken together, our study highlights a critical role of T cell-derived IFN-γ for Th1-Treg polarization in inflamed brain to ameliorate EAE.
Collapse
Affiliation(s)
- Masaaki Okamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Ayumi Kuratani
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Osaka University, Suita, Osaka565-0871, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita879-5593, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita879-5593, Japan
- Division of Pathophysiology, Research Center for GLOBAL and LOCAL Infectious Diseases, Oita University, Oita879-5593, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka565-0871, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka565-0871, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka565-0871, Japan
- Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka565-0871, Japan
| |
Collapse
|
30
|
Wang X, Li Y, Jin J, Chai X, Ma Z, Duan J, Zhang G, Huang T, Zhang X, Zhang T, Wu H, Cao Y, Su B. Severe acute respiratory syndrome coronavirus 2-specific T-cell responses are induced in people living with human immunodeficiency virus after booster vaccination. Chin Med J (Engl) 2024; 137:2734-2744. [PMID: 39028115 PMCID: PMC11611240 DOI: 10.1097/cm9.0000000000003176] [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: 04/27/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND T-cell-mediated immunity is crucial for the effective clearance of viral infection, but the T-cell-mediated immune responses that are induced by booster doses of inactivated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in people living with human immunodeficiency virus (PLWH) remain unclear. METHODS Forty-five PLWH who had received antiretroviral therapy (ART) for more than two years and 29 healthy controls (HCs) at Beijing Youan Hospital were enrolled to assess the dynamic changes in T-cell responses between the day before the third vaccine dose (week 0) and 4 or 12 weeks (week 4 or week 12) after receiving the third dose of inactivated SARS-CoV-2 vaccine. Flow cytometry, enzyme-linked immunospot (ELISpot), and multiplex cytokines profiling were used to assess T-cell responses at the three timepoints in this study. RESULTS The results of the ELISpot and activation-induced marker (AIM) assays showed that SARS-CoV-2-specific T-cell responses were increased in both PLWH and HCs after the third dose of the inactivated SARS-CoV-2 vaccine, and a similar magnitude of immune response was induced against the Omicron (B.1.1.529) variant compared to the wild-type strain. In detail, spike-specific T-cell responses (measured by the ELISpot assay for interferon γ [IFN-γ] release) in both PLWH and HCs significantly increased in week 4, and the spike-specific T-cell responses in HCs were significantly stronger than those in PLWH 4 weeks after the third vaccination. In the AIM assay, spike-specific CD4 + T-cell responses peaked in both PLWH and HCs in week 12. Additionally, significantly higher spike-specific CD8 + T-cell responses were induced in PLWH than in HCs in week 12. In PLWH, the release of the cytokines interleukin-2 (IL-2), tumour necrosis factor-alpha (TNF-α), and IL-22 by peripheral blood mononuclear cells (PBMCs) that were stimulated with spike peptides increased in week 12. In addition, the levels of IL-4 and IL-5 were higher in PLWH than in HCs in week 12. Interestingly, the magnitude of SARS-CoV-2-specific T-cell responses in PLWH was negatively associated with the extent of CD8 + T-cell activation and exhaustion. In addition, positive correlations were observed between the magnitude of spike-specific T-cell responses (determined by measuring IFN-γ release by ELISpot) and the amounts of IL-4, IL-5, IL-2 and IL-17F. CONCLUSIONS Our findings suggested that SARS-CoV-2-specific T-cell responses could be enhanced by the booster dose of inactivated COVID-19 vaccines and further illustrate the importance of additional vaccination for PLWH.
Collapse
Affiliation(s)
- Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Yongzheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100069, China
| | - Junyan Jin
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xiaoran Chai
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100069, China
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Zhenglai Ma
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Junyi Duan
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Guanghui Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tao Huang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Yunlong Cao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100069, China
- Changping Laboratory, Beijing 102299, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
31
|
Li Y, Chen P, Huang X, Huang H, Ma Q, Lin Z, Qiu L, Ou C, Liu W. Pathogenic Th17 cells are a potential therapeutic target for tacrolimus in AChR-myasthenia gravis patients. J Neuroimmunol 2024; 396:578464. [PMID: 39393213 DOI: 10.1016/j.jneuroim.2024.578464] [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: 04/18/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/13/2024]
Abstract
In our study, we investigated the impact of tacrolimus (TAC) on CD4+ T cell subsets in 41 AChR-MG patients over 12 weeks. Twenty-seven patients were classified as the response group (RG) (improved myasthenia gravis composite scores ≥3), while 14 were non-response. We found that TAC treatment significantly reduced Th17 and pathogenic Th17 cells, along with IL-17 levels in RG, while Th1 and Tfh cells slightly decreased without affecting Th2 or Treg subsets. This indicates that TAC's clinical benefits may be due to its inhibitory effect on the Th17 response, enhancing our insight into its immunomodulatory mechanisms in MG management.
Collapse
Affiliation(s)
- Yingkai Li
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Neuromuscular division, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Pei Chen
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xin Huang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Hao Huang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Qian Ma
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhongqiang Lin
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Li Qiu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Changyi Ou
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Weibin Liu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
32
|
Sakai Y, Yamada S, Inoue M, Shiga T, Konagayoshi K, Kasai K, Kimura A, Murakami K. Efficacy of a DNA vaccine encoding the E2 glycoprotein of bovine viral diarrhea virus 1 fused to mouse lysosome-associated membrane protein 1. Vet Microbiol 2024; 298:110283. [PMID: 39488135 DOI: 10.1016/j.vetmic.2024.110283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/04/2024]
Abstract
The E2 protein of bovine viral diarrhea virus (BVDV) is a known protective antigen and a major target for DNA vaccines. DNA vaccines have various advantages; however, their immunogenicity needs to be enhanced by using adjuvants or drug delivery systems. In this study, we used mouse lysosome-associated membrane protein 1 (mLAMP1) as a molecular adjuvant and developed a DNA vaccine encoding an mLAMP1-BVDV E2 chimeric protein (pVax-mLAMP1-E2). We constructed DNA plasmids in which the E2 gene was inserted within the hinge region (H) or membrane proximal domain (D) of the mLAMP1 gene. Transfection of these plasmids into cultured cells led to high expression of E2 antigen from pVax-mLAMP1-E2 (H). Intradermal immunization of mice with pVax-mLAMP1-E2 (H) induced sufficient neutralizing antibodies and splenocytes with E2 antigen-specific IFN-γ production compared with pVax-mLAMP1-E2 (D). However, the immunogenicity of pVax mLAMP1-E2 (H) in mice did not differ from that of a control plasmid without the LAMP1 molecule (pVax-E2). In cattle, geometric mean serum neutralizing antibody titers after intradermal or intramuscular injection tended to be higher with pVax-mLAMP1-E2 (H) than with pVax that expressed E2 without mLAMP1. In addition, E2 antigen-specific IFN-γ production in peripheral blood mononuclear cells from cattle immunized intradermally with pVax-mLAMP1-E2 (H) was not significantly different from that of pVax-E2. These results suggest that mLAMP1 fusion antigens effectively induce humoral and cellular immunity in mice and cattle, especially when the antigen is inserted in the hinge region of mLAMP1. The LAMP1-E2 fusion antigen may be a useful candidate for a BVDV DNA vaccine in cattle.
Collapse
Affiliation(s)
- Yusuke Sakai
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Nippon Zenyaku Kogyo Co., Ltd., Koriyama, Fukushima 963-0196, Japan
| | - Shinji Yamada
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan; Farm Animal Clinic and Disease Control Center, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Maho Inoue
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Toshinori Shiga
- Nippon Zenyaku Kogyo Co., Ltd., Koriyama, Fukushima 963-0196, Japan
| | | | - Kei Kasai
- Nippon Zenyaku Kogyo Co., Ltd., Koriyama, Fukushima 963-0196, Japan
| | - Atsushi Kimura
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Farm Animal Clinic and Disease Control Center, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Kenji Murakami
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan; Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan; Farm Animal Clinic and Disease Control Center, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan.
| |
Collapse
|
33
|
Breidenbach JD, French BW, Stanoszek LM, Lavik JP, Maddipati KR, Premathilaka SH, Baliu-Rodriguez D, Timalsina B, Aradhyula V, Patel SC, Lad A, Syed I, Kleinhenz AL, Blomquist TM, Gohara A, Dube P, Zhang S, Faleel D, Khalaf FK, Isailovic D, Wooten RM, Willey JC, Hammersley JR, Modyanov NN, Malhotra D, Dworkin LD, Kennedy DJ, Haller ST. Aerosolized Harmful Algal Bloom Toxin Microcystin-LR Induces Type 1/Type 17 Inflammation of Murine Airways. Toxins (Basel) 2024; 16:470. [PMID: 39591225 PMCID: PMC11598155 DOI: 10.3390/toxins16110470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Harmful algal blooms are increasing globally and pose serious health concerns releasing cyanotoxins. Microcystin-LR (MC-LR), one of the most frequently produced cyanotoxins, has recently been detected in aerosols generated by the normal motions of affected bodies of water. MC-LR aerosol exposure has been linked to a pro-inflammatory influence on the airways of mice; however, little is understood about the underlying mechanism or the potential consequences. This study aimed to investigate the pro-inflammatory effects of aerosolized MC-LR on murine airways. C57BL/6 and BALB/c mice were exposed to MC-LR aerosols, as these strains are predisposed to type 1/type 17 and type 2 immune responses, respectively. Exposure to MC-LR induced granulocytic inflammation in C57BL/6 but not BALB/c mice, as observed by increased expression of cytokines MIP-1α, CXCL1, CCL2, and GM-CSF compared with their respective vehicle controls. Furthermore, the upregulation of interleukins IL-17A and IL-12 is consistent with Th1- and Th17-driven type 1/type 17 inflammation. Histological analysis confirmed inflammation in the C57BL/6 lungs, with elevated neutrophils and macrophages in the bronchoalveolar lavage fluid and increased pro-inflammatory and pro-resolving oxidized lipids. In contrast, BALB/c mice showed no significant airway inflammation. These results highlight the ability of aerosolized MC-LR to trigger harmful airway inflammation, requiring further research, particularly into populations with predispositions to type 1/type 17 inflammation.
Collapse
Affiliation(s)
- Joshua D. Breidenbach
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (I.S.); (R.M.W.)
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Benjamin W. French
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Lauren M. Stanoszek
- Department of Pathology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (L.M.S.); (T.M.B.); (A.G.)
| | - John-Paul Lavik
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Krishna Rao Maddipati
- Department of Pathology, Lipidomics Core Facility, Wayne State University, Detroit, MI 48202, USA;
| | - Sanduni H. Premathilaka
- Department of Chemistry and Biochemistry, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43606, USA; (S.H.P.); (D.B.-R.); (D.I.)
| | - David Baliu-Rodriguez
- Department of Chemistry and Biochemistry, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43606, USA; (S.H.P.); (D.B.-R.); (D.I.)
| | - Bivek Timalsina
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (I.S.); (R.M.W.)
| | - Vaishnavi Aradhyula
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Shivani C. Patel
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Apurva Lad
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Irum Syed
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (I.S.); (R.M.W.)
| | - Andrew L. Kleinhenz
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Thomas M. Blomquist
- Department of Pathology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (L.M.S.); (T.M.B.); (A.G.)
| | - Amira Gohara
- Department of Pathology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (L.M.S.); (T.M.B.); (A.G.)
| | - Prabhatchandra Dube
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Shungang Zhang
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Dhilhani Faleel
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Fatimah K. Khalaf
- Department of Medicine, College of Medicine, University of Alkafeel, Najaf 54001, Iraq;
| | - Dragan Isailovic
- Department of Chemistry and Biochemistry, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43606, USA; (S.H.P.); (D.B.-R.); (D.I.)
| | - R. Mark Wooten
- Department of Medical Microbiology and Immunology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (I.S.); (R.M.W.)
| | - James C. Willey
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Jeffrey R. Hammersley
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Nikolai N. Modyanov
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA;
| | - Deepak Malhotra
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Lance D. Dworkin
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - David J. Kennedy
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| | - Steven T. Haller
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43614, USA; (J.D.B.); (B.W.F.); (B.T.); (V.A.); (S.C.P.); (A.L.); (A.L.K.); (P.D.); (S.Z.); (D.F.); (J.C.W.); (J.R.H.); (D.M.); (L.D.D.)
| |
Collapse
|
34
|
Mi Z, Wang Z, Wang Y, Xue X, Liao X, Wang C, Sun L, Lin Y, Wang J, Guo D, Liu T, Liu J, Modlin RL, Liu H, Zhang F. Cellular and molecular determinants of bacterial burden in leprosy granulomas revealed by single-cell multimodal omics. EBioMedicine 2024; 108:105342. [PMID: 39321499 PMCID: PMC11462173 DOI: 10.1016/j.ebiom.2024.105342] [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: 05/09/2024] [Revised: 08/23/2024] [Accepted: 09/01/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Which cell populations that determine the fate of bacteria in infectious granulomas remain unclear. Leprosy, a granulomatous disease with a strong genetic predisposition, caused by Mycobacterium leprae infection, exhibits distinct sub-types with varying bacterial load and is considered an outstanding disease model for studying host-pathogen interactions. METHODS We performed single-cell RNA and immune repertoire sequencing on 11 healthy controls and 20 patients with leprosy, and integrated single-cell data with genome-wide genetic data on leprosy. Multiplex immunohistochemistry, and in vitro and in vivo infection experiments were conducted to confirm the multimodal omics findings. FINDINGS Lepromatous leprosy (L-LEP) granulomas with high bacterial burden were characterised by exhausted CD8+ T cells, and high RGS1 expression in CD8+ T cells was associated with L-LEP. By contrast, tuberculoid leprosy (T-LEP) granulomas with low bacterial burden displayed enrichment in resident memory IFNG+ CD8+ T cells (CD8+ Trm) with high GNLY expression. This enrichment was potentially attributable to the communication between IL1B macrophages and CD8+ Trm via CXCL10-CXCR3 signalling. Additionally, IL1B macrophages in L-LEP exhibited anti-inflammatory phenotype, with high APOE expression contributing to high bacterial burden. Conversely, IL1B macrophages in T-LEP were distinguished by interferon-γ induced GBP family genes. INTERPRETATION The state of IL1B macrophages and functional CD8+ T cells, as well as the relationship between them, is crucial for controlling bacterial persistence within granulomas. These insights may indicate potential targets for host-directed immunotherapy in granulomatous diseases caused by mycobacteria and other intracellular bacteria. FUNDING The Key research and development program of Shandong Province (2021LCZX07), Natural Science Foundation of Shandong Province (ZR2023MH046), Youth Science Foundation Cultivation Funding Plan of Shandong First Medical University (Shandong Academy of Medical Sciences) (202201-123), National Natural Science Foundation of China (82471800, 82230107, 82273545, 82304039), the China Postdoctoral Science Foundation (2023M742162), Shandong Province Taishan Scholar Project (tspd20230608), Joint Innovation Team for Clinical & Basic Research (202410), Central guidance for local scientific and technological development projects of Shandong Province (YDZX2023058).
Collapse
Affiliation(s)
- Zihao Mi
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Zhenzhen Wang
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Yi Wang
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Xiaotong Xue
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Xiaojie Liao
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Chuan Wang
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Lele Sun
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Yingjie Lin
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Jianwen Wang
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Dianhao Guo
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Tingting Liu
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China
| | - Jianjun Liu
- Laboratory of Human Genomics, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA.
| | - Hong Liu
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China.
| | - Furen Zhang
- Hospital for Skin Diseases, Shandong First Medical University, Shandong, China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Shandong, China.
| |
Collapse
|
35
|
García-Patiño MG, Marcial-Medina MC, Ruiz-Medina BE, Licona-Limón P. IL-17 in skin infections and homeostasis. Clin Immunol 2024; 267:110352. [PMID: 39218195 DOI: 10.1016/j.clim.2024.110352] [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: 05/10/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Interleukin (IL) 17 is a proinflammatory cytokine belonging to a structurally related group of cytokines known as the IL-17 family. It has been profoundly studied for its contribution to the pathology of autoimmune diseases. However, it also plays an important role in homeostasis and the defense against extracellular bacteria and fungi. IL-17 is important for epithelial barriers, including the skin, where some of its cellular targets reside. Most of the research work on IL-17 has focused on its effects in the skin within the context of autoimmune diseases or sterile inflammation, despite also having impact on other skin conditions. In recent years, studies on the role of IL-17 in the defense against skin pathogens and in the maintenance of skin homeostasis mediated by the microbiota have grown in importance. Here we review and discuss the cumulative evidence regarding the main contribution of IL-17 in the maintenance of skin integrity as well as its protective or pathogenic effects during some skin infections.
Collapse
Affiliation(s)
- M G García-Patiño
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - M C Marcial-Medina
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - B E Ruiz-Medina
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - P Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
36
|
Hashempour A, Khodadad N, Bemani P, Ghasemi Y, Akbarinia S, Bordbari R, Tabatabaei AH, Falahi S. Design of multivalent-epitope vaccine models directed toward the world's population against HIV-Gag polyprotein: Reverse vaccinology and immunoinformatics. PLoS One 2024; 19:e0306559. [PMID: 39331650 PMCID: PMC11432917 DOI: 10.1371/journal.pone.0306559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/18/2024] [Indexed: 09/29/2024] Open
Abstract
Significant progress has been made in HIV-1 research; however, researchers have not yet achieved the objective of eradicating HIV-1 infection. Accordingly, in this study, eucaryotic and procaryotic in silico vaccines were developed for HIV-Gag polyproteins from 100 major HIV subtypes and CRFs using immunoinformatic techniques to simulate immune responses in mice and humans. The epitopes located in the conserved domains of the Gag polyprotein were evaluated for allergenicity, antigenicity, immunogenicity, toxicity, homology, topology, and IFN-γ induction. Adjuvants, linkers, CTLs, HTLs, and BCL epitopes were incorporated into the vaccine models. Strong binding affinities were detected between HLA/MHC alleles, TLR-2, TLR-3, TLR-4, TLR-7, and TLR-9, and vaccine models. Immunological simulation showed that innate and adaptive immune cells elicited active and consistent responses. The human vaccine model was matched with approximately 93.91% of the human population. The strong binding of the vaccine to MHC/HLA and TLR molecules was confirmed through molecular dynamic stimulation. Codon optimization ensured the successful translation of the designed constructs into human cells and E. coli hosts. We believe that the HIV-1 Gag vaccine formulated in our research can reduce the challenges faced in developing an HIV-1 vaccine. Nevertheless, experimental verification is necessary to confirm the effectiveness of these vaccines in these models.
Collapse
Affiliation(s)
- Ava Hashempour
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nastaran Khodadad
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Peyman Bemani
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shokufeh Akbarinia
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Bordbari
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Hossein Tabatabaei
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahab Falahi
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
37
|
Massaut KB, Vitola HRS, Gonçalves VS, Leite FPL, Jardim RD, Moreira ÂN, da Silva WP, Fiorentini ÂM. Administration of Lacticaseibacillus casei CSL3 in Swiss Mice with Immunosuppression Induced by Cyclophosphamide: Effects on Immunological, Biochemical, Oxidative Stress, and Histological Parameters. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10362-9. [PMID: 39313704 DOI: 10.1007/s12602-024-10362-9] [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] [Accepted: 09/08/2024] [Indexed: 09/25/2024]
Abstract
The study aimed to evaluate the effects of supplementation with Lacticaseibacillus casei CSL3 in Swiss mice immunosuppressed with cyclophosphamide on immunological, biochemical, oxidative stress, and histological parameters. The animals were distributed into four groups (control, CSL3, cyclophosphamide, and CSL3 + cyclophosphamide), where two groups were treated with L. casei CSL3 (10 log CFU mL-1) for 30 days, and two groups received chemotherapy (days 27 and 30-total dose of 250 mg kg-1). Counts of lactic acid bacteria (LAB) and bile-resistant LAB in stool samples; blood count (erythrogram, leukogram, and platelets); serum total cholesterol levels; catalase enzyme activity; and thiobarbituric acid reactive substances (TBARS) levels in liver, kidney, and brain; IL-4 expression; IL-23, TNF-α, NF-κβ in the spleen; and histological changes in the liver, kidneys, and intestine were evaluated. The CSL3 + cyclophosphamide group showed a significant increase in bile-resistant LAB counts in feces (p = 0.0001), leukocyte counts, and expression of IL-23, TNF-α, and NF-κβ (p < 0.05) significantly reduced total cholesterol levels (p = 0.001) and protected liver damage of supplemented animals. For oxidative stress damage, the bacterium did not influence the results. It is concluded that the bacterium is safe at a concentration of 10 log CFU mL-1 and has probiotic potential due to its positive influence on the immune response and lipid metabolism.
Collapse
Affiliation(s)
- Khadija Bezerra Massaut
- Laboratory of Food Microbiology, Department of Agroindustrial Science and Technology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Helena Reissing Soares Vitola
- Laboratory of Food Microbiology, Department of Agroindustrial Science and Technology, Federal University of Pelotas, Pelotas, RS, Brazil
| | | | | | - Rodrigo Desessards Jardim
- Histology Laboratory, Institute of Biological Sciences, Federal University of Rio Grande, Rio Grande, RS, Brazil
| | - Ângela Nunes Moreira
- Laboratory of Applied Immunology, Biotechnology Unit, Technology Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Wladimir Padilha da Silva
- Laboratory of Food Microbiology, Department of Agroindustrial Science and Technology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Ângela Maria Fiorentini
- Laboratory of Food Microbiology, Department of Agroindustrial Science and Technology, Federal University of Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
38
|
Abacar K, Macleod T, Direskeneli H, McGonagle D. How underappreciated autoinflammatory (innate immunity) mechanisms dominate disparate autoimmune disorders. Front Immunol 2024; 15:1439371. [PMID: 39372419 PMCID: PMC11449752 DOI: 10.3389/fimmu.2024.1439371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Historically inflammation against self was considered autoimmune which stems back to the seminal observations by Ehrlich who described serum factors, now known to be autoantibodies produced by B lineage cells that mediate "horror autotoxicus". The 20th century elucidation of B- and T-cell adaptive immune responses cemented the understanding of the key role of adaptive immune responses in mediating pathology against self. However, Mechnikov shared the Nobel Prize for the discovery of phagocytosis, the most rudimentary aspect of innate immunity. Fast forward some 100 years and an immunogenetic understanding of innate immunity led to the categorising of innate immunopathology under the umbrella term 'auto inflammation' and terminology such as "horror autoinflammaticus" to highlight the schism from the classical adaptive immune understanding of autoimmunity. These concepts lead to calls for a two-tiered classification of inflammation against self, but just as innate and adaptive immunity are functionally integrated, so is immunopathology in many settings and the concept of an autoimmune to autoinflammation continuum emerged with overlaps between both. Herein we describe several historically designated disorders of adaptive immunity where innate immunity is key, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic juvenile idiopathic arthritis (sJIA) and adult-onset Still's disease (AOSD) where the immunopathology phenotype is strongly linked to major histocompatibility complex (MHC) class II associations and responds to drugs that target T-cells. We also consider MHC-I-opathies including psoriasis and Behcet's disease(BD) that are increasingly viewed as archetype CD8 T-cell related disorders. We also briefly review the key role of barrier dysfunction in eczema and ulcerative colitis (UC) where innate tissue permeability barrier dysfunction and microbial dysbiosis contributes to prominent adaptive immune pathological mechanisms. We also highlight the emerging roles of intermediate populations of lymphocytes including gamma delta (γδ) and mucosal-associated invariant T (MAIT) cells that represent a blend of adaptive immune plasticity and innate immune rapid responders that may also determine site specific patterns of inflammation.
Collapse
Affiliation(s)
- Kerem Abacar
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Tom Macleod
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
| | - Haner Direskeneli
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Dennis McGonagle
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| |
Collapse
|
39
|
Mondell E, Nino G, Hong X, Wang X, Gutierrez MJ. Immune Biomarkers at Birth Predict Lower Respiratory Tract Infection Risk in a Large Birth Cohort. Pathogens 2024; 13:765. [PMID: 39338956 PMCID: PMC11435078 DOI: 10.3390/pathogens13090765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Lower respiratory tract infections (LRTIs) remain the leading cause of infant morbidity and mortality worldwide and affect long-term respiratory health. Identifying immunological determinants of LRTI susceptibility may help stratify disease risk and identify therapies. This study aimed to identify neonatal immunological factors predicting LRTI risk in infancy. Cord blood plasma from 191 neonates from the Boston Birth Cohort was analyzed for 28 soluble immune factors. LRTI was defined as bronchiolitis, bronchitis, or pneumonia during the first year of life. Welch's t-test demonstrated significantly higher log10 transformed concentrations of IL-17 and IFNγ in the LRTI group compared to neonates without LRTI in the first year of life (p < 0.05). Risk associations were determined using multivariate survival models. There were 29 infants with LRTIs. High cord blood levels of IFNγ (aHR = 2.35, 95% CI 1.07-5.17), TNF-β (aHR = 2.86, 95% CI 1.27-6.47), MIP-1α (aHR = 2.82, 95% CI 1.22-6.51), and MIP-1β (aHR = 2.34, 95% CI 1.05-5.20) were associated with a higher risk of LRTIs. RANTES was associated with a lower risk (aHR = 0.43, 95% CI 0.19-0.97). Soluble immune factors linked to antiviral immunity (IFNγ) and cytokines mediating inflammatory responses (TNF-β), and cell homing (MIP-1α/b), at birth were associated with an increased risk of LRTIs during infancy.
Collapse
Affiliation(s)
- Ethan Mondell
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Gustavo Nino
- Division of Pulmonary and Sleep Medicine, Children’s National Hospital, George Washington University, Washington, DC 20010, USA;
- Center for Genetic Medicine Research, Children’s Research Institute, Washington, DC 20010, USA
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (X.H.); (X.W.)
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (X.H.); (X.W.)
- Division of General Pediatrics and Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Maria J. Gutierrez
- Division of Pediatric Allergy, Immunology and Rheumatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
40
|
Cui Y, Hackett RG, Ascue J, Muralidaran V, Patil D, Kang J, Kaufman SS, Khan K, Kroemer A. Innate and Adaptive Immune Responses in Intestinal Transplant Rejection: Through the Lens of Inflammatory Bowel and Intestinal Graft-Versus-Host Diseases. Gastroenterol Clin North Am 2024; 53:359-382. [PMID: 39068000 DOI: 10.1016/j.gtc.2024.01.002] [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] [Indexed: 07/30/2024]
Abstract
Intestinal transplantation is a life-saving procedure utilized for patients failing total parenteral nutrition. However, intestinal transplantattion remains plagued with low survival rates and high risk of allograft rejection. The authors explore roles of innate (macrophages, natural killer cells, innate lymphoid cells) and adaptive immune cells (Th1, Th2, Th17, Tregs) in inflammatory responses, particularly inflammatory bowel disease and graft versus host disease, and correlate these findings to intestinal allograft rejection, highlighting which effectors exacerbate or suppress intestinal rejection. Better understanding of this immunology can open further investigation into potential biomolecular targets to develop improved therapeutic treatment options and immunomonitoring techniques to combat allograft rejection and enhance patient lives.
Collapse
Affiliation(s)
- Yuki Cui
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Ryan G Hackett
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jhalen Ascue
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Vinona Muralidaran
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Digvijay Patil
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Stuart S Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
41
|
Hazzard AA, McCrorey M, Salman T, Johnson DE, Luo Z, Fu X, Keegan AP, Benitez A, Fitting S, Jiang W. Cannabis use, oral dysbiosis, and neurological disorders. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:183-193. [PMID: 39741560 PMCID: PMC11683879 DOI: 10.1515/nipt-2024-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 01/03/2025]
Abstract
Cannabis (marijuana) is a leafy plant that has medical, recreational, and other uses. Cannabis is socially accepted and widely used throughout the United States. Though cannabis use is increasingly gaining popularity, studies detail the deleterious effects of chronic cannabis smoking on mental health, as well as the immunosuppressive properties of cannabinoids. Additionally, oral dysbiosis induced by cannabis smoking serves as a novel catalyst for neurological abnormalities, potentially possible through microbial translocation via the oral-brain axis. This review summarizes the effects and link of smoking cannabis on neurological abnormalities, immunity, and oral microbiome.
Collapse
Affiliation(s)
- Amber A. Hazzard
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Marice McCrorey
- Department of Molecular and Cellular Biology and Pathobiology, Medical University of South Carolina, Charleston, SC, USA
| | - Tabinda Salman
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Douglas E. Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaoyu Fu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Andrew P. Keegan
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | - Andreana Benitez
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
- Divison of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
42
|
Ahamed F, Eppler N, Jones E, Zhang Y. Understanding Macrophage Complexity in Metabolic Dysfunction-Associated Steatotic Liver Disease: Transitioning from the M1/M2 Paradigm to Spatial Dynamics. LIVERS 2024; 4:455-478. [PMID: 39328386 PMCID: PMC11426415 DOI: 10.3390/livers4030033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses metabolic dysfunction-associated fatty liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), with MASH posing a risk of progression to cirrhosis and hepatocellular carcinoma (HCC). The global prevalence of MASLD is estimated at approximately a quarter of the population, with significant healthcare costs and implications for liver transplantation. The pathogenesis of MASLD involves intrahepatic liver cells, extrahepatic components, and immunological aspects, particularly the involvement of macrophages. Hepatic macrophages are a crucial cellular component of the liver and play important roles in liver function, contributing significantly to tissue homeostasis and swift responses during pathophysiological conditions. Recent advancements in technology have revealed the remarkable heterogeneity and plasticity of hepatic macrophage populations and their activation states in MASLD, challenging traditional classification methods like the M1/M2 paradigm and highlighting the coexistence of harmful and beneficial macrophage phenotypes that are dynamically regulated during MASLD progression. This complexity underscores the importance of considering macrophage heterogeneity in therapeutic targeting strategies, including their distinct ontogeny and functional phenotypes. This review provides an overview of macrophage involvement in MASLD progression, combining traditional paradigms with recent insights from single-cell analysis and spatial dynamics. It also addresses unresolved questions and challenges in this area.
Collapse
Affiliation(s)
- Forkan Ahamed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Natalie Eppler
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Elizabeth Jones
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| |
Collapse
|
43
|
Gao Y, Zhou Y, Lin Z, Chen F, Wu H, Peng C, Xie Y. Prioritizing drug targets in systemic lupus erythematosus from a genetic perspective: a druggable genome-wide Mendelian randomization study. Clin Rheumatol 2024; 43:2843-2856. [PMID: 38997544 PMCID: PMC11330408 DOI: 10.1007/s10067-024-07059-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/05/2024] [Accepted: 07/04/2024] [Indexed: 07/14/2024]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease with an unsatisfactory state of treatment. We aim to explore novel targets for SLE from a genetic standpoint. METHODS Cis-expression quantitative trait loci (eQTLs) for whole blood from 31,684 samples provided by the eQTLGen Consortium as well as two large SLE cohorts were utilized for screening and validating genes causally associated with SLE. Colocalization analysis was employed to further investigate whether changes in the expression of risk genes, as indicated by GWAS signals, influence the occurrence and development of SLE. Targets identified for drug development were evaluated for potential side effects using a phenome-wide association study (PheWAS). Based on the multiple databases, we explored the interactions between drugs and genes for drug prediction and the assessment of current medications. RESULTS The analysis comprised 5427 druggable genes in total. The two-sample Mendelian randomization (MR) in the discovery phase identified 20 genes causally associated with SLE and validated 8 genes in the replication phase. Colocalization analysis ultimately identified five genes (BLK, HIST1H3H, HSPA1A, IL12A, NEU1) with PPH4 > 0.8. PheWAS further indicated that drugs acting on BLK and IL12A are less likely to have potential side effects, while HSPA1A and NEU1 were associated with other traits. Four genes (BLK, HSPA1A, IL12A, NEU1) have been targeted for drug development in autoimmune diseases and other conditions. CONCLUSIONS .This study identified five genes as therapeutic targets for SLE. Repurposing and developing drugs targeting these genes is anticipated to improve the existing treatment state for SLE. Key Points • We identified five gene targets of priority for the treatment of SLE, with BLK and IL12A indicating fewer side effects. • Among the existing drugs that target these candidate genes, Ustekinumab, Ebdarokimab, and Briakinumab (targeting the IL12 gene) and CD24FC (targeting HSPA1A) may potentially be repurposed for the treatment of SLE.
Collapse
Affiliation(s)
- Yuan Gao
- The First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Youtao Zhou
- The First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Zikai Lin
- Nanshan College, Guangzhou Medical University, Guangzhou, China
| | - Fengzhen Chen
- The First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Haiyang Wu
- The Second Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Chusheng Peng
- The First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Yingying Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 151, Yanjiang West Road, Yuexiu District, Guangzhou, China.
| |
Collapse
|
44
|
Hashempour A, Khodadad N, Akbarinia S, Ghasabi F, Ghasemi Y, Nazar MMKA, Falahi S. Reverse vaccinology approaches to design a potent multiepitope vaccine against the HIV whole genome: immunoinformatic, bioinformatics, and molecular dynamics approaches. BMC Infect Dis 2024; 24:873. [PMID: 39198721 PMCID: PMC11360854 DOI: 10.1186/s12879-024-09775-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Substantial advances have been made in the development of promising HIV vaccines to eliminate HIV-1 infection. For the first time, one hundred of the most submitted HIV subtypes and CRFs were retrieved from the LANL database, and the consensus sequences of the eleven HIV proteins were obtained to design vaccines for human and mouse hosts. By using various servers and filters, highly qualified B-cell epitopes, as well as HTL and CD8 + epitopes that were common between mouse and human alleles and were also located in the conserved domains of HIV proteins, were considered in the vaccine constructs. With 90% coverage worldwide, the human vaccine model covers a diverse allelic population, making it widely available. Codon optimization and in silico cloning in prokaryotic and eukaryotic vectors guarantee high expression of the vaccine models in human and E. coli hosts. Molecular dynamics confirmed the stable interaction of the vaccine constructs with TLR3, TLR4, and TLR9, leading to a substantial immunogenic response to the designed vaccine. Vaccine models effectively target the humoral and cellular immune systems in humans and mice; however, experimental validation is needed to confirm these findings in silico.
Collapse
Affiliation(s)
- Ava Hashempour
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nastaran Khodadad
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Shokufeh Akbarinia
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzane Ghasabi
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Shahab Falahi
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
45
|
Gordon S, Roberti A, Kaufmann SHE. Mononuclear Phagocytes, Cellular Immunity, and Nobel Prizes: A Historic Perspective. Cells 2024; 13:1378. [PMID: 39195266 PMCID: PMC11352343 DOI: 10.3390/cells13161378] [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: 07/19/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
The mononuclear phagocyte system includes monocytes, macrophages, some dendritic cells, and multinuclear giant cells. These cell populations display marked heterogeneity depending on their differentiation from embryonic and bone marrow hematopoietic progenitors, tissue location, and activation. They contribute to tissue homeostasis by interacting with local and systemic immune and non-immune cells through trophic, clearance, and cytocidal functions. During evolution, they contributed to the innate host defense before effector mechanisms of specific adaptive immunity emerged. Mouse macrophages appear at mid-gestation and are distributed throughout the embryo to facilitate organogenesis and clear cells undergoing programmed cell death. Yolk sac, AGM, and fetal liver-derived tissue-resident macrophages persist throughout postnatal and adult life, supplemented by bone marrow-derived blood monocytes, as required after injury and infection. Nobel awards to Elie Metchnikoff and Paul Ehrlich in 1908 drew attention to cellular phagocytic and humoral immunity, respectively. In 2011, prizes were awarded to Jules Hoffmann and Bruce Beutler for contributions to innate immunity and to Ralph Steinman for the discovery of dendritic cells and their role in antigen presentation to T lymphocytes. We trace milestones in the history of mononuclear phagocyte research from the perspective of Nobel awards bearing directly and indirectly on their role in cellular immunity.
Collapse
Affiliation(s)
- Siamon Gordon
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Annabell Roberti
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, 10117 Berlin, Germany;
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX 77843, USA
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
46
|
Sun X, Gu R, Bai J. Differentiation and regulation of CD4 + T cell subsets in Parkinson's disease. Cell Mol Life Sci 2024; 81:352. [PMID: 39153043 PMCID: PMC11335276 DOI: 10.1007/s00018-024-05402-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and its hallmark pathological features are the loss of dopaminergic (DA) neurons in the midbrain substantia nigra pars compacta (SNpc) and the accumulation of alpha-synuclein (α-syn). It has been shown that the integrity of the blood-brain barrier (BBB) is damaged in PD patients, and a large number of infiltrating T cells and inflammatory cytokines have been detected in the cerebrospinal fluid (CSF) and brain parenchyma of PD patients and PD animal models, including significant change in the number and proportion of different CD4+ T cell subsets. This suggests that the neuroinflammatory response caused by CD4+ T cells is an important risk factor for the development of PD. Here, we systematically review the differentiation of CD4+ T cell subsets, and focus on describing the functions and mechanisms of different CD4+ T cell subsets and their secreted cytokines in PD. We also summarize the current immunotherapy targeting CD4+ T cells with a view to providing assistance in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
- Southwest United Graduate School, Kunming, 650500, China
| | - Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
- Southwest United Graduate School, Kunming, 650500, China.
| |
Collapse
|
47
|
Sanna FC, Benešová I, Pervan P, Krenz A, Wurzel A, Lohmayer R, Mühlbauer J, Wöllner A, Köhl N, Menevse AN, Stamova S, Volpin V, Beckhove P, Xydia M. IL-2 and TCR stimulation induce expression and secretion of IL-32β by human T cells. Front Immunol 2024; 15:1437224. [PMID: 39211051 PMCID: PMC11357969 DOI: 10.3389/fimmu.2024.1437224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
IL-32 expression is important for pathogen clearance but detrimental in chronic inflammation, autoimmunity, and cancer. T cells are major IL-32 producers in these diseases and key mediators of pathogen and tumor elimination but also autoimmune destruction. However, their contribution to IL-32 biology during immune responses is hardly understood due to several isoforms with divergent inflammatory properties. Here, we identified IL-32β as the predominant isoform in various T cell subsets of healthy individuals and breast cancer patients with the highest levels detected in intratumoral regulatory T cells. We show that IL-32β is induced by IL-2 but IL-32β release requires T Cell Receptor rather than IL2R stimulation. Using inhibitors of protein secretion pathways and serial (ultra)centrifugation of T cell supernatants, we demonstrate that T cells actively secrete IL-32β unconventionally, as a free protein and, to a minor degree, through exosomes. Thus, our data identify activated T cells as major IL-32β secretors in health and cancer.
Collapse
Affiliation(s)
| | - Iva Benešová
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Philip Pervan
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Adriana Krenz
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Alexander Wurzel
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Robert Lohmayer
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Algorithmic Bioinformatics, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Jasmin Mühlbauer
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Amélie Wöllner
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Nina Köhl
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Ayse Nur Menevse
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Slava Stamova
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Valentina Volpin
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Philipp Beckhove
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Maria Xydia
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| |
Collapse
|
48
|
Enriquez GF, Macchiaverna NP, Garbossa G, Quebrada Palacio LP, Ojeda BL, Bua J, Gaspe MS, Cimino R, Gürtler RE, Postan M, Cardinal MV. Humans seropositive for Trypanosoma cruzi co-infected with intestinal helminths have higher infectiousness, parasitaemia and Th2-type response in the Argentine Chaco. Parasit Vectors 2024; 17:340. [PMID: 39135121 PMCID: PMC11320973 DOI: 10.1186/s13071-024-06401-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The Gran Chaco ecoregion is a well-known hotspot of several neglected tropical diseases (NTDs) including Chagas disease, soil-transmitted helminthiasis and multiparasitic infections. Interspecific interactions between parasite species can modify host susceptibility, pathogenesis and transmissibility through immunomodulation. Our objective was to test the association between human co-infection with intestinal parasites and host parasitaemia, infectiousness to the vector and immunological profiles in Trypanosoma cruzi-seropositive individuals residing in an endemic region of the Argentine Chaco. METHODS We conducted a cross-sectional serological survey for T. cruzi infection along with an intestinal parasite survey in two adjacent rural villages. Each participant was tested for T. cruzi and Strongyloides stercoralis infection by serodiagnosis, and by coprological tests for intestinal parasite detection. Trypanosoma cruzi bloodstream parasite load was determined by quantitative PCR (qPCR), host infectiousness by artificial xenodiagnosis and serum human cytokine levels by flow cytometry. RESULTS The seroprevalence for T. cruzi was 16.1% and for S. stercoralis 11.5% (n = 87). We found 25.3% of patients with Enterobius vermicularis. The most frequent protozoan parasites were Blastocystis spp. (39.1%), Giardia lamblia (6.9%) and Cryptosporidium spp. (3.4%). Multiparasitism occurred in 36.8% of the examined patients. Co-infection ranged from 6.9% to 8.1% for T. cruzi-seropositive humans simultaneously infected with at least one protozoan or helminth species, respectively. The relative odds of being positive by qPCR or xenodiagnosis (i.e. infectious) of 28 T. cruzi-seropositive patients was eight times higher in people co-infected with at least one helminth species than in patients with no such co-infection. Trypanosoma cruzi parasite load and host infectiousness were positively associated with helminth co-infection in a multiple regression analysis. Interferon-gamma (IFN-γ) response, measured in relation to interleukin (IL)-4 among humans infected with T. cruzi only, was 1.5-fold higher than for T. cruzi-seropositive patients co-infected with helminths. The median concentration of IL-4 was significantly higher in T. cruzi-seropositive patients with a positive qPCR test than in qPCR-negative patients. CONCLUSIONS Our results show a high level of multiparasitism and suggest that co-infection with intestinal helminths increased T. cruzi parasitaemia and upregulated the Th2-type response in the study patients.
Collapse
Affiliation(s)
- Gustavo Fabián Enriquez
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina.
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Natalia Paula Macchiaverna
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Graciela Garbossa
- Laboratorio de Parasitología Clínica y Ambiental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET-UBA), Instituto de Investigaciones en Salud Pública, Buenos Aires, Argentina
| | - Luz Piedad Quebrada Palacio
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Institute of Virology, Helmholtz Centre Munich, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Bárbara Leonor Ojeda
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
| | - Jacqueline Bua
- Instituto Nacional de Parasitología Dr. M. Fatala Chabén, Administración Nacional de Laboratorios e Institutos de Salud Dr. C.G. Malbrán, Buenos Aires, Argentina
| | - María Sol Gaspe
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rubén Cimino
- Instituto de Investigaciones de Enfermedades Tropicales (IIET). Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-CCT Salta, Universidad Nacional de Salta, Sede Regional Orán, Salta, Argentina
- Facultad de Ciencias Naturales, Cátedra de Química Biológica, Universidad Nacional de Salta, Salta, Argentina
| | - Ricardo Esteban Gürtler
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Miriam Postan
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Marta Victoria Cardinal
- Universidad de Buenos Aires., Facultad de Ciencias Exactas y Naturales. Departamento de Ecología, Genética y Evolución. Laboratorio de Eco-Epidemiología., Buenos Aires, Argentina
- Instituto de Ecología, Genética y Evolución (IEGEBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
49
|
Cui X, Vervaeke P, Gao Y, Opsomer L, Sun Q, Snoeck J, Devriendt B, Zhong Z, Sanders NN. Immunogenicity and biodistribution of lipid nanoparticle formulated self-amplifying mRNA vaccines against H5 avian influenza. NPJ Vaccines 2024; 9:138. [PMID: 39097672 PMCID: PMC11298010 DOI: 10.1038/s41541-024-00932-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/17/2024] [Indexed: 08/05/2024] Open
Abstract
This study reports on the immunogenicity and biodistribution of H5 hemagglutinin (HA)-based self-amplifying (sa) mRNA vaccines in mice. Four sa-mRNA vaccines encoding either a secreted full-length HA, a secreted HA head domain, a secreted HA stalk domain, or a full-length membrane-anchored HA were investigated. All vaccines elicited an adaptive immune response. However, the full-length HA sa-RNA vaccines demonstrated superior performance compared to head and stalk domain vaccines. The antibody titers positively correlated with the vaccine dose. Cellular immune responses and antigen-specific IgA antibodies in the lungs were also observed. The comparison of the sa-mRNA vaccines encoding the secreted and membrane-anchored full-length HA revealed that anchoring of the HA to the membrane significantly enhanced the antibody and cellular responses. In addition to the injection site, the intramuscularly injected sa-mRNA-LNPs were also detected in the draining lymph nodes, spleen, and to a lesser extent, in the lung, kidney, liver, and heart.
Collapse
Affiliation(s)
- Xiaole Cui
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Pieter Vervaeke
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Ya Gao
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, B-9820, Merelbeke, Belgium
| | - Lisa Opsomer
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Qing Sun
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Janne Snoeck
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Bert Devriendt
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, B-9820, Merelbeke, Belgium
| | - Zifu Zhong
- Department of Pharmaceutics, Ghent University, Ghent, Belgium.
- Cancer Research Institute (CRIG), Ghent University, 9000, Ghent, Belgium.
| | - Niek N Sanders
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium.
- Cancer Research Institute (CRIG), Ghent University, 9000, Ghent, Belgium.
| |
Collapse
|
50
|
Luo J, Chen W, Liu W, Jiang S, Ye Y, Shrimanker R, Hynes G, Klenerman P, Pavord ID, Xue L. IL-5 antagonism reverses priming and activation of eosinophils in severe eosinophilic asthma. Mucosal Immunol 2024; 17:524-536. [PMID: 38493955 PMCID: PMC11649845 DOI: 10.1016/j.mucimm.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Eosinophils are key effector cells mediating airway inflammation and exacerbation in patients with severe eosinophilic asthma. They are present in increased numbers and activation states in the airway mucosa and lumen. Interleukin-5 (IL-5) is the key eosinophil growth factor that is thought to play a role in eosinophil priming and activation. However, the mechanism of these effects is still not fully understood. The anti-IL-5 antibody mepolizumab reduces eosinophil counts in the airway modestly but has a large beneficial effect on the frequency of exacerbations of severe eosinophilic asthma, suggesting that reduction in eosinophil priming and activation is of central mechanistic importance. In this study, we used the therapeutic effect of mepolizumab and single-cell ribonucleic acid sequencing to investigate the mechanism of eosinophil priming and activation by IL-5. We demonstrated that IL-5 is a dominant driver of eosinophil priming and plays multifaceted roles in eosinophil function. It enhances eosinophil responses to other stimulators of migration, survival, and activation by activating phosphatidylinositol-3-kinases, extracellular signal-regulated kinases, and p38 mitogen-activated protein kinases signaling pathways. It also enhances the pro-fibrotic roles of eosinophils in airway remodeling via transforming growth factor-β pathway. These findings provide a mechanistic understanding of eosinophil priming in severe eosinophilic asthma and the therapeutic effect of anti-IL-5 approaches in the disease.
Collapse
Affiliation(s)
- Jian Luo
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom.
| | - Wentao Chen
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Wei Liu
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Division of Pulmonary Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Jiang
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuan Ye
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Rahul Shrimanker
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Gareth Hynes
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Translational Gastroenterology Unit and Peter Medawar Building, University of Oxford, Oxford, United Kingdom
| | - Ian D Pavord
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Luzheng Xue
- Respiratory Medicine Unit and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|