1
|
Jiao L, Song Z, Zhou Y, Zhu T, Yu R, Wang Z, Qiu Y, Miao J, Zhang S, Liu Z, Wang D. Naringenin as a phytogenic adjuvant systematically enhances the protective efficacy of H9N2 inactivated vaccine through coordinated innate-adaptive immune priming in chickens. Poult Sci 2025; 104:105257. [PMID: 40344923 DOI: 10.1016/j.psj.2025.105257] [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: 03/17/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025] Open
Abstract
Although inactivated vaccines remain the primary strategy for preventing and controlling avian influenza virus, they fail to induce durable and systemic immune protection. Adjuvants are crucial for enhancing antigen immunogenicity and improving immune responses. In this study, we evaluated the adjuvant activity of naringenin (Nar) for H9N2 inactivated vaccine by detecting humoral immunity, cellular immunity, and viral challenge. The results demonstrated that Nar/H9N2 co-administration significantly increased IgG levels and hemagglutination inhibition (HI) titers. Nar/H9N2 promoted the formation of high-affinity antibodies by upregulating the expression of genes associated with B cell activation and germinal centers (GCs) formation, thus facilitating humoral immune responses. Concurrently, Nar/H9N2 vaccine enhanced T cell proliferation, CD4+and CD8+T cell differentiation, and the expression of Th1/Th2 cytokines, thereby promoting cellular immunity. Crucially, compared to the inactivated H9N2 vaccine alone, viral challenge experiments confirmed that Nar-adjuvanted immunization confers superior protection, markedly reducing viral shedding and minimizing damage to the trachea and lungs. These findings elucidate the capacity of naringenin to synchronize multifaceted immune activation through GCs optimization and T-cell modulation, establishing Nar as a viable candidate for poultry vaccine adjuvants.
Collapse
Affiliation(s)
- Lina Jiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zuchen Song
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yantong Zhou
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianyu Zhu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ruihong Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zheng Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yawei Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Shun Zhang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
2
|
Sadighi Akha AA, Csomós K, Ujházi B, Walter JE, Kumánovics A. Evolving Approach to Clinical Cytometry for Immunodeficiencies and Other Immune Disorders. Immunol Allergy Clin North Am 2025; 45:205-221. [PMID: 40287169 DOI: 10.1016/j.iac.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Primary immunodeficiencies were initially identified on the basis of recurrent, severe or unusual infections. Subsequently, it was noted that these diseases can also manifest with autoimmunity, autoinflammation, allergy, lymphoproliferation and malignancy, hence a conceptual change and their renaming as inborn errors of immunity. Ongoing advances in flow cytometry provide the opportunity to expand or modify the utility and scope of existing laboratory tests in this field to mirror this conceptual change. Here we have used the B cell subset, variably known as CD21low B cells, age-associated B cells and T-bet+ B cells, as an example to demonstrate this possibility.
Collapse
Affiliation(s)
- Amir A Sadighi Akha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Krisztián Csomós
- Division of Pediatric Allergy/Immunology, University of South Florida, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Boglárka Ujházi
- Division of Pediatric Allergy/Immunology, University of South Florida, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Jolán E Walter
- Division of Pediatric Allergy/Immunology, University of South Florida, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Attila Kumánovics
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Kardava L, Lim J, Buckner CM, Lopes de Assis F, Zhang X, Wang W, Melnyk ML, El Merhebi O, Trihemasava K, Teng IT, Carroll R, Jethmalani Y, Castro M, Lin BC, Praiss LH, Seamon CA, Kwong PD, Koup RA, Serebryannyy L, Nickle DC, Chun TW, Moir S. Phenotypic heterogeneity defines B cell responses to repeated SARS-CoV-2 exposures through vaccination and infection. Cell Rep 2025; 44:115557. [PMID: 40222009 PMCID: PMC12080740 DOI: 10.1016/j.celrep.2025.115557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/29/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Long-lived humoral memory is key to durable immunity against pathogens yet remains challenging to define due to heterogeneity among antigen-reactive B cells. We addressed this gap through longitudinal sampling over the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccinations with or without breakthrough infection. High-dimensional phenotypic profiling performed on ∼72 million B cells showed that receptor-binding domain (RBD) reactivity was associated with five distinct immunoglobulin G (IgG) B cell populations. Two expressed the activation marker CD71, both correlated with neutralizing antibodies, yet the one lacking the memory marker CD27 was induced by vaccination and blunted by infection. Two were resting memory populations; one lacking CD73 arose early and contributed to cross-reactivity; the other, expressing CD73, arose later and correlated with neutralizing antibodies. The fifth, a rare germinal center-like population, contributed to recall responses and was highly cross reactive. Overall, robust and distinct responses to booster vaccination overcame the superiority of hybrid immunity provided by breakthrough infection.
Collapse
Affiliation(s)
- Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - James Lim
- Monoceros Biosystems, San Diego, CA 29130, USA
| | - Clarisa M Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Felipe Lopes de Assis
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaozhen Zhang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Wei Wang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mattie L Melnyk
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Omar El Merhebi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Krittin Trihemasava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - I-Ting Teng
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Robin Carroll
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Mike Castro
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Bob C Lin
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Lauren H Praiss
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Catherine A Seamon
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA; Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Richard A Koup
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - David C Nickle
- Monoceros Biosystems, San Diego, CA 29130, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
4
|
Perugino CA, Liu H, Feldman J, Marbourg J, Guy TV, Hui A, Ingram N, Liebaert J, Chaudhary N, Tao W, Jacob-Dolan C, Hauser BM, Mian Z, Nathan A, Zhao Z, Kaseke C, Tano-Menka R, Getz MA, Senjobe F, Berrios C, Ofoman O, Manickas-Hill Z, Wesemann DR, Lemieux JE, Goldberg MB, Nündel K, Moormann A, Marshak-Rothstein A, Larocque RC, Ryan ET, Iafrate JA, Lingwood D, Gaiha G, Charles R, Balazs AB, Pandit A, Naranbhai V, Schmidt AG, Pillai S. Two distinct durable human class-switched memory B cell populations are induced by vaccination and infection. Cell Rep 2025; 44:115472. [PMID: 40173042 DOI: 10.1016/j.celrep.2025.115472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/21/2025] [Accepted: 03/05/2025] [Indexed: 04/04/2025] Open
Abstract
Memory lymphocytes are durable cells that persist in the absence of antigen, but few human B cell subsets have been characterized in terms of durability. The relative durability of eight non-overlapping human B cell sub-populations covering 100% of all human class-switched B cells was interrogated. Only two long-lived B cell populations persisted in the relative absence of antigen. In addition to canonical germinal center-derived switched-memory B cells with an IgD-CD27+CXCR5+ phenotype, a second, non-canonical, but distinct memory population of IgD-CD27-CXCR5+ DN1 B cells was also durable, exhibited a unique TP63-linked transcriptional and anti-apoptotic signature, had low levels of somatic hypermutation, but was more clonally expanded than canonical switched-memory B cells. DN1 B cells likely evolved to preserve immunological breadth and may represent the human counterparts of rodent extrafollicular memory B cells that, unlike canonical memory B cells, can enter germinal centers and facilitate B cell and antibody evolution.
Collapse
Affiliation(s)
- Cory A Perugino
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Division of Rheumatology Allergy and Immunology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hang Liu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jess Marbourg
- Abbvie Cambridge Research Center, Cambridge, MA 02139, USA
| | - Thomas V Guy
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Anson Hui
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Nicole Ingram
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Julian Liebaert
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Neha Chaudhary
- Abbvie Cambridge Research Center, Cambridge, MA 02139, USA
| | - Weiyang Tao
- Abbvie Cambridge Research Center, Cambridge, MA 02139, USA
| | - Catherine Jacob-Dolan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Monash University, Melbourne, VIC 3800, Australia
| | - Blake M Hauser
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zayd Mian
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Anusha Nathan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zezhou Zhao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Clarety Kaseke
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rhoda Tano-Menka
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Matthew A Getz
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Fernando Senjobe
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Cristhian Berrios
- Department of Pathology, Massachusetts Hospital, Boston, MA 02114, USA
| | - Onosereme Ofoman
- Department of Pathology, Massachusetts Hospital, Boston, MA 02114, USA
| | | | - Duane R Wesemann
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jacob E Lemieux
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Marcia B Goldberg
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Kerstin Nündel
- University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ann Moormann
- University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Regina C Larocque
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA
| | - Edward T Ryan
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA
| | - John A Iafrate
- MGH Cancer Center, Massachusetts Hospital, Boston, MA 02114, USA
| | - Daniel Lingwood
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Gaurav Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Gastrointestinal Unit, Massachusetts Hospital, Boston, MA 02114, USA
| | - Richelle Charles
- Infectious Diseases Division, Massachusetts Hospital, Boston, MA 02114, USA
| | | | | | - Vivek Naranbhai
- MGH Cancer Center, Massachusetts Hospital, Boston, MA 02114, USA; Monash University, Melbourne, VIC 3800, Australia; Center for the AIDS Programme of Research in South Africa, Durban 4001, South Africa
| | - Aaaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
5
|
Chu Q, Li K, He Q, Ren L, Wang J, Wang S, Liu X, Liu Y, He J, Li D, Shao Y. Efficient boosting of Omicron-reactive memory B cells after breakthrough infection protects from repeated exposure. iScience 2025; 28:112278. [PMID: 40264792 PMCID: PMC12013488 DOI: 10.1016/j.isci.2025.112278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/17/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Exploring the impact of persistent mutations in SARS-CoV-2 variants and reduced immunity on breakthrough infections (BTIs) is crucial, particularly in understanding how antigen-specific memory B cells (MBCs) respond to new variants. We followed 107 participants who received the ancestral inactivated vaccine and experienced one or two Omicron BTIs over six months. Using flow cytometry, SARS-CoV-2 antigen probes, single-cell RNA sequencing, and B cell receptor (BCR) profiling, we assessed MBCs and immune diversity. Our findings revealed that although neutralizing antibody levels decreased over time, the number of specific MBCs remained stable and matured progressively. Notably, pre-existing Omicron-specific MBCs played a key role in preventing secondary Omicron infections. Differential gene analysis showed enrichment in antigen processing and immune regulation pathways, while clonal lineage analysis revealed more B cell expansion and V(D)J gene-specific rearrangements in high neutralization samples. These results emphasize MBCs' critical role in long-term immunity and inform future vaccination strategies.
Collapse
Affiliation(s)
- Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Kang Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Qianxin He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Li Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jiguo Wang
- Toroivd Technology Company Limited, Shanghai 200439, China
| | - Shuo Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiaojing Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ying Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jiangshan He
- College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China
| | - Dan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yiming Shao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Changping Laboratory, Beijing 102299, China
| |
Collapse
|
6
|
Cao L, Dai H, Wei S, Ba Y, Chen F, Chen Y, Yu C, Zhang S, Chen E, Zhang H. Endoplasmic reticulum stress-related prognosis signature characterizes the immune landscape and predicts the prognosis of colon adenocarcinoma. Front Genet 2025; 16:1516232. [PMID: 40236629 PMCID: PMC11996786 DOI: 10.3389/fgene.2025.1516232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Background Colon adenocarcinoma (COAD) is characterized by high mortality and poor prognosis. Endoplasmic reticulum stress-related gene (ERSG) plays an indispensable role in the progression and immunotherapy of COAD. In this study, we evaluated the prognostic value of ERSGs in COAD. Methods We constructed and validated the ERSG-related prognostic signature based on public databases using univariate Cox analysis, Kaplan-Meier survival analysis, the LASSO method, and multivariate Cox analysis. In addition, TCGA-COAD, the Human Protein Atlas, and quantitative real-time PCR (q-PCR) were used to detect the mRNA and protein expressions of ERSGs in normal and cancer tissues/cells. The immunotherapeutic cohort was used to evaluate the predictive value of the ERSG signature for immunotherapeutic sensitivity. Results The ERSG signature, consisted of HSPA1A, SERPINA1, and DAPK1, could predict the prognosis of patients with COAD. Clinicopathologic characteristics were significantly correlated with risk scores. There were significant differences in the proportion of tumor-infiltrating immune cells, the TP53 mutation rate, the expression of immune checkpoint-related genes, and IC50 of the chemotherapeutic drugs between the low- and high-risk groups. Compared with normal tissues, the mRNA and protein expressions of three ERSGs were decreased in cancer tissues. Compared with NCM460, the mRNA levels of HSPA1A and DAPK1 were decreased in the majority of COAD cell lines, whereas the mRNA level of SERPINA1 was increased in HCT116 and SW480, and reduced in SW620. The ERSG signature could be used as a predictor of immunotherapeutic outcomes. Conclusion The ERSG signature has a predictive value in the prognosis and immunotherapeutic sensitivity in COAD, helping guide the personalized treatment.
Collapse
Affiliation(s)
- Lichao Cao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, China
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, China
- Department of Research and Development, Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, China
| | - Haoyang Dai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, China
- School of Medicine, Northwest University, Xi’an, China
| | - Shangqing Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, China
- School of Medicine, Northwest University, Xi’an, China
| | - Ying Ba
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, China
- Department of Research and Development, Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, China
| | - Fang Chen
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, China
- Department of Research and Development, Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, China
| | - Yingying Chen
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, China
- Department of Research and Development, Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, China
| | - Chendi Yu
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, China
- Department of Research and Development, Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, China
| | - Shenrui Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, China
- Department of Research and Development, Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, China
| | - Erfei Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an, China
- School of Medicine, Northwest University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Hezi Zhang
- Shenzhen Nucleus Gene Technology Co., Ltd., Shenzhen, China
- Shanghai Nucleus Biotechnology Co., Ltd., Shanghai, China
- Department of Research and Development, Shenzhen Nucleus Huaxi Medical Laboratory, Shenzhen, China
| |
Collapse
|
7
|
Castañeda J, Poblete L, Rosemblatt MV, Sauma D, Rosemblatt M, Bono MR, Nuñez S. Early-life homeostatic differentiation of thymus-resident B cells into memory B cells. Front Immunol 2025; 16:1567788. [PMID: 40226628 PMCID: PMC11985456 DOI: 10.3389/fimmu.2025.1567788] [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: 01/28/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
The thymus contains various antigen-presenting cells, including B cells, which remain activated even under steady-state conditions, suggesting ongoing local stimulation. In this study, we identify class-switched memory B cells in the thymus. Some of these cells switch their immunoglobulin to IgG2b and IgA, and express typical memory markers CD73 and PD-L2. Memory B cell differentiation in the thymus begins in neonatal mice, preceding the appearance of class-switched B cells in other peripheral lymphoid organs. Notably, exposure to environmental antigens does not influence their differentiation. Additionally, cognate interaction with CD4+ positive thymocytes is crucial for the development of memory B cells in the thymus. Our findings demonstrate that the thymus supports the local differentiation of memory B cells through a steady-state process, independent of foreign antigen stimulation and driven by interactions with developing T cells.
Collapse
Affiliation(s)
- Justine Castañeda
- Escuela de Postgrado, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lilian Poblete
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Mariana V. Rosemblatt
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - Mario Rosemblatt
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - María Rosa Bono
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - Sarah Nuñez
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| |
Collapse
|
8
|
Cancro MP. B cells and aging: a historical perspective. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf025. [PMID: 40107285 DOI: 10.1093/jimmun/vkaf025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/21/2025] [Indexed: 03/22/2025]
Affiliation(s)
- Michael P Cancro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
9
|
Ouyang Q, Cui J, Wang Y, Liu K, Zhan Y, Zhuo W, Chen J, Zhou H, Luo C, Xia J, Wang L, Guo C, Zhang J, Liu Z, Yin J. eIF3a function in immunity and protection against severe sepsis by regulating B cell quantity and function through m 6A modification. Acta Pharm Sin B 2025; 15:1571-1588. [PMID: 40370535 PMCID: PMC12069248 DOI: 10.1016/j.apsb.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/06/2024] [Accepted: 11/29/2024] [Indexed: 05/16/2025] Open
Abstract
eIF3a is a N 6-methyladenosine (m6A) reader that regulates mRNA translation by recognizing m6A modifications of these mRNAs. It has been suggested that eIF3a may play an important role in regulating translation initiation via m6A during infection when canonical cap-dependent initiation is inhibited. However, the death of animal model studies impedes our understanding of the functional significance of eIF3a in immunity and regulation in vivo. In this study, we investigated the in vivo function of eIF3a using eIF3a knockout and knockdown mouse models and found that eIF3a deficiency resulted in splenic tissue structural disruption and multi-organ damage, which contributed to severe sepsis induced by Lipopolysaccharide (LPS). Ectopic eIF3a overexpression in the eIF3a knockdown mice rescued mice from LPS-induced severe sepsis. We further showed that eIF3a maintains a functional and healthy immune system by regulating B cell function and quantity through m6A modification of mRNAs. These findings unveil a novel mechanism underlying sepsis, implicating the pivotal role of B cells in this complex disease process regulated by eIF3a. Furthermore, eIF3a may be used to develop a potential strategy for treating sepsis.
Collapse
Affiliation(s)
- Qianying Ouyang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Lymphoma & Hematology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jiajia Cui
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Geratic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ke Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yan Zhan
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Zhuo
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Pharmacy, Hunan Traditional Chinese Medical College, Zhuzhou 412000, China
| | - Juan Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chenhui Luo
- Scientific Research Office, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jianming Xia
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650102, China
| | - Liansheng Wang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chengxian Guo
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha 410017, China
| | - Jianting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiye Yin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Central South University, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
10
|
Fu W, Xie Q, Yu P, Liu S, Xu L, Ye X, Zhao W, Wang Q, Pan Y, Zhang Z, Wang Z. Pig jejunal single-cell RNA landscapes revealing breed-specific immunology differentiation at various domestication stages. Front Immunol 2025; 16:1530214. [PMID: 40151618 PMCID: PMC11947726 DOI: 10.3389/fimmu.2025.1530214] [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: 11/18/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
Background Domestication of wild boars into local and intensive pig breeds has driven adaptive genomic changes, resulting in significant phenotypic differences in intestinal immune function. The intestine relies on diverse immune cells, but their evolutionary changes during domestication remain poorly understood at single-cell resolution. Methods We performed single-cell RNA sequencing (scRNA-seq) and marker gene analysis on jejunal tissues from wild boars, a Chinese local breed (Jinhua), and an intensive breed (Duroc). Then, we developed an immune cell evaluation system that includes immune scoring, gene identification, and cell communication analysis. Additionally, we mapped domestication-related clustering relationships, highlighting changes in gene expression and immune function. Results We generated a single-cell atlas of jejunal tissues, analyzing 26,246 cells and identifying 11 distinct cell lineages, including epithelial and plasma cells, and discovered shared and unique patterns in intestinal nutrition and immunity across breeds. Immune cell evaluation analysis confirmed the conservation and heterogeneity of immune cells, manifested by highly conserved functions of immune cell subgroups, but wild boars possess stronger immune capabilities than domesticated breeds. We also discovered four patterns of domestication-related breed-specific genes related to metabolism, immune surveillance, and cytotoxic functions. Lastly, we identified a unique population of plasma cells with distinctive antibody production in Jinhua pig population. Conclusions Our findings provide valuable single-cell insights into the cellular heterogeneity and immune function evolution in the jejunum during pig at various domestication stages. The single-cell atlas also serves as a resource for comparative studies and supports breeding programs aimed at enhancing immune traits in pigs.
Collapse
Affiliation(s)
- Wenyu Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qinqin Xie
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Pengfei Yu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Shuang Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lingyao Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaowei Ye
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wei Zhao
- SciGene Biotechnology Co., Ltd, Hefei, China
| | - Qishan Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Building 11, Yongyou Industrial Park, Yazhou Bay Science and Technology City, Yazhou District, Sanya, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
| | - Yuchun Pan
- Hainan Institute of Zhejiang University, Building 11, Yongyou Industrial Park, Yazhou Bay Science and Technology City, Yazhou District, Sanya, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Hainan Yazhou Bay Seed Lab, Yongyou Industrial Park, Yazhou Bay Sci-Tech City, Sanya, China
| | - Zhe Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
| | - Zhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
| |
Collapse
|
11
|
Knox JJ, Karolyi K, Monslow J, Cromley D, Rader DJ, Puré E, Cancro MP. T-bet-expressing B cells promote atherosclerosis in apolipoprotein E-deficient mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae027. [PMID: 40073097 PMCID: PMC11952879 DOI: 10.1093/jimmun/vkae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/18/2024] [Indexed: 03/14/2025]
Abstract
The humoral immune system influences the development of atherosclerosis, but the contributions of specific memory B cell subsets and IgG isotypes are poorly understood. We assessed the relationship between atherosclerosis and age-associated B cells (ABCs), a T-bet-expressing memory B cell subset that is enriched for IgG2c production and implicated in humoral autoimmunity. We found increased numbers of splenic CD11c+ ABCs in 6-mo-old, chow-fed Apoe-/- mice versus C57BL/6 control mice, which were exacerbated by high-fat diet. Deletion of T-bet in the B lineage in high-fat diet-fed Apoe-/- mice reduced aortic lesion area, and this correlated with decreased splenic CD11c+ B cells and reduced serum oxidized low-density lipoprotein-specific IgG2c. Our findings suggest that T-bet-expressing B cells are atherogenic agents in the Apoe-/- model and indicate that interventions to inhibit a T-bet-driven humoral response may improve atherosclerotic disease.
Collapse
Affiliation(s)
- James J Knox
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katalin Karolyi
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - James Monslow
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Debra Cromley
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael P Cancro
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Kirchenbaum GA, Pawelec G, Lehmann PV. The Importance of Monitoring Antigen-Specific Memory B Cells, and How ImmunoSpot Assays Are Suitable for This Task. Cells 2025; 14:223. [PMID: 39937014 PMCID: PMC11816810 DOI: 10.3390/cells14030223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/10/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
Determining an individual's humoral immune reactivity to a pathogen, autoantigen, or environmental agent is traditionally accomplished through the assessment of specific antibody levels in blood. However, in many instances, titers of specific antibodies decline over time and thus do not faithfully reveal prior antigen exposure or establishment of immunological memory. To estimate an individual's humoral immune competence, it is therefore necessary to assess functional B cell memory. Here, we describe novel B cell ELISPOT and FluoroSpot assays (collectively referred to as ImmunoSpot) that can be rapidly developed and validated to characterize the memory B cell (Bmem) repertoire specific for any desired antigen ex vivo and at single-cell resolution. Moreover, multiplexed variants of the B cell FluoroSpot assay enable high-throughput testing of antigen-specific B cells secreting distinct antibody classes and/or IgG subclasses, with minimal cell material requirements. B cell ImmunoSpot assays also enable measurement of affinity distributions within the antigen-specific Bmem compartment and permit cross-reactivity measurements that can provide insights into Bmem established against future pathogen variants. Collectively, the ImmunoSpot® system presented here is highly reproducible, and can be readily validated for regulated tests. The newly gained ability to monitor the antigen-specific Bmem compartment should catalyze a more comprehensive understanding of humoral immunity in health and disease.
Collapse
Affiliation(s)
- Greg A. Kirchenbaum
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA;
| | - Graham Pawelec
- Department of Immunology, University of Tübingen, D-72076 Tübingen, Germany;
- Health Sciences North Research Institute, Sudbury, ON P3E 2H3, Canada
| | - Paul V. Lehmann
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA;
| |
Collapse
|
13
|
Seguin C, Seif M, Jacoberger-Foissac C, Gentine P, Wantz M, Frisch B, Heurtault B, Fournel S. NOD1 Agonist Induces Proliferation and Plasma Cell Differentiation of Mouse B Cells Especially CD23 high B Cells. Immunol Invest 2025; 54:202-216. [PMID: 39560184 DOI: 10.1080/08820139.2024.2428788] [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: 11/20/2024]
Abstract
BACKGROUND Like innate cells, B cells also express Pattern Recognition Receptors (PRRs) to detect danger signal such as tissue damage or pathogen intrusion. Production of specific antibodies by plasma cells results from the activation and differentiation of B cells following three signals: (i) antigen recognition by B Cell Receptors, (ii) recognition of danger and (iii) T-cell help. However, it is unclear whether T-cell help is dispensable for B cell activation and differentiation or not. Few studies have investigated the role of cytosolic PRRs such as NOD1 in B cell differentiation. METHODS We used splenic C57BL6J B cells to evaluate NOD1 expression and then assessed the effect of stimulation with C12-iE-DAP, a NOD1 ligand, with or without CD40L as a T-cell help signal on B-cell responses globally or according to their CD23 expression level. RESULTS We showed that murine B cells express NOD1 and that the presence of C12-iE-DAP induces activation, proliferation and initiates differentiation in plasma cells even in the absence of a T-dependent signal. Surprisingly, CD23high B cells are more sensitive than CD23low B cells to stimulation. CONCLUSION Our results suggest that the NLR pathway could induce antibody development during infections and be exploited to develop more effective vaccination.
Collapse
Affiliation(s)
- Cendrine Seguin
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Michelle Seif
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Célia Jacoberger-Foissac
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Philippe Gentine
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - May Wantz
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Benoit Frisch
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Béatrice Heurtault
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| | - Sylvie Fournel
- Biomaterials and Bioengineering, Faculté de Pharmacie, INSERM UMR_S1121 CNRS EMR7003 Université de Strasbourg, Illkirch Cedex, France
| |
Collapse
|
14
|
Liang M, Dong Q, Wu W, Fan J. Short-Chain Fatty Acids: Promising Therapeutic Targets for Respiratory Syncytial Virus Infection. Clin Rev Allergy Immunol 2025; 68:8. [PMID: 39873814 DOI: 10.1007/s12016-024-09018-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2024] [Indexed: 01/30/2025]
Abstract
The intestinal microbiota is a complex community of organisms present in the human gastrointestinal tract, some of which can produce short-chain fatty acids (SCFAs) through the fermentation of dietary fiber. SCFAs play a major role in mediating the intestinal microbiota's regulation of host immunity and intestinal homeostasis. Respiratory syncytial virus (RSV) can cause an imbalance between anti-inflammatory and proinflammatory responses in the host. In addition, changes in SCFA levels and the structure of the intestinal microbiota have been observed after RSV infection. Therefore, there may be a link between SCFAs and RSV infection, and SCFAs are expected to be therapeutic targets for RSV infection.
Collapse
Affiliation(s)
- Mingxin Liang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Qinqin Dong
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Weiyi Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Juan Fan
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
15
|
Ressler JM, Plaschka M, Silmbrod R, Bachmayr V, Shaw LE, Silly T, Zila N, Stepan A, Kusienicka A, Tschandl P, Tittes J, Roka F, Haslik W, Petzelbauer P, Koenig F, Kunstfeld R, Farlik M, Halbritter F, Weninger W, Hoeller C. Efficacy and tolerability of neoadjuvant therapy with Talimogene laherparepvec in cutaneous basal cell carcinoma: a phase II trial (NeoBCC trial). NATURE CANCER 2025; 6:51-66. [PMID: 39820126 PMCID: PMC11779647 DOI: 10.1038/s43018-024-00879-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/13/2024] [Indexed: 01/19/2025]
Abstract
We present a single-arm, phase II, neoadjuvant trial with the oncolytic virus talimogene laherparepvec (T-VEC) in 18 patients with difficult-to-resect cutaneous basal cell carcinomas. The primary end point, defined as the proportion of patients, who after six cycles of T-VEC (13 weeks), become resectable without the need for plastic reconstructive surgery, was already achieved after stage I (9 of 18 patients; 50.0%); thus the study was discontinued for early success. The objective response rate was 55.6% and the complete pathological response rate was 33.3%. Secondary end points included safety, relapse-free survival and overall survival, time to occurrence of new basal cell carcinomas and biological read outs. Only mild adverse events occurred. The 6-month relapse-free survival and overall survival rates were 100%. In two patients a new basal cell carcinoma was diagnosed. T-VEC led to a significant increase in cytotoxic T cells (P = 0.0092), B cells (P = 0.0004) and myeloid cells (P = 0.0042) and a decrease in regulatory T cells (P = 0.0290) within the tumor microenvironment. Together, neoadjuvant T-VEC represents a viable treatment option for patients with difficult-to-resect basal cell carcinomas (EudraCT no. 2018-002165-19).
Collapse
Affiliation(s)
| | - Maud Plaschka
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Rita Silmbrod
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Victoria Bachmayr
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Lisa Ellen Shaw
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Thomas Silly
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Nina Zila
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
- University of Applied Sciences FH Campus Wien, Division of Biomedical Science, Vienna, Austria
| | - Andreas Stepan
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Anna Kusienicka
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Philipp Tschandl
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Julia Tittes
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Florian Roka
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Werner Haslik
- Medical University of Vienna, Department of Obstetrics and Gynecology, Vienna, Austria
| | - Peter Petzelbauer
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
- Medical University of Vienna, SERD Skin and Endothelium Research Division, Vienna, Austria
| | - Franz Koenig
- Medical University of Vienna, Center of Medical Statistics, Informatics and Intelligent Systems, Vienna, Austria
| | - Rainer Kunstfeld
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Matthias Farlik
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | | | - Wolfgang Weninger
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| | - Christoph Hoeller
- Medical University of Vienna, Department of Dermatology, Vienna, Austria
| |
Collapse
|
16
|
Kometani K, Yorimitsu T, Jo N, Yamaguchi E, Kikuchi O, Fukahori M, Sawada T, Tsujimoto Y, Sunami A, Li M, Ito T, Pretemer Y, Gao Y, Hidaka Y, Yamamoto M, Kaku N, Nakagama Y, Kido Y, Grifoni A, Sette A, Nagao M, Morita S, Nakajima TE, Muto M, Hamazaki Y. Booster COVID-19 mRNA vaccination ameliorates impaired B-cell but not T-cell responses in older adults. Front Immunol 2024; 15:1455334. [PMID: 39717779 PMCID: PMC11663736 DOI: 10.3389/fimmu.2024.1455334] [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: 06/26/2024] [Accepted: 11/05/2024] [Indexed: 12/25/2024] Open
Abstract
Age-associated differences in the effect of repetitive vaccination, particularly on memory T-cell and B-cell responses, remain unclear. While older adults (aged ≥65 years) exhibited enhanced IgG responses following COVID-19 mRNA booster vaccination, they produced fewer spike-specific circulating follicular helper T cells-1 than younger adults. Similarly, the cytotoxic CD8+ T-cell response remained diminished with reduced PD-1 expression even after booster vaccination compared with that in younger adults, suggesting impaired memory T-cell activation in older adults. In contrast, although B-cell responses in older adults were weaker than those in younger adults in the primary response, the responses were significantly enhanced upon booster vaccination, reaching levels comparable with that observed in younger adults. Therefore, while booster vaccination ameliorates impaired humoral immunity in older adults by efficiently stimulating memory B-cell responses, it may less effectively enhance T-cell-mediated cellular immunity. Our study provides insights for the development of effective therapeutic and vaccine strategies for the most vulnerable older population.
Collapse
Affiliation(s)
- Kohei Kometani
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takaaki Yorimitsu
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norihide Jo
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Alliance Laboratory for Advanced Medical Research, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Erina Yamaguchi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Osamu Kikuchi
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Clinical Bio-Resource Center, Kyoto University Hospital, Kyoto, Japan
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University, Kyoto, Japan
| | - Masaru Fukahori
- Department of Early Clinical Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy (Ki-CONNECT), Kyoto University Hospital, Kyoto, Japan
| | - Takeshi Sawada
- Department of Early Clinical Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy (Ki-CONNECT), Kyoto University Hospital, Kyoto, Japan
| | - Yoshitaka Tsujimoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ayana Sunami
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Laboratory of Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mengqian Li
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takeshi Ito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yann Pretemer
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuxian Gao
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Laboratory of Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yu Hidaka
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Yamamoto
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Natsuko Kaku
- Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Yu Nakagama
- Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Yasutoshi Kido
- Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, United States
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takako E. Nakajima
- Department of Early Clinical Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy (Ki-CONNECT), Kyoto University Hospital, Kyoto, Japan
| | - Manabu Muto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Clinical Bio-Resource Center, Kyoto University Hospital, Kyoto, Japan
- Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy (Ki-CONNECT), Kyoto University Hospital, Kyoto, Japan
| | - Yoko Hamazaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Laboratory of Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto University Immunomonitoring Center, Kyoto, Japan
| |
Collapse
|
17
|
Zhang P, Ruan C, Yang G, Guan Y, Zhu Y, Li Q, Dai X, An Y, Shi X, Huang P, Chen Y, He Z, Du Z, Liu C. PGRN Inhibits Early B-cell Activation and IgE Production Through the IFITM3-STAT1 Signaling Pathway in Asthma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403939. [PMID: 39412083 PMCID: PMC11615816 DOI: 10.1002/advs.202403939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/23/2024] [Indexed: 12/06/2024]
Abstract
Progranulin (PGRN) plays a critical role in bronchial asthma and the function of various immune cells. However, the mechanisms by which PGRN influences B-cell receptor (BCR) signaling and immunoglobulin E(IgE) production are not fully understood. The study aimed to elucidate the molecular mechanisms through which PGRN affects BCR signaling, B-cell differentiation, and IgE production. A PGRN knockout mouse model, along with techniques including flow cytometry, the creation of a bone marrow chimeric mouse model, total internal reflection fluorescence (TIRF), and Western blot (WB) analysis is employed, to investigate the link between PGRN and various aspects of B-cell biology. It is discovered that the absence of PGRN in mice alters peripheral B-cell subpopulations, promotes IgE class switching in a cell-intrinsic manner, and affects B-cell subpopulations. Additionally, PGRN modulates B-cell functions by regulating BCR signaling pathways, metabolic processes, and the actin cytoskeleton during early B-cell activation. Significantly, PGRN deficiency results in diminished production of NP-specific antibodies. Moreover, it is found that PGRN inhibits B-cell activation and IgE production through the PGRN-IFITM3-STAT1 signaling pathway. The findings provide new strategies for the targeted treatment of bronchial asthma, highlighting the crucial role of PGRN in B-cell signaling and IgE production.
Collapse
Affiliation(s)
- Pingping Zhang
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Changshun Ruan
- Chongqing Key Laboratory of Child Infection and ImmunityChildren's Hospital of Chongqing Medical UniversityChongqing400014China
| | - Guangli Yang
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Yaning Guan
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Yin Zhu
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Qian Li
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Xin Dai
- Zhanjiang Institute of Clinical MedicineZhanjiang Central HospitalGuangdong Medical UniversityZhanjiang524037China
- Department of HematologyCentral People's Hospital of ZhanjiangZhanjiang524037China
| | - Yang An
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Xiaoqi Shi
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Pei Huang
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Yan Chen
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Zhixu He
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Zuochen Du
- Department of PediatricsAffiliated Hospital of Zunyi Medical UniversityZunyi563000China
- Department of PediatricsGuizhou Children's HospitalZunyi563000China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical UniversityZunyi563099China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic MedicineTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science and Technology Wuhan Hubei ChinaHubei430074China
| |
Collapse
|
18
|
Li H, Lin S, Wang Y, Shi Y, Fang X, Wang J, Cui H, Bian Y, Qi X. Immunosenescence: A new direction in anti-aging research. Int Immunopharmacol 2024; 141:112900. [PMID: 39137628 DOI: 10.1016/j.intimp.2024.112900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
The immune system is a major regulatory system of the body, that is composed of immune cells, immune organs, and related signaling factors. As an organism ages, observable age-related changes in the function of the immune system accumulate in a process described as 'immune aging. Research has shown that the impact of aging on immunity is detrimental, with various dysregulated responses that affect the function of immune cells at the cellular level. For example, increased aging has been shown to result in the abnormal chemotaxis of neutrophils and decreased phagocytosis of macrophages. Age-related diminished functionality of immune cell types has direct effects on host fitness, leading to poorer responses to vaccination, more inflammation and tissue damage, as well as autoimmune disorders and the inability to control infections. Similarly, age impacts the function of the immune system at the organ level, resulting in decreased hematopoietic function in the bone marrow, a gradual deficiency of catalase in the thymus, and thymic atrophy, resulting in reduced production of related immune cells such as B cells and T cells, further increasing the risk of autoimmune disorders in the elderly. As the immune function of the body weakens, aging cells and inflammatory factors cannot be cleared, resulting in a cycle of increased inflammation that accumulates over time. Cumulatively, the consequences of immune aging increase the likelihood of developing age-related diseases, such as Alzheimer's disease, atherosclerosis, and osteoporosis, among others. Therefore, targeting the age-related changes that occur within cells of the immune system might be an effective anti-aging strategy. In this article, we summarize the relevant literature on immune aging research, focusing on its impact on aging, in hopes of providing new directions for anti-aging research.
Collapse
Affiliation(s)
- Hanzhou Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Union Medical Center, Tianjin, China
| | - Shan Lin
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuming Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuexuan Shi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xixing Fang
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jida Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huantian Cui
- Yunnan University of Chinese Medicine, Yunnan, China.
| | - Yuhong Bian
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xin Qi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Union Medical Center, Tianjin, China.
| |
Collapse
|
19
|
Lam N, Lee Y, Farber DL. A guide to adaptive immune memory. Nat Rev Immunol 2024; 24:810-829. [PMID: 38831162 DOI: 10.1038/s41577-024-01040-6] [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: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Immune memory - comprising T cells, B cells and plasma cells and their secreted antibodies - is crucial for human survival. It enables the rapid and effective clearance of a pathogen after re-exposure, to minimize damage to the host. When antigen-experienced, memory T cells become activated, they proliferate and produce effector molecules at faster rates and in greater magnitudes than antigen-inexperienced, naive cells. Similarly, memory B cells become activated and differentiate into antibody-secreting cells more rapidly than naive B cells, and they undergo processes that increase their affinity for antigen. The ability of T cells and B cells to form memory cells after antigen exposure is the rationale behind vaccination. Understanding immune memory not only is crucial for the design of more-efficacious vaccines but also has important implications for immunotherapies in infectious disease and cancer. This 'guide to' article provides an overview of the current understanding of the phenotype, function, location, and pathways for the generation, maintenance and protective capacity of memory T cells and memory B cells.
Collapse
Affiliation(s)
- Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
20
|
Kang S, Wu Q, Shen J, Wu C. CD27 is not an ideal marker for human memory B cells and can be modulated by IL-21 upon stimulated by Anti-CD40. Sci Rep 2024; 14:23742. [PMID: 39390111 PMCID: PMC11467254 DOI: 10.1038/s41598-024-75636-2] [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: 02/01/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
B cells play a key role in humoral immune responses by producing antibodies. Although there are numerous research on memory B cells definition markers and cytokines on B cell development, different studies have yielded contradictory conclusions due to species studied, the different cells and stimulating agents used. In the current study, we conducted a detailed characterization of B cells in human CBMCs, PBMCs and tonsil, including expression of Igs, activation and memory markers. Furthermore, we found that considerable amounts of IgA and IgG were expressed by CD27- B cells. These "Atypical" memory B cells corresponded to approximately 50% of IgG+ and IgA+B cells in blood, this proportion even reached 90% in tonsil. In addition, we investigated the effect of IL-21 and TGF-β1 on the membrane-bound form and secreted form of Igs using PBMCs and purified blood B cells. There were actual differences between the effect of cytokines on Igs secretion and surface expression. Our study will be helpful to advance the knowledge and understanding of humoral memory.
Collapse
Affiliation(s)
- Shuangpeng Kang
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, People's Republic of China.
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, 1501 Leifeng Road, 410219, Changsha, People's Republic of China.
| | - Qiongli Wu
- Shenzhen Experimental Education School, Shenzhen, People's Republic of China
| | - Juan Shen
- Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Changyou Wu
- Clinical Research Center of Clifford Hospital, Guangzhou, People's Republic of China.
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, 510080, Guangzhou, People's Republic of China.
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, 1501 Leifeng Road, 410219, Changsha, People's Republic of China.
| |
Collapse
|
21
|
Xiao R, Han J, Deng Y, Zhang L, Qian Y, Tian N, Yang Z, Zhang L. AGTR1: a potential biomarker associated with the occurrence and prognosis of lung adenocarcinoma. Front Oncol 2024; 14:1441235. [PMID: 39450258 PMCID: PMC11499140 DOI: 10.3389/fonc.2024.1441235] [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: 05/30/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Lung adenocarcinoma, a disease with complex pathogenesis, high mortality and poor prognosis, is one of the subtypes of lung cancer. Hence, it is very crucial to find novel biomarkers as diagnostic and therapeutic targets for LUAD. Methods GSE10072 was used for DEGs and WGCNA, and the intersection genes were subjected to enrichment analysis through Metascape and GSEA. Key genes were screened by three machine learning methods. Further, the reliability of key genes was identified by ROC, COX regression analysis and qRT-PCR. CIBERSORT and Spearman analysis were used for understanding the relationships of LUAD, immunity and key genes. In addition, ceRNA networks and potential drugs of key genes were constructed and predicted. Results After overlapping 631 DEGs and key module genes, 623 intersection genes were obtained. Subsequently, DUOX1, CD36, AGTR1, FHL5 and SSR4 were further selected using three machine learning methods. Reliability analysis demonstrated that AGTR1 possesses important predictive value for the occurrence and prognosis of LUAD. The enrichment analysis showed that AGTR1 was significantly enriched in the GPCR-related pathways. Immune infiltration analysis showed that the development of LUAD was related to the changes of immune cells such as M2 macrophages and neutrophils, which were regulated by AGTR1. Further, AGTR1 is also involved in regulating immune chemokines, checkpoints and immune regulatory factors such as PECAM1, ADARB1, SPP1 and ENO1, all of them playing important roles in immune cell regulation, tumor cell proliferation and migration. Further, the drug-gene interaction network screened out 13 potential drugs such as Benazepril, Valsartan, Eprosartan, and so on. Discussion AGTR1 is a potential biomarker for the occurrence and progression of LUAD, closely related to tumor immunity, proliferation and migration. It can serve as a new target for the diagnosis and treatment of LUAD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Zhang
- College of Life Science/Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Liu JC, Zeng Q, Duan YG, Yeung WSB, Li RHW, Ng EHY, Cheung KW, Zhang Q, Chiu PCN. B cells: roles in physiology and pathology of pregnancy. Front Immunol 2024; 15:1456171. [PMID: 39434884 PMCID: PMC11491347 DOI: 10.3389/fimmu.2024.1456171] [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: 06/28/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
B cells constitute a diverse and adaptable immune cell population with functions that can vary according to the environment and circumstances. The involvement of B cells in pregnancy, as well as the associated molecular pathways, has yet to be investigated. This review consolidates current knowledge on B cell activities and regulation during pregnancy, with a particular focus on the roles of various B cell subsets and the effects of B cell-derived factors on pregnancy outcomes. Moreover, the review examines the significance of B cell-associated autoantibodies, cytokines, and signaling pathways in relation to pregnancy complications such as pregnancy loss, preeclampsia, and preterm birth.
Collapse
Affiliation(s)
- Jin-Chuan Liu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qunxiong Zeng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Raymond H. W. Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ka-Wang Cheung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
23
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
24
|
Liu Y, Tian T, Li XC, Chen YM, Li H, Li YL, He WT, Chen H. Efficacy of natural duct specimen extraction versus conventional laparoscopic surgery for rectal cancer: a single-centre retrospective analysis. Am J Cancer Res 2024; 14:4472-4483. [PMID: 39417176 PMCID: PMC11477820 DOI: 10.62347/xzhw4521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVE Rectal cancer has a high incidence and its onset age is getting younger. Currently, conventional laparoscopic surgery can no longer meet the clinical requirements for surgical incisions. Natural orifice specimen extraction surgery (NOSES) is less invasive, but there have been few studies on the effectiveness of this procedure for rectal cancer. Therefore, this study aimed to explore the efficacy of NOSES and conventional laparoscopic surgery in rectal cancer treatment. METHODS In this retrospective analysis, we collected clinical data of 150 rectal cancer patients. Patients who received NOSES were included in a NOSES group and those underwent routine laparoscopic surgery were in a control group. Then, the observation group was matched with the control group at a ratio of 1:1 by using the propensity score matching method. We compared the surgical indicators, postoperative recovery indicators, physical indicators, pain, surgical stress-related indicators, inflammation indicators, immune indicators, quality of life, and postoperative complications between the two groups. RESULTS We found that compared with the control group, the NOSES group had a shorter exhaust start time, getting out-of-bed activity time, length of hospital stay, bowel sound recovery time, and gastrointestinal peristalsis time. The Pittsburgh Sleep Quality Index (PSQI) and Positive and Negative Affect Schedule (PANAS) scores decreased in both groups after surgery, with the NOSES group showing a more significant reduction. The Visual Analogue Scale (VAS) scores decreased in both groups, and the NOSES group had lower VAS scores. Additionally, the NOSES group exhibited a significant interaction effect with time (intergroup effect: F = 497.800; time effect: F = 163.100; interaction effect: F = 5.307). Superoxide dismutase (SOD) levels decreased and malondialdehyde (MDA) levels increased in both groups postoperatively; however, the NOSES group had higher SOD levels and lower MDA levels. All the above comparisons were statistically significant (P < 0.05). There was no statistically significant difference in the total complication rates between the NOSES group and the control group (Z = -0.768, P = 0.442; χ2 = 2.333, P = 0.127). CONCLUSION Compared to conventional laparoscopic surgery, NOSES results in less pain and injury, a more stable mood, faster recovery, and comparable safety.
Collapse
Affiliation(s)
- Yang Liu
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| | - Ting Tian
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| | - Xin-Chun Li
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| | - Yan-Min Chen
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| | - Hong Li
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| | - Yu-Lin Li
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| | - Wen-Tao He
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| | - Hua Chen
- Department of General Surgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City) Changde 415000, Hunan, China
| |
Collapse
|
25
|
Hillion S, Miranda A, Le Dantec C, Boudigou M, Le Pottier L, Cornec D, Torres RM, Pelanda R. Maf expression in B cells restricts reactive plasmablast and germinal center B cell expansion. Nat Commun 2024; 15:7982. [PMID: 39266537 PMCID: PMC11393457 DOI: 10.1038/s41467-024-52224-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 08/29/2024] [Indexed: 09/14/2024] Open
Abstract
Precise regulation of B cell differentiation is essential for an effective adaptive immune response. Here, we show that B cell development in mice with B cell-specific Maf deletion is unaffected, but marginal zone B cells, germinal centre B cells, and plasmablasts are significantly more frequent in the spleen of naive Maf-deficient mice compared to wild type controls. In the context of a T cell-dependent immunization, Maf deletion causes increased proliferation of germinal centre B cells and extrafollicular plasmablasts. This is accompanied by higher production of antigen-specific IgG1 antibodies with minimal modification of early memory B cells, but a reduction in plasma cell numbers. Single-cell RNA sequencing shows upregulation of genes associated with DNA replication and cell cycle progression, confirming the role of Maf in cell proliferation. Subsequent pathway analysis reveals that Maf influences cellular metabolism, transporter activity, and mitochondrial proteins, which have been implicated in controlling the germinal centre reaction. In summary, our findings demonstrate that Maf acts intrinsically in B cells as a negative regulator of late B cell differentiation, plasmablast proliferation and germinal centre B cell formation.
Collapse
Affiliation(s)
- Sophie Hillion
- LBAI, UMR1227, Univ Brest, Inserm, and CHU de Brest, Brest, France.
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Anjelica Miranda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | | | | | | | - Divi Cornec
- LBAI, UMR1227, Univ Brest, Inserm, and CHU de Brest, Brest, France
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
26
|
Liu M. Effector and regulatory B-cell imbalance in systemic sclerosis: cooperation or competition? Clin Rheumatol 2024; 43:2783-2789. [PMID: 39080112 PMCID: PMC11330388 DOI: 10.1007/s10067-024-07086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/18/2024]
Abstract
B cells play a central role in the pathogenesis of systemic sclerosis (SSc). Most B-cell studies have focused on their pathological role as antibody producers. However, in addition to immunoglobulin secretion, these cells have a wide range of functions in the immune response, including antigen presentation to T cells and cytokine production. Importantly, not all B-cell subsets promote the immune response. Regulatory B cells (Bregs) attenuate inflammation and contribute to the maintenance of immune tolerance. However, effector B cells (Beffs) positively modulate the immune response through the production of various cytokines. In SSc, Bregs are insufficient and/or dysfunctional. B-cell-targeting biologics have been trialled with promising results in the treatment of SSc. These therapies can affect Bregs or Beffs, which can potentially limit their long-term efficacy. Future strategies might involve the modulation of effector B cells in combination with the stimulation of regulatory subsets. Additionally, the monitoring of individual B-cell subsets in patients may lead to the discovery of novel biomarkers that could help predict disease relapse or progression. The purpose of this review is to summarize the relevant literatures and explain how Bregs and Beffs jointly participate in the pathogenesis of SSc.
Collapse
Affiliation(s)
- Mengguo Liu
- Department of Dermatology, Huashan Hospital, Fudan University, the 12Th Urumqi Road, Shanghai, 200040, China.
| |
Collapse
|
27
|
Ahmed Z, Xiang W, Wang F, Nawaz M, Kuthu ZH, Lei C, Xu D. Whole-genome resequencing deciphers patterns of genetic diversity, phylogeny, and evolutionary dynamics in Kashmir cattle. Anim Genet 2024; 55:511-526. [PMID: 38726735 DOI: 10.1111/age.13434] [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: 04/02/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 07/04/2024]
Abstract
Kashmir cattle, which were kept by local pastoralists for centuries, are exceptionally resilient and adaptive to harsh environments. Despite its significance, the genomic characteristics of this cattle breed remain elusive. This study utilized whole genome sequences of Kashmir cattle (n = 20; newly sequenced) alongside published whole genomes of 32 distinct breeds and seven core cattle populations (n = 135). The analysis identified ~25.87 million biallelic single nucleotide polymorphisms in Kashmir cattle, predominantly in intergenic and intron regions. Population structure analyses revealed distinct clustering patterns of Kashmir cattle with proximity to the South Asian, African and Chinese indicine cattle populations. Genetic diversity analysis of Kashmir cattle demonstrated lower inbreeding and greater nucleotide diversity than analyzed global breeds. Homozygosity runs indicated less consanguineous mating in Kashmir cattle compared with European taurine breeds. Furthermore, six selection sweep detection methods were used within Kashmir cattle and other cattle populations to identify genes associated with vital traits, including immunity (BOLA-DQA5, BOLA-DQB, TNFAIP8L, FCRL4, AOAH, HIF1AN, FBXL3, MPEG1, CDC40, etc.), reproduction (GOLGA4, BRWD1, OSBP2, LEO1 ADCY5, etc.), growth (ADPRHL1, NRG2, TCF12, TMOD4, GBP4, IGF2, RSPO3, SCD, etc.), milk composition (MRPS30 and CSF1) and high-altitude adaptation (EDNRA, ITPR2, AGBL4 and SCG3). These findings provide essential genetic insights into the characteristics and establish the foundation for the scientific conservation and utilization of Kashmir cattle breed.
Collapse
Affiliation(s)
- Zulfiqar Ahmed
- College of Animal Science and Technology, Huazhong Agriculture University, Wuhan, China
- NCLBG&G, Department of Livestock and Poultry Production, Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Azad Jammu and Kashmir, Pakistan
| | - Weixuan Xiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Fuwen Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Mohsin Nawaz
- NCLBG&G, Department of Livestock and Poultry Production, Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Azad Jammu and Kashmir, Pakistan
| | - Zulfiqar Hussan Kuthu
- NCLBG&G, Department of Livestock and Poultry Production, Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Azad Jammu and Kashmir, Pakistan
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Dequan Xu
- College of Animal Science and Technology, Huazhong Agriculture University, Wuhan, China
| |
Collapse
|
28
|
Han J, Zhao B, Han X, Sun T, Yue M, Hou M, Wu J, Tu M, An Y. Comprehensive Analysis of a Six-Gene Signature Predicting Survival and Immune Infiltration of Liposarcoma Patients and Deciphering Its Therapeutic Significance. Int J Mol Sci 2024; 25:7792. [PMID: 39063036 PMCID: PMC11277418 DOI: 10.3390/ijms25147792] [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: 06/04/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND As a common soft tissue sarcoma, liposarcoma (LPS) is a heterogeneous malignant tumor derived from adipose tissue. Due to the high risk of metastasis and recurrence, the prognosis of LPS remains unfavorable. To improve clinical treatment, a robust risk prediction model is essential to evaluate the prognosis of LPS patients. METHODS By comprehensive analysis of data derived from GEO datasets, differentially expressed genes (DEGs) were obtained. Univariate and Lasso Cox regressions were subsequently employed to reveal distant recurrence-free survival (DRFS)-associated DEGs and develop a prognostic gene signature, which was assessed by Kaplan-Meier survival and ROC curve. GSEA and immune infiltration analyses were conducted to illuminate molecular mechanisms and immune correlations of this model in LPS progression. Furthermore, a correlation analysis was involved to decipher the therapeutic significance of this model for LPS. RESULTS A six-gene signature was developed to predict DRFS of LPS patients and showed higher precision performance in more aggressive LPS subtypes. Then, a nomogram was further established for clinical application based on this risk model. Via GSEA, the high-risk group was significantly enriched in cell cycle-related pathways. In the LPS microenvironment, neutrophils, memory B cells and resting mast cells exhibited significant differences in cell abundance between high-risk and low-risk patients. Moreover, this model was significantly correlated with therapeutic targets. CONCLUSION A prognostic six-gene signature was developed and significantly associated with cell cycle pathways and therapeutic target genes, which could provide new insights into risk assessment of LPS progression and therapeutic strategies for LPS patients to improve their prognosis.
Collapse
Affiliation(s)
- Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Man Yue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Mengwen Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Jialin Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| |
Collapse
|
29
|
Noble J, Cabezas L, Truffot A, Dumolard L, Jouve T, Malvezzi P, Rostaing L, Dard C, Saas P, Cravedi P, Macek-Jilkova Z. Glycolysis Changes in Alloreactive Memory B Cells in Highly Sensitized Kidney Transplant Recipients Undergoing Desensitization Therapy. Transpl Int 2024; 37:13029. [PMID: 39081904 PMCID: PMC11287219 DOI: 10.3389/ti.2024.13029] [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: 03/23/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
Despite the growing use of desensitization strategies, hyperimmune patients remain at high risk of antibody-mediated rejection suggesting that, even when donor-specific antibodies (DSA) are effectively depleted, anti-donor specific B cells persist. We included 10 highly sensitized recipients that underwent desensitization with plasmapheresis and B cell depletion prior to kidney transplantation. We quantified changes in DSA (luminex), total B-cell subsets (flow cytometry), anti-donor HLA B cells (fluorospot), and single-cell metabolism in serially collected samples before desensitization, at the time of transplant, and at 6 and 12 months thereafter. Desensitization was associated with a decrease in DSA and total memory B cell and naive B cell percentage, while plasma cells and memory anti-donor HLA circulating B cells persisted up to 12 months after transplant. At 12-month post-transplantation, memory B cells increased their glycolytic capacity, while proliferative KI67+ plasma cells modified their metabolism by increasing fatty acid and amino acid oxidation capacity and decreasing their glucose dependence. Despite effective DSA depletion, anti-donor B cells persist in kidney transplant recipients. Due to the reliance of these cells on glycolysis, glycolysis-targeting therapies might represent a valuable treatment strategy.
Collapse
Affiliation(s)
- Johan Noble
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lara Cabezas
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Aurelie Truffot
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Virology Department, University Hospital Grenoble, Grenoble, France
| | - Lucile Dumolard
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Thomas Jouve
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Paolo Malvezzi
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
| | - Lionel Rostaing
- Nephrology, Hemodialysis Apheresis and Kidney Transplantation, Department, CHU Grenoble Alpes, Grenoble, France
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| | - Céline Dard
- EFS, Recherche et Développement, Grenoble, France
| | - Philippe Saas
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- EFS, Recherche et Développement, Grenoble, France
| | - Paolo Cravedi
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zuzana Macek-Jilkova
- University Grenoble Alpes, CNRS, Inserm, CHU Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
- Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
30
|
Lenert ME, Green AR, Merriwether EN, Burton MD. B-cell and plasma cell activation in a mouse model of chronic muscle pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100169. [PMID: 39507010 PMCID: PMC11539501 DOI: 10.1016/j.ynpai.2024.100169] [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: 06/02/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024]
Abstract
Fibromyalgia (FM) is a complex chronic musculoskeletal pain disorder with an elusive pathogenesis, with a strong implication of immune interactions. We recently found that IL-5 and the adaptive immune system mediates pain outcomes in fibromyalgia (FM) patients and preclinical models of FM-like chronic widespread pain (CWP). However, there is an active debate if FM/CWP has an autoimmune etiology. Preclinical models of CWP utilize a repeated insult paradigm, which resembles a primary, then secondary response similarly observed in the antibody response, in which the subsequent event causes a potentiated pain response. Recent translational studies have implicated immunoglobulins (Ig) and B-cells in FM/CWP pathophysiology. To understand if these are involved in preclinical models of CWP, we performed comprehensive B-cell phenotyping in the bone marrow, circulation, and popliteal (draining) lymph nodes in the two-hit acidic saline model of CWP. We found increased MHC class II-expressing B-cells in peripheral blood, increased activated plasma cells in peripheral blood, and increased memory B-cells in the bone marrow. Interestingly, acidic pH (4.0) injected mice have reduced levels of IgG1, independent of treatment with IL-5. We have demonstrated that the acidic saline model of CWP induces T-cell mediated activation of B-cells, increased active plasma cells, and increased memory B-cells in female mice.
Collapse
Affiliation(s)
- Melissa E. Lenert
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Audrey R. Green
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Ericka N. Merriwether
- Inclusive and Translational Research in Pain Lab Department of Physical Therapy Steinhardt School of Culture, Education, and Human Development New York University 380 Second Avenue, 4th Floor New York, NY 10012, USA
| | - Michael D. Burton
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| |
Collapse
|
31
|
Elsner RA, Smita S, Shlomchik MJ. IL-12 induces a B cell-intrinsic IL-12/IFNγ feed-forward loop promoting extrafollicular B cell responses. Nat Immunol 2024; 25:1283-1295. [PMID: 38862796 PMCID: PMC11992614 DOI: 10.1038/s41590-024-01858-1] [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] [Received: 05/12/2023] [Accepted: 04/26/2024] [Indexed: 06/13/2024]
Abstract
While some infections elicit germinal centers, others produce only extrafollicular responses. The mechanisms controlling these dichotomous fates are poorly understood. We identify IL-12 as a cytokine switch, acting directly on B cells to promote extrafollicular and suppress germinal center responses. IL-12 initiates a B cell-intrinsic feed-forward loop between IL-12 and IFNγ, amplifying IFNγ production, which promotes proliferation and plasmablast differentiation from mouse and human B cells, in synergy with IL-12. IL-12 sustains the expression of a portion of IFNγ-inducible genes. Together, they also induce unique gene changes, reflecting both IFNγ amplification and cooperative effects between both cytokines. In vivo, cells lacking both IL-12 and IFNγ receptors are more impaired in plasmablast production than those lacking either receptor alone. Further, B cell-derived IL-12 enhances both plasmablast responses and T helper 1 cell commitment. Thus, B cell-derived IL-12, acting on T and B cells, determines the immune response mode, with implications for vaccines, pathogen protection and autoimmunity.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Shuchi Smita
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
32
|
Lin DL, Magnaye KM, Porsche CE, Levan SR, Rackaityte E, Özçam M, Lynch SV. 12,13-diHOME Promotes Inflammatory Macrophages and Epigenetically Modifies Their Capacity to Respond to Microbes and Allergens. J Immunol Res 2024; 2024:2506586. [PMID: 38974097 PMCID: PMC11227377 DOI: 10.1155/2024/2506586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/10/2024] [Accepted: 05/25/2024] [Indexed: 07/09/2024] Open
Abstract
Elevated infant fecal concentrations of the bacterial-derived lipid 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) increase the risk for childhood atopy and asthma. However, the mechanisms by which this lipid contributes to disease development are largely unknown. We hypothesized that macrophages, which are key to both antimicrobial and antigen responses, are functionally and epigenetically modified by 12,13-diHOME leading to short- and long-term dysfunction with consequences for both antimicrobial and antigenic responses. Macrophages exposed to 12,13-diHOME are skewed toward inflammatory IL-1β highCD206low cells, a phenomenon that is further amplified in the presence of common microbial-, aero-, and food-allergens. These IL-1β highCD206low macrophages also exhibit reduced bacterial phagocytic capacity. In primary immune cell coculture assays involving peanut allergen stimulation, 12,13-diHOME promotes both IL-1β and IL-6 production, memory B cell expansion, and increased IgE production. Exposure to 12,13-diHOME also induces macrophage chromatin remodeling, specifically diminishing access to interferon-stimulated response elements resulting in reduced interferon-regulated gene expression upon bacterial lipopolysaccharide stimulation. Thus 12,13-diHOME reprograms macrophage effector function, B-cell interactions and promotes epigenetic modifications that exacerbate inflammatory response to allergens and mutes antimicrobial response along the interferon axis. These observations offer plausible mechanisms by which this lipid promotes early-life pathogenic microbiome development and innate immune dysfunction associated with childhood allergic sensitization.
Collapse
Affiliation(s)
- Din L. Lin
- Division of GastroenterologyDepartment of MedicineUniversity of California, San Francisco, CA 94143, USA
| | - Kevin M. Magnaye
- Division of GastroenterologyDepartment of MedicineUniversity of California, San Francisco, CA 94143, USA
| | - Cara E. Porsche
- Division of GastroenterologyDepartment of MedicineUniversity of California, San Francisco, CA 94143, USA
| | - Sophia R. Levan
- Division of GastroenterologyDepartment of MedicineUniversity of California, San Francisco, CA 94143, USA
| | - Elze Rackaityte
- Division of GastroenterologyDepartment of MedicineUniversity of California, San Francisco, CA 94143, USA
| | - Mustafa Özçam
- Division of GastroenterologyDepartment of MedicineUniversity of California, San Francisco, CA 94143, USA
| | - Susan V. Lynch
- Division of GastroenterologyDepartment of MedicineUniversity of California, San Francisco, CA 94143, USA
| |
Collapse
|
33
|
Ma C, Hoffmann FW, Shay AE, Koo I, Green KA, Green WR, Hoffmann PR. Upregulated selenoprotein I during lipopolysaccharide-induced B cell activation promotes lipidomic changes and is required for effective differentiation into IgM-secreting plasma B cells. J Leukoc Biol 2024; 116:6-17. [PMID: 38289835 PMCID: PMC11212798 DOI: 10.1093/jleuko/qiae024] [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: 09/13/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
The mechanisms driving metabolic reprogramming during B cell activation are unclear, particularly roles for enzymatic pathways involved in lipid remodeling. We found that murine B cell activation with lipopolysaccharide (LPS) led to a 1.6-fold increase in total lipids that included higher levels of phosphatidylethanolamine (PE) and plasmenyl PE. Selenoprotein I (SELENOI) is an ethanolamine phospholipid transferase involved in the synthesis of both PE and plasmenyl PE, and SELENOI expression was also upregulated during activation. Selenoi knockout (KO) B cells exhibited decreased levels of plasmenyl PE, which plays an important antioxidant role. Lipid peroxidation was measured and found to increase ∼2-fold in KO vs. wild-type (WT) B cells. Cell death was not impacted by KO in LPS-treated B cells and proliferation was only slightly reduced, but differentiation into CD138 + Blimp-1+ plasma B cells was decreased ∼2-fold. This led to examination of B cell receptors important for differentiation that recognize the ligand B cell activating factor, and levels of TACI (transmembrane activator, calcium-modulator, and cytophilin ligand interactor) (CD267) were significantly decreased on KO B cells compared with WT control cells. Vaccination with ovalbumin/adjuvant led to decreased ovalbumin-specific immunoglobulin M (IgM) levels in sera of KO mice compared with WT mice. Real-time polymerase chain reaction analyses revealed a decreased switch from surface to secreted IgM in spleens of KO mice induced by vaccination or LP-BM5 retrovirus infection. Overall, these findings detail the lipidomic response of B cells to LPS activation and reveal the importance of upregulated SELENOI for promoting differentiation into IgM-secreting plasma B cells.
Collapse
Affiliation(s)
- Chi Ma
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, United States
| | - FuKun W Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, United States
| | - Ashley E Shay
- Huck Institutes of the Life Sciences, The Pennsylvania State University, 101 Huck Life Sciences Building, University Park, PA 16802, United States
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 107 Animal, Veterinary, and Biomedical Sciences Building, University Park, PA 16802, United States
| | - Kathy A Green
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, One Medical Center Drive HB7556, Lebanon, NH 03756, United States
| | - William R Green
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, One Medical Center Drive HB7556, Lebanon, NH 03756, United States
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI 96813, United States
| |
Collapse
|
34
|
Li Y, Jin H, Li Q, Shi L, Mao Y, Zhao L. The role of RNA methylation in tumor immunity and its potential in immunotherapy. Mol Cancer 2024; 23:130. [PMID: 38902779 PMCID: PMC11188252 DOI: 10.1186/s12943-024-02041-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
RNA methylation, a prevalent post-transcriptional modification, has garnered considerable attention in research circles. It exerts regulatory control over diverse biological functions by modulating RNA splicing, translation, transport, and stability. Notably, studies have illuminated the substantial impact of RNA methylation on tumor immunity. The primary types of RNA methylation encompass N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), and N7-methylguanosine (m7G), and 3-methylcytidine (m3C). Compelling evidence underscores the involvement of RNA methylation in regulating the tumor microenvironment (TME). By affecting RNA translation and stability through the "writers", "erasers" and "readers", RNA methylation exerts influence over the dysregulation of immune cells and immune factors. Consequently, RNA methylation plays a pivotal role in modulating tumor immunity and mediating various biological behaviors, encompassing proliferation, invasion, metastasis, etc. In this review, we discussed the mechanisms and functions of several RNA methylations, providing a comprehensive overview of their biological roles and underlying mechanisms within the tumor microenvironment and among immunocytes. By exploring how these RNA modifications mediate tumor immune evasion, we also examine their potential applications in immunotherapy. This review aims to provide novel insights and strategies for identifying novel targets in RNA methylation and advancing cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Yan Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Haoer Jin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qingling Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Liangrong Shi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yitao Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Luqing Zhao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
35
|
Prakash S, Kumbhojkar N, Gottlieb AP, Park KS, Kapate N, Mitragotri S. Polymer Micropatches as B-Cell Engagers. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28184-28192. [PMID: 38770711 DOI: 10.1021/acsami.4c04385] [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: 05/22/2024]
Abstract
B cells, despite their several unique functionalities, remain largely untapped for use as an adoptive cell therapy and are limited to in vitro use for antibody production. B cells can be easily sourced, they possess excellent lymphoid-homing capabilities, and they can act as antigen-presenting cells (APCs), offering an alternative to dendritic cells (DCs), which have shown limited efficacy in the clinical setting. Soluble factors such as IL-4 and anti-CD40 antibody can enhance the activation, survival, and antigen-presenting capabilities of B cells; however, it is difficult to attain sufficiently high concentrations of these biologics to stimulate B cells in vivo. Micropatches as Cell Engagers (MACE) are polymeric microparticles, surface functionalized with anti-CD40 and anti-IgM, which can attach to B cells and simultaneously engage multiple B-cell receptors (BCR) and CD40 receptors. Stimulation of these receptors through MACE, unlike free antibodies, enhanced the display of costimulatory molecules on the B-cell surface, increased B-cell viability, and improved antigen presentation by B cells to T cells in vitro. B-cell activation by MACE further synergized with soluble IL-4 and anti-CD40. MACE also elicited T-cell chemokine secretion by B cells. Upon intravenous adoptive transfer, MACE-bound B cells homed to the spleen and lymph nodes, key sites for antigen presentation to T cells. Adoptive transfer of MACE-B cells pulsed with the CD4+ and CD8+ epitopes of ovalbumin significantly delayed tumor progression in a murine subcutaneous EG7-OVA tumor model, demonstrating the functional benefit conferred to B cells by MACE.
Collapse
Affiliation(s)
- Supriya Prakash
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
| | - Ninad Kumbhojkar
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
| | - Alexander P Gottlieb
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
| | - Kyung-Soo Park
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
| | - Neha Kapate
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
- Harvard-MIT Program in Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Samir Mitragotri
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
| |
Collapse
|
36
|
Shen X, Wang S, Hao Y, Fu Y, Ren L, Li D, Tang W, Li J, Chen R, Zhu M, Wang S, Liu Y, Shao Y. DNA vaccine prime and replicating vaccinia vaccine boost induce robust humoral and cellular immune responses against MERS-CoV in mice. Virol Sin 2024; 39:490-500. [PMID: 38768713 PMCID: PMC11279798 DOI: 10.1016/j.virs.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 05/15/2024] [Indexed: 05/22/2024] Open
Abstract
As of December 2022, 2603 laboratory-identified Middle East respiratory syndrome coronavirus (MERS-CoV) infections and 935 associated deaths, with a mortality rate of 36%, had been reported to the World Health Organization (WHO). However, there are still no vaccines for MERS-CoV, which makes the prevention and control of MERS-CoV difficult. In this study, we generated two DNA vaccine candidates by integrating MERS-CoV Spike (S) gene into a replicating Vaccinia Tian Tan (VTT) vector. Compared to homologous immunization with either vaccine, mice immunized with DNA vaccine prime and VTT vaccine boost exhibited much stronger and durable humoral and cellular immune responses. The immunized mice produced robust binding antibodies and broad neutralizing antibodies against the EMC2012, England1 and KNIH strains of MERS-CoV. Prime-Boost immunization also induced strong MERS-S specific T cells responses, with high memory and poly-functional (CD107a-IFN-γ-TNF-α) effector CD8+ T cells. In conclusion, the research demonstrated that DNA-Prime/VTT-Boost strategy could elicit robust and balanced humoral and cellular immune responses against MERS-CoV-S. This study not only provides a promising set of MERS-CoV vaccine candidates, but also proposes a heterologous sequential immunization strategy worthy of further development.
Collapse
MESH Headings
- Animals
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Middle East Respiratory Syndrome Coronavirus/immunology
- Middle East Respiratory Syndrome Coronavirus/genetics
- Immunity, Cellular
- Antibodies, Viral/blood
- Mice
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Immunity, Humoral
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Female
- Coronavirus Infections/prevention & control
- Coronavirus Infections/immunology
- Mice, Inbred BALB C
- CD8-Positive T-Lymphocytes/immunology
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Immunization, Secondary
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
Collapse
Affiliation(s)
- Xiuli Shen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shuhui Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yanling Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yuyu Fu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Li Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Dan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Wenqi Tang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jing Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ran Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Meiling Zhu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shuo Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ying Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Yiming Shao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
37
|
Liang X, Guo X, Jin H, Shen L, Ding L, Guan X, Kou Y, Wu Y, Guo H. Changes in the intestinal microbiota of multiple myeloma patients living in high‑altitude and cold regions analyzed using 16s rRNA high‑throughput sequencing. Exp Ther Med 2024; 27:269. [PMID: 38756900 PMCID: PMC11097272 DOI: 10.3892/etm.2024.12557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
Multiple myeloma (MM) is a plasma cell clonal disease and these plasma cells can survive in the gut. The intestinal microbiota is a complex ecosystem and its dysfunction can release persistent stimulus signals that trigger genetic mutations and clonal evolution in the gut. The present study analyzed the intestinal microbiota in fecal samples of MM patients in high-altitude and cold regions of China using 16s rRNA sequencing and analyzed significantly enriched species at the phylum and genus levels. Although no significant difference in the alpha diversity was observed between the MM and control groups, a significant difference was noted in the beta diversity. A total of 15 significant differential bacteria at the genus level were found between the two groups, among which Bacteroides, Streptococcus, Lactobacillus and Alistipes were significantly enriched in the MM group. The present study also constructed a disease diagnosis model using Random Forest analysis and verified its accuracy using receiver operating characteristic analysis. In addition, using correlation analysis, it demonstrated that the composition of the intestinal microbiota in patients with MM was associated with complement levels. Notably, the present study predicted that the signaling and metabolic pathways of the intestinal microbiota affected MM progression through Kyoto Encyclopedia of Genes and Genomes functional analysis. The present study provides a new approach for the prevention and treatment of MM, in which the intestinal microbiota may become a novel therapeutic target for MM.
Collapse
Affiliation(s)
- Xiaofei Liang
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Xuyang Guo
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Huixin Jin
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Lijuan Shen
- Department of Laboratory Medicine, Qiqihar MingZhu Hospital, Qiqihar, Heilongjiang 161000, P.R. China
| | - Ling Ding
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Xin Guan
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Yujie Kou
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Yi Wu
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Haipeng Guo
- Department of Laboratory Medicine, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| |
Collapse
|
38
|
Shaghayegh G, Cooksley C, Bouras G, Panchatcharam BS, Feizi S, Javadian S, Ramezanpour M, Fenix KA, Wormald PJ, Psaltis AJ, Vreugde S. S. aureus biofilm properties correlate with immune B cell subset frequencies and severity of chronic rhinosinusitis. Clin Immunol 2024; 263:110221. [PMID: 38636891 DOI: 10.1016/j.clim.2024.110221] [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: 12/20/2023] [Revised: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Staphylococcus aureus mucosal biofilms are associated with recalcitrant chronic rhinosinusitis (CRS). However, S. aureus colonisation of sinus mucosa is frequent in the absence of mucosal inflammation. This questions the relevance of S. aureus biofilms in CRS etiopathogenesis. This study aimed to investigate whether strain-level variation in in vitro-grown S. aureus biofilm properties relates to CRS disease severity, in vitro toxicity, and immune B cell responses in sinonasal tissue from CRS patients and non-CRS controls. S. aureus clinical isolates, tissue samples, and matched clinical datasets were collected from CRS patients with nasal polyps (CRSwNP), CRS without nasal polyps (CRSsNP), and controls. B cell responses in tissue samples were characterised by FACS. S. aureus biofilms were established in vitro, followed by measuring their properties of metabolic activity, biomass, colony-forming units, and exoprotein production. S. aureus virulence was evaluated using whole-genome sequencing, mass spectrometry and application of S. aureus biofilm exoproteins to air-liquid interface cultures of primary human nasal epithelial cells (HNEC-ALI). In vitro S. aureus biofilm properties were correlated with increased CRS severity scores, infiltration of antibody-secreting cells and loss of regulatory B cells in tissue samples. Biofilm exoproteins from S. aureus with high biofilm metabolic activity had enriched virulence genes and proteins, and negatively affected the barrier function of HNEC-ALI cultures. These findings support the notion of strain-level variation in S. aureus biofilms to be critical in the pathophysiology of CRS.
Collapse
Affiliation(s)
- Gohar Shaghayegh
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Clare Cooksley
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - George Bouras
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Beula Subashini Panchatcharam
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Sholeh Feizi
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Shari Javadian
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Mahnaz Ramezanpour
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Kevin Aaron Fenix
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Peter-John Wormald
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Alkis James Psaltis
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia
| | - Sarah Vreugde
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Department of Surgery-Otolaryngology, Head and Neck Surgery, University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, South Australia, Australia.
| |
Collapse
|
39
|
Deng Y, Song L, Huang J, Zhou W, Liu Y, Lu X, Zhao H, Liu D. Astragalus polysaccharides ameliorates experimental colitis by regulating memory B cells metabolism. Chem Biol Interact 2024; 394:110969. [PMID: 38522565 DOI: 10.1016/j.cbi.2024.110969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
It is well-established that the reduced Memory B cells (MBCs) play an important role in the pathogenesis of ulcerative colitis (UC), rendering them a potential therapeutic target for UC intervention. Astragalus polysaccharide (APS), a primary active constituent derived from the classic traditional Chinese medicine Astragalus membranaceus (AM), has been used for centuries in the treatment of UC in both human and animal subjects due to its renowned immunomodulatory properties. However, it is unknown whether APS can regulate MBCs to alleviate experimental colitis. In the present investigation, the murine colitis was successfully induced using dextran sulphate sodium (DSS) and subsequently treated with APS for a duration of 7 days. APS exhibited significant efficacy in reducing the disease activity index (DAI), colonic weight index, the index of colonic weight/colonic length. Furthermore, APS mitigated colonic pathological injuries, restored the colonic length, elevated the immunoglobulin A (IgA), transforming growth factor-β1 (TGF-β1) and interleukin (IL)-10 levels, while concurrently suppressing IgG, IgM, IL-6, tumor necrosis factor alpha (TNF-α) levels. Crucially, the quantities of MBCs, IgA+MBCs and forkhead box P3 (Foxp3+) MBCs were notably increased along with a concurrent decrease in IgG1+MBCs, IG2a+MBCs, IgG2b+MBCs after APS administration in colitis mice. Additionally, the Mitotracker red expressions of MBCs and their subgroups demonstrated a significantly up-regulation. Meanwhile, the transcriptomics analysis identified mitochondrial metabolism as the predominant and pivotal mechanism underlying APS-mediated mitigation of DSS-induced colitis. Key differentially expressed genes, including B-cell linker (BLNK), aldehyde dehydrogenase 1A1 (ALDH1A1), B-cell lymphoma 6 (BCL-6), B-lymphocyte-induced maturation protein 1 (Blimp-1), paired box gene 5 (PAX5), purinergic 2 × 7 receptor (P2X7R), B Cell activation factor (BAFF), B Cell activation factor receptor (BAFFR), CD40, nuclear factor kappa-B (NF-κB), IL-6 and so on were implicated in this process. These mRNA expressions were validated through quantitative polymerase chain reaction (qPCR) and immunohistochemistry. These findings revealed that APS effectively restored MBCs and their balance to ameliorate DSS-induced colitis, which was potentially realized via promoting mitochondrial metabolism to maintain MBCs activation.
Collapse
Affiliation(s)
- Yifei Deng
- Clinical Medical School, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Lizhao Song
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Jiaqi Huang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Wen Zhou
- Nanchang Medical College, Nanchang, 330052, Jiangxi Province, China
| | - Yali Liu
- Nanchang Medical College, Nanchang, 330052, Jiangxi Province, China
| | - Xiuyun Lu
- Nanchang Medical College, Nanchang, 330052, Jiangxi Province, China.
| | - Haimei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China; Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Duanyong Liu
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China; School of Nursing, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| |
Collapse
|
40
|
Walzik D, Belen S, Wilisch K, Kupjetz M, Kirschke S, Esser T, Joisten N, Schenk A, Proschinger S, Zimmer P. Impact of exercise on markers of B cell-related immunity: A systematic review. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 13:339-352. [PMID: 37832643 PMCID: PMC11116964 DOI: 10.1016/j.jshs.2023.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND B cells represent a crucial component of adaptive immunity that ensures long-term protection from infection by generating pathogen-specific immunoglobulins. Exercise alters B cell counts and immunoglobulin levels, but evidence-based conclusions on potential benefits for adaptive immunity are lacking. This systematic review assessed current literatures on the impact of acute exercise and exercise training on B cells, immunoglobulins, and markers of secretory immunity in human biofluids. METHODS According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, MEDLINE, Web of Science, and Embase were searched on March 8, 2023. Non-randomized controlled trials and crossover trials investigating the impact of acute exercise or exercise training on B cell counts and proportions, immunoglobulin levels, salivary flow rate, or secretory immunoglobulin A secretion rate were included. Quality and reporting of exercise training studies were assessed using the Tool for the Assessment of Study Quality and reporting in Exercise. Study characteristics, outcome measures, and statistically significant changes were summarized tabularly. RESULTS Of the 67 eligible studies, 22 applied acute exercise and 45 applied exercise training. All included outcomes revealed significant alterations over time in acute exercise and exercise training context, but only a few investigations showed significant differences compared to control conditions. Secretory and plasma immunoglobulin A levels were most consistently increased in response to exercise training. CONCLUSION B cell-related outcomes are altered by acute exercise and exercise training, but evidence-based conclusions cannot be drawn with high confidence due to the large heterogeneity in populations and exercise modalities. Well-designed trials with large sample sizes are needed to clarify how exercise shapes B cell-related immunity.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Sergen Belen
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Karen Wilisch
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Marie Kupjetz
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Silvana Kirschke
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Tobias Esser
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Alexander Schenk
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Sebastian Proschinger
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, Dortmund 44227, Germany.
| |
Collapse
|
41
|
Li Y, Yang R, Yin F, Zhang H, Zhai G, Sun S, Tian B, Zeng Q. Correlation between 146S Antigen Content in Foot-and-Mouth Disease Inactivated Vaccines and Immunogenicity Level and Vaccine Potency Alternative Test Methods. Vet Sci 2024; 11:168. [PMID: 38668435 PMCID: PMC11053669 DOI: 10.3390/vetsci11040168] [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: 03/08/2024] [Revised: 03/30/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
To investigate the association between 146S antigen contents in FMD inactivated vaccines and levels of antiviral immunity, this study vaccinated 30 kg pigs with three batches of FMD types O and A bivalent inactivated vaccines. Antibody titers and interferon-gamma (IFN-γ) secretion levels were measured on days 7, 14, 21, and 28 after primary immunization and on days 14 and 28 following booster immunization to assess associations between 146S contents and both antibody titers and IFN-γ secretion levels. Furthermore, 30 kg pigs were vaccinated with 46 batches of FMD type O inactivated vaccines and challenged on day 28, after which PD50 values were determined to evaluate the association between 146S content and PD50. The findings suggested that antibody titers and IFN-γ secretion levels at specific time points after immunization were positively associated with 146S contents. Additionally, 146S content showed a positive correlation with PD50, with greater PD50 values recorded for 146S contents ranging from 4.72 to 16.55 µg/dose. This investigation established a significant association between the 146S content in FMD inactivated vaccines and induced immune response against FMDV, thereby emphasizing its critical role in vaccine quality control. The determination of 146S content could serve as a new method for potency testing, offering an alternative to animal challenge tests.
Collapse
Affiliation(s)
- Yongxia Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
- China Agricultural Veterinary Biotechnology Co., Ltd., Lanzhou 730046, China
| | - Ruai Yang
- China Agricultural Veterinary Biotechnology Co., Ltd., Lanzhou 730046, China
| | - Fu Yin
- China Agricultural Veterinary Biotechnology Co., Ltd., Lanzhou 730046, China
| | - Haisheng Zhang
- China Agricultural Veterinary Biotechnology Co., Ltd., Lanzhou 730046, China
| | - Guoyuan Zhai
- China Agricultural Veterinary Biotechnology Co., Ltd., Lanzhou 730046, China
| | - Shiqi Sun
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Bo Tian
- China Agricultural Veterinary Biotechnology Co., Ltd., Lanzhou 730046, China
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| |
Collapse
|
42
|
Yi XY, Hou XR, Huang ZX, Zhu P, Liu BY. Immunization with a peptide mimicking lipoteichoic acid induces memory B cells in BALB/c mice. BMC Infect Dis 2024; 24:371. [PMID: 38566017 PMCID: PMC10986077 DOI: 10.1186/s12879-024-09262-8] [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: 06/16/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND There is an urgent clinical need for developing novel immunoprophylaxis and immunotherapy strategies against Staphylococcus aureus (S. aureus). In our previous work, immunization with a tetra-branched multiple antigenic peptide, named MAP2-3 that mimics lipoteichoic acid, a cell wall component of S. aureus, successfully induced a humoral immune response and protected BALB/c mice against S. aureus systemic infection. In this study, we further investigated whether vaccination with MAP2-3 can elicit immunologic memory. METHODS BALB/c mice were immunized with MAP2-3 five times. After one month of the last vaccination, mice were challenged with heat-killed S. aureus via intraperitoneal injection. After a 7-day inoculation, the percentage of plasma cells, memory B cells, effector memory T cells, and follicular helper T cells were detected by flow cytometry. The levels of IL-6, IL-21, IL-2, and IFN-γ were measured by real-time PCR and ELISA. Flow cytometry results were compared by using one-way ANOVA or Mann-Whitney test, real-time PCR results were compared by using one-way ANOVA, and ELISA results were compared by using one-way ANOVA or student's t-test. RESULTS The percentage of plasma cells and memory B cells in the spleen and bone marrow from the MAP2-3 immunized mice was significantly higher than that from the control mice. The percentage of effector memory T cells in spleens and lymphoid nodes as well as follicular helper T cells in spleens from the MAP2-3 immunized mice were also higher. Moreover, the levels of IL-6 and IL-21, two critical cytokines for the development of memory B cells, were significantly higher in the isolated splenocytes from immunized mice after lipoteichoic acid stimulation. CONCLUSIONS Immunization with MAP2-3 can efficiently induce memory B cells and memory T cells.
Collapse
Affiliation(s)
- Xia-Yu Yi
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
- Department of Clinical Laboratory, The First People's Hospital of Wuhu, Wuhu, Anhui, P.R. China
| | - Xiao-Rui Hou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Zhao-Xia Huang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, P.R. China
| | - Ping Zhu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Bei-Yi Liu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
43
|
Tan C, Zhou J, Wu A, Li C. In silico development of a novel anti-mutation, multi-epitope mRNA vaccine against MPXV variants of emerging lineage and sub-lineages by using immunoinformatics approaches. J Biomol Struct Dyn 2024:1-18. [PMID: 38450722 DOI: 10.1080/07391102.2024.2325109] [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: 10/26/2023] [Accepted: 02/24/2024] [Indexed: 03/08/2024]
Abstract
Over the past year, an unexpected surge in human monkeypox (hMPX) cases has been observed. This outbreak differs from previous ones, displaying distinct epidemiological characteristics and transmission patterns, believed to be influenced by a newly emerging monkeypox virus (MPXV) lineage. Notably, this emerging MPXV lineage has exhibited several non-synonymous mutations, some of which are linked to immunomodulatory activities and antigenic characteristics that aid in host detection. However, specific treatments or vaccines for human monkeypox are currently lacking. Hence, we aim to develop a multi-epitope mRNA vaccine by using immunoinformatics approaches against the MPXV, particularly its emerging variants. Six proteins (A29L, A35R, B6R, M1R, H3L, and E8L) were chosen for epitope and mutation site identification. Seventeen top-performing epitopes and eight epitopes containing mutation sites were selected and combined with adjuvants, the PADRE sequence, and linkers for vaccine development. The molecular and physical properties of the designed vaccine (WLmpx) were favorable. Immunological characteristics of WLmpx were assessed through molecular docking, molecular dynamics (MD) simulations, and immune simulations. Finally, the vaccine sequence was utilized to formulate an mRNA-based vaccine. The informatics-based predicted results indicated that the designed vaccine exhibits significant potential in eliciting high-level humoral and cellular immune responses, but further validation through in vivo and vitro studies is warranted.
Collapse
Affiliation(s)
- Caixia Tan
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, China
| | - Jingxiang Zhou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, China
| | - Anhua Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, China
| | - Chunhui Li
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, China
| |
Collapse
|
44
|
Qi Y, Si H, Jin X, Guo Y, Xia J, He J, Deng X, Deng M, Yao W, Hao C. Expression levels of key immune indicators and immune checkpoints in manganese-exposed rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116029. [PMID: 38290312 DOI: 10.1016/j.ecoenv.2024.116029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/01/2024]
Abstract
Manganese is essential trace elements, to participate in the body a variety of biochemical reactions, has important physiological functions, such as stimulate the immune cell proliferation, strengthen the cellular immunity, etc. However, excessive manganese exposure can cause damage to multiple systems of the body.The immune system is extremely vulnerable to external toxicants, however manganese research on the immune system are inadequate and biomarkers are lacking. Therefore, here we applied a manganese-exposed rat model to make preliminary observations on the immunotoxic effects of manganese. We found that manganese exposure inhibited humoral immune function in rats by decreasing peripheral blood IgG (ImmunoglobulinG, IgG), IgM (ImmunoglobulinM, IgM) and complement C3 levels; It also regulates rat cellular immune activity by influencing peripheral blood, spleen, and thymus T cell numbers and immune organ ICs (Immune Checkpoints, ICs) and cytokine expression. Furthermore, it was revealed that the impact of manganese exposure on the immune function of rats exhibited a correlation with both the dosage and duration of exposure. Notably, prolonged exposure to high doses of manganese had the most pronounced influence on rat immune function, primarily manifesting as immunosuppression.The above findings suggest that manganese exposure leads to impaired immune function and related changes in immune indicators, or may provide clues for the discovery of its biomarkers.
Collapse
Affiliation(s)
- Yuanmeng Qi
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China
| | - Huifang Si
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China; Prevention and Infection Control Section, Xi'an Union Hospital, 710199 Xi'an, Shaanxi Province, China
| | - Xiaofei Jin
- Department of Ultrasound, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450015 Henan, China
| | - Yonghua Guo
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China
| | - Jiarui Xia
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China
| | - Jing He
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China
| | - Xuedan Deng
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China
| | - Meng Deng
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China
| | - Wu Yao
- Department of Occupational and Environment Health, College of Public Health, Zhengzhou University, 450001 Henan, China.
| | - Changfu Hao
- Department of Child and Adolescence Health, College of Public Health, Zhengzhou University, 450001 Henan, China.
| |
Collapse
|
45
|
Kortner TM, Afanasyev S, Koppang EO, Bjørgen H, Krogdahl Å, Krasnov A. A comprehensive transcriptional body map of Atlantic salmon unveils the vital role of the intestine in the immune system and highlights functional specialization within its compartments. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109422. [PMID: 38307300 DOI: 10.1016/j.fsi.2024.109422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/04/2024]
Abstract
The intestine is a barrier organ that plays an important role in the immune system of Atlantic salmon. The immune functions are distributed among the diffuse gut lymphoid tissue containing diverse immune cells, and other cell types. Comparison of intestinal transcriptomes with those of other organs and tissues offers an opportunity to elucidate the specific roles of the intestine and its relationship with other parts of the body. In this work, a meta-analysis was performed on a large volume of data obtained using a genome-wide DNA oligonucleotide microarray. The intestine ranks third by the expression level of immune genes after the spleen and head kidney. The activity of antigen presentation and innate antiviral immunity is higher in the intestine than in any other tissue. By comparing transcriptome profiles, intestine shows the greatest similarity with the gill, head kidney, spleen, epidermis, and olfactory rosette (descending order), which emphasizes the integrity of the peripheral mucosal system and its strong connections with the major lymphoid organs. T cells-specific genes dominate among the genes co-expressed in these tissues. The transcription signature of CD8+ (86 genes, r > 0.9) includes a master gene of immune tolerance foxp3 and other negative regulators. Different segments of the intestine were compared in a separate experiment, in which expression gradients along the intestine were found across several functional groups of genes. The expression of luminal and intracellular (lysosome) proteases is markedly higher in pyloric caeca and distal intestine respectively. Steroid metabolism and cytochromes P450 are highly expressed in pyloric caeca and mid intestine while the distal intestine harbors genes related to vitamin and iron metabolism. The expression of genes for antigen presenting proteins and immunoglobulins shows a gradual increase towards the distal intestine.
Collapse
Affiliation(s)
- Trond M Kortner
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Ås, Norway.
| | - Sergey Afanasyev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia
| | - Erling Olaf Koppang
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Ås, Norway
| | - Håvard Bjørgen
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Ås, Norway
| | - Åshild Krogdahl
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Ås, Norway
| | | |
Collapse
|
46
|
Zhu Y, Jiang Q, Lei C, Yu Q, Qiu L. The response of CD27 +CD38 + plasmablasts, CD24 hiCD38 hi transitional B cells, CXCR5 -ICOS +PD-1 + Tph, Tph2 and Tfh2 subtypes to allergens in children with allergic asthma. BMC Pediatr 2024; 24:154. [PMID: 38424520 PMCID: PMC10902953 DOI: 10.1186/s12887-024-04622-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Allergic asthma is a type I allergic reaction mediated by serum Immunoglobulin E (IgE). B cell-mediated humoral immune response to allergens in the pathophysiology of allergic asthma have not been thoroughly elucidated. Peripheral helper T cells (Tph) and follicular helper T cells (Tfh) promote B cell differentiation and antibody production in inflamed tissues. OBJECTIVE To investigate the roles of B cell subsets, Tph cell subsets and Tfh cell subsets in allergic immune responses. METHODS Circulating B cell subsets, Tph cell subsets and Tfh cell subsets in 33 children with allergic asthma and 17 healthy children were analyzed using multicolor flow cytometry. The level of serum total IgE was also assessed. RESULTS Our study found that CD27+CD38+ plasmablasts and CD24hiCD38hi transitional B cells increased and were correlated with serum total IgE level, CD27- naive B cells and CD24hiCD27+ B cells decreased in children with allergic asthma. CXCR5- Tph, CXCR5-ICOS+ Tph, CXCR5-ICOS+PD-1+ Tph, CXCR5+ICOS+ Tfh and CXCR5+ICOS+PD-1+ Tfh increased in children with allergic asthma. Further analysis showed increased Tph2, Tph17, Tfh2 and Tfh17 subtypes while decreased Tph1 and Tfh1 subtypes in children with allergic asthma. Most interestingly, Tph2 or Tfh2 subtypes had a positive correlation with serum total IgE level. CONCLUSION Overall, these results provide insight into the allergens elicited B, Tph or Tfh cell response and identify heretofore unappreciated CD24hiCD38hi transitional B cells, CD24hiCD27+ B cells, CXCR5- Tph, CXCR5-ICOS+PD-1+ Tph, Tph2 subtypes and Tfh2 subtypes response to allergens.
Collapse
Affiliation(s)
- Yunying Zhu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital),Hangzhou Medical College, Hangzhou, 310014, China
- Schoolcollege of Medical Technology of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Qian Jiang
- Department of Clinical Laboratory, Ningbo Puji Hospital (Ningbo Second Hospital West Hospital), Ningbo, 315099, Zhejiang, China
| | - Chenshuang Lei
- Department of Clinical Laboratory, Wenzhou Central Hospital, Wenzhou, 325099, China
| | - Qinhua Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital),Hangzhou Medical College, Hangzhou, 310014, China
| | - Liannv Qiu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital(Affiliated People's Hospital),Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
47
|
Song Y, Mehl F, Zeichner SL. Vaccine Strategies to Elicit Mucosal Immunity. Vaccines (Basel) 2024; 12:191. [PMID: 38400174 PMCID: PMC10892965 DOI: 10.3390/vaccines12020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccines are essential tools to prevent infection and control transmission of infectious diseases that threaten public health. Most infectious agents enter their hosts across mucosal surfaces, which make up key first lines of host defense against pathogens. Mucosal immune responses play critical roles in host immune defense to provide durable and better recall responses. Substantial attention has been focused on developing effective mucosal vaccines to elicit robust localized and systemic immune responses by administration via mucosal routes. Mucosal vaccines that elicit effective immune responses yield protection superior to parenterally delivered vaccines. Beyond their valuable immunogenicity, mucosal vaccines can be less expensive and easier to administer without a need for injection materials and more highly trained personnel. However, developing effective mucosal vaccines faces many challenges, and much effort has been directed at their development. In this article, we review the history of mucosal vaccine development and present an overview of mucosal compartment biology and the roles that mucosal immunity plays in defending against infection, knowledge that has helped inform mucosal vaccine development. We explore new progress in mucosal vaccine design and optimization and novel approaches created to improve the efficacy and safety of mucosal vaccines.
Collapse
Affiliation(s)
- Yufeng Song
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Frances Mehl
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Steven L. Zeichner
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
48
|
Heryanto YD, Zhang YZ, Imoto S. Predicting cell types with supervised contrastive learning on cells and their types. Sci Rep 2024; 14:430. [PMID: 38172501 PMCID: PMC10764802 DOI: 10.1038/s41598-023-50185-2] [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: 09/09/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) is a powerful technique that provides high-resolution expression profiling of individual cells. It significantly advances our understanding of cellular diversity and function. Despite its potential, the analysis of scRNA-seq data poses considerable challenges related to multicollinearity, data imbalance, and batch effect. One of the pivotal tasks in single-cell data analysis is cell type annotation, which classifies cells into discrete types based on their gene expression profiles. In this work, we propose a novel modeling formalism for cell type annotation with a supervised contrastive learning method, named SCLSC (Supervised Contrastive Learning for Single Cell). Different from the previous usage of contrastive learning in single cell data analysis, we employed the contrastive learning for instance-type pairs instead of instance-instance pairs. More specifically, in the cell type annotation task, the contrastive learning is applied to learn cell and cell type representation that render cells of the same type to be clustered in the new embedding space. Through this approach, the knowledge derived from annotated cells is transferred to the feature representation for scRNA-seq data. The whole training process becomes more efficient when conducting contrastive learning for cell and their types. Our experiment results demonstrate that the proposed SCLSC method consistently achieves superior accuracy in predicting cell types compared to five state-of-the-art methods. SCLSC also performs well in identifying cell types in different batch groups. The simplicity of our method allows for scalability, making it suitable for analyzing datasets with a large number of cells. In a real-world application of SCLSC to monitor the dynamics of immune cell subpopulations over time, SCLSC demonstrates a capability to discriminate cell subtypes of CD19+ B cells that were not present in the training dataset.
Collapse
Affiliation(s)
- Yusri Dwi Heryanto
- The Institute of Medical science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yao-Zhong Zhang
- The Institute of Medical science, The University of Tokyo, Tokyo, 108-8639, Japan.
| | - Seiya Imoto
- The Institute of Medical science, The University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
49
|
Jucá PM, de Almeida Duque É, Covre LHH, Mariano KAA, Munhoz CD. Microglia and Systemic Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:287-302. [PMID: 39207698 DOI: 10.1007/978-3-031-55529-9_16] [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: 09/04/2024]
Abstract
Microglia are specialized immune cells that reside in the central nervous system (CNS) and play a crucial role in maintaining the homeostasis of the brain microenvironment. While traditionally regarded as a part of the innate immune system, recent research has highlighted their role in adaptive immunity. The CNS is no longer considered an immune-privileged organ, and increasing evidence suggests bidirectional communication between the immune system and the CNS. Microglia are sensitive to systemic immune signals and can respond to systemic inflammation by producing various inflammatory cytokines and chemokines. This response is mediated by activating pattern recognition receptors (PRRs), which recognize pathogen- and danger-associated molecular patterns in the systemic circulation. The microglial response to systemic inflammation has been implicated in several neurological conditions, including depression, anxiety, and cognitive impairment. Understanding the complex interplay between microglia and systemic immunity is crucial for developing therapeutic interventions to modulate immune responses in the CNS.
Collapse
Affiliation(s)
- Paloma Marinho Jucá
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | - Érica de Almeida Duque
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | - Luiza Helena Halas Covre
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | | | - Carolina Demarchi Munhoz
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil.
| |
Collapse
|
50
|
Borlongan MC, Saha D, Wang H. Tumor Microenvironment: A Niche for Cancer Stem Cell Immunotherapy. Stem Cell Rev Rep 2024; 20:3-24. [PMID: 37861969 DOI: 10.1007/s12015-023-10639-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Tumorigenic Cancer Stem Cells (CSCs), often called tumor-initiating cells (TICs), represent a unique subset of cells within the tumor milieu. They stand apart from the bulk of tumor cells due to their exceptional self-renewal, metastatic, and differentiation capabilities. Despite significant progress in classifying CSCs, these cells remain notably resilient to conventional radiotherapy and chemotherapy, contributing to cancer recurrence. In this review, our objective is to explore novel avenues of research that delve into the distinctive characteristics of CSCs within their surrounding tumor microenvironment (TME). We will start with an overview of the defining features of CSCs and then delve into their intricate interactions with cells from the lymphoid lineage, namely T cells, B cells, and natural killer (NK) cells. Furthermore, we will discuss their dynamic interplay with myeloid lineage cells, including macrophages, neutrophils, and myeloid-derived suppressor cells (MDSCs). Moreover, we will illuminate the crosstalk between CSCs and cells of mesenchymal origin, specifically fibroblasts, adipocytes, and endothelial cells. Subsequently, we will underscore the pivotal role of CSCs within the context of the tumor-associated extracellular matrix (ECM). Finally, we will highlight pre-clinical and clinical studies that target CSCs within the intricate landscape of the TME, including CAR-T therapy, oncolytic viruses, and CSC-vaccines, with the ultimate goal of uncovering novel avenues for CSC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Mia C Borlongan
- College of Medicine, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA
| | - Dipongkor Saha
- Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
| | - Hongbin Wang
- College of Medicine, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
- Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
- Master Program of Pharmaceutical Sciences College of Graduate Studies, Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, Department of Basic Science College of Medicine, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
| |
Collapse
|