1
|
Uemura I, Takahashi-Suzuki N, Satoh T. Impact of afatinib on intestinal and salivary IgA: Immune response alterations linked to gastrointestinal side effects. Immunol Lett 2025; 275:107024. [PMID: 40228698 DOI: 10.1016/j.imlet.2025.107024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Afatinib, an oral molecular-targeted anticancer agent, is effective but causes significant gastrointestinal side effects. These effects are associated with EGFR inhibition in intestinal cells and changes in the microbiota. OBJECTIVE To investigate the effects of afatinib on intestinal mucosal immunity in rats, focusing on IgA levels in the intestine and saliva, and to understand the innate and acquired immune responses to these side effects. METHODS Male Wistar rats received afatinib (5.2 mg/kg) daily for 24 h (Day 1) and for 2 weeks (Day 14). Gene expression in the intestine was analyzed using quantitative polymerase chain reaction. IgA levels in the intestine and saliva were measured using enzyme-linked immunosorbent assay. RESULTS Afatinib suppressed α-defensin 5 and pIgR in the jejunum and ileum, indicating reduced innate immunity. It increased IgA levels in the intestine and saliva, suggesting altered acquired immunity. Salivary IgA levels significantly correlated with intestinal IgA levels. CONCLUSIONS Afatinib affects gastrointestinal mucosal immunity, suppresses innate defense, and alters IgA production. Salivary IgA could serve as a marker for monitoring these effects, aiding cancer therapy management.
Collapse
Affiliation(s)
- Ippei Uemura
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine-ku, Sapporo, Hokkaido 006-8585, Japan.
| | - Natsuko Takahashi-Suzuki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine-ku, Sapporo, Hokkaido 006-8585, Japan.
| | - Takashi Satoh
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine-ku, Sapporo, Hokkaido 006-8585, Japan.
| |
Collapse
|
2
|
Son SU, Suh HJ, Shin KS. Effects of novel sulfated-rhamnoglucuronan isolated from Korean seaweed Ulva pertusa on IgA-mediated intestinal-immune system activation in naïve C3H/HeN mice. Int J Biol Macromol 2025; 314:144170. [PMID: 40373895 DOI: 10.1016/j.ijbiomac.2025.144170] [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: 04/01/2025] [Accepted: 05/11/2025] [Indexed: 05/17/2025]
Abstract
A previous study identified a novel sulfated-rhamnoglucuronan structure in Ulva pertusa polysaccharide (UPP). The present study evaluates the intestinal immunostimulatory effects of UPP in naïve C3H/HeN mice. Oral administration of UPP significantly enhanced the secretion of IgA and associated cytokines, including interleukin (IL)-6, IL-10, and transforming growth factor (TGF)-β, from Peyer's patch (PP) cells in vivo. Additionally, the supernatant from PP cell cultures stimulated the proliferation of bone marrow cells. Histological analysis revealed strong stimulation of PP by UPP, as evidenced by hematoxylin and eosin staining. Serum analysis indicated that UPP triggered TGF-β secretion, which subsequently promoted IgA production. Furthermore, UPP administration enhanced IgA-related cytokine production in ileum tissues and was linked to the activation of the mitogen-activated protein kinase and nuclear factor kappa B signaling pathways. The secretion of A proliferation-inducing ligand (tumor necrosis factor superfamily member 13; TNFSF13) and B-cell activating factor (TNFSF13B) was also upregulated in the ileum. Additionally, fecal analysis demonstrated an increase in short-chain fatty acids, including acetic, propionic, and butyric acids, in a dose- and administration period-dependent manner. These findings suggest that UPP administration contributes IgA-related immune responses and intestinal immune system modulation via PP cells.
Collapse
Affiliation(s)
- Seung-U Son
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea; Precision Nutrition Research Group, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea.
| | - Hyung Joo Suh
- Transdisciplinary Major in Learning Health System, Department of Integrated Biomedical and Life Science, Korea University, Seoul 02841, Republic of Korea.
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea.
| |
Collapse
|
3
|
Kwon DI, Mao T, Israelow B, Santos Guedes de Sá K, Dong H, Iwasaki A. Mucosal unadjuvanted booster vaccines elicit local IgA responses by conversion of pre-existing immunity in mice. Nat Immunol 2025:10.1038/s41590-025-02156-0. [PMID: 40360777 DOI: 10.1038/s41590-025-02156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025]
Abstract
Mucosal delivery of vaccine boosters induces robust local protective immune responses even without any adjuvants. Yet, the mechanisms by which antigen alone induces mucosal immunity in the respiratory tract remain unclear. Here we show that an intranasal booster with an unadjuvanted recombinant SARS-CoV-2 spike protein, after intramuscular immunization with 1 μg of mRNA-LNP vaccine encoding the full-length SARS-CoV-2 spike protein (Pfizer/BioNTech BNT162b2), elicits protective mucosal immunity by retooling the lymph node-resident immune cells. On intranasal boosting, peripheral lymph node-primed B cells rapidly migrated to the lung through CXCR3-CXCL9 and CXCR3-CXCL10 signaling and differentiated into antigen-specific IgA-secreting plasma cells. Memory CD4+ T cells in the lung served as a natural adjuvant for developing mucosal IgA by inducing the expression of chemokines CXCL9 and CXCL10 for memory B cell recruitment. Furthermore, CD40 and TGFβ signaling had important roles in mucosal IgA development. Repeated mucosal boosting with an unadjuvanted protein amplified anamnestic IgA responses in both the upper and the lower respiratory tracts. These findings help explain why nasal boosters do not require an adjuvant to induce robust mucosal immunity at the respiratory mucosa and can be used to design safe and effective vaccines against respiratory pathogens.
Collapse
Affiliation(s)
- Dong-Il Kwon
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Keyla Santos Guedes de Sá
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Huiping Dong
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
4
|
Dang S, Zhang X, Zhang Y, Zhang H. New thoughts on the intestinal microbiome-B cell-IgA axis and therapies in IgA nephropathy. Autoimmun Rev 2025; 24:103835. [PMID: 40360014 DOI: 10.1016/j.autrev.2025.103835] [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: 03/21/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
IgA nephropathy (IgAN), as the most common chronic glomerulonephritis worldwide, is often triggered by mucosal infections and follows a chronic progression, with the majority of patients ultimately progressing to end-stage renal disease (ESRD) during their lifetimes. Since the mystery of its complete pathogenesis has not been fully solved, the resulting lack of effective early diagnosis and treatment greatly affects the prognosis of patients. Given the well-defined pathological feature of IgA deposition in the mesangial region, the source and role of pathogenic IgA has been focused on. Starting from the microbiology and immunity of the gut, we systematically review both the physiological and the pathological process of microbiome-B cell-IgA axis, from microbial-induced IgA production to the role of IgA in the intestinal immune milieu, and ultimately end up with the various aspects of microbiome-B cell-IgA axis in the pathogenesis of IgAN as well as the corresponding therapeutic initiatives available. Our retrospective review helps researchers to systematically understand the complex role between intestinal flora dysbiosis and pathogenic IgA in IgAN. This understanding provides a foundation for in-depth explorations to uncover more detailed pathogenic mechanisms and to develop more precise and effective diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Shaoqing Dang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangyu Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yuemiao Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Alimohammadi M, Kahkesh S, Abbasi A, Hashemi M, Khoshnazar SM, Taheriazam A, Hushmandi K. LncRNAs and IgA nephropathy: underlying molecular pathways and clinical applications. Clin Exp Med 2025; 25:140. [PMID: 40328979 PMCID: PMC12055897 DOI: 10.1007/s10238-025-01660-9] [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: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025]
Abstract
IgA nephropathy (IgAN), also known as Berger's disease, is a prevalent kidney disorder caused by the accumulation of IgA antibodies in the glomerular tissue. Long noncoding RNAs (lncRNAs), a class of noncoding RNAs longer than 200 nucleotides, play crucial roles in regulating various cellular and molecular processes, including translation, chromatin remodeling, and transcriptional efficiency. Research has highlighted the significant impact of lncRNA imbalances on the development and progression of kidney diseases, including IgAN. These molecules influence several key signaling pathways, such as PI3K/AKT/mTOR, PTEN, Notch, JNK, and immune-related pathways, with their dysregulation contributing to IgAN pathogenesis. This review aims to provide a comprehensive analysis of the molecular signaling pathways involving lncRNAs in IgAN, underscoring their potential as biomarkers for screening, diagnosis, and prevention. Furthermore, it explores the therapeutic potential of lncRNAs as precise targets for personalized treatment strategies.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Amirhosein Abbasi
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Mima A, Nakamoto T, Matsuki T, Kido S, Saito Y, Morikawa T, Matsumoto K, Gotoda H, Lee S. IgA Nephropathy Associated With Infliximab Treatment in Patients With Crohn's Disease: Study of IgA1 and IgA2 Expression in Glomeruli. In Vivo 2025; 39:1731-1738. [PMID: 40295023 PMCID: PMC12041970 DOI: 10.21873/invivo.13975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND/AIM It is well known that infliximab is an anti-tumor necrosis factor chimeric factor that is effective in treating inflammatory bowel diseases, such as Crohn's disease. Recently, there have been reports of new onset or flare-ups of immunoglobulin A (IgA) nephropathy during infliximab therapy for Crohn's disease. Inflammatory bowel disease-associated IgA nephropathy has been associated with IgA2; However, its activation by infliximab is still unknown. CASE REPORT We report our experience with two patients who experienced acute exacerbations of pre-existing abnormal urinalysis and renal dysfunction 1-18 years following infliximab treatment for Crohn's disease. Renal biopsies at the time of renal disease flare-up revealed IgA nephropathy in one patient and mesangial proliferative nephropathy in the other. Immunostaining results showed no clear predominance of intraglomerular expression of IgA2, and the patient diagnosed with IgA nephropathy entered remission with high dose methylprednisolone pulse therapy and oral corticosteroids, without the need for tonsillectomy. In contrast, the patient with mesangial proliferative nephritis had many devastated glomeruli, thus corticosteroids were not administrated, and the patient was followed up. CONCLUSION The clinical course of our patients, along with similar cases reported in the literature, indicates that infliximab therapy for Crohn's disease is linked to a relatively high risk of new-onset IgA nephropathy or disease relapse. This report is notable because it is the first to compare the expression of IgA1 and IgA2 in glomeruli in nephritis associated with infliximab therapy.
Collapse
Affiliation(s)
- Akira Mima
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takahiro Nakamoto
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takamasa Matsuki
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Suguru Kido
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yuta Saito
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takaaki Morikawa
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Keishi Matsumoto
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Hidemasa Gotoda
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shinji Lee
- Department of Nephrology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
7
|
Gong Y, Xu R, Gao G, Li S, Liu Y. The role of fatty acid metabolism on B cells and B cell-related autoimmune diseases. Inflamm Res 2025; 74:75. [PMID: 40299047 DOI: 10.1007/s00011-025-02042-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Fatty acid metabolism plays a critical role in regulating immune cell function, including B cells, which are central to humoral immunity and the pathogenesis of autoimmune diseases. Emerging evidence suggests that fatty acid metabolism influences B cell development, activation, differentiation, and antibody production, thereby impacting B cell-related autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS). In this review, we discuss the mechanisms by which fatty acid metabolism modulates B cell biology, including energy provision, membrane composition, and signaling pathways. We highlight how alterations in fatty acid synthesis, oxidation, and uptake affect B cell function and contribute to autoimmune pathogenesis. Additionally, we explore the therapeutic potential of targeting fatty acid metabolism in B cells to treat autoimmune diseases. Understanding the interplay between fatty acid metabolism and B cell immunity may provide novel insights into the development of precision therapies for B cell-mediated autoimmune disorders.
Collapse
Affiliation(s)
- Yanmei Gong
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ruiqi Xu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Guohui Gao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Simiao Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ying Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China, China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, Shandong, China.
| |
Collapse
|
8
|
Li X, Chen M, Chen T, Xie L, Luo Q, Fan X, Yin Y, Meng S, Jin Z, He Y, Wen Y. The intricate interplay among microbiota, mucosal immunity, and viral infection in the respiratory tract. J Transl Med 2025; 23:488. [PMID: 40301955 PMCID: PMC12042608 DOI: 10.1186/s12967-025-06433-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/26/2025] [Indexed: 05/01/2025] Open
Abstract
The mucosal system serves as the primary barrier against respiratory diseases and plays a crucial role in combating viral infections through mucosal immunity. The resident microbial community constitutes the main component of the mucosal system and exerts a significant inhibitory impact on the invasion of exogenous agents. However, the precise relationship between resident microbiota, mucosal immunity, and viral infections remains incomplete. This review aims to summarize the regulatory interactions between the resident microbiota of the mucosal system and innate immune components such as mucosal immunity and trained immunity. By clarifying these complex relationships, this review seeks to identify potential targets for augmenting respiratory disease prevention strategies and developing novel vaccine formulations. Furthermore, we propose the possibility of integrating the fields of microbiome-based therapeutics and vaccine development to create multifunctional vaccine formulations capable of targeting mucosal immunity induction. Such an approach holds great potential in offering novel pathways and strategies for the prevention and treatment of respiratory diseases.
Collapse
Affiliation(s)
- Xinyue Li
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Maohua Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Tingting Chen
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China.
| | - Lingxin Xie
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Qian Luo
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyue Fan
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Yin
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Siqin Meng
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Zhixing Jin
- Pathogen Biology and Immunology Laboratory, Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, China
| | - Yonglin He
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Yao Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 You Yi Road, Chongqing, PR China.
| |
Collapse
|
9
|
Zhang J, Luo W, Cui Y, Sun B. B-cell epitope peptide immunotherapy alleviates chitin-binding protein-induced type 2 airway inflammation in a Blomia tropicalis-murine model. Respir Res 2025; 26:129. [PMID: 40205365 PMCID: PMC11983821 DOI: 10.1186/s12931-025-03207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Peptide immunotherapy (PIT) offers a safe and effective treatment with minimal side effects. This study aims to identify B-cell epitopes of a novel allergen from Blomia tropicalis (B. tropicalis), specifically the Chitin-binding domain type 2 (ChtBD2) protein, and evaluate the therapeutic effects of peptide treatment in a murine model. METHODS Using Alphafold2, the 3D structure of ChtBD2 was constructed. AI-based and traditional computational tools predicted the predominant B-cell epitopes. Twelve synthesized peptides were assessed for allergenicity and immunogenicity. A murine model of B. tropicalis-induced allergic airway inflammation mimicking human atopic asthma was developed and analyzed. RESULTS Predominant B-cell epitopes of ChtBD2 were identified as promising IgE-binding domains. Peptide 1 (PT1: 1-15) showed significant IgE-binding activity and the highest inhibition rate in competitive IgE-binding assays. PT1 upregulated IL-4, IL-13, and CD63 in B. tropicalis-sensitized patients' PBMCs and basophils, respectively. Notably, IT groups showed reduced lung cellular infiltration and type 2 cytokine expression in BALF. Specific IgE levels were reduced, with a decline in the IgG1/IgG2a ratio. CONCLUSIONS This study represents the first AI-facilitated development of a B-cell epitope-based ChtBD2 PIT, showing promise as an immunotherapy for B. tropicalis-allergic patients with reduced allergenicity and high immunogenicity in inducing IgG-blocking antibodies. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- Guangzhou Laboratory, Guangzhou, China
| | - Wenting Luo
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- Guangzhou Laboratory, Guangzhou, China
| | - YuBao Cui
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China.
| | - Baoqing Sun
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
- Guangzhou Laboratory, Guangzhou, China.
| |
Collapse
|
10
|
Nie W, Tong X, Pung C, Li J, Ye H, Huang X. Insights into the relationship between the acetylation of Dendrobium officinale polysaccharides and the ability to promote sIgA secretion. Int J Biol Macromol 2025; 304:140764. [PMID: 39924036 DOI: 10.1016/j.ijbiomac.2025.140764] [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: 12/08/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
The acetyl group is a significant reactive component of Dendrobium officinale polysaccharide (DOP). In this study, we prepared DOPs with different degrees of acetyl substitution and investigated how the acetyl group, a naturally occurring characteristic of DOP, influences the immunomodulatory activity and the production of secretory IgA (sIgA) in the small intestine. Physical property measurements revealed significant changes in surface morphology and solubility of DOP caused by the addition or removal of acetyl groups. In vivo studies have demonstrated that DOP can mitigate Cyclophosphamide-induced immunosuppression by enhancing the immune organ index, promoting immunoglobulin secretion, and increasing the population of immune cells. Additionally, DOP can enhance sIgA production through multiple pathways, including enhanced IgA+ B cell class switch recombination, gut homing of IgA+ plasma cells, and upregulation of factors involved in sIgA composition and secretion. Correlation analysis revealed strong, piecewise-specific correlations between DOP acetylation and sIgA production at varying intervals of acetyl substitution. Based on this, we propose a theoretical framework in which the acetylation of DOP and the secretion of small intestinal sIgA demonstrate a "piecewise correlation". This framework illustrates the influence of DOP acetylation on immunomodulatory activity and provides a theoretical basis for enhancing the added value of Dendrobium officinale resources.
Collapse
Affiliation(s)
- Wenlei Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Xuecong Tong
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Chewhui Pung
- School of Chemistry, Chemical Engineering and Biotechnology. Nanyang Technological University, Singapore
| | - Jia Li
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Hui Ye
- School of Chemistry, Chemical Engineering and Biotechnology. Nanyang Technological University, Singapore
| | - Xiaojun Huang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
11
|
Kreimeyer H, Llorente C, Schnabl B. Influence of Alcohol on the Intestinal Immune System. Alcohol Res 2025; 45:03. [PMID: 40151622 PMCID: PMC11913448 DOI: 10.35946/arcr.v45.1.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Alcohol misuse is associated with disruption of the microbial homeostasis (dysbiosis) and microbial overgrowth in the gut, gut barrier disruption, and translocation of microbes into the systemic circulation. It also induces changes in regulatory mechanisms of the gut, which is the largest peripheral immune organ. The gut-liver axis is important for health and disease, and alterations in the intestinal immune system contribute to alcohol-associated liver disease (ALD). Understanding these changes might help discover new targets for drugs and therapeutic approaches. SEARCH METHODS A systematic literature search was conducted in PubMed, Medline, and Embase of manuscripts published between January 2000 and November 2023 using the terms ("alcohol" or "ethanol") AND ("immune" or "immunol") AND ("intestine," "colon," or "gut"). Eligible manuscripts included studies and reviews that discussed the effects of ethanol on immune cells in the intestine. SEARCH RESULTS A total of 506 publications were found in the databases on November 20, 2023. After excluding duplicates and research not covering ALD (415 articles), 91 studies were reviewed. Also included were manuscripts covering specific immune cells in the context of ALD. DISCUSSION AND CONCLUSIONS Balancing immune tolerance vs. initiating an immune response challenges the intestinal immune system. Alcohol induces disruption of the intestinal barrier, which is accompanied by a thicker mucus layer and reduced anti-microbial peptides. This leads to longer attachment of bacteria to epithelial cells and consequently greater translocation into the circulation. Bacterial translocation activates the immune system, reducing the activity of regulatory T cells and inducing T helper 17 response via a variety of pathways. The role of innate immune cells, especially Type 3 innate lymphoid cells, and of specific B- and T-cell subsets in ALD remains elusive. Gut dysbiosis, translocation of viable bacteria and bacterial products into the circulation, and changes in the intestinal barrier have been linked to immune deficiency and infections in patients with cirrhosis. Modifying the intestinal immune system could reduce intestinal inflammation and alcohol-induced liver injury. Understanding the underlying pathophysiology can help to detect new targets for drugs and design therapeutic strategies.
Collapse
Affiliation(s)
- Henriette Kreimeyer
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California
- Department of Medicine, U.S. Department of Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
12
|
Sakamoto Y, Niwa M, Muramatsu K, Shimo S. Effect of high-fat diet on IgA + cells and BAFF/APRIL in small intestinal villous lamina propria of mice. Cell Immunol 2025; 409-410:104911. [PMID: 39842230 DOI: 10.1016/j.cellimm.2024.104911] [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: 04/11/2024] [Revised: 08/14/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025]
Abstract
Obesity exacerbates susceptibility to infectious diseases. We investigated the effects of a high-fat diet (HFD) on intestinal immunity, particularly immunoglobulin (Ig)A-producing cells, B-cell activating factor (BAFF), and a proliferation-inducing ligand (APRIL) localization. Mice (4- to 20-weeks old) were fed HFD or standard chow diet, and their jejunum and ileum were fixed using the in vivo cryotechnique. Immunohistochemistry was performed for IgA, BAFF, and APRIL. In the HFD group, IgA+, IgA+CD22+ (p < 0.001), and IgA+CD138- (p = 0.007) cell counts were diminished in the middle sections of the lamina propria of jejunal villi, and BAFF levels were significantly reduced in jejunal villi. The HFD effects on IgA+ cell distribution seem to be confined to jejunal villi, hinting at localized vulnerabilities in intestinal immunity during obesity. Moreover, in the HFD group, IgA+ B-cell counts were reduced in the middle jejunum, indicating inhibition of the IgA+ B-cells through a T-cell-independent pathway.
Collapse
Affiliation(s)
- Yuta Sakamoto
- Faculty of Health Sciences, Department of Rehabilitation, Health Science University, 7187 Kodachi, Fujikawaguchiko-Machi, Minamitsuru-Gun, Yamanashi, Japan; Graduate School of Health Sciences, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, Japan.
| | - Masatoshi Niwa
- Faculty of Health Sciences, Department of Rehabilitation, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, Japan.
| | - Ken Muramatsu
- Faculty of Health Sciences, Department of Rehabilitation, Kyorin University, 5-4-1 Shimorenjaku, Mitaka-shi, Tokyo, Japan.
| | - Satoshi Shimo
- Faculty of Health Sciences, Department of Rehabilitation, Health Science University, 7187 Kodachi, Fujikawaguchiko-Machi, Minamitsuru-Gun, Yamanashi, Japan.
| |
Collapse
|
13
|
Popova A, Slisere B, Racenis K, Kuzema V, Karklins R, Saulite M, Seilis J, Saulite AJ, Vasilvolfa A, Vaivode K, Pjanova D, Kroica J, Cernevskis H, Lejnieks A, Petersons A, Oleinika K. IgA class-switched CD27-CD21+ B cells in IgA nephropathy. Nephrol Dial Transplant 2025; 40:505-515. [PMID: 39020236 PMCID: PMC11879059 DOI: 10.1093/ndt/gfae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Immunoglobulin A nephropathy (IgAN) is characterized by the production of galactose-deficient IgA1 (GdIgA1) antibodies. As the source of pathogenic antibodies, B cells are central to IgAN pathogenesis, but the B cell activation pathways as well as the potential B cell source of dysregulated IgA secretion remain unknown. METHODS We carried out flow cytometry analysis of peripheral blood B cells in patients with IgAN and control subjects with a focus on IgA-expressing B cells to uncover the pathways of B cell activation in IgAN and how these could give rise to pathogenic GdIgA1 antibodies. RESULTS In addition to global changes in the B cell landscape-expansion of naïve and reduction in memory B cells-IgAN patients present with an increased frequency of IgA-expressing B cells that lack the classical memory marker CD27, but are CD21+. IgAN patients furthermore have an expanded population of IgA+ antibody-secreting cells, which correlate with serum IgA levels. Both IgA+ plasmabalsts and CD27- B cells co-express GdIgA1. Implicating dysregulation at mucosal surfaces as the driver of such B cell differentiation, we found a correlation between lipopolysaccharide in the serum and IgA+CD27- B cell frequency. CONCLUSION We propose that dysregulated immunity in the mucosa may drive de novo B cell activation within germinal centres, giving rise to IgA+CD27- B cells and subsequently IgA-producing plasmablasts. These data integrate B cells into the paradigm of IgAN pathogenesis and allow further investigation of this pathway to uncover biomarkers and develop therapeutic interventions.
Collapse
Affiliation(s)
- Anna Popova
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Biology and Microbiology, Riga Stradins University, Riga, Latvia
- Department of Internal Medicine, University of Latvia, Riga, Latvia
| | - Baiba Slisere
- Joint Laboratory, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Doctoral Studies, Riga Stradins University, Riga, Latvia
| | - Karlis Racenis
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Biology and Microbiology, Riga Stradins University, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Viktorija Kuzema
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Roberts Karklins
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Mikus Saulite
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Janis Seilis
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Anna Jana Saulite
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Aiga Vasilvolfa
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Internal Medicine, University of Latvia, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Kristine Vaivode
- Institute of Microbiology and Virology, Riga Stradins University, Riga, Latvia
| | - Dace Pjanova
- Institute of Microbiology and Virology, Riga Stradins University, Riga, Latvia
| | - Juta Kroica
- Department of Biology and Microbiology, Riga Stradins University, Riga, Latvia
| | - Harijs Cernevskis
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Aivars Lejnieks
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
- Riga East Clinical University Hospital, Riga, Latvia
| | - Aivars Petersons
- Department of Nephrology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Kristine Oleinika
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical, School, Boston, MA, USA
| |
Collapse
|
14
|
Armstrong D, Chang CY, Hong MJ, Green L, Shen Y, Hudson W, Mauk KE, Song LZ, Jammi S, Casal B, Burns B, Creighton CJ, Carisey A, Zhang XHF, McKenna NJ, Kang SW, Lee HS, Decker W, Corry DB, Kheradmand F. MAGE-A4 induces non-small cell lung cancer and tumor-promoting plasma cell accumulation. SCIENCE ADVANCES 2025; 11:eads4227. [PMID: 39937892 PMCID: PMC11817953 DOI: 10.1126/sciadv.ads4227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025]
Abstract
Adaptive immunity is critical in eliminating tumors, but cancer-intrinsic factors can subvert this function. Melanoma antigen-A4 (MAGE-A4), a cancer-testis antigen, is expressed in solid tumors and correlates with poor survival, but its role in tumorigenesis and antitumor immunity remains unclear. We found that expression of MAGE-A4 was highly associated with the loss of PTEN, a tumor suppressor, in human non-small cell lung cancers (NSCLC). Here, we show that constitutive expression of human MAGE-A4 with Pten loss in mouse airway epithelia results in metastatic adenocarcinoma. Tumors showed distinct enrichment in IgA+ CD138+ CXCR4+ plasma cells (PCs) and increased expression of CXCL12 in endothelial cells. Consistently, human NSCLC expressing MAGE-A4 showed increased CD138+ IgA+ PCs surrounding tumors. Abrogation of PCs decreased tumor burden, increased activated T cell infiltration, and reduced CD163+CD206+ macrophages in the MAGE-A4-induced lung tumors. These findings suggest MAGE-A4 promotes NSCLC tumorigenesis, in part, through the recruitment and retention of IgA+ PCs in the lungs.
Collapse
Affiliation(s)
- Dominique Armstrong
- Translation Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cheng-Yen Chang
- Translation Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Monica J. Hong
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Linda Green
- Department of Pathology, Michael E. DeBakey VA, Houston, TX 77030, USA
| | - Yichao Shen
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - William Hudson
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kelsey E. Mauk
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li-Zhen Song
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sheetal Jammi
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Benjamin Casal
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brianna Burns
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chad J. Creighton
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandre Carisey
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- William T Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Xiang H.-F. Zhang
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Neil J. McKenna
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sung Wook Kang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hyun-Sung Lee
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - William Decker
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David B. Corry
- Translation Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, Houston, TX 77030, USA
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Farrah Kheradmand
- Translation Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, Houston, TX 77030, USA
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
15
|
Yang B, Guo Y, Liu L, Huang T, Zhao B, Bai W, Zhang G, Zhu C, Dong J. The LIM-domain-only protein LMO2 and its binding partner LDB1 are differentially required for class switch recombination. Proc Natl Acad Sci U S A 2025; 122:e2412376122. [PMID: 39847321 PMCID: PMC11789033 DOI: 10.1073/pnas.2412376122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/08/2024] [Indexed: 01/24/2025] Open
Abstract
The LIM-domain-only protein LMO2 interacts with LDB1 in context-dependent multiprotein complexes and plays key roles in erythropoiesis and T cell leukemogenesis, but whether they have any roles in B cells is unclear. Through a CRISPR/Cas9-based loss-of-function screening, we identified LMO2 and LDB1 as factors for class switch recombination (CSR) in murine B cells. LMO2 contributes to CSR at least in part by promoting end joining of DNA double-strand breaks (DSBs) and inhibiting end resection. Although LDB1 stabilizes LMO2 proteins, it is not required for end joining but functions as a positive regulator of AID transcription independent of LMO2, and this function of LDB1 requires its dimerization domain. Moreover, LDB1 directly binds to and promotes the looping of the AID promoter to upstream enhancers through dimerization. Our study revealed the mechanistically separated roles of LMO2 and LDB1 in different steps of CSR for antibody diversification.
Collapse
Affiliation(s)
- Beibei Yang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong518107, China
- Department of Immunology and Microbiology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| | - Yao Guo
- Department of Immunology and Microbiology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| | - Lilong Liu
- Department of Immunology and Microbiology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| | - Ting Huang
- Department of Immunology and Microbiology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| | - Bo Zhao
- Department of Immunology and Microbiology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| | - Wanyu Bai
- Department of Immunology and Microbiology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| | - Guigen Zhang
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| | - Chengming Zhu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong518107, China
| | - Junchao Dong
- Department of Immunology and Microbiology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong510000, China
| |
Collapse
|
16
|
Chin A, Small A, Wong SW, Wechalekar MD. T Cell Dysregulation in Rheumatoid Arthritis: from Genetic Susceptibility to Established Disease. Curr Rheumatol Rep 2025; 27:14. [PMID: 39862300 PMCID: PMC11762599 DOI: 10.1007/s11926-025-01180-1] [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] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
PURPOSE OF REVIEW Rheumatoid arthritis (RA) is a complex autoimmune disease characterized by chronic inflammation of the synovial tissue, where T cells play a central role in pathogenesis. Recent research has identified T peripheral helper (Tph) cells as critical mediators of local B cell activation in inflamed tissues. This review synthesizes the latest advancements in our understanding the of the role of T cells in RA, from initiation to established disease. RECENT FINDINGS We explore recent advances regarding the genetic and epigenetic factors that predispose individuals to RA, the mechanisms of T cell activation and differentiation, and the interactions between T cells and other immune and stromal cells within the synovial microenvironment. The emergence of Tph cells as key drivers of RA pathobiology is highlighted, along with their potential as therapeutic targets. We also discuss the heterogeneity of T cell responses and their interplay with synovial cells, while addressing critical research gaps such as the drivers of T cell recruitment and the plasticity of synovial phenotypes. A deeper understanding of T cell dynamics in RA will provide valuable insights for developing targeted therapies to modulate T cell-mediated inflammation and improve patient outcomes.
Collapse
Affiliation(s)
- Athena Chin
- Department of Rheumatology, Flinders Medical Centre, Adelaide, SA, Australia
| | - Annabelle Small
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Soon Wei Wong
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Mihir D Wechalekar
- Department of Rheumatology, Flinders Medical Centre, Adelaide, SA, Australia.
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia.
| |
Collapse
|
17
|
Lin Y, Liao X, Cao X, Zhang Z, Wang X, He X, Liao H, Ju B, Qi F, Xu H, Ren Z, Wang Y, Hu Z, Yang J, Fu YX, Zhao J, Zhang Z, Peng H. Sequential intranasal booster triggers class switching from intramuscularly primed IgG to mucosal IgA against SARS-CoV-2. J Clin Invest 2025; 135:e175233. [PMID: 39808503 PMCID: PMC11870729 DOI: 10.1172/jci175233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
The persistent emergence of COVID-19 variants and recurrent waves of infection worldwide underscores the urgent need for vaccines that effectively reduce viral transmission and prevent infections. Current intramuscular (IM) COVID-19 vaccines inadequately protect the upper respiratory mucosa. In response, we have developed a nonadjuvanted, IFN-armed SARS-CoV-2 fusion protein vaccine with IM priming and intranasal (IN) boost sequential immunization. Our study showed that this sequential vaccination strategy of the IM+IN significantly enhanced both upper respiratory and systemic antiviral immunity in a mouse model, characterized by the rapid increase in systemic and mucosal T and B cell responses, particularly the mucosal IgA antibody response. The IN boost triggered a swift secondary immune response, rapidly inducing antigen-specific IgA+ B cells. Further B cell receptor-seq (BCR-seq) analysis indicated that these IgA+ B cells primarily arose through direct class switching from preexisting IgG+ B cells in draining lymph nodes. Notably, our clinical studies revealed that the IN boost after IM vaccination elicited a robust systemic IgA antibody response in humans, as measured in serum. Thus, we believe that our cytokine-armed protein vaccine presents a promising strategy for inducing rapid and potent mucosal protection against respiratory viral infections.
Collapse
Affiliation(s)
- Yifan Lin
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xuezhi Cao
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiuye Wang
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Xiaomeng He
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | | | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hairong Xu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | | | | | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Jincun Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hua Peng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Jafari N, Abediankenari S. Role of microRNAs in immunoregulatory functions of epithelial cells. BMC Immunol 2024; 25:84. [PMID: 39707170 PMCID: PMC11662810 DOI: 10.1186/s12865-024-00675-3] [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: 07/16/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
Epithelial cells (ECs) provide the first line of defense against microbial threats and environmental challenges. They participate in the host's immune responses via the expression and secretion of various immune-related molecules such as cytokines and chemokines, as well as interaction with immune cells. A growing body of evidence suggests that the dysregulated function of ECs can be involved in the pathophysiology of a broad range of infectious, autoimmune, and inflammatory diseases, including inflammatory bowel disease (IBD), asthma, multiple sclerosis, and rheumatoid arthritis. To maintain a substantial immunoregulatory function of ECs, precise expression of different molecules and their regulatory effects are indispensable. MicroRNAs (miRNAs, miRs) are small non-coding RNAs that regulate gene expression commonly at post-transcriptional level through degradation of target messenger RNAs (mRNAs) or suppression of protein translation. MiRNAs implicate as critical regulators in many cellular processes, including apoptosis, growth, differentiation, and immune response. Due to the crucial roles of miRNAs in such a vast range of biological processes, they have become the spotlight of biological research for more than two decades, but we are still at the beginning stages of the use of miRNA-based therapies in the improvement of human health. Hence, in the present paper, attempts are made to provide a comprehensive overview with regard to the roles of miRNAs in the immunoregulatory functions of ECs. A better understanding of the molecular mechanisms through which immunoregulatory properties of ECs are manifested, could aid the development of efficient strategies to prevent and treat multiple human diseases.
Collapse
Affiliation(s)
- Narjes Jafari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
19
|
Nejabat S, Khomartash MS, Mohammadimehr M, Adloo Z, Zanchi FB, Ghorbani M, Nezafat N. Immunoinformatics approach: Developing a multi-epitope vaccine with novel carriers targeting monkeypox virus. FASEB J 2024; 38:e70257. [PMID: 39679938 DOI: 10.1096/fj.202400757rr] [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: 04/04/2024] [Revised: 10/12/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
Since May 2022, the global spread of monkeypox virus (MPXV) has presented a significant threat to public health. Despite this, there are limited preventive measures available. In this study, different computational tools were employed to design a multi-epitope vaccine targeting MPXV. Three key MPXV proteins, M1R, B6R, and F3L, were chosen for epitope selection, guided by bioinformatic analyses to identify immunodominant epitopes for T- and B-cell activation. To enhance immune stimulation and facilitate targeted delivery of the vaccine to specific cells, the selected epitopes were linked to novel carriers, including the extracellular domain of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), a 12-mer Clec9a binding peptide (CBP-12), and a Toll-like receptor 2 (TLR2) peptide ligand. The designed vaccine construct exhibited strong antigenicity along with nonallergenic and nontoxic properties, with favorable physicochemical characteristics. The validated vaccine's tertiary structure underwent evaluation for interactions with CD80/86, Clec9a, and TLR2 through molecular docking and molecular dynamics simulation. The results ensured the vaccine's stability and high affinity for the aforementioned receptors. In silico immune simulations studies revealed robust innate and adaptive immune responses, including enhanced mucosal immunity essential for protection against MPXV. Ultimately, the DNA sequence of the vaccine construct was synthesized and successfully cloned into the pET-22b(+) vector. Our study, through integration of computational predictions, suggests the proposed vaccine's potential efficacy in safeguarding against MPXV; however, further in vitro and in vivo validations are imperative to assess real-world effectiveness and safety.
Collapse
Affiliation(s)
- Sajjad Nejabat
- Science and Technology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mojgan Mohammadimehr
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Laboratory Sciences, Faculty of Paramedicine, AJA University of Medical Sciences, Tehran, Iran
| | - Zahra Adloo
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fernando Berton Zanchi
- Laboratório de Bioinformática e Química Medicinal (LABIOQUIM), Fundação Oswaldo Cruz Rondônia, Porto Velho, Brazil
| | - Mahdi Ghorbani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
20
|
Cao EY, Burrows K, Chiaranunt P, Popovic A, Zhou X, Xie C, Thakur A, Britton G, Spindler M, Ngai L, Tai SL, Dasoveanu DC, Nguyen A, Faith JJ, Parkinson J, Gommerman JL, Mortha A. The protozoan commensal Tritrichomonas musculis is a natural adjuvant for mucosal IgA. J Exp Med 2024; 221:e20221727. [PMID: 39535524 PMCID: PMC11561467 DOI: 10.1084/jem.20221727] [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: 10/08/2022] [Revised: 08/29/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Immunoglobulin (Ig) A supports mucosal immune homeostasis and host-microbiota interactions. While commensal bacteria are known for their ability to promote IgA, the role of non-bacterial commensal microbes in the induction of IgA remains elusive. Here, we demonstrate that permanent colonization with the protozoan commensal Tritrichomonas musculis (T.mu) promotes T cell-dependent, IgA class-switch recombination, and intestinal accumulation of IgA-secreting plasma cells (PC). T.mu colonization specifically drives the expansion of T follicular helper cells and a unique ICOS+ non-Tfh cell population, accompanied by an increase in germinal center B cells. Blockade of ICOS:ICOSL co-stimulation or MHCII-expression on B cells is central for the induction of IgA following colonization by T.mu, implicating a previously underappreciated mode of IgA induction following protozoan commensal colonization. Finally, T.mu further improves the induction of IgA-secreting PC specific to orally ingested antigens and their peripheral dissemination, identifying T.mu as a "natural adjuvant" for IgA. Collectively, these findings propose a protozoa-driven mode of IgA induction to support intestinal immune homeostasis.
Collapse
Affiliation(s)
- Eric Yixiao Cao
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Kyle Burrows
- Department of Immunology, University of Toronto, Toronto, Canada
| | | | - Ana Popovic
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Xueyang Zhou
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Cong Xie
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Ayushi Thakur
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Graham Britton
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Spindler
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Siu Ling Tai
- Department of Immunology, University of Toronto, Toronto, Canada
| | | | - Albert Nguyen
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Jeremiah J. Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Parkinson
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | | | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, Canada
| |
Collapse
|
21
|
Tejedor Vaquero S, Neuman H, Comerma L, Marcos-Fa X, Corral-Vazquez C, Uzzan M, Pybus M, Segura-Garzón D, Guerra J, Perruzza L, Tachó-Piñot R, Sintes J, Rosenstein A, Grasset EK, Iglesias M, Gonzalez Farré M, Lop J, Patriaca-Amiano ME, Larrubia-Loring M, Santiago-Diaz P, Perera-Bel J, Berenguer-Molins P, Martinez Gallo M, Martin-Nalda A, Varela E, Garrido-Pontnou M, Grassi F, Guarner F, Mehandru S, Márquez-Mosquera L, Mehr R, Cerutti A, Magri G. Immunomolecular and reactivity landscapes of gut IgA subclasses in homeostasis and inflammatory bowel disease. J Exp Med 2024; 221:e20230079. [PMID: 39560666 PMCID: PMC11577441 DOI: 10.1084/jem.20230079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/13/2024] [Accepted: 09/24/2024] [Indexed: 11/20/2024] Open
Abstract
The human gut includes plasma cells (PCs) expressing immunoglobulin A1 (IgA1) or IgA2, two structurally distinct IgA subclasses with elusive regulation, function, and reactivity. We show here that intestinal IgA1+ and IgA2+ PCs co-emerged early in life, comparably accumulated somatic mutations, and were enriched within short-lived CD19+ and long-lived CD19- PC subsets, respectively. IgA2+ PCs were extensively clonally related to IgA1+ PCs and a subset of them presumably emerged from IgA1+ precursors. Of note, secretory IgA1 (SIgA1) and SIgA2 dually coated a large fraction of mucus-embedded bacteria, including Akkermansia muciniphila. Disruption of homeostasis by inflammatory bowel disease (IBD) was associated with an increase in actively proliferating IgA1+ plasmablasts, a depletion in long-lived IgA2+ PCs, and increased SIgA1+SIgA2+ gut microbiota. Such increase featured enhanced IgA1 reactivity to pathobionts, including Escherichia coli, combined with depletion of beneficial A. muciniphila. Thus, gut IgA1 and IgA2 emerge from clonally related PCs and show unique changes in both frequency and reactivity in IBD.
Collapse
Affiliation(s)
- Sonia Tejedor Vaquero
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Hadas Neuman
- Computational Immunology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Laura Comerma
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Xavi Marcos-Fa
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Celia Corral-Vazquez
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Mathieu Uzzan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Marc Pybus
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Daniel Segura-Garzón
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Joana Guerra
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Lisa Perruzza
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Roser Tachó-Piñot
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Jordi Sintes
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Adam Rosenstein
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Emilie K. Grasset
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Mar Iglesias
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | - Joan Lop
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | | | | | - Júlia Perera-Bel
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Pau Berenguer-Molins
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Monica Martinez Gallo
- Immunology Division, Vall d’Hebron University Hospital and Translational Immunology Research Group, Vall d’Hebron Research Institute (VHIR), Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Andrea Martin-Nalda
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d’Hebron University Hospital, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Barcelona, Spain
| | - Encarna Varela
- Department of Gastroenterology, Vall d’Hebron Research Institute, Barcelona, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases, Instituto Carlos III, Madrid, Spain
| | | | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Francisco Guarner
- Department of Gastroenterology, Vall d’Hebron Research Institute, Barcelona, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases, Instituto Carlos III, Madrid, Spain
| | - Saurabh Mehandru
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Lucia Márquez-Mosquera
- Department of Gastroenterology, Hospital del Mar Medical Research Institute Barcelona, Barcelona, Spain
| | - Ramit Mehr
- Computational Immunology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Andrea Cerutti
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
- Catalan Institute for Research and Advanced Studies, Barcelona, Spain
| | - Giuliana Magri
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| |
Collapse
|
22
|
Yu L, Lin F, Yu Y, Deng X, Shi X, Lu X, Lu Y, Wang D. Rehmannia glutinosa polysaccharides enhance intestinal immunity of mice through regulating the microbiota. Int J Biol Macromol 2024; 283:137878. [PMID: 39571844 DOI: 10.1016/j.ijbiomac.2024.137878] [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: 04/03/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
The Rehmannia glutinosa polysaccharides (RGP) have various benefits such as enhancing immune cell activity, decreasing oxidative stress and delaying or inhibiting tumor occurrence. Although much research has been directed at understanding the role of RGP, its influence on gut immunity is largely understudied. Here, we aimed to dissect the immune-regulating effects of RGP in the mice intestines. In vivo experiments involving the oral administration of RGP to mice at dosages of 100, 200, and 400 mg/kg over seven consecutive days revealed that RGP therapy significantly increased the percentages of CD3+ T lymphocytes and CD19+ B lymphocytes in intestines and improved the integrity of the mucosal barrier. Moreover, RGP modified the gut microbiota composition by enhancing the abundance of beneficial bacteria like Lactobacillus and Akkermansia. Fecal microbiota transplantation (FMT) experiments further revealed that RGP modulated the host's intestinal immunological function by altering the gut microbiota composition. These findings indicate that RGP may control the immunological function of the intestines.
Collapse
Affiliation(s)
- Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Fangzhu Lin
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yaming Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangwen Deng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaofeng Shi
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuanqi Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
23
|
Liu Y, Wu F, Zhang M, Jin Y, Yuan X, Hao Y, Chen L, Fang B. 2'-Fucosyllactose and 3'-Sialyllactose Reduce Mortality in Neonatal Enteroaggregative Escherichia coli Infection by Improving the Construction of Intestinal Mucosal Immunity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:26165-26177. [PMID: 39535070 DOI: 10.1021/acs.jafc.4c06524] [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: 11/16/2024]
Abstract
Human milk oligosaccharides could prevent pathogenic bacterial infections in neonates; however, direct in vivo anti-infection evidence was still lacking. Here, we systematically evaluated the effects of 2'-fucosyllactose (2'-FL) and 3'-sialyllactose (3'-SL) on the structural development and functional maturation in neonates and their defense against enteroaggregative Escherichia coli infection. It was found that supplementation with 2'-FL and 3'-SL improved the resistance of weaned mice to enteroaggregative E. coli. The mechanism related to the promotion of 2'-FL and 3'-SL in the maturation of intestinal mucosal immunity by promoting stem cell differentiation, mucus layer integrity, and tight junction formation. 2'-FL and 3'-SL significantly increased the ratio of Th1 and Treg cells in the lamina propria, contents of short-chain fatty acids, as well as the serum content of IgA. This study lays a theoretical basis for the application of 2'-FL and 3'-SL in infant formula, as well as the development of intestinal health products.
Collapse
Affiliation(s)
- Yaqiong Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Fang Wu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Ming Zhang
- School of Food Science and Chemical Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Yutong Jin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Xinlei Yuan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300450, China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Lishui Chen
- Food Laboratory of Zhongyuan, Luohe, Henan 462300, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| |
Collapse
|
24
|
OKADA H, TSUDA M, KOJIMA N, WATANABE H, HARATA G, MIYAZAWA K, KYOUI D, HACHIMURA S, TAKAHASHI Y, TAKAHASHI K, HOSONO A. The constitutive presence of commensal bacteria contributes to the abundance of cecal IgG2b + B cells and the supply of serum IgG2b reactive to commensal bacteria in adult mice. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 44:128-136. [PMID: 40171386 PMCID: PMC11957761 DOI: 10.12938/bmfh.2024-083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/10/2024] [Indexed: 04/03/2025]
Abstract
Immunoglobulin (Ig) G isotypes in the sera of healthy mice and humans react to commensal bacteria. We previously reported that BALB/c mice with normal gut microbiota possessed abundant B cells that produced IgG2b reactive to commensal bacteria in cecal patches (CePs), indicating a potential source of a systemic pool of commensal bacteria-reactive IgG2b. Mice housed under germ-free conditions demonstrate the importance of the gut microbiota in driving cecal IgG2b responses. However, it is unclear whether the constitutive presence of the gut microbiota and specific bacterial taxa are important for IgG2b responses in adult mice. In this study, we showed that elimination of the gut microbiota by mixed antibiotic treatment in adult mice decreased the abundance of IgG2b+ B cells, follicular helper T (Tfh) cells in CePs, and the serum levels of commensal bacteria-reactive IgG2b. Reduced IgG2b responses have also been observed in mice with an altered gut microbiota following treatment with ampicillin or vancomycin. Changes in the diversity and composition of the cecal microbiota, particularly a decrease in Lachnospiraceae, Muribaculaceae, Ruminococcaceae, and Bacteroidaceae abundance at the family level, were observed in these mice. In addition, depletion of CD4+ T cells by the injection of neutralizing antibodies in adult mice reduced IgG2b responses. Our results suggest that specific gut bacteria susceptible to ampicillin and vancomycin play roles in providing an abundance of Tfh cells to help the generation of IgG2b+ B cells in CePs in adult mice, which may contribute to the supply of systemic commensal bacteria-reactive IgG2b.
Collapse
Affiliation(s)
- Hiraku OKADA
- Department of Food Science and Technology, College of
Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa 252-0880,
Japan
| | - Masato TSUDA
- Department of Food Science and Technology, College of
Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa 252-0880,
Japan
| | - Natsuki KOJIMA
- Department of Food Science and Technology, College of
Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa 252-0880,
Japan
| | - Hirofumi WATANABE
- Department of Food Science and Technology, College of
Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa 252-0880,
Japan
| | - Gaku HARATA
- Technical Research Laboratory, Takanashi Milk Products Co.,
Ltd., 5 Honjuku-cho, Asahi-ku Yokohama-shi, Kanagawa 241-0023, Japan
| | - Kenji MIYAZAWA
- Technical Research Laboratory, Takanashi Milk Products Co.,
Ltd., 5 Honjuku-cho, Asahi-ku Yokohama-shi, Kanagawa 241-0023, Japan
| | - Daisuke KYOUI
- Department of Food Science and Technology, College of
Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa 252-0880,
Japan
| | - Satoshi HACHIMURA
- Research Center for Food Safety, Graduate School of
Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo
113-8657, Japan
| | - Yoshimasa TAKAHASHI
- Research Center for Drug and Vaccine Development, National
Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Kyoko TAKAHASHI
- Department of Zoological Science, College of Bioresource
Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa 252-0880, Japan
| | - Akira HOSONO
- Department of Food Science and Technology, College of
Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa 252-0880,
Japan
| |
Collapse
|
25
|
Soliman MG, Martinez-Serra A, Antonello G, Dobricic M, Wilkins T, Serchi T, Fenoglio I, Monopoli MP. Understanding the role of biomolecular coronas in human exposure to nanomaterials. ENVIRONMENTAL SCIENCE. NANO 2024; 11:4421-4448. [PMID: 39263008 PMCID: PMC11382216 DOI: 10.1039/d4en00488d] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/17/2024] [Indexed: 09/13/2024]
Abstract
Nanomaterials (NMs) are increasingly used in medical treatments, electronics, and food additives. However, nanosafety-the possible adverse effects of NMs on human health-is an area of active research. This review provides an overview of the influence of biomolecular coronas on NM transformation following various exposure routes. We discuss potential exposure pathways, including inhalation and ingestion, describing the physiology of exposure routes and emphasising the relevance of coronas in these environments. Additionally, we review other routes to NM exposure, such as synovial fluid, blood (translocation and injection), dermal and ocular exposure, as well as the dose and medium impact on NM interactions. We emphasize the need for an in-depth characterisation of coronas in different biological media, highlighting the need and opportunity to study lung and gastric fluids to understand NM behaviour and potential toxicity. Future research aims to predict better in vivo outcomes and address the complexities of NM interactions with biological systems.
Collapse
Affiliation(s)
- Mahmoud G Soliman
- Chemistry Department, RCSI (Royal College of Surgeons in Ireland) 123 St Stephen Green Dublin 2 Ireland
- Physics Department, Faculty of Science, Al-Azhar University Cairo Egypt
| | - Alberto Martinez-Serra
- Chemistry Department, RCSI (Royal College of Surgeons in Ireland) 123 St Stephen Green Dublin 2 Ireland
| | - Giulia Antonello
- Department of Chemistry, University of Torino 10125 Torino Italy
| | - Marko Dobricic
- Chemistry Department, RCSI (Royal College of Surgeons in Ireland) 123 St Stephen Green Dublin 2 Ireland
| | - Terence Wilkins
- School of Chemical & Process Innovation, University of Leeds Engineering Building Leeds LS2 9JT UK
| | - Tommaso Serchi
- Environmental Research and Innovation Department (Luxembourg Institute of Science and Technology) 41, Rue du Brill L4422 Belvaux GD Luxembourg
| | - Ivana Fenoglio
- Department of Chemistry, University of Torino 10125 Torino Italy
| | - Marco P Monopoli
- Chemistry Department, RCSI (Royal College of Surgeons in Ireland) 123 St Stephen Green Dublin 2 Ireland
| |
Collapse
|
26
|
Gleeson PJ, Camara NOS, Launay P, Lehuen A, Monteiro RC. Immunoglobulin A Antibodies: From Protection to Harmful Roles. Immunol Rev 2024; 328:171-191. [PMID: 39578936 PMCID: PMC11659943 DOI: 10.1111/imr.13424] [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: 09/25/2024] [Revised: 10/15/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024]
Abstract
Immunoglobulin A (IgA) is the most abundantly produced antibody in humans. IgA is a unique class of immunoglobulin due to its multiple molecular forms, and a defining difference between the two subclasses: IgA1 has a long hinge-region that is heavily O-glycosylated, whereas the IgA2 hinge-region is shorter but resistant to bacterial proteases prevalent at mucosal sites. IgA is essential for immune homeostasis and education. Mucosal IgA plays a crucial role in maintaining the integrity of the mucosal barrier by immune exclusion of pathobionts while facilitating colonization with certain commensals; a large part of the gut microbiota is coated with IgA. In the circulation, monomeric IgA that has not been engaged by antigen plays a discrete role in dampening inflammatory responses. Protective and harmful roles of IgA have been studied over several decades, but a new understanding of the complex role of this immunoglobulin in health and disease has been provided by recent studies. Here, we discuss the physiological and pathological roles of IgA with a special focus on the gut, kidneys, and autoimmunity. We also discuss new IgA-based therapeutic approaches.
Collapse
Affiliation(s)
- Patrick J. Gleeson
- Center for Research on InflammationParis Cité UniversityParisFrance
- INSERMParisFrance
- CNRSParisFrance
- Inflamex Laboratory of ExcellenceParisFrance
- Nephrology DepartmentBichat HospitalParisFrance
| | - Niels O. S. Camara
- Department of Immunology, Institute of Biomedical SciencesUniversity of Sao PauloSao PauloBrazil
| | - Pierre Launay
- Center for Research on InflammationParis Cité UniversityParisFrance
- INSERMParisFrance
- CNRSParisFrance
- Inflamex Laboratory of ExcellenceParisFrance
| | - Agnès Lehuen
- Inflamex Laboratory of ExcellenceParisFrance
- Cochin Institute, INSERM, CNRSParis Cité UniversityParisFrance
| | - Renato C. Monteiro
- Center for Research on InflammationParis Cité UniversityParisFrance
- INSERMParisFrance
- CNRSParisFrance
- Inflamex Laboratory of ExcellenceParisFrance
| |
Collapse
|
27
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
28
|
Zhong H, Wang F, Tang C, Li J, Cheng JH. Combination of Structural Analysis and Proteomics Strategy Revealed the Mechanism of Ultrasound-Assisted Cold Plasma Regulating Shrimp Allergy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39356241 DOI: 10.1021/acs.jafc.4c06388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Allergic incidents of crustacean aquatic products occur frequently, and tropomyosin (TM) is the main allergen. Therefore, it is worthwhile to develop technologies to efficiently reduce the allergenicity of TM. In this study, ultrasound-assisted cold plasma (UCP) treatment was used to regulate shrimp allergy. The remarkable changes in TM structure were substantiated by alteration in secondary structure, reduction in sulfhydryl content, change in surface hydrophobicity, and disparity in surface morphology. The IgE and IgG binding ability of TM significantly decreased by 52.40% and 46.51% due to UCP treatment. In the Balb/c mouse model, mice in the UCP group showed most prominent mitigation of allergic symptoms, proved by lower allergy score, changes in levels of TM-specific antibodies, and restoration of Th1/Th2 cytokine imbalance. Using a proteomics approach, 439 differentially expressed proteins (DEPs) in the TM group (vs phosphate-buffered saline group) and 170 DEPs in the UCP group (vs TM group) were determined. Subsequent analysis demonstrated that Col6a5, Col6a6, and Epx were potential biomarkers of TM allergy. Moreover, Col6a5, Col6a6, Dcn, and Kng1 might be the target proteins of UCP treatment, while PI3K/Akt/mTOR might be the regulated signaling pathway. These findings proved that UCP treatment has great potential in reducing TM allergenicity and provide new insights into the development of hypoallergenic shrimp products.
Collapse
Affiliation(s)
- Hangyu Zhong
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Fengqi Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Caidie Tang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Jilin Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Jun-Hu Cheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Academy of Contemporary Food Engineering, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
- Engineering and Technological Research Centre of Guangdong Province on Intelligent Sensing and Process Control of Cold Chain Foods, & Guangdong Province Engineering Laboratory for Intelligent Cold Chain Logistics Equipment for Agricultural Products, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| |
Collapse
|
29
|
Duan KL, Wang TX, You JW, Wang HN, Wang ZQ, Huang ZX, Zhang JY, Sun YP, Xiong Y, Guan KL, Ye D, Chen L, Liu R, Yuan HX. PCK2 maintains intestinal homeostasis and prevents colitis by protecting antibody-secreting cells from oxidative stress. Immunology 2024; 173:339-359. [PMID: 38934051 DOI: 10.1111/imm.13827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Maintaining intracellular redox balance is essential for the survival, antibody secretion, and mucosal immune homeostasis of immunoglobulin A (IgA) antibody-secreting cells (ASCs). However, the relationship between mitochondrial metabolic enzymes and the redox balance in ASCs has yet to be comprehensively studied. Our study unveils the pivotal role of mitochondrial enzyme PCK2 in regulating ASCs' redox balance and intestinal homeostasis. We discover that PCK2 loss, whether globally or in B cells, exacerbates dextran sodium sulphate (DSS)-induced colitis due to increased IgA ASC cell death and diminished antibody production. Mechanistically, the absence of PCK2 diverts glutamine into the TCA cycle, leading to heightened TCA flux and excessive mitochondrial reactive oxygen species (mtROS) production. In addition, PCK2 loss reduces glutamine availability for glutathione (GSH) synthesis, resulting in a decrease of total glutathione level. The elevated mtROS and reduced GSH expose ASCs to overwhelming oxidative stress, culminating in cell apoptosis. Crucially, we found that the mitochondria-targeted antioxidant Mitoquinone (Mito-Q) can mitigate the detrimental effects of PCK2 deficiency in IgA ASCs, thereby alleviating colitis in mice. Our findings highlight PCK2 as a key player in IgA ASC survival and provide a potential new target for colitis treatment.
Collapse
Affiliation(s)
- Kun-Long Duan
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tian-Xiang Wang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jian-Wei You
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hai-Ning Wang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhi-Qiang Wang
- Department of Immunology, School of Basic Medical Sciences, Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zi-Xuan Huang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jin-Ye Zhang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yi-Ping Sun
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yue Xiong
- Cullgen Inc., San Diego, California, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Dan Ye
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital of Fudan University, Key Laboratory of Metabolism and Molecular Medicine (Ministry of Education), Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Li Chen
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Ronghua Liu
- Shanghai Fifth People's Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hai-Xin Yuan
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
30
|
Matsui K, Hempel HA, Shelton G, Ocampo R, Kemp TJ, Pan Y, Pinto LA. Reproducibility Assessment of Enzyme-Linked Immunosorbent Assays to Detect Anti-HPV16 L1-Specific IgG1, IgG3, IgA, and IgM Antibodies. Vaccines (Basel) 2024; 12:1108. [PMID: 39460275 PMCID: PMC11511443 DOI: 10.3390/vaccines12101108] [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: 08/09/2024] [Revised: 09/01/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Enzyme-linked immunosorbent assays (ELISAs) have been used to measure anti-human-papillomavirus (HPV) immunoglobulin IgG. The goal of this study was to evaluate the reproducibility of ELISAs measuring different HPV immunoglobulin isotypes, IgG1, 2, 3, and 4, IgA, and IgM, against HPV16. METHODS Seventy-two serum samples collected from participants in the Costa Rica HPV Vaccine Trial (CVT) and immunized with bivalent HPV vaccine (2vHPV) were used for reproducibility assessment. IgG2 and IgG4 levels were too low to be detected. Levels of IgG1, IgG3, IgA, and IgM were measured, and the data were used to calculate intraclass correlation coefficients (ICCs) and coefficients of variation (CVs). RESULTS CVs were assessed between technicians (12.8-22.7%) and across days (6.2-30.6%). The overall CVs ranged from 7.7-31.1%. IgM ELISA showed higher CVs (15.8-31.1%) than IgG1, IgG3, and IgA (6.2-22.7%). All ICC values were >98.7%. IgG3 was detected in all samples, while IgG1 and IgA had >86.3% detectability and IgM had 62.1% detectability. Pearson correlational analyses between different antibodies all showed significant correlations (p ≤ 0.001), except when comparing IgGs or IgA to IgM (p = 0.29-0.53). CONCLUSIONS Our data showed that these ELISAs are reproducible and detect isotype antibodies to HPV16 L1 across a range of concentrations in 2vHPV-vaccinated participants.
Collapse
Affiliation(s)
- Ken Matsui
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Heidi Anne Hempel
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Gloriana Shelton
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
- Digital Transformation Solutions/Civil Health, Leidos, Bethesda, MD 20817, USA
| | - Rebecca Ocampo
- Agencia Costarricense de Investigaciones Biomédicas (ACIB-FUNIN), San José 10108, Costa Rica
| | - Troy J. Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Yuanji Pan
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Ligia A. Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| |
Collapse
|
31
|
Crone L, Sobek J, Müller N, Restin T, Bassler D, Paganini D, Zimmermann MB, Zarnovican P, Routier FH, Romero-Uruñuela T, Izquierdo L, Hennet T. Inter-individual and inter-regional variability of breast milk antibody reactivity to bacterial lipopolysaccharides. Front Immunol 2024; 15:1404192. [PMID: 39308863 PMCID: PMC11412857 DOI: 10.3389/fimmu.2024.1404192] [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: 03/20/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Breast milk is a vital source of nutrients, prebiotics, probiotics, and protective factors, including antibodies, immune cells and antimicrobial proteins. Using bacterial lipopolysaccharide arrays, we investigated the reactivity and specificity of breast milk antibodies towards microbial antigens, comparing samples from rural Kenya and urban Switzerland. Results showed considerable variability in antibody reactivity both within and between these locations. Kenyan breast milk demonstrated broad reactivity to bacterial lipopolysaccharides, likely due to increased microbial exposure. Antibodies primarily recognized the O-antigens of lipopolysaccharides and showed strong binding to specific carbohydrate motifs. Notably, antibodies against specific Escherichia coli O-antigens showed cross-reactivity with parasitic pathogens like Leishmania major and Plasmodium falciparum, thus showing that antibodies reacting against lipopolysaccharide O-antigens can recognize a wide range of antigens beyond bacteria. The observed diversity in antigen recognition highlights the significance of breast milk in safeguarding infants from infections, particularly those prevalent in specific geographic regions. The findings also offer insights for potential immunobiotic strategies to augment natural antibody-mediated defense against diverse pathogens.
Collapse
Affiliation(s)
- Lisa Crone
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Jens Sobek
- Functional Genomics Center Zurich, Eidgenössische Technische Hochschule (ETH) Zurich and University of Zurich, Zurich, Switzerland
| | - Nicole Müller
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Tanja Restin
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Department of Neonatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Dirk Bassler
- Department of Neonatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Daniela Paganini
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Michael B. Zimmermann
- Medical Research Council (MRC) Translational Immune Discovery Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Patricia Zarnovican
- Department of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | | | - Tais Romero-Uruñuela
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Luis Izquierdo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Makita Y, Reich HN. Pathogenic Immunoglobulin A-Producing Cells in Immunoglobulin A Nephropathy. J Clin Med 2024; 13:5255. [PMID: 39274468 PMCID: PMC11396043 DOI: 10.3390/jcm13175255] [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: 06/25/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/16/2024] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is the most prevalent primary glomerular disease worldwide and it remains a leading cause of kidney failure. Clinical manifestations of IgA are exacerbated by infections, and emerging data suggest that aberrant mucosal immune responses are important contributors to the immunopathogenesis of this disease. However, the exact stimuli, location and mechanism of nephritis-inducing IgA production remains unclear. In this focused review we explore recent developments in our understanding of the contribution of the mucosal immune system and mucosal-derived IgA-producing cells to the development of IgAN.
Collapse
Affiliation(s)
- Yuko Makita
- Division of Nephrology, University Health Network, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan
- Toronto General Hospital Research Institute, Toronto, ON M5G 2C4, Canada
| | - Heather N Reich
- Division of Nephrology, University Health Network, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Toronto General Hospital Research Institute, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
33
|
Seefeld ML, Templeton EL, Lehtinen JM, Sinclair N, Yadav D, Hartwell BL. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front Immunol 2024; 15:1419527. [PMID: 39286244 PMCID: PMC11403286 DOI: 10.3389/fimmu.2024.1419527] [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: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.
Collapse
Affiliation(s)
- Madison L Seefeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Erin L Templeton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Justin M Lehtinen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Noah Sinclair
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Daman Yadav
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Brittany L Hartwell
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
34
|
Miyazawa H, Matsuda Y, Shirahashi T, Toma T, Wada T. Autosomal dominant TCF3 deficiency with absent B cells and a unique immunoglobulin profile. Pediatr Allergy Immunol 2024; 35:e14246. [PMID: 39295294 DOI: 10.1111/pai.14246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/21/2024]
Affiliation(s)
- Hanae Miyazawa
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Matsuda
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tetsujiro Shirahashi
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tomoko Toma
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Taizo Wada
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
35
|
Novak J, Reily C, Steers NJ, Schumann T, Rizk DV, Julian BA, Kiryluk K, Gharavi AG, Green TJ. Emerging Biochemical and Immunologic Mechanisms in the Pathogenesis of IgA Nephropathy. Semin Nephrol 2024; 44:151565. [PMID: 40087124 PMCID: PMC11972156 DOI: 10.1016/j.semnephrol.2025.151565] [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] [Indexed: 03/16/2025]
Abstract
IgA nephropathy is a mesangioproliferative glomerular disease with significant morbidity and mortality. Most patients with IgA nephropathy develop kidney failure in their lifetime, reducing their life expectancy by a decade. Since its first description in 1968, it has been established that kidneys of IgA nephropathy patients are injured as "innocent bystanders" by nephritogenic IgA1-containing immune complexes. Results from clinical, biochemical, immunologic, and genetic studies suggest a multistep pathogenetic mechanism. In genetically predisposed individuals, this process results in formation of circulating immune complexes due to the binding of IgG/IgA autoantibodies to the polymeric IgA1 molecules with incomplete O-glycosylation. This event is followed by the addition of other proteins, such as complement C3, resulting in the formation of nephritogenic immune complexes. These complexes are not effectively removed from the circulation, and some of them pass through the fenestration of glomerular endothelial cells to enter the mesangial space and activate mesangial cells. It is thought that the process is initiated by soluble immune complexes and that their accumulation results in the formation of immunodeposits that further amplify glomerular injury. Here we summarize current understanding of the pathogenesis of IgA nephropathy and discuss experimental model systems that can inform development of new therapeutic strategies and targets.
Collapse
Affiliation(s)
- Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL.
| | - Colin Reily
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL; Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Nicholas J Steers
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | | | - Dana V Rizk
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Bruce A Julian
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Todd J Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
36
|
Gleeson PJ, Monteiro RC. The Role of Mucosal Immunity: What Can We Learn From Animal and Human Studies? Semin Nephrol 2024; 44:151566. [PMID: 40082160 DOI: 10.1016/j.semnephrol.2025.151566] [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: 03/16/2025]
Abstract
Immunoglobulin A (IgA) is a key actor in the mucosal immune system, which moderates interactions between the host and environmental factors such as food antigens and commensal microorganisms. The pathogenesis of IgA nephropathy (IgAN) involves a multistep process starting with deglycosylation of mucosally derived, polymeric IgA1 (dg-IgA1) that reaches the circulation. Modified O-glycans on dg-IgA1 are targeted by IgG-autoantibodies, leading to the formation of circulating immune complexes that deposit in the glomerular mesangium. Infections of mucosal surfaces trigger flares of primary IgAN, while inflammatory bowel disease and liver cirrhosis are important causes of secondary IgAN, supporting a mucosal source of nephritogenic IgA1. In the presence of microbial pathogens or food antigens, activated dendritic cells in the gut mucosa induce T-cell-dependent or T-cell-independent B-cell differentiation into IgA-secreting plasma cells. Herein we review the literature concerning mucosal immune function and how it is altered in this disease. We discuss recent evidence supporting a causal role of gut microbiota dysbiosis in IgAN pathogenesis.
Collapse
Affiliation(s)
- Patrick J Gleeson
- Paris Cité University, Center for Research on Inflammation, Paris, France; Inserm, UMR1149; CNRS EMR8252; Inflamex Laboratory of Excellence; Nephrology Department.
| | - Renato C Monteiro
- Paris Cité University, Center for Research on Inflammation, Paris, France; Inserm, UMR1149; CNRS EMR8252; Inflamex Laboratory of Excellence; Immunology laboratory of Bichat hospital, Paris, France
| |
Collapse
|
37
|
Ong SC, Julian BA. Post-transplant IgA Nephropathy. Semin Nephrol 2024; 44:151570. [PMID: 40087123 DOI: 10.1016/j.semnephrol.2025.151570] [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: 03/16/2025]
Abstract
Immunoglobulin A (IgA) nephropathy is the most common glomerulonephritis in many countries. Most patients progress to kidney failure for which kidney transplantation is the optimal therapy. Unfortunately, IgA nephropathy commonly recurs post transplant and shortens allograft survival. Multiple recipient and donor characteristics have been associated with the risk of recurrence, although these have varied between different cohorts. The clinical expression of post-transplant IgA nephropathy is modified by immunosuppression. Biomarkers have been identified and studied in native-kidney IgA nephropathy but need validation in transplantation. Treatment of recurrent IgA nephropathy hinges on supportive measures derived largely from evidence in native-kidney IgA nephropathy. The improved understanding of the autoimmune mechanisms of disease in native-kidney IgA nephropathy has led to promising new targets for treatment, which may in turn be deployed in post-transplant IgA nephropathy.
Collapse
Affiliation(s)
- Song C Ong
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Bruce A Julian
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
38
|
Filippone EJ, Gulati R, Farber JL. Contemporary review of IgA nephropathy. Front Immunol 2024; 15:1436923. [PMID: 39188719 PMCID: PMC11345586 DOI: 10.3389/fimmu.2024.1436923] [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: 05/22/2024] [Accepted: 07/02/2024] [Indexed: 08/28/2024] Open
Abstract
IgA nephropathy (IgAN) is considered the most common primary glomerulonephritis worldwide with a predilection for Asian-Pacific populations and relative rarity in those of African descent. Perhaps 20%-50% of patients progress to kidney failure. The pathogenesis is incompletely understood. Mesangial deposition of immune complexes containing galactose-deficient IgA1 complexed with anti-glycan IgG or IgA antibodies results in mesangial cell activation and proliferation, inflammatory cell recruitment, complement activation, and podocyte damage. Diagnosis requires a biopsy interpreted by the Oxford criteria. Additional pathologic features include podocytopathy, thrombotic microangiopathy, and C4d staining. Biomarkers predicting adverse outcomes include proteinuria, reduced GFR, hypertension, and pathology. Acceptable surrogate endpoints for therapeutic trials include ongoing proteinuria and rate of eGFR decline. The significance of persisting hematuria remains uncertain. The mainstay of therapy is supportive, consisting of lifestyle modifications, renin-angiotensin inhibition (if hypertensive or proteinuric), sodium-glucose-transporter 2 inhibition (if GFR reduced or proteinuric), and endothelin-receptor antagonism (if proteinuric). Immunosuppression should be considered for those at high risk after maximal supportive care. Corticosteroids are controversial with the most positive results observed in Chinese. They carry a high risk of serious side effects. Similarly, mycophenolate may be most effective in Chinese. Other immunosuppressants are of uncertain benefit. Tonsillectomy appears efficacious in Japanese. Active areas of investigation include B-cell inhibition with agents targeting the survival factors BAFF and APRIL and complement inhibition with agents targeting the alternate pathway (Factors B and D), the lectin pathway (MASP-2), and the common pathway (C3 and C5). Hopefully soon, the who and the how of immunosuppression will be clarified, and kidney failure can be forestalled.
Collapse
Affiliation(s)
- Edward J. Filippone
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - Rakesh Gulati
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - John L. Farber
- Department of Pathology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
39
|
Fekrvand S, Abolhassani H, Rezaei N. An overview of early genetic predictors of IgA deficiency. Expert Rev Mol Diagn 2024; 24:715-727. [PMID: 39087770 DOI: 10.1080/14737159.2024.2385521] [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: 03/09/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Inborn errors of immunity (IEIs) refer to a heterogeneous category of diseases with defects in the number and/or function of components of the immune system. Immunoglobulin A (IgA) deficiency is the most prevalent IEI characterized by low serum level of IgA and normal serum levels of IgG and/or IgM. Most of the individuals with IgA deficiency are asymptomatic and are only identified through routine laboratory tests. Others may experience a wide range of clinical features including mucosal infections, allergies, and malignancies as the most important features. IgA deficiency is a multi-complex disease, and the exact pathogenesis of it is still unknown. AREAS COVERED This review compiles recent research on genetic and epigenetic factors that may contribute to the development of IgA deficiency. These factors include defects in B-cell development, IgA class switch recombination, synthesis, secretion, and the long-term survival of IgA switched memory B cells and plasma cells. EXPERT OPINION A better and more comprehensive understanding of the cellular pathways involved in IgA deficiency could lead to personalized surveillance and potentially curative strategies for affected patients, especially those with severe symptoms.
Collapse
Affiliation(s)
- Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Clinical Immunology, Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
40
|
Liu J, Zhang K, Zhang X, Guan F, Zeng H, Kubo M, Lee P, Candotti F, James LK, Camara NOS, Benlagha K, Lei J, Forsman H, Yang L, Xiao W, Liu Z, Liu C. Immunoglobulin class-switch recombination: Mechanism, regulation, and related diseases. MedComm (Beijing) 2024; 5:e662. [PMID: 39144468 PMCID: PMC11322596 DOI: 10.1002/mco2.662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2024] [Accepted: 06/30/2024] [Indexed: 08/16/2024] Open
Abstract
Maturation of the secondary antibody repertoire requires class-switch recombination (CSR), which switches IgM to other immunoglobulins (Igs), and somatic hypermutation, which promotes the production of high-affinity antibodies. Following immune response or infection within the body, activation of T cell-dependent and T cell-independent antigens triggers the activation of activation-induced cytidine deaminase, initiating the CSR process. CSR has the capacity to modify the functional properties of antibodies, thereby contributing to the adaptive immune response in the organism. Ig CSR defects, characterized by an abnormal relative frequency of Ig isotypes, represent a rare form of primary immunodeficiency. Elucidating the molecular basis of Ig diversification is essential for a better understanding of diseases related to Ig CSR defects and could provide clues for clinical diagnosis and therapeutic approaches. Here, we review the most recent insights on the diversification of five Ig isotypes and choose several classic diseases, including hyper-IgM syndrome, Waldenström macroglobulinemia, hyper-IgD syndrome, selective IgA deficiency, hyper-IgE syndrome, multiple myeloma, and Burkitt lymphoma, to illustrate the mechanism of Ig CSR deficiency. The investigation into the underlying mechanism of Ig CSR holds significant potential for the advancement of increasingly precise diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Jia‐Chen Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Ke Zhang
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xu Zhang
- Department of RespiratoryThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Fei Guan
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Hu Zeng
- Department of ImmunologyMayo Clinic College of Medicine and ScienceRochesterUSA
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), RIKEN Yokohama InstituteYokohamaJapan
| | - Pamela Lee
- Department of Paediatrics and Adolescent MedicineLKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Fabio Candotti
- Division of Immunology and AllergyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | | | | | - Kamel Benlagha
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| | - Jia‐Hui Lei
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Huamei Forsman
- Department of Rheumatology and Inflammation ResearchInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Lu Yang
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Wei Xiao
- Department of RespiratoryThe First Affiliated Hospital of Yangtze UniversityJingzhouChina
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji Hospital, Tongji Medical College, HuazhongUniversity of Science and TechnologyWuhanChina
| | - Chao‐Hong Liu
- Department of Pathogen BiologySchool of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
41
|
Guo X, Hu J, Yin G, Cai Y, Gao Z, Liu Y, Zhong M, Wang R, Feng X. The Immunomodulatory Function of Assembled Composite Nanopolypeptide Containing Bursal-Derived BP7 (CNPB7) in Promoting the Mucosal Immune Response within Poultry Immunization. Vaccines (Basel) 2024; 12:834. [PMID: 39203960 PMCID: PMC11360326 DOI: 10.3390/vaccines12080834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Mucosal immunity is the main defense line against respiratory disease pathogens. Newcastle disease and avian infectious bronchitis are common respiratory diseases in poultry. However, the mucosal immune response is not sufficiently activated and thus fails to achieve the ideal immune protection. Therefore, it is important to develop a suitable mucosal immune adjuvant to enhance the immune response of live vaccines. Here, the bursal-derived peptide BP7, β-glucan, and hyaluronic acid were selected as the adjuvant to be assembled into the composite nanopolypeptide adjuvant (CNPB7) with ultrasonic dispersion technology. The results showed that after optimizing assembly conditions, the optimal average particle size of nanoparticle CNPB7 was 514.9 nm and PDI was 0.298. To evaluate the non-specific immune responses of nanoparticle CNPB7, the chickens were immunized only with nanoparticle CNPB7. It was confirmed that nanoparticle CNPB7 enhanced the expression of CD3, CD4, CD80, and CD86 factors in the spleen lymphocyte from the chicken immunized with nanoparticle CNPB7. To investigate the mucosal immune response of nanoparticle CNPB7, the chickens were orally immunized with Newcastle disease virus (NDV)-infectious bronchitis virus (IBV) dual vaccines and CNPB7. The results proved that the levels of immunoglobulin SIgA, IL-4, IFN-γ, and IL-13 in the mucus samples from the respiratory and digestive tract in chicken immunized with nanoparticle CNPB7 and vaccines were significantly increased, compared to that of vaccine control. Finally, it was observed that nanoparticle CNPB7 promoted specific increased antibody productions against NDV and IBV in the immunized chicken. These results proved that the assembled nanoparticle CNPB7 could enhance the vaccination efficacy in chicken, which provided the experimental basis for the development of new adjuvants, and offered technical support for preventing virus transmission of avian diseases.
Collapse
Affiliation(s)
- Xinyu Guo
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jianing Hu
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Guihu Yin
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yiqin Cai
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zichen Gao
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ye Liu
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Meng Zhong
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruiying Wang
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiuli Feng
- Key Laboratory of Animal Microbiology of China’s Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.G.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
42
|
Armstrong D, Chang CY, Hong MJ, Green L, Hudson W, Shen Y, Song LZ, Jammi S, Casal B, Creighton CJ, Carisey A, Zhang XHF, McKenna NJ, Kang SW, Lee HS, Corry DB, Kheradmand F. MAGE-A4-Responsive Plasma Cells Promote Non-Small Cell Lung Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602985. [PMID: 39071307 PMCID: PMC11275715 DOI: 10.1101/2024.07.10.602985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Adaptive immunity is critical to eliminate malignant cells, while multiple tumor-intrinsic factors can alter this protective function. Melanoma antigen-A4 (MAGE-A4), a cancer-testis antigen, is expressed in several solid tumors and correlates with poor survival in non-small cell lung cancer (NSCLC), but its role in altering antitumor immunity remains unclear. We found that expression of MAGE-A4 was highly associated with the loss of PTEN , a tumor suppressor, in human NSCLC. Here we show that constitutive expression of human MAGE-A4 combined with the loss of Pten in mouse airway epithelial cells results in metastatic adenocarcinoma enriched in CD138 + CXCR4 + plasma cells, predominantly expressing IgA. Consistently, human NSCLC expressing MAGE-A4 showed increased CD138 + IgA + plasma cell density surrounding tumors. The abrogation of MAGE-A4-responsive plasma cells (MARPs) decreased tumor burden, increased T cell infiltration and activation, and reduced CD163 + CD206 + macrophages in mouse lungs. These findings suggest MAGE-A4 promotes NSCLC tumorigenesis, in part, through the recruitment and retention of IgA + MARPs in the lungs.
Collapse
|
43
|
Kögl T, Chang HF, Staniek J, Chiang SC, Thoulass G, Lao J, Weißert K, Dettmer-Monaco V, Geiger K, Manna PT, Beziat V, Momenilandi M, Tu SM, Keppler SJ, Pattu V, Wolf P, Kupferschmid L, Tholen S, Covill LE, Ebert K, Straub T, Groß M, Gather R, Engel H, Salzer U, Schell C, Maier S, Lehmberg K, Cornu TI, Pircher H, Shahrooei M, Parvaneh N, Elling R, Rizzi M, Bryceson YT, Ehl S, Aichele P, Ammann S. Patients and mice with deficiency in the SNARE protein SYNTAXIN-11 have a secondary B cell defect. J Exp Med 2024; 221:e20221122. [PMID: 38722309 PMCID: PMC11082451 DOI: 10.1084/jem.20221122] [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: 06/30/2022] [Revised: 03/08/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
SYNTAXIN-11 (STX11) is a SNARE protein that mediates the fusion of cytotoxic granules with the plasma membrane at the immunological synapses of CD8 T or NK cells. Autosomal recessive inheritance of deleterious STX11 variants impairs cytotoxic granule exocytosis, causing familial hemophagocytic lymphohistiocytosis type 4 (FHL-4). In several FHL-4 patients, we also observed hypogammaglobulinemia, elevated frequencies of naive B cells, and increased double-negative DN2:DN1 B cell ratios, indicating a hitherto unrecognized role of STX11 in humoral immunity. Detailed analysis of Stx11-deficient mice revealed impaired CD4 T cell help for B cells, associated with disrupted germinal center formation, reduced isotype class switching, and low antibody avidity. Mechanistically, Stx11-/- CD4 T cells exhibit impaired membrane fusion leading to reduced CD107a and CD40L surface mobilization and diminished IL-2 and IL-10 secretion. Our findings highlight a critical role of STX11 in SNARE-mediated membrane trafficking and vesicle exocytosis in CD4 T cells, important for successful CD4 T cell-B cell interactions. Deficiency in STX11 impairs CD4 T cell-dependent B cell differentiation and humoral responses.
Collapse
Affiliation(s)
- Tamara Kögl
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Hsin-Fang Chang
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Julian Staniek
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center— University of Freiburg, Freiburg, Germany
| | - Samuel C.C. Chiang
- Division of Bone Marrow Transplantation and Immune Deficiency, and Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
- Department of Medicine, Center for Hematology and Regenerative Medicine Huddinge, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Gudrun Thoulass
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Jessica Lao
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Kristoffer Weißert
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Viviane Dettmer-Monaco
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Transfusion Medicine and Gene Therapy—University of Freiburg, Freiburg, Germany
| | - Kerstin Geiger
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Transfusion Medicine and Gene Therapy—University of Freiburg, Freiburg, Germany
| | - Paul T. Manna
- Department of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Vivien Beziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
| | - Szu-Min Tu
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Selina J. Keppler
- Division of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Varsha Pattu
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Philipp Wolf
- Department of Urology, Faculty of Medicine, Medical Center—University of Freiburg, Freiburg, Germany
| | - Laurence Kupferschmid
- Institute of Medical Microbiology and Hygiene, University Medical Center, Freiburg, Germany
| | - Stefan Tholen
- Department of Pathology, Institute of Surgical Pathology, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Laura E. Covill
- Department of Medicine, Center for Hematology and Regenerative Medicine Huddinge, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Karolina Ebert
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Tobias Straub
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Miriam Groß
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Ruth Gather
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Helena Engel
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Ulrich Salzer
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center— University of Freiburg, Freiburg, Germany
| | - Christoph Schell
- Department of Pathology, Institute of Surgical Pathology, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Sarah Maier
- Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Lehmberg
- Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatjana I. Cornu
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Institute for Transfusion Medicine and Gene Therapy—University of Freiburg, Freiburg, Germany
| | - Hanspeter Pircher
- Institute for Immunology, Center for Microbiology and Hygiene, Medical Center—University of Freiburg, Freiburg, Germany
| | - Mohammad Shahrooei
- Department of Microbiology, Immunology, and Transplantation, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
- Dr. Shahrooei Laboratory, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Parvaneh
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Roland Elling
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty for Medicine, Center for Pediatrics and Adolescent Medicine, Medical Center—University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center— University of Freiburg, Freiburg, Germany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Clinical Immunology, Medical Center—University of Freiburg, Freiburg, Germany
| | - Yenan T. Bryceson
- Department of Medicine, Center for Hematology and Regenerative Medicine Huddinge, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Broegelmann Laboratory, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Stephan Ehl
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Peter Aichele
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| | - Sandra Ammann
- Faculty of Medicine, Institute for Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center—University of Freiburg, Freiburg, Germany
| |
Collapse
|
44
|
Li Z, Lin A, Gao Z, Jiang A, Xiong M, Song J, Liu Z, Cheng Q, Zhang J, Luo P. B-cell performance in chemotherapy: Unravelling the mystery of B-cell therapeutic potential. Clin Transl Med 2024; 14:e1761. [PMID: 38997802 PMCID: PMC11245406 DOI: 10.1002/ctm2.1761] [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: 04/15/2024] [Revised: 06/02/2024] [Accepted: 06/30/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND AND MAIN BODY The anti-tumour and tumour-promoting roles of B cells in the tumour microenvironment (TME) have gained considerable attention in recent years. As essential orchestrators of humoral immunity, B cells potentially play a crucial role in anti-tumour therapies. Chemotherapy, a mainstay in cancer treatment, influences the proliferation and function of diverse B-cell subsets and their crosstalk with the TME. Modulating B-cell function by targeting B cells or their associated cells may enhance chemotherapy efficacy, presenting a promising avenue for future targeted therapy investigations. CONCLUSION This review explores the intricate interplay between chemotherapy and B cells, underscoring the pivotal role of B cells in chemotherapy treatment. We summarise promising B-cell-related therapeutic targets, illustrating the immense potential of B cells in anti-tumour therapy. Our work lays a theoretical foundation for harnessing B cells in chemotherapy and combination strategies for cancer treatment. KEY POINTS Chemotherapy can inhibit B-cell proliferation and alter subset distributions and functions, including factor secretion, receptor signalling, and costimulation. Chemotherapy can modulate complex B-cell-T-cell interactions with variable effects on anti-tumour immunity. Targeting B-cell surface markers or signalling improves chemotherapy responses, blocks immune evasion and inhibits tumour growth. Critical knowledge gaps remain regarding B-cell interactions in TME, B-cell chemoresistance mechanisms, TLS biology, heterogeneity, spatial distributions, chemotherapy drug selection and B-cell targets that future studies should address.
Collapse
Affiliation(s)
- Zizhuo Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhifei Gao
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Minying Xiong
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiapeng Song
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Hua K, Liu D, Xu Q, Peng Y, Sun Y, He R, Luo R, Jin H. The role of hormones in the regulation of lactogenic immunity in porcine and bovine species. Domest Anim Endocrinol 2024; 88:106851. [PMID: 38733944 DOI: 10.1016/j.domaniend.2024.106851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Colostrum and milk offer a complete diet and vital immune protection for newborn mammals with developing immune systems. High immunoglobulin levels in colostrum serve as the primary antibody source for newborn piglets and calves. Subsequent milk feeding support continued local antibody protection against enteric pathogens, as well as maturation of the developing immune system and provide nutrients for newborn growth. Mammals have evolved hormonal strategies that modulate the levels of immunoglobulins in colostrum and milk to facilitate effective lactational immunity. In addition, hormones regulate the gut-mammary gland-secretory immunoglobulin A (sIgA) axis in pregnant mammals, controlling the levels of sIgA in milk, which serves as the primary source of IgA for piglets and helps them resist pathogens such as PEDV and TGEV. In the present study, we review the existing studies on the interactions between hormones and the gut-mammary-sIgA axis/lactogenic immunity in mammals and explore the potential mechanisms of hormonal regulation that have not been studied in detail, to draw attention to the role of hormones in influencing the immune response of pregnant and lactating mammals and their offspring, and highlight the effect of hormones in regulating sIgA-mediated anti-infection processes in colostrum and milk. Discussion of the relationship between hormones and lactogenic immunity may lead to a better way of improving lactogenic immunity by determining a better injection time and developing new vaccines.
Collapse
Affiliation(s)
- Kexin Hua
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Dan Liu
- China Institute of Veterinary Drug Control, Beijing 100081, PR China
| | - Qianshuai Xu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Yuna Peng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Yu Sun
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Rongrong He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China.
| |
Collapse
|
46
|
Raposo B, Klareskog L, Robinson WH, Malmström V, Grönwall C. The peculiar features, diversity and impact of citrulline-reactive autoantibodies. Nat Rev Rheumatol 2024; 20:399-416. [PMID: 38858604 DOI: 10.1038/s41584-024-01124-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2024] [Indexed: 06/12/2024]
Abstract
Since entering the stage 25 years ago as a highly specific serological biomarker for rheumatoid arthritis, anti-citrullinated protein antibodies (ACPAs) have been a topic of extensive research. This hallmark B cell response arises years before disease onset, displays interpatient autoantigen variability, and is associated with poor clinical outcomes. Technological and scientific advances have revealed broad clonal diversity and intriguing features including high levels of somatic hypermutation, variable-domain N-linked glycosylation, hapten-like peptide interactions, and clone-specific multireactivity to citrullinated, carbamylated and acetylated epitopes. ACPAs have been found in different isotypes and subclasses, in both circulation and tissue, and are secreted by both plasmablasts and long-lived plasma cells. Notably, although some disease-promoting features have been reported, results now demonstrate that certain monoclonal ACPAs therapeutically block arthritis and inflammation in mouse models. A wealth of functional studies using patient-derived polyclonal and monoclonal antibodies have provided evidence for pathogenic and protective effects of ACPAs in the context of arthritis. To understand the roles of ACPAs, one needs to consider their immunological properties by incorporating different facets such as rheumatoid arthritis B cell biology, environmental triggers and chronic antigen exposure. The emerging picture points to a complex role of citrulline-reactive autoantibodies, in which the diversity and dynamics of antibody clones could determine clinical progression and manifestations.
Collapse
Affiliation(s)
- Bruno Raposo
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Vivianne Malmström
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Caroline Grönwall
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
47
|
Flores-Hermenegildo JM, Hernández-Cázares FDJ, Pérez-Pérez D, Romero-Ramírez H, Rodríguez-Alba JC, Licona-Limon P, Kilimann MW, Santos-Argumedo L, López-Herrera G. Lrba participates in the differentiation of IgA+ B lymphocytes through TGFβR signaling. Front Immunol 2024; 15:1386260. [PMID: 38975349 PMCID: PMC11224471 DOI: 10.3389/fimmu.2024.1386260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Introduction Lrba is a cytoplasmic protein involved in vesicular trafficking. Lrba-deficient (Lrba-/-) mice exhibit substantially higher levels of IgA in both serum and feces than wild-type (WT) mice. Transforming growth factor β1 (TGFβ1) and its receptors (TGFβR I and II) is essential for differentiating IgA+ B cells. Furthermore, increased IgA production suggests a potential connection between Lrba and the TGFβR signaling pathway in IgA production. However, the specific function of Lrba in B cell biology remains unknown. Aim Given the increased IgA levels in Lrba-/- mice, the goal in this work was to explore the lymph organs where the switch to IgA occurs, and if TGFβR function is affected. Methods Non-immunized Lrba-/- mice were compared with Lrba+/+ mice. IgA levels in the serum and feces, as well as during peripheral B cell development, were determined. IgA+ B cells and plasma cells were assessed in the small intestine and secondary lymphoid organs, such as the spleen, mesenteric lymph nodes, and Peyer's patches. The TGFβR signaling pathway was evaluated by determining the expression of TGFβR on B cells. Additionally, SMAD2 phosphorylation was measured under basal conditions and in response to recombinant TGFβ. Finally, confocal microscopy was performed to investigate a possible interaction between Lrba and TGFβR in B cells. Results Lrba-/- mice exhibited significantly higher levels of circulating IgA, IgA+ B, and plasma cells than in peripheral lymphoid organs those in WT mice. TGFβR expression on the membrane of B cells was similar in both Lrba-/- and Lrba+/+ mice. However, intracellular TGFβR expression was reduced in Lrba-/- mice. SMAD2 phosphorylation showed increased levels under basal conditions; stimulation with recombinant TGFβ elicited a poorer response than in that in Lrba+/+ B cells. Finally, we found that Lrba colocalizes with TGFβR in B cells. Conclusion Lrba is essential in controlling TGFβR signaling, subsequently regulating SMAD2 phosphorylation on B cells. This mechanism may explain the increased differentiation of IgA+ B cells and production of IgA-producing plasma cells.
Collapse
Affiliation(s)
- José Mizael Flores-Hermenegildo
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría (INP), Ciudad de México, Mexico
| | - Felipe de Jesús Hernández-Cázares
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | - Daniela Pérez-Pérez
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría (INP), Ciudad de México, Mexico
- Programa de Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Héctor Romero-Ramírez
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | - Juan Carlos Rodríguez-Alba
- Unidad de Neuroinmunología y Neurooncología, Instituto Nacional de Neurología y Neurocirugia (NINN), Ciudad de México, Mexico
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca (UABJO), Ciudad de Oaxaca, Mexico
| | - Paula Licona-Limon
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Manfred W. Kilimann
- Department of Molecular Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | - Gabriela López-Herrera
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría (INP), Ciudad de México, Mexico
| |
Collapse
|
48
|
Shiraishi T, Matsuzaki C, Chiou TY, Kumeta H, Kawada M, Yamamoto K, Takahashi T, Yokota SI. Lipoteichoic acid composed of poly-glycerolphosphate containing l-lysine and involved in immunoglobulin A-inducing activity in Apilactobacillus genus. Int J Biol Macromol 2024; 271:132540. [PMID: 38782319 DOI: 10.1016/j.ijbiomac.2024.132540] [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: 12/21/2023] [Revised: 04/20/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Lipoteichoic acid (LTA) in the gram-positive bacterial cell wall acts as an immunomodulatory factor in host cells. The chemical structures vary among bacterial species and strains, and may be related to biological activities. In our previous work, much higher immunoglobulin A (IgA)-inducing activity was observed in cells of the Apilactobacillus genus (Apilactobacillus kosoi 10HT, Apilactobacillus apinorum JCM 30765T, and Apilactobacillus kunkeei JCM 16173T) than other lactic acid bacteria, and their LTA was responsible for the activity. In the present study, we elucidated the chemical structures of LTA from these Apilactobacillus strains to explore the structure-function relationship of the IgA-inducing activity. The 1H-nuclear magnetic resonance spectra suggested that their LTA structures were similar. All have a poly-glycerolphosphate main chain, which comprised 12 to 20 average number of the repeating units, with partial substitutions of glucose(α1-, glucosyl(α1-2)glucose(α1- (α-linked-kojibiose), and l-lysine at the C-2 hydroxy group of the glycerol residue. l-Lysine is a substituent never seen before in LTA, and is a probable characteristic of the Apilactobacillus genus. Removal of l-lysine residue from LTA by mild alkaline treatment decreased IgA induction in murine Peyer's patch experiments. The novel l-lysine residue in Apilactobacillus LTA plays a crucial role in the remarkably high IgA-inducing activity.
Collapse
Affiliation(s)
- Tsukasa Shiraishi
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan.
| | - Chiaki Matsuzaki
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Tai-Ying Chiou
- School of Regional Innovation and Social Design Engineering, Kitami Institute of Technology, Kitami, Hokkaido 090-8507, Japan
| | - Hiroyuki Kumeta
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Manami Kawada
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Ishikawa 921-8836, Japan
| | - Kenji Yamamoto
- Center for Innovative and Joint Research, Wakayama University, Wakayama, Wakayama 640-8510, Japan
| | - Tomoya Takahashi
- ARSOA Research & Development Center, Arsoa Keioh Group Corporation, Hokuto, Yamanashi 408-8522, Japan
| | - Shin-Ichi Yokota
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| |
Collapse
|
49
|
Zhao B, Xia Z, Yang B, Guo Y, Zhou R, Gu M, Liu M, Li Q, Bai W, Huang J, Zhang X, Zhu C, Leung KT, Chen C, Dong J. USP7 promotes IgA class switching through stabilizing RUNX3 for germline transcription activation. Cell Rep 2024; 43:114194. [PMID: 38735043 DOI: 10.1016/j.celrep.2024.114194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/04/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024] Open
Abstract
Class switch recombination (CSR) diversifies the effector functions of antibodies and involves complex regulation of transcription and DNA damage repair. Here, we show that the deubiquitinase USP7 promotes CSR to immunoglobulin A (IgA) and suppresses unscheduled IgG switching in mature B cells independent of its role in DNA damage repair, but through modulating switch region germline transcription. USP7 depletion impairs Sα transcription, leading to abnormal activation of Sγ germline transcription and increased interaction with the CSR center via loop extrusion for unscheduled IgG switching. Rescue of Sα transcription by transforming growth factor β (TGF-β) in USP7-deleted cells suppresses Sγ germline transcription and prevents loop extrusion toward IgG CSR. Mechanistically, USP7 protects transcription factor RUNX3 from ubiquitination-mediated degradation to promote Sα germline transcription. Our study provides evidence for active transcription serving as an anchor to impede loop extrusion and reveals a functional interplay between USP7 and TGF-β signaling in promoting RUNX3 expression for efficient IgA CSR.
Collapse
Affiliation(s)
- Bo Zhao
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhigang Xia
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Beibei Yang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yao Guo
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ruizhi Zhou
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Mingyu Gu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Meiling Liu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qingcheng Li
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Wanyu Bai
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Junbin Huang
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xuefei Zhang
- Biomedical Pioneering Innovation Center, Innovation Center for Genomics, Peking University, Beijing 100871, China
| | - Chengming Zhu
- Center for Scientific Research, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Kam Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chun Chen
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Junchao Dong
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
50
|
Xie Y, Huang Y, Li ZY, Jiang W, Shi NX, Lu Y, Cao G, Yin Z, Lin XJ. Interleukin-21 receptor signaling promotes metabolic dysfunction-associated steatohepatitis-driven hepatocellular carcinoma by inducing immunosuppressive IgA + B cells. Mol Cancer 2024; 23:95. [PMID: 38720319 PMCID: PMC11077880 DOI: 10.1186/s12943-024-02001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/13/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Dysregulation of immune surveillance is tightly linked to the development of metabolic dysfunction-associated steatohepatitis (MASH)-driven hepatocellular carcinoma (HCC); however, its underlying mechanisms remain unclear. Herein, we aimed to determine the role of interleukin-21 receptor (IL-21R) in MASH-driven HCC. METHODS The clinical significance of IL-21R was assessed in human HCC specimens using immunohistochemistry staining. Furthermore, the expression of IL-21R in mice was assessed in the STAM model. Thereafter, two different MASH-driven HCC mouse models were applied between IL-21R-deficient mice and wild type controls to explore the role of IL-21R in MASH-driven HCC. To further elucidate the potential mechanisms by which IL-21R affected MASH-driven HCC, whole transcriptome sequencing, flow cytometry and adoptive lymphocyte transfer were performed. Finally, flow cytometry, enzyme-linked immunosorbent assay, immunofluorescent staining, chromatin immunoprecipitation assay and western blotting were conducted to explore the mechanism by which IL-21R induced IgA+ B cells. RESULTS HCC patients with high IL-21R expression exhibited poor relapse-free survival, advanced TNM stage and severe steatosis. Additionally, IL-21R was demonstrated to be upregulated in mouse liver tumors. Particularly, ablation of IL-21R impeded MASH-driven hepatocarcinogenesis with dramatically reduction of lipid accumulation. Moreover, cytotoxic CD8+ T lymphocyte activation was enhanced in the absence of IL-21R due to the reduction of immunosuppressive IgA+ B cells. Mechanistically, the IL-21R-STAT1-c-Jun/c-Fos regulatory axis was activated in MASH-driven HCC and thus promoted the transcription of Igha, resulting in the induction of IgA+ B cells. CONCLUSIONS IL-21R plays a cancer-promoting role by inducing IgA+ B cells in MASH-driven hepatocarcinogenesis. Targeting IL-21R signaling represents a potential therapeutic strategy for cancer therapy.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- B-Lymphocytes/metabolism
- B-Lymphocytes/immunology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/genetics
- Cell Line, Tumor
- Disease Models, Animal
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Fatty Liver/etiology
- Gene Expression Regulation, Neoplastic
- Immunoglobulin A/metabolism
- Interleukin-21 Receptor alpha Subunit/metabolism
- Interleukin-21 Receptor alpha Subunit/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/etiology
- Liver Neoplasms/immunology
- Liver Neoplasms/genetics
- Receptors, Interleukin-21/metabolism
- Receptors, Interleukin-21/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Ying Xie
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China
| | - Yu Huang
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China
| | - Zhi-Yong Li
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China
| | - Weihua Jiang
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China
| | - Nan-Xi Shi
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China
| | - Yuanzhi Lu
- Department of Pathology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Guangchao Cao
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China
| | - Zhinan Yin
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China.
| | - Xue-Jia Lin
- The Biomedical Translational Research Institute, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, China.
| |
Collapse
|