51
|
Zhang R, Wang M, Cheng A, Yang Q, Wu Y, Jia R, Chen S, Zhu D, Liu M, Zhao X, Zhang S, Huang J, Ou X, Mao S, Gao Q, Yu Y, Zhang L, Liu Y, Tian B, Pan L. Molecular cloning of duck CD40 and its immune function research. Poult Sci 2021; 100:101100. [PMID: 33975048 PMCID: PMC8122164 DOI: 10.1016/j.psj.2021.101100] [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: 01/13/2021] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 11/20/2022] Open
Abstract
Cosignal molecules are cell surface molecules that transduce signals to other cells to modulate immune response positively (costimulate) or negatively (cosuppress). Costimulatory signals are key factors in determining whether T/B cells are capable of responding to specific antigens and ultimately mediating an appropriate immune response. In this study, the cDNA sequence containing the complete coding frame of the costimulatory molecule duck CD40 gene was cloned and reported for the first time, and its mediated antiviral innate immune was verified in vitro. Results suggested duck CD40 molecule plays an important role in the innate immune responsiveness against some viruses. These data will be beneficial for the further understand of the avian immune system.
Collapse
Affiliation(s)
- Rujuan Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China.
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Dekang Zhu
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Xinxin Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Shaqiu Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Juan Huang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Xumin Ou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Sai Mao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Qun Gao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Yanling Yu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Ling Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Yunya Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Bin Tian
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| | - Leichang Pan
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang 611130, People's Republic of China
| |
Collapse
|
52
|
Holz LE, Chua YC, de Menezes MN, Anderson RJ, Draper SL, Compton BJ, Chan STS, Mathew J, Li J, Kedzierski L, Wang Z, Beattie L, Enders MH, Ghilas S, May R, Steiner TM, Lange J, Fernandez-Ruiz D, Valencia-Hernandez AM, Osmond TL, Farrand KJ, Seneviratna R, Almeida CF, Tullett KM, Bertolino P, Bowen DG, Cozijnsen A, Mollard V, McFadden GI, Caminschi I, Lahoud MH, Kedzierska K, Turner SJ, Godfrey DI, Hermans IF, Painter GF, Heath WR. Glycolipid-peptide vaccination induces liver-resident memory CD8 + T cells that protect against rodent malaria. Sci Immunol 2021; 5:5/48/eaaz8035. [PMID: 32591409 DOI: 10.1126/sciimmunol.aaz8035] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 05/22/2020] [Indexed: 12/29/2022]
Abstract
Liver resident-memory CD8+ T cells (TRM cells) can kill liver-stage Plasmodium-infected cells and prevent malaria, but simple vaccines for generating this important immune population are lacking. Here, we report the development of a fully synthetic self-adjuvanting glycolipid-peptide conjugate vaccine designed to efficiently induce liver TRM cells. Upon cleavage in vivo, the glycolipid-peptide conjugate vaccine releases an MHC I-restricted peptide epitope (to stimulate Plasmodium-specific CD8+ T cells) and an adjuvant component, the NKT cell agonist α-galactosylceramide (α-GalCer). A single dose of this vaccine in mice induced substantial numbers of intrahepatic malaria-specific CD8+ T cells expressing canonical markers of liver TRM cells (CD69, CXCR6, and CD101), and these cells could be further increased in number upon vaccine boosting. We show that modifications to the peptide, such as addition of proteasomal-cleavage sequences or epitope-flanking sequences, or the use of alternative conjugation methods to link the peptide to the glycolipid improved liver TRM cell generation and led to the development of a vaccine able to induce sterile protection in C57BL/6 mice against Plasmodium berghei sporozoite challenge after a single dose. Furthermore, this vaccine induced endogenous liver TRM cells that were long-lived (half-life of ~425 days) and were able to maintain >90% sterile protection to day 200. Our findings describe an ideal synthetic vaccine platform for generating large numbers of liver TRM cells for effective control of liver-stage malaria and, potentially, a variety of other hepatotropic infections.
Collapse
Affiliation(s)
- Lauren E Holz
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Yu Cheng Chua
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Maria N de Menezes
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Regan J Anderson
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Sarah L Draper
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Susanna T S Chan
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Juby Mathew
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Jasmine Li
- Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Zhongfang Wang
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Matthias H Enders
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia.,LIMES Institute, University of Bonn, Bonn, Germany
| | - Sonia Ghilas
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Rose May
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Thiago M Steiner
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Joshua Lange
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Ana Maria Valencia-Hernandez
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Taryn L Osmond
- Malaghan Institute of Medical Research, Wellington, New Zealand.,Avalia Immunotherapies Limited, Lower Hutt, New Zealand
| | | | - Rebecca Seneviratna
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Catarina F Almeida
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Kirsteen M Tullett
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Patrick Bertolino
- Centenary Institute, The University of Sydney and AW Morrow Gastroenterology and Liver Centre, Liver Immunology Program, Newtown, NSW, Australia
| | - David G Bowen
- Centenary Institute, The University of Sydney and AW Morrow Gastroenterology and Liver Centre, Liver Immunology Program, Newtown, NSW, Australia
| | - Anton Cozijnsen
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Vanessa Mollard
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | | | - Irina Caminschi
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Mireille H Lahoud
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Turner
- Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand. .,Avalia Immunotherapies Limited, Lower Hutt, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Wellington, New Zealand
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand. .,Avalia Immunotherapies Limited, Lower Hutt, New Zealand
| | - William R Heath
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
53
|
Rogers KJ, Shtanko O, Stunz LL, Mallinger LN, Arkee T, Schmidt ME, Bohan D, Brunton B, White JM, Varga SM, Butler NS, Bishop GA, Maury W. Frontline Science: CD40 signaling restricts RNA virus replication in Mϕs, leading to rapid innate immune control of acute virus infection. J Leukoc Biol 2021; 109:309-325. [PMID: 32441445 PMCID: PMC7774454 DOI: 10.1002/jlb.4hi0420-285rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 01/03/2023] Open
Abstract
Many acute viral infections target tissue Mϕs, yet the mechanisms of Mϕ-mediated control of viruses are poorly understood. Here, we report that CD40 expressed by peritoneal Mϕs restricts early infection of a broad range of RNA viruses. Loss of CD40 expression enhanced virus replication as early as 12-24 h of infection and, conversely, stimulation of CD40 signaling with an agonistic Ab blocked infection. With peritoneal cell populations infected with the filovirus, wild-type (WT) Ebola virus (EBOV), or a BSL2 model virus, recombinant vesicular stomatitis virus encoding Ebola virus glycoprotein (rVSV/EBOV GP), we examined the mechanism conferring protection. Here, we demonstrate that restricted virus replication in Mϕs required CD154/CD40 interactions that stimulated IL-12 production through TRAF6-dependent signaling. In turn, IL-12 production resulted in IFN-γ production, which induced proinflammatory polarization of Mϕs, protecting the cells from infection. These CD40-dependent events protected mice against virus challenge. CD40-/- mice were exquisitely sensitive to intraperitoneal challenge with a dose of rVSV/EBOV GP that was sublethal to CD40+/+ mice, exhibiting viremia within 12 h of infection and rapidly succumbing to infection. This study identifies a previously unappreciated role for Mϕ-intrinsic CD40 signaling in controlling acute virus infection.
Collapse
Affiliation(s)
- Kai J. Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Laura L. Stunz
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Laura N. Mallinger
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Tina Arkee
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Megan E. Schmidt
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Dana Bohan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Bethany Brunton
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Judith M. White
- Department of Cell Biology, University of Virginia, Charlottesville, VA, United States
| | - Steve M. Varga
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Noah S. Butler
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| | - Gail A. Bishop
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
- Iowa City VA Health Care System, Iowa City, Iowa City, IA, United States
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
54
|
Functional Role of B Cells in Atherosclerosis. Cells 2021; 10:cells10020270. [PMID: 33572939 PMCID: PMC7911276 DOI: 10.3390/cells10020270] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of blood vessels, and both innate and adaptive immune responses are involved in its development. The impact of B cells on atherosclerosis has been demonstrated in numerous studies and B cells have been found in close proximity to atherosclerotic plaques in humans and mice. B cells exert both atheroprotective and pro-atherogenic functions, which have been associated with their B cell subset attribution. While B1 cells and marginal zone B cells are considered to protect against atherosclerosis, follicular B cells and innate response activator B cells have been shown to promote atherosclerosis. In this review, we shed light on the role of B cells from a different, functional perspective and focus on the three major B cell functions: antibody production, antigen presentation/T cell interaction, and the release of cytokines. All of these functions have the potential to affect atherosclerosis by multiple ways and are dependent on the cellular milieu and the activation status of the B cell. Moreover, we discuss B cell receptor signaling and the mechanism of B cell activation under atherosclerosis-prone conditions. By summarizing current knowledge of B cells in and beyond atherosclerosis, we are pointing out open questions and enabling new perspectives.
Collapse
|
55
|
An Overview of Advances in Cell-Based Cancer Immunotherapies Based on the Multiple Immune-Cancer Cell Interactions. Methods Mol Biol 2021; 2097:139-171. [PMID: 31776925 DOI: 10.1007/978-1-0716-0203-4_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumors have a complex ecosystem in which behavior and fate are determined by the interaction of diverse cancerous and noncancerous cells at local and systemic levels. A number of studies indicate that various immune cells participate in tumor development (Fig. 1). In this review, we will discuss interactions among T lymphocytes (T cells), B cells, natural killer (NK) cells, dendritic cells (DCs), tumor-associated macrophages (TAMs), neutrophils, and myeloid-derived suppressor cells (MDSCs). In addition, we will touch upon attempts to either use or block subsets of immune cells to target cancer.
Collapse
|
56
|
Chen H, Song Y, Deng C, Xu Y, Xu H, Zhu X, Song G, Tang Q, Lu J, Wang J. Comprehensive analysis of immune infiltration and gene expression for predicting survival in patients with sarcomas. Aging (Albany NY) 2020; 13:2168-2183. [PMID: 33316779 PMCID: PMC7880383 DOI: 10.18632/aging.202229] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/31/2020] [Indexed: 12/24/2022]
Abstract
Tumor microenvironments are strongly related to tumor development, and immune-infiltrating cells and immune-related molecules are potential prognostic markers. However, the shortcomings of traditional measurement methods limit the accurate evaluation of various components in tumor microenvironments. With the rapid advancement of Next-Generation RNA Sequencing technology, dedicated and in-depth analyses of immune filtration within the tumor microenvironment has been achieved. In this study, we combined the bioinformatics analysis methods ESTIMATE, CIBERSORT, and ssGSEA to characterize the immune infiltration of sarcomas and to identify specific immunomodulators of different pathological subtypes. We further extracted a functional enrichment of significant immune-related genes related to improved prognosis, including NR1H3, VAMP5, GIMAP2, GBP2, HLA-E and CRIP1. Overall, the immune microenvironment is an important prognostic determinant of sarcomas and may be a potential resource for developing effective immunotherapy.
Collapse
Affiliation(s)
- Hongmin Chen
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Yijiang Song
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Chuangzhong Deng
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Yanyang Xu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Huaiyuan Xu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Xiaojun Zhu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Guohui Song
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Jin Wang
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| |
Collapse
|
57
|
Skin-Associated B Cells in the Pathogenesis of Cutaneous Autoimmune Diseases-Implications for Therapeutic Approaches. Cells 2020; 9:cells9122627. [PMID: 33297481 PMCID: PMC7762338 DOI: 10.3390/cells9122627] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
B lymphocytes are crucial mediators of systemic immune responses and are known to be substantial in the pathogenesis of autoimmune diseases with cutaneous manifestations. Amongst them are lupus erythematosus, dermatomyositis, systemic sclerosis and psoriasis, and particularly those driven by autoantibodies such as pemphigus and pemphigoid. However, the concept of autoreactive skin-associated B cells, which may reside in the skin and locally contribute to chronic inflammation, is gradually evolving. These cells are believed to differ from B cells of primary and secondary lymphoid organs and may provide additional features besides autoantibody production, including cytokine expression and crosstalk to autoreactive T cells in an antigen-presenting manner. In chronically inflamed skin, B cells may appear in tertiary lymphoid structures. Those abnormal lymph node-like structures comprise a network of immune and stromal cells possibly enriched by vascular structures and thus constitute an ideal niche for local autoimmune responses. In this review, we describe current considerations of different B cell subsets and their assumed role in skin autoimmunity. Moreover, we discuss traditional and B cell-associated approaches for the treatment of autoimmune skin diseases, including drugs targeting B cells (e.g., CD19- and CD20-antibodies), plasma cells (e.g., proteasome inhibitors, CXCR4 antagonists), activated pathways (such as BTK- and PI3K-inhibitors) and associated activator molecules (BLyS, APRIL).
Collapse
|
58
|
Daub S, Lutgens E, Münzel T, Daiber A. CD40/CD40L and Related Signaling Pathways in Cardiovascular Health and Disease-The Pros and Cons for Cardioprotection. Int J Mol Sci 2020; 21:E8533. [PMID: 33198327 PMCID: PMC7697597 DOI: 10.3390/ijms21228533] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023] Open
Abstract
The CD40-CD40 ligand (CD40L) dyad represents a scientific and clinical field that has raised many controversies in the past and cannot be clearly defined as being an either beneficial or harmful pathway. Being crucially involved in physiological immunological processes as well as pathological inflammatory reactions, the signaling pathway has been recognized as a key player in the development of both autoimmune and cardiovascular disease. Even though the possibilities of a therapeutic approach to the dyad were recognized decades ago, due to unfortunate events, detailed in this review, pharmacological treatment targeting the dyad, especially in patients suffering from atherosclerosis, is not available. Despite the recent advances in the treatment of classical cardiovascular risk factors, such as arterial hypertension and diabetes mellitus, the treatment of the associated low-grade inflammation that accounts for the progression of atherosclerosis is still challenging. Low-grade inflammation can be detected in a significant portion of patients that suffer from cardiovascular disease and it is therefore imperative to develop new therapeutic strategies in order to combat this driver of atherosclerosis. Of note, established cardiovascular drugs such as angiotensin-converting enzyme inhibitors or statins have proven beneficial cardiovascular effects that are also related to their pleiotropic immunomodulatory properties. In this review, we will discuss the setbacks encountered as well as new avenues discovered on the path to a different, inflammation-centered approach for the treatment of cardiovascular disease with the CD40-CD40L axis as a central therapeutic target.
Collapse
Affiliation(s)
- Steffen Daub
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (S.D.); (T.M.)
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands;
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, 80336 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany and Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (S.D.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, 55131 Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (S.D.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, 55131 Mainz, Germany
| |
Collapse
|
59
|
van der Poel CE, Bajic G, Macaulay CW, van den Broek T, Ellson CD, Bouma G, Victora GD, Degn SE, Carroll MC. Follicular Dendritic Cells Modulate Germinal Center B Cell Diversity through FcγRIIB. Cell Rep 2020; 29:2745-2755.e4. [PMID: 31775042 PMCID: PMC7015177 DOI: 10.1016/j.celrep.2019.10.086] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/05/2019] [Accepted: 10/22/2019] [Indexed: 01/02/2023] Open
Abstract
Follicular dendritic cells (FDCs), a rare and enigmatic stromal cell type in the B cell follicles of secondary lymphoid organs, store and present antigen to B cells. While essential for germinal center (GC) responses, their exact role during GC B cell selection remains unknown. FDCs upregulate the inhibitory IgG Fc receptor FcγRIIB during GC formation. We show that the stromal deficiency of FcγRIIB does not affect GC B cell frequencies compared to wild-type mice. However, in the absence of FcγRIIB on FDCs, GCs show aberrant B cell selection during autoreactive and selective foreign antigen responses. These GCs are more diverse as measured by the AidCreERT2 -confetti system and show the persistence of IgM+ clones with decreased numbers of IgH mutations. Our results show that FDCs can modulate GC B cell diversity by the upregulation of FcγRIIB. Permissive clonal selection and subsequent increased GC diversity may affect epitope spreading during autoimmunity and foreign responses. van der Poel et al. show that follicular dendritic cells (FDCs) can regulate germinal center diversity through FcγRIIB. In the absence of this receptor, germinal centers appear more diverse. In addition, the loss of FcγRIIB on FDCs leads to the persistence of IgM clones with decreased levels of somatic hypermutation.
Collapse
Affiliation(s)
- Cees E van der Poel
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Goran Bajic
- Laboratory of Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles W Macaulay
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Boston University, Boston, MA 02215, USA
| | - Theo van den Broek
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | - Søren E Degn
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
60
|
Misawa T, SoRelle JA, Choi JH, Yue T, Wang KW, McAlpine W, Wang J, Liu A, Tabeta K, Turer EE, Evers B, Nair-Gill E, Poddar S, Su L, Ou F, Yu L, Russell J, Ludwig S, Zhan X, Hildebrand S, Li X, Tang M, Murray AR, Moresco EMY, Beutler B. Mutual inhibition between Prkd2 and Bcl6 controls T follicular helper cell differentiation. Sci Immunol 2020; 5:5/43/eaaz0085. [PMID: 31980486 DOI: 10.1126/sciimmunol.aaz0085] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022]
Abstract
T follicular helper cells (TFH) participate in germinal center (GC) development and are necessary for B cell production of high-affinity, isotype-switched antibodies. In a forward genetic screen, we identified a missense mutation in Prkd2, encoding the serine/threonine kinase protein kinase D2, which caused elevated titers of immunoglobulin E (IgE) in the serum. Subsequent analysis of serum antibodies in mice with a targeted null mutation of Prkd2 demonstrated polyclonal hypergammaglobulinemia of IgE, IgG1, and IgA isotypes, which was exacerbated by the T cell-dependent humoral response to immunization. GC formation and GC B cells were increased in Prkd2-/- spleens. These effects were the result of excessive cell-autonomous TFH development caused by unrestricted Bcl6 nuclear translocation in Prkd2-/- CD4+ T cells. Prkd2 directly binds to Bcl6, and Prkd2-dependent phosphorylation of Bcl6 is necessary to constrain Bcl6 to the cytoplasm, thereby limiting TFH development. In response to immunization, Bcl6 repressed Prkd2 expression in CD4+ T cells, thereby committing them to TFH development. Thus, Prkd2 and Bcl6 form a mutually inhibitory positive feedback loop that controls the stable transition from naïve CD4+ T cells to TFH during the adaptive immune response.
Collapse
Affiliation(s)
- Takuma Misawa
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jeffrey A SoRelle
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jin Huk Choi
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tao Yue
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kuan-Wen Wang
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - William McAlpine
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jianhui Wang
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aijie Liu
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Koichi Tabeta
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| | - Emre E Turer
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bret Evers
- Division of Neuropathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Evan Nair-Gill
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Subhajit Poddar
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lijing Su
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Feiya Ou
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Liyang Yu
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jamie Russell
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sara Ludwig
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoming Zhan
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sara Hildebrand
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaohong Li
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Miao Tang
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anne R Murray
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eva Marie Y Moresco
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
61
|
Abstract
Therapeutic targeting of immune checkpoints has garnered significant attention in the area of cancer immunotherapy, in which efforts have focused in particular on cytotoxic T lymphocyte antigen 4 (CTLA4) and PD1, both of which are members of the CD28 family. In autoimmunity, these same pathways can be targeted to opposite effect: to curb the over-exuberant immune response. The CTLA4 checkpoint serves as an exemplar, whereby CTLA4 activity is blocked by antibodies in cancer immunotherapy and augmented by the provision of soluble CTLA4 in autoimmunity. Here, we review the targeting of co-stimulatory molecules in autoimmune diseases, focusing in particular on agents directed at members of the CD28 or tumour necrosis factor receptor families. We present the state of the art in co-stimulatory blockade approaches, including rational combinations of immune inhibitory agents, and discuss the future opportunities and challenges in this field.
Collapse
|
62
|
Serrat N, Guerrero-Hernández M, Matas-Céspedes A, Yahiaoui A, Valero JG, Nadeu F, Clot G, Di Re M, Corbera-Bellalta M, Magnano L, Rivas-Delgado A, Enjuanes A, Beà S, Cid MC, Campo E, Montero J, Hodson DJ, López-Guillermo A, Colomer D, Tannheimer S, Pérez-Galán P. PI3Kδ inhibition reshapes follicular lymphoma-immune microenvironment cross talk and unleashes the activity of venetoclax. Blood Adv 2020; 4:4217-4231. [PMID: 32898249 PMCID: PMC7479943 DOI: 10.1182/bloodadvances.2020001584] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 07/23/2020] [Indexed: 12/17/2022] Open
Abstract
Despite idelalisib approval in relapsed follicular lymphoma (FL), a complete characterization of the immunomodulatory consequences of phosphatidylinositol 3-kinase δ (PI3Kδ) inhibition, biomarkers of response, and potential combinatorial therapies in FL remain to be established. Using ex vivo cocultures of FL patient biopsies and follicular dendritic cells (FDCs) to mimic the germinal center (n = 42), we uncovered that PI3Kδ inhibition interferes with FDC-induced genes related to angiogenesis, extracellular matrix formation, and transendothelial migration in a subset of FL samples, defining an 18-gene signature fingerprint of idelalisib sensitivity. A common hallmark of idelalisib found in all FL cases was its interference with the CD40/CD40L pathway and induced proliferation, together with the downregulation of proteins crucial for B-T-cell synapses, leading to an inefficient cross talk between FL cells and the supportive T-follicular helper cells (TFH). Moreover, idelalisib downmodulates the chemokine CCL22, hampering the recruitment of TFH and immunosupressive T-regulatory cells to the FL niche, leading to a less supportive and tolerogenic immune microenvironment. Finally, using BH3 profiling, we uncovered that FL-FDC and FL-macrophage cocultures augment tumor addiction to BCL-XL and MCL-1 or BFL-1, respectively, limiting the cytotoxic activity of the BCL-2 inhibitor venetoclax. Idelalisib restored FL dependence on BCL-2 and venetoclax activity. In summary, idelalisib exhibits a patient-dependent activity toward angiogenesis and lymphoma dissemination. In all FL cases, idelalisib exerts a general reshaping of the FL immune microenvironment and restores dependence on BCL-2, predisposing FL to cell death, providing a mechanistic rationale for investigating the combination of PI3Kδ inhibitors and venetoclax in clinical trials.
Collapse
Affiliation(s)
- Neus Serrat
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Martina Guerrero-Hernández
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Alba Matas-Céspedes
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Anella Yahiaoui
- Department of Biomarker Sciences, Gilead Sciences, Inc., Seattle, WA
| | - Juan G Valero
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Ferran Nadeu
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Guillem Clot
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Miriam Di Re
- Department of Haematology, Wellcome Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Marc Corbera-Bellalta
- Vasculitis Research Unit, Department of Autoimmune Diseases, Clinical Institute of Medicine and Dermatology, Hospital Clinic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS-CRB CELLEX), Barcelona, Spain
| | - Laura Magnano
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Department of Hematology and
| | - Alfredo Rivas-Delgado
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Department of Hematology and
| | - Anna Enjuanes
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Silvia Beà
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Maria C Cid
- Vasculitis Research Unit, Department of Autoimmune Diseases, Clinical Institute of Medicine and Dermatology, Hospital Clinic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS-CRB CELLEX), Barcelona, Spain
| | - Elías Campo
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic-IDIBAPS, Barcelona, Spain; and
| | - Joan Montero
- Department of Nanobioengineering, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Daniel J Hodson
- Department of Haematology, Wellcome Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Armando López-Guillermo
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Department of Hematology and
| | - Dolors Colomer
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic-IDIBAPS, Barcelona, Spain; and
| | - Stacey Tannheimer
- Department of Biomarker Sciences, Gilead Sciences, Inc., Seattle, WA
| | - Patricia Pérez-Galán
- Department of Hematology-Oncology, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| |
Collapse
|
63
|
Fibroblasts as a source of self-antigens for central immune tolerance. Nat Immunol 2020; 21:1172-1180. [PMID: 32839611 DOI: 10.1038/s41590-020-0756-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
Fibroblasts are one of the most common but also neglected types of stromal cells, the heterogeneity of which underlies the specific function of tissue microenvironments in development and regeneration. In the thymus, autoreactive T cells are thought to be negatively selected by reference to the self-antigens expressed in medullary epithelial cells, but the contribution of other stromal cells to tolerance induction has been poorly examined. In the present study, we report a PDGFR+ gp38+ DPP4- thymic fibroblast subset that is required for T cell tolerance induction. The deletion of the lymphotoxin β-receptor in thymic fibroblasts caused an autoimmune phenotype with decreased expression of tissue-restricted and fibroblast-specific antigens, offering insight into the long-sought target of lymphotoxin signaling in the context of the regulation of autoimmunity. Thus, thymic medullary fibroblasts play an essential role in the establishment of central tolerance by producing a diverse array of self-antigens.
Collapse
|
64
|
Asmiyou A, Bakr AM, Shahin DA, Wahba Y. CD40 and CD72 expression and prognostic values among children with systemic lupus erythematosus: a case-control study. Lupus 2020; 29:1270-1276. [PMID: 32700598 DOI: 10.1177/0961203320941931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic disease with proven interactions between immune system components, including both humoral- and cell-mediated immunity, as well as co-stimulatory and inhibitory molecules such as CD40 and CD72. Here, we investigated CD40 and CD72 expression on B cells of SLE children and assessed their prognostic values. We conducted a preliminary case-control study in Mansoura University Children's Hospital, Egypt from September 2018 to January 2020 including 27 SLE children and 27 healthy controls. We assessed cases during initial flare and after remission. Flow cytometry analysis was carried out for all participants for CD40 and CD72 expression of B cells. During flare, SLE cases had statistically significant higher CD40 and lower CD72 expression in comparison with controls (p < 0.001). After remission, the number of CD40+ B cells significantly decreased (p < 0.001), while the number of CD72+ B cells significantly increased (p < 0.001) in comparison with flare. We reported non-significant positive correlations between CD40 expression and SLE Disease Activity Index (SLEDAI; p = 0.347 during flare and p = 0.653 after remission) and negative correlations between CD72 expression and SLEDAI (p = 0.34 during flare and p = 0.044 after remission). No significant differences were detected between renal histopathology classes with regard to CDs expression on B cells (p = 0.45 for CD40 and p = 0.63 for CD72). In conclusion, CD40+ B cells and CD72+ B cells could be considered as markers of paediatric SLE flare and remission, respectively.
Collapse
Affiliation(s)
- Abtisam Asmiyou
- Paediatric Department, Faculty of Medicine, Mansoura University, Egypt
| | - Ashraf M Bakr
- Paediatric Department, Faculty of Medicine, Mansoura University, Egypt
| | - Doaa A Shahin
- Clinical Pathology (Haematology) Department, Faculty of Medicine, Mansoura University, Egypt
| | - Yahya Wahba
- Paediatric Department, Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
65
|
Lee MJ, Jo H, Park SH, Ko MK, Kim SM, Kim B, Park JH. Advanced Foot-And-Mouth Disease Vaccine Platform for Stimulation of Simultaneous Cellular and Humoral Immune Responses. Vaccines (Basel) 2020; 8:E254. [PMID: 32481687 PMCID: PMC7349985 DOI: 10.3390/vaccines8020254] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/16/2020] [Accepted: 05/25/2020] [Indexed: 12/16/2022] Open
Abstract
Currently available commercial foot-and-mouth disease (FMD) vaccines have various limitations, such as the slow induction and short-term maintenance of antibody titers. Therefore, a novel FMD vaccine that can rapidly induce high neutralizing antibody titers to protect the host in early stages of an FMD virus infection, maintain high antibody titers for long periods after one vaccination dose, and confer full protection against clinical symptoms by simultaneously stimulating cellular and humoral immunity is needed. Here, we developed immunopotent FMD vaccine strains A-3A and A-HSP70, which elicit strong initial cellular immune response and induce humoral immune response, including long-lasting memory response. We purified the antigen (inactivated virus) derived from these immunopotent vaccine strains, and evaluated the immunogenicity and efficacy of the vaccines containing these antigens in mice and pigs. The immunopotent vaccine strains A-3A and A-HSP70 demonstrated superior immunogenicity compared with the A strain (backbone strain) in mice. The oil emulsion-free vaccine containing A-3A and A-HSP70 antigens effectively induced early, mid-term, and long-term immunity in mice and pigs by eliciting robust cellular and humoral immune responses through the activation of co-stimulatory molecules and the secretion of proinflammatory cytokines. We successfully derived an innovative FMD vaccine formulation to create more effective FMD vaccines.
Collapse
Affiliation(s)
- Min Ja Lee
- Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Korea; (H.J.); (S.H.P.); (M.-K.K.); (S.-M.K.); (B.K.)
| | | | | | | | | | | | - Jong-Hyeon Park
- Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Korea; (H.J.); (S.H.P.); (M.-K.K.); (S.-M.K.); (B.K.)
| |
Collapse
|
66
|
Karnell JL, Albulescu M, Drabic S, Wang L, Moate R, Baca M, Oganesyan V, Gunsior M, Thisted T, Yan L, Li J, Xiong X, Eck SC, de Los Reyes M, Yusuf I, Streicher K, Müller-Ladner U, Howe D, Ettinger R, Herbst R, Drappa J. A CD40L-targeting protein reduces autoantibodies and improves disease activity in patients with autoimmunity. Sci Transl Med 2020; 11:11/489/eaar6584. [PMID: 31019027 DOI: 10.1126/scitranslmed.aar6584] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 11/06/2018] [Accepted: 03/25/2019] [Indexed: 12/23/2022]
Abstract
The CD40/CD40L axis plays a central role in the generation of humoral immune responses and is an attractive target for treating autoimmune diseases in the clinic. Here, we report the generation and clinical results of a CD40L binding protein, VIB4920, which lacks an Fc domain, therefore avoiding platelet-related safety issues observed with earlier monoclonal antibody therapeutics that targeted CD40L. VIB4920 blocked downstream CD40 signaling events, resulting in inhibition of human B cell activation and plasma cell differentiation, and did not induce platelet aggregation in preclinical studies. In a phase 1 study in healthy volunteers, VIB4920 suppressed antigen-specific IgG in a dose-dependent fashion after priming and boosting with the T-dependent antigen, KLH. Furthermore, VIB4920 significantly reduced circulating Ki67+ dividing B cells, class-switched memory B cells, and a plasma cell gene signature after immunization. In a phase 1b proof-of-concept study in patients with rheumatoid arthritis, VIB4920 significantly decreased disease activity, achieving low disease activity or clinical remission in more than 50% of patients in the two higher-dose groups. Dose-dependent decreases in rheumatoid factor autoantibodies and Vectra DA biomarker score provide additional evidence that VIB4920 effectively blocked the CD40/CD40L pathway. VIB4920 demonstrated a good overall safety profile in both clinical studies. Together, these data demonstrate the potential of VIB4920 to significantly affect autoimmune disease and humoral immune activation and to support further evaluation of this molecule in inflammatory conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Li Yan
- Viela Bio, Gaithersburg, MD 20878, USA
| | - Jing Li
- MedImmune, San Francisco, CA 94080, USA
| | | | | | | | | | | | - Ulf Müller-Ladner
- Department of Rheumatology and Clinical Immunology, Campus Kerchoff, Justus-Liebig University Giessen, 61231 Bad Nauheim, Germany
| | - David Howe
- MedImmune, Granta Park, Cambridge CB21 6GH, UK
| | | | | | | |
Collapse
|
67
|
Cooper L, Good-Jacobson KL. Dysregulation of humoral immunity in chronic infection. Immunol Cell Biol 2020; 98:456-466. [PMID: 32275789 DOI: 10.1111/imcb.12338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Chronic viral infections disrupt the ability of the humoral immune response to produce neutralizing antibody or form effective immune memory, preventing viral clearance and making vaccine design difficult. Multiple components of the B-cell response are affected by pathogens that are not cleared from the host. Changes in the microenvironment shift production of B cells to short-lived plasma cells early in the response. Polyclonal B cells are recruited into both the plasma cell and germinal center compartments, inhibiting the formation of a targeted, high-affinity response. Finally, memory B cells shift toward an "atypical" phenotype, which may in turn result in changes to the functional properties of this population. While similar properties of B-cell dysregulation have been described across different types of persistent infections, key questions about the underlying mechanisms remain. This review will discuss the recent advances in this field, as well as highlight the critical questions about the interplay between viral load, microenvironment, the polyclonal response and atypical memory B cells that are yet to be answered. Design of new preventative treatments will rely on identifying the extrinsic and intrinsic modulators that push B cells toward an ineffective response, and thus identify new ways to guide them back onto the best path for clearance of virus and formation of effective immune memory.
Collapse
Affiliation(s)
- Lucy Cooper
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Kim L Good-Jacobson
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
68
|
Pucino V, Gardner DH, Fisher BA. Rationale for CD40 pathway blockade in autoimmune rheumatic disorders. THE LANCET. RHEUMATOLOGY 2020; 2:e292-e301. [PMID: 38273474 DOI: 10.1016/s2665-9913(20)30038-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/15/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
Abstract
CD40 and its ligand CD40L (CD154) belong to the tumor necrosis factor receptor superfamily and are expressed by a variety of immune and non-immune cells. CD40L plays a central role in co-stimulation and regulation of the immune response via activation of cells expressing CD40. Imbalance of the CD40-CD40L co-stimulatory pathway has been reported in many autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, and Sjögren's syndrome, thus supporting its role in the breach of immune tolerance that is typical of these diseases. Targeting CD40-CD40L signalling might represent a novel therapeutic option for several autoimmune disorders.
Collapse
Affiliation(s)
- Valentina Pucino
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK; National Institute for Health Research, Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - David H Gardner
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK; National Institute for Health Research, Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| |
Collapse
|
69
|
Cui ZW, Zhang XY, Wu CS, Zhang YA, Zhou Y, Zhang XJ. Membrane IgM + plasma cells in grass carp (Ctenopharyngodon idella): Insights into the conserved evolution of IgM + plasma cells in vertebrates. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 106:103613. [PMID: 31935401 DOI: 10.1016/j.dci.2020.103613] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 06/10/2023]
Abstract
Teleost fish are the most primitive bony vertebrates that contain B cells; thus, comparative analysis of teleost naïve/mature B cells and plasma cells can provide helpful evidence for understanding the evolution paradigms of these two B-cell subpopulations in vertebrates. In this study, we developed monoclonal antibody against grass carp IgM and identified two different IgM+ cell subsets: IgM+ lymphocytes (Lym), resembling naïve/mature B cells, and IgM+ myeloid cells (Mye), resembling plasma cells. Like plasma cells in mammals, the size of IgM+ Mye is significantly larger than that of IgM+ Lym, as revealed by flow cytometric analysis and transmission electron microscopy. The IgM+ Mye were further verified as plasma cells because they showed gene expression patterns similar with those of human plasma cells and a great capacity to secrete IgM. Like mammalian IgM+ and IgA+ plasma cells, not IgG+ plasma cells, grass carp IgM+ Mye also expressed membrane immunoglobulins, a feature conserved in IgM+ plasma cells in vertebrates. Furthermore, recombinant CD40L or IL-21 alone could induce the plasma cell generation and IgM secretion, while the combination of CD40L and IL-21 had greater effect on IgM secretion, but not on plasma cell generation. This study fills an important gap in the knowledge of plasma cells in teleost fish and provides critical insights into the conserved evolution of IgM+ plasma cells in vertebrates.
Collapse
Affiliation(s)
- Zheng-Wei Cui
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Fishery Drug Development, Ministry of Agriculture, China, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China; Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430070, China
| | - Xiang-Yang Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430070, China
| | - Chang-Song Wu
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China; Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430070, China
| | - Yang Zhou
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Xu-Jie Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
70
|
Sufficiency of CD40 activation and immune checkpoint blockade for T cell priming and tumor immunity. Proc Natl Acad Sci U S A 2020; 117:8022-8031. [PMID: 32213589 DOI: 10.1073/pnas.1918971117] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Innate immune receptors such as toll-like receptors (TLRs) provide critical molecular links between innate cells and adaptive immune responses. Here, we studied the CD40 pathway as an alternative bridge between dendritic cells (DCs) and adaptive immunity in cancer. Using an experimental design free of chemo- or radiotherapy, we found CD40 activation with agonistic antibodies (⍺CD40) produced complete tumor regressions in a therapy-resistant pancreas cancer model, but only when combined with immune checkpoint blockade (ICB). This effect, unachievable with ICB alone, was independent of TLR, STING, or IFNAR pathways. Mechanistically, αCD40/ICB primed durable T cell responses, and efficacy required DCs and host expression of CD40. Moreover, ICB drove optimal generation of polyfunctional T cells in this "cold" tumor model, instead of rescuing T cell exhaustion. Thus, immunostimulation via αCD40 is sufficient to synergize with ICB for priming. Clinically, combination αCD40/ICB may extend efficacy in patients with "cold" and checkpoint-refractory tumors.
Collapse
|
71
|
Fisher BA, Szanto A, Ng WF, Bombardieri M, Posch MG, Papas AS, Farag AM, Daikeler T, Bannert B, Kyburz D, Kivitz AJ, Carsons SE, Isenberg DA, Barone F, Bowman SJ, Espié P, Floch D, Dupuy C, Ren X, Faerber PM, Wright AM, Hockey HU, Rotte M, Milojevic J, Avrameas A, Valentin MA, Rush JS, Gergely P. Assessment of the anti-CD40 antibody iscalimab in patients with primary Sjögren's syndrome: a multicentre, randomised, double-blind, placebo-controlled, proof-of-concept study. THE LANCET. RHEUMATOLOGY 2020; 2:e142-e152. [PMID: 38263652 DOI: 10.1016/s2665-9913(19)30135-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND Primary Sjögren's syndrome is an autoimmune disease that presents as dryness of the mouth and eyes due to impairment of the exocrine glands. To our knowledge, no systemic therapies for primary Sjögren's syndrome have shown efficacy. CD40-CD154-mediated T cell-B cell interactions in primary Sjögren's syndrome contribute to aberrant lymphocyte activation in inflamed tissue, leading to sialadenitis and other tissue injury. Therefore, we investigated the safety and preliminary efficacy of iscalimab (CFZ533), a novel anti-CD40 monoclonal antibody, in patients with primary Sjögren's syndrome. METHODS This multicentre, randomised, double-blind, placebo-controlled, proof-of-concept study took place at ten investigational sites across Europe (UK, n=4; Germany, Switzerland, and Hungary, n=1 each) and the USA (n=3). Eligible patients were aged 18-75 years and fulfilled the 2002 American European consensus group diagnostic classification criteria for primary Sjögren's syndrome. In the double-blind phase of the trial, patients were randomly assigned (2:1) via computer-generated unique randomisation numbers to receive subcutaneous iscalimab (3 mg/kg) or placebo at weeks 0, 2, 4, and 8 (cohort 1) or intravenous iscalimab (10 mg/kg) or placebo at weeks 0, 2, 4, and 8 (cohort 2). Randomisation was stratified according to baseline intake of oral corticosteroids. At week 12, patients in both cohorts received open-label iscalimab (same dose and route) for 12 weeks. The primary objectives of the study were to assess the safety, tolerability, and efficacy of multiple doses of iscalimab in the two sequential dose cohorts. Safety and tolerability were assessed by adverse events and efficacy of iscalimab versus placebo was assessed by clinical disease activity, as measured by the change in European League Against Rheumatism Sjögren's syndrome disease activity index (ESSDAI) score after 12 weeks of treatment. Analyses were done on a per-protocol basis. The trial was registered with ClinicalTrials.gov, NCT02291029. FINDINGS Between Oct 22, 2014, and June 28, 2016, we assessed 82 patients for eligibility (25 for cohort 1 and 57 for cohort 2). 38 patients were excluded because of ineligibility. In cohort 1, 12 patients were randomly assigned to receive either 3 mg/kg doses of iscalimab (n=8) or placebo (n=4), and in cohort 2, 32 patients were randomly assigned to receive either intravenous 10 mg/kg doses of iscalimab (n=21) or placebo (n=11). Adverse events were similar between iscalimab treatment groups and placebo groups, with adverse events occurring in all patients in cohort 1, and in 52% and 64% of the iscalimab and placebo groups, respectively, in cohort 2. Two serious adverse events were reported (one case of bacterial conjunctivitis in cohort 1 and one case of atrial fibrillation in cohort 2), which were unrelated to treatment with iscalimab. Intravenous treatment with iscalimab resulted in a mean reduction of 5·21 points (95% CI 0·96-9·46; one-sided p=0·0090) in ESSDAI score compared with placebo. There was no signficiant difference in ESSDAI score between subcutaneous iscalimab and placebo. INTERPRETATION To our knowledge, this is the first randomised, placebo-controlled proof-of-concept study of a new investigational drug for primary Sjögren's syndrome that indicates preliminary efficacy. Our data suggest a role of CD40-CD154 interactions in primary Sjögren's syndrome pathology and the therapeutic potential for CD40 blockade in this disease should be investigated further. FUNDING Novartis Pharma.
Collapse
Affiliation(s)
- Benjamin A Fisher
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Antonia Szanto
- Division of Clinical Immunology, Department of Internal Medicine, University of Debrecen, Debrecen, Hungary
| | - Wan-Fai Ng
- NIHR Newcastle Biomedical Research Centre and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK; Clinical Research Facility, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michele Bombardieri
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Athena S Papas
- Division of Oral Medicine, Tufts University School of Dental Medicine, Tufts University, Boston, USA
| | - Arwa M Farag
- Division of Oral Medicine, Tufts University School of Dental Medicine, Tufts University, Boston, USA; Department of Oral Diagnostic Science, Faculty of Dentistry, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Thomas Daikeler
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - Bettina Bannert
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - Diego Kyburz
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - Alan J Kivitz
- Department of Rheumatology, Altoona Center for Clinical Research, Duncansville, PA, USA
| | - Steven E Carsons
- Division of Rheumatology, Allergy and Immunology NYU Winthrop Hospital, NYU Long Island School of Medicine, New York, NY, USA
| | - David A Isenberg
- Centre for Rheumatology, Department of Medicine, University College London Hospital, London, UK
| | - Francesca Barone
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Simon J Bowman
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Pascal Espié
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - David Floch
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Cyrielle Dupuy
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Xiaohui Ren
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Petra M Faerber
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | - Michael Rotte
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Julie Milojevic
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | - James S Rush
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Peter Gergely
- Novartis Institutes for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
72
|
Expression of CD40L by the ALVAC-Simian Immunodeficiency Virus Vector Abrogates T Cell Responses in Macaques. J Virol 2020; 94:JVI.01933-19. [PMID: 31896599 DOI: 10.1128/jvi.01933-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022] Open
Abstract
Immunization with recombinant ALVAC/gp120 alum vaccine provided modest protection from human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) acquisition in humans and macaques. Vaccine-mediated protection was associated with the elicitation of IgG against the envelope V2 loop and of envelope-specific CD4+ T cell responses. We hypothesized that the simultaneous expression of the costimulatory molecule CD40L (CD154) by the ALVAC-HIV vector could increase both protective humoral and cellular responses. We engineered an ALVAC-SIV coexpressing CD40L with SIVmac251 (ALVAC-SIV/CD40L) gag, pol, and env genes. We compared its immunogenicity in macaques with that of a canonical ALVAC-SIV, with both given as a vector-prime/gp120 in alum boost strategy. The ALVAC-SIV/CD40L was superior to the ALVAC-SIV regimen in inducing binding and tier 1 neutralizing antibodies against the gp120. The increase in humoral responses was associated with the expression of the membrane-bound form of the CD40L by CD4+ T cells in lymph nodes. Unexpectedly, the ALVAC-SIV/CD40L vector had a blunting effect on CD4+ Th1 helper responses and instead favored the induction of myeloid-derived suppressor cells, the immune-suppressive interleukin-10 (IL-10) cytokine, and the down-modulatory tryptophan catabolism. Ultimately, this strategy failed to protect macaques from SIV acquisition. Taken together, these results underlie the importance of balanced vaccine-induced activating versus suppressive immune responses in affording protection from HIV.IMPORTANCE CD40-CD40 ligand (CD40L) interaction is crucial for inducing effective cytotoxic and humoral responses against pathogens. Because of its immunomodulatory function, CD40L has been used to enhance immune responses to vaccines, including candidate vaccines for HIV. The only successful vaccine ever tested in humans utilized a strategy combining canarypox virus-based vector (ALVAC) together with an envelope protein (gp120) adjuvanted in alum. This strategy showed limited efficacy in preventing HIV-1/SIV acquisition in humans and macaques. In both species, protection was associated with vaccine-induced antibodies against the HIV envelope and CD4+ T cell responses, including type 1 antiviral responses. In this study, we tested whether augmenting CD40L expression by coexpressing it with the ALVAC vector could increase the protective immune responses. Although coexpression of CD40L did increase humoral responses, it blunted type 1 CD4+ T cell responses against the SIV envelope protein and failed to protect macaques from viral infection.
Collapse
|
73
|
Davis JM, Crowson CS, Knutson KL, Achenbach SJ, Strausbauch MA, Therneau TM, Matteson EL, Gabriel SE, Wettstein PJ. Longitudinal relationships between rheumatoid factor and cytokine expression by immunostimulated peripheral blood lymphocytes from patients with rheumatoid arthritis: New insights into B-cell activation. Clin Immunol 2020; 211:108342. [PMID: 31926330 PMCID: PMC7045286 DOI: 10.1016/j.clim.2020.108342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/20/2019] [Accepted: 01/04/2020] [Indexed: 01/16/2023]
Abstract
To identify associations between immunostimulated cytokine production and disease characteristics, peripheral blood lymphocytes were collected from 155 adult patients with rheumatoid arthritis (RA) before and after a 5-year interval. The lymphocytes were activated in vitro with T-cell stimulants, cytosine-phosphate-guanine (CpG) oligonucleotide, and medium alone (negative control). Expression of 17 cytokines was evaluated with immunoassays, and factor analysis was used to reduce data complexity and identify cytokine combinations indicative of cell types preferentially activated by each immunostimulant. The findings showed that the highest numbers of correlations were between cytokine levels and rheumatoid factor (RF) positivity and between cytokine levels and disease duration. Scores for cytokines driven by CpG and medium alone were negatively associated with RF positivity and disease duration at baseline but positively associated with both at 5 years. Our findings suggest that RF expression sustained over time increases activation of B cells and monocytes without requirements for T-cell functions.
Collapse
Affiliation(s)
- John M Davis
- Division of Rheumatology, Mayo Clinic, Rochester, MN, United States of America.
| | - Cynthia S Crowson
- Division of Rheumatology, Mayo Clinic, Rochester, MN, United States of America; Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL, United States of America
| | - Sara J Achenbach
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Michael A Strausbauch
- Immunochemical Core Laboratory, Mayo Clinic, Rochester, MN, United States of America
| | - Terry M Therneau
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Eric L Matteson
- Division of Rheumatology, Mayo Clinic, Rochester, MN, United States of America
| | - Sherine E Gabriel
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Peter J Wettstein
- Department of Surgery, Mayo Clinic, Rochester, MN, United States of America
| |
Collapse
|
74
|
Espié P, He Y, Koo P, Sickert D, Dupuy C, Chokoté E, Schuler R, Mergentaler H, Ristov J, Milojevic J, Verles A, Groenewegen A, Auger A, Avrameas A, Rotte M, Colin L, Tomek CS, Hernandez-Illas M, Rush JS, Gergely P. First-in-human clinical trial to assess pharmacokinetics, pharmacodynamics, safety, and tolerability of iscalimab, an anti-CD40 monoclonal antibody. Am J Transplant 2020; 20:463-473. [PMID: 31647605 DOI: 10.1111/ajt.15661] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 01/25/2023]
Abstract
Iscalimab is a fully human, CD40 pathway blocking, nondepleting monoclonal antibody being developed as an immunosuppressive agent. We describe a first-in-human, randomized, double-blind, placebo-controlled study investigating the safety, tolerability, pharmacokinetics, and pharmacodynamics of iscalimab in healthy subjects and rheumatoid arthritis patients. Healthy subjects (n = 56) received single doses of intravenous iscalimab (0.03, 0.1, 0.3, 1, or 3 mg/kg), or subcutaneous iscalimab (3 mg/kg), or placebo. Rheumatoid arthritis patients (n = 20) received single doses of intravenous iscalimab (10 or 30 mg/kg) or placebo. Iscalimab exhibited target-mediated drug disposition resulting in dose-dependent and nonlinear pharmacokinetics. Complete (≥90%) CD40 receptor occupancy on whole blood B cells was observed at plasma concentrations >0.3-0.4 µg/mL. In subjects receiving 3 mg/kg iscalimab, antibody responses to keyhole limpet hemocyanin were transiently suppressed. CD40 occupancy by iscalimab prevented ex vivo human rCD154-induced expression of CD69 on B cells in whole blood. All doses were generally safe and well tolerated, with no clinically relevant changes in any safety parameters, including no evidence of thromboembolic events. Iscalimab appears to be a promising blocker of the CD40-CD154 costimulatory pathway with potential use in transplantation and other autoimmune diseases.
Collapse
Affiliation(s)
- Pascal Espié
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - YanLing He
- Novartis Institutes for BioMedical Research, Inc., Cambridge, Massachusetts, USA
| | - Phillip Koo
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Denise Sickert
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Cyrielle Dupuy
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Edwige Chokoté
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Roland Schuler
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Jacinda Ristov
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Julie Milojevic
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Aurelie Verles
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Anita Auger
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Michael Rotte
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Laurence Colin
- Novartis Institutes for BioMedical Research, Inc., Cambridge, Massachusetts, USA
| | | | | | - James S Rush
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Peter Gergely
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
75
|
Impact of CD40 gene polymorphisms on the risk of immune thrombocytopenic purpura. Gene 2020; 736:144419. [PMID: 32018016 DOI: 10.1016/j.gene.2020.144419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 01/19/2023]
Abstract
OBJECTIVES To evaluate the relationship between two common single nucleotide polymorphisms (SNPs) of CD40 gene (rs1883832 C/T and rs4810485 G/T) and the risk of immune thrombocytopenia (ITP) in the Egyptian population. METHODS A case-control study was conducted retrospectively on 101 cases with ITP and 97 healthy subjects. Two SNPs of CD40 gene (rs1883832 C/T and rs4810485 G/T) were genotyped via Taqman allele discrimination real-time PCR. The frequencies of different genetic models of both SNPs were calculated and compared between ITP cases and controls. Linkage analysis was performed between the studied SNPs. Odds ratio (OR) and 95% confidence interval (CI) were assessed using multinomial logistic regression analysis to determine the association of CD40 gene SNPs genotypes, alleles, and haplotypes with the risk of ITP. The odds ratio was further adjusted to the confounders for risk stratification. RESULTS CD40 (rs1883832) TT genotype carriers have a significantly higher risk of ITP when compared to CC genotype carriers (adjusted OR: 3.792, 95%CI: 1.252-11.49, P = 0.018). T allele also represents 1.711-fold increased risk of ITP which is more evident in males (P = 0.016). No significant difference was observed in the frequency of CD40 (rs4810485 G/T) genetic models between cases and controls. Linkage disequilibrium was found between the two SNPs and revealed four main haplotypes (C-G; C-T; T-G; T-T) with a significantly higher frequency of T-G haplotype in ITP cases than in healthy controls which confers an increased risk of ITP development (OR: 2.349, 95%CI: 1.271-4.339, P = 0.006). CONCLUSIONS CD40 gene SNP rs1883832 is associated with an increased risk of ITP development in the Egyptian population, while the SNP rs4810485 has no association. Moreover, T-G haplotype is a risk genetic model for ITP.
Collapse
|
76
|
Tay C, Kanellakis P, Hosseini H, Cao A, Toh BH, Bobik A, Kyaw T. B Cell and CD4 T Cell Interactions Promote Development of Atherosclerosis. Front Immunol 2020; 10:3046. [PMID: 31998318 PMCID: PMC6965321 DOI: 10.3389/fimmu.2019.03046] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022] Open
Abstract
Interaction between B and CD4 T cells is crucial for their optimal responses in adaptive immunity. Immune responses augmented by their partnership promote chronic inflammation. Here we report that interaction between B and CD4 T cells augments their atherogenicity to promote lipid-induced atherosclerosis. Genetic deletion of the gene encoding immunoglobulin mu (μ) heavy chain (μMT) in ApoE−/− mice resulted in global loss of B cells including those in atherosclerotic plaques, undetectable immunoglobulins and impaired germinal center formation. Despite unaffected numbers in the circulation and peripheral lymph nodes, CD4 T cells were also reduced in spleens as were activated and memory CD4 T cells. In hyperlipidemic μMT−/− ApoE−/− mice, B cell deficiency decreased atherosclerotic lesions, accompanied by absence of immunoglobulins and reduced CD4 T cell accumulation in lesions. Adoptive transfer of B cells deficient in either MHCII or co-stimulatory molecule CD40, molecules required for B and CD4 T cell interaction, into B cell-deficient μMT−/− ApoE−/− mice failed to increase atherosclerosis. In contrast, wildtype B cells transferred into μMT−/− ApoE−/− mice increased atherosclerosis and increased CD4 T cells in lesions including activated and memory CD4 T cells. Transferred B cells also increased their expression of atherogenic cytokines IL-1β, TGF-β, MCP-1, M-CSF, and MIF, with partial restoration of germinal centers and plasma immunoglobulins. Our study demonstrates that interaction between B and CD4 T cells utilizing MHCII and CD40 is essential to augment their function to increase atherosclerosis in hyperlipidemic mice. These findings suggest that targeting B cell and CD4 T cell interaction may be a therapeutic strategy to limit atherosclerosis progression.
Collapse
Affiliation(s)
- Christopher Tay
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Hamid Hosseini
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
77
|
Biram A, Davidzohn N, Shulman Z. T cell interactions with B cells during germinal center formation, a three-step model. Immunol Rev 2019; 288:37-48. [PMID: 30874355 DOI: 10.1111/imr.12737] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/04/2019] [Indexed: 12/25/2022]
Abstract
Establishment of effective immunity against invading microbes depends on continuous generation of antibodies that facilitate pathogen clearance. Long-lived plasma cells with the capacity to produce high affinity antibodies evolve in germinal centers (GCs), where B cells undergo somatic hypermutation and are subjected to affinity-based selection. Here, we focus on the cellular interactions that take place early in the antibody immune response during GC colonization. Clones bearing B-cell receptors with different affinities and specificities compete for entry to the GC, at the boundary between the B-cell and T-cell zones in lymphoid organs. During this process, B cells compete for interactions with T follicular helper cells, which provide selection signals required for differentiation into GC cells and antibody secreting cells. These cellular engagements are long-lasting and depend on activation of adhesion molecules that support persistent interactions and promote transmission of signals between the cells. Here, we discuss how interactions between cognate T and B cells are primarily maintained by three types of molecular interactions: homophilic signaling lymphocytic activation molecule (SLAM) interactions, T-cell receptor: peptide-loaded major histocompatibility class II (pMHCII), and LFA-1:ICAMs. These essential components support a three-step process that controls clonal selection for entry into the antibody affinity maturation response in the GC, and establishment of long-lasting antibody-mediated immunity.
Collapse
Affiliation(s)
- Adi Biram
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Natalia Davidzohn
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
78
|
Activation-induced surface proteins in the identification of antigen-responsive CD4 T cells. Immunol Lett 2019; 219:1-7. [PMID: 31881234 DOI: 10.1016/j.imlet.2019.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/09/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023]
Abstract
Identification of antigen specificity of CD4 T cells is instrumental in understanding adaptive immune responses in health and disease. The high diversity of CD4 T cell repertoire combined with the functional heterogeneity of the compartment poses a challenge to the assessment of CD4 T cell responses. In spite of that, multiple technologies allow direct or indirect interrogation of antigen specificity of CD4 T cells. In the last decade, multiple surface proteins have been established as cytokine-independent surrogates of in vitro CD4 T cell activation, and have found applications in the live identification and isolation of antigen-responsive CD4 T cells. Here we review the current knowledge of the surface proteins that permit identification of viable antigen-responsive CD4 T cells with high specificity, including those capable of identifying specialized CD4 T subsets such as germinal center follicular helper T cells and CD4 regulatory T cells.
Collapse
|
79
|
Zhu Z, Shukla A, Ramezani-Rad P, Apgar JR, Rickert RC. The AKT isoforms 1 and 2 drive B cell fate decisions during the germinal center response. Life Sci Alliance 2019; 2:e201900506. [PMID: 31767615 PMCID: PMC6878223 DOI: 10.26508/lsa.201900506] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
The PI3K pathway is integral for the germinal center (GC) response. However, the contribution of protein kinase B (AKT) as a PI3K effector in GC B cells remains unknown. Here, we show that mice lacking the AKT1 and AKT2 isoforms in B cells failed to form GCs, which undermined affinity maturation and antibody production in response to immunization. Upon B-cell receptor stimulation, AKT1/2-deficient B cells showed poor survival, reduced proliferation, and impaired mitochondrial and metabolic fitness, which collectively halted GC development. By comparison, Foxo1 T24A mutant, which cannot be inactivated by AKT1/2 phosphorylation and is sequestered in the nucleus, significantly enhanced antibody class switch recombination via induction of activation-induced cytidine deaminase (AID) expression. By contrast, repression of FOXO1 activity by AKT1/2 promoted IRF4-driven plasma cell differentiation. Last, we show that T-cell help via CD40, but not enforced expression of Bcl2, rescued the defective GC response in AKT1/2-deficient animals by restoring proliferative expansion and energy production. Overall, our study provides mechanistic insights into the key role of AKT and downstream pathways on B cell fate decisions during the GC response.
Collapse
Affiliation(s)
- Zilu Zhu
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- National Cancer Institute-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ashima Shukla
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- National Cancer Institute-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Parham Ramezani-Rad
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- National Cancer Institute-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - John R Apgar
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- National Cancer Institute-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Robert C Rickert
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- National Cancer Institute-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
80
|
Lu J, Wu J, Xie F, Tian J, Tang X, Guo H, Ma J, Xu P, Mao L, Xu H, Wang S. CD4 + T Cell-Released Extracellular Vesicles Potentiate the Efficacy of the HBsAg Vaccine by Enhancing B Cell Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1802219. [PMID: 31832305 PMCID: PMC6891927 DOI: 10.1002/advs.201802219] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 09/09/2019] [Indexed: 05/15/2023]
Abstract
T cells secrete bioactive extracellular vesicles (EVs), but the potential biological effects of CD4+ T cell EVs are not clear. The main purpose of this study is to investigate the effects of CD4+ T cell-derived EVs on B cell responses and examine their role in antigen-mediated humoral immune responses. In this study, CD4+ T cell EVs are purified from activated CD4+ T cells in vitro. After immunization with the Hepatitis B surface antigen (HBsAg) vaccine, CD4+ T cell EVs-treated mice show stronger humoral immune responses, which is indicated by a greater Hepatitis B surface antibody (HBsAb) level in serum and a greater proportion of plasma cells in bone marrow. In addition, it is found that EVs released from activated CD4+ T cells play an important role in B cell responses in vitro, which significantly promote B cell activation, proliferation, and antibody production. Interestingly, antigen-specific CD4+ T cell EVs are found to be more efficient than control EVs in enhancing B cell responses. Furthermore, it is shown that CD40 ligand (CD40L) is involved in CD4+ T cell EVs-mediated B cell responses. Overall, the results have demonstrated that CD4+ T cell EVs enhance B cell responses and serve as a novel immunomodulator to promote antigen-specific humoral immune responses.
Collapse
Affiliation(s)
- Jian Lu
- Department of Laboratory MedicineThe Affiliated People's HospitalJiangsu UniversityZhenjiang212002China
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| | - Jing Wu
- Department of Laboratory MedicineThe Affiliated People's HospitalJiangsu UniversityZhenjiang212002China
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| | - Feiting Xie
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| | - Jie Tian
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| | - Xinyi Tang
- Department of Laboratory MedicineThe Affiliated People's HospitalJiangsu UniversityZhenjiang212002China
| | - Hongye Guo
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| | - Jie Ma
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| | - Ping Xu
- Department of Laboratory MedicineThe Fifth People's Hospital of SuzhouSuzhou215131China
| | - Lingxiang Mao
- Department of Laboratory MedicineThe Affiliated People's HospitalJiangsu UniversityZhenjiang212002China
| | - Huaxi Xu
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| | - Shengjun Wang
- Department of Laboratory MedicineThe Affiliated People's HospitalJiangsu UniversityZhenjiang212002China
- Department of ImmunologyJiangsu Key Laboratory of Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiang212013China
| |
Collapse
|
81
|
Geginat J, Vasco M, Gerosa M, Tas SW, Pagani M, Grassi F, Flavell RA, Meroni P, Abrignani S. IL-10 producing regulatory and helper T-cells in systemic lupus erythematosus. Semin Immunol 2019; 44:101330. [PMID: 31735515 DOI: 10.1016/j.smim.2019.101330] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) is a highly heterogeneous autoimmune disease characterised by the production of pathogenic autoantibodies against nuclear self-antigens. The anti-inflammatory and tolerogenic cytokine Interleukin-10 appears to play a paradoxical pathogenic role in SLE and is therefore currently therapeutically targeted in clinical trials. It is generally assumed that the pathogenic effect of IL-10 in SLE is due to its growth and differentiation factor activity on autoreactive B-cells, but effects on other cells might also play a role. To date, a unique cellular source of pathogenic IL-10 in SLE has not been identified. In this review, we focus on the contribution of different CD4+T-cell subsets to IL-10 and autoantibody production in SLE. In particular, we discuss that IL-10 produced by different subsets of adaptive regulatory T-cells, follicular helper T-cells and extra-follicular B-helper T-cells is likely to have different effects on autoreactive B-cell responses. A better understanding of the role of IL-10 in B-cell responses and lupus would allow to identify the most promising therapies for individual SLE patients in the future.
Collapse
Affiliation(s)
- J Geginat
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy.
| | - M Vasco
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - M Gerosa
- DISCCO, Department of Clinical Science and Community Health, University of Milan, Italy; ASST Istituto G. Pini, Milan, Italy
| | - S W Tas
- Amsterdam UMC, University of Amsterdam, Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute and Amsterdam Rheumatology & immunology Center (ARC), Academic Medical Center, Amsterdam, the Netherlands
| | - M Pagani
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy; Amsterdam UMC, University of Amsterdam, Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute and Amsterdam Rheumatology & immunology Center (ARC), Academic Medical Center, Amsterdam, the Netherlands; Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | - F Grassi
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy; Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - R A Flavell
- Department of Immunobiology, and Howard Hughes Medical Institute, School of Medicine, Yale University, New Haven, USA
| | - Pl Meroni
- Istituto Auxologico Italiano, Milano, Italy
| | - S Abrignani
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy; DISCCO, Department of Clinical Science and Community Health, University of Milan, Italy
| |
Collapse
|
82
|
Mugue N, Terekhanova N, Afanasyev S, Krasnov A. Transcriptome sequencing of hybrid bester sturgeon: Responses to poly (I:C) in the context of comparative immunogenomics. FISH & SHELLFISH IMMUNOLOGY 2019; 93:888-894. [PMID: 31425830 DOI: 10.1016/j.fsi.2019.08.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 06/10/2023]
Abstract
Sturgeons represent a substantial scientific interest due to their high economic value, endangered status and also as the most primitive group of ray-finned fishes. Rapid progress in knowledge of sturgeon immunity was achieved recently with use of RNA sequencing. We report transcriptome sequencing of gill, head kidney, and spleen of bester sturgeon (a hybrid of beluga Huso huso and sterlet Acipenser ruthenus) injected with synthetic double-stranded RNA (polyI:C). The composition of transcriptome and responses to treatment were examined in the context of comparative genomics with focus on immune genes. Sturgeon transcripts matched to 21.5 k different proteins (blastx). With reference to Atlantic salmon, the functional groups and pathways of the immune system were uniformly represented: at average 36.5 ± 0.8% genes were found. Immune genes comprise a significant fraction of transcriptome. Among twenty genes with highest transcription levels, five are specialized immune genes and two encode heme and iron binding proteins (serotransferrin and hemopexin) also known as acute phase proteins. Challenge induced multiple functional groups including apoptosis, cell cycle and a number of metabolic pathways. Treatment stimulated innate antiviral immunity, which is well conserved between sturgeon and salmon, the most responsive genes were mx, rsad2 (viperin), interferon induced protein 44 and protein with tetratricopeptide repeats 5, cd87 and receptor transporting protein 3. Results added to knowledge of immune phylogeny. Gain and loss of genes was assessed by comparison with genomes from different phylogenetic groups. Among differentially expressed genes, percentage of acquired and lost genes was much lower in comparison with genes present in all vertebrates. Innate antiviral immunity was subject to the greatest changes in evolution of jawed vertebrates. A significant fraction of genes (15%) was lost in mammals and only half of genes is annotated in public databases as involved in antiviral responses. Change of function may have an important role in evolution of immunity together with gain and loss of genes.
Collapse
Affiliation(s)
- Nikolai Mugue
- Russian Federal Research Institute of Fisheries and Oceanography, Moscow, Russia; N. K. Koltzov Institute of Developmental Biology RAS, Moscow, Russia.
| | | | - Sergey Afanasyev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia.
| | - Aleksei Krasnov
- Nofima AS, Norwegian Institute of Food, Fisheries & Aquaculture Research, Ås, Norway.
| |
Collapse
|
83
|
Bae SC, Lee YH. Association between CD40 polymorphisms and systemic lupus erythematosus and correlation between soluble CD40 and CD40 ligand levels in the disease: a meta-analysis. Lupus 2019; 28:1452-1459. [DOI: 10.1177/0961203319878822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Objective The aim of this study was to systematically review evidence regarding the association between CD40 polymorphisms and systemic lupus erythematosus and between soluble CD40 (sCD40) and CD40 ligand (sCD40L) levels and systemic lupus erythematosus. Methods We performed a meta-analysis on the association between CD40 rs4810495, rs1883832, and rs376545 polymorphisms and systemic lupus erythematosus risk and sCD40/sCD40L levels in patients with systemic lupus erythematosus and controls. Results Fourteen studies were included. Ethnicity-specific meta-analysis indicated a significant association between the T allele of CD40 rs4810485 polymorphism and systemic lupus erythematosus in Europeans (odds ratio = 0.715, 95% confidence interval = 0.641–0.832, p < 0.001) and a trend toward an association between the T allele and systemic lupus erythematosus in Asians (odds ratio = 1.255, 95% confidence interval = 0.978–1.810, p = 0.074). Furthermore, a significant association was reported between systemic lupus erythematosus and the C allele of CD40 rs1883832 polymorphism (odds ratio = 1.235, 95% confidence interval = 1.087–1.405, p = 0.001) and A allele of CD40 rs3765456 polymorphism and systemic lupus erythematosus in Asians (odds ratio = 1.184, 95% confidence interval = 1.040–1.348, p = 0.011). sCD40 and sCD40L levels were significantly higher in SLE than in controls (standardized mean difference = 1.564, 95% confidence interval = 0.256–2.872, p = 0.019 and standardized mean difference = 1.499, 95% confidence interval = 1.031–1.967, p < 0.001, respectively). Stratification based on ethnicity revealed higher sCD40L levels in the systemic lupus erythematosus group among European, Asian, North American, and Arab populations. Conclusions Our meta-analyses found associations between CD40 rs4810495, rs1883832, and rs376545 polymorphisms and systemic lupus erythematosus susceptibility and significantly higher sCD40 and sCD40L levels in patients with systemic lupus erythematosus than in controls.
Collapse
Affiliation(s)
- S -C Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| | - Y H Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
84
|
Skarlis C, Marketos N, Mavragani CP. Biologics in Sjögren's syndrome. Pharmacol Res 2019; 147:104389. [DOI: 10.1016/j.phrs.2019.104389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 12/20/2022]
|
85
|
Targeting the CD40-CD154 Signaling Pathway for Treatment of Autoimmune Arthritis. Cells 2019; 8:cells8080927. [PMID: 31426619 PMCID: PMC6721639 DOI: 10.3390/cells8080927] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 12/14/2022] Open
Abstract
Full activation of T lymphocytes requires signals from both T cell receptors and costimulatory molecules. In addition to CD28, several T cell molecules could deliver costimulatory signals, including CD154, which primarily interacts with CD40 on B-cells. CD40 is a critical molecule regulating several B-cell functions, such as antibody production, germinal center formation and cellular proliferation. Upregulated expression of CD40 and CD154 occurs in immune effector cells and non-immune cells in different autoimmune diseases. In addition, therapeutic benefits have been observed by blocking the CD40-CD154 interaction in animals with collagen-induced arthritis. Given the therapeutic success of the biologics abatacept, which blocks CD28 costimulation, and rituximab, which deletes B cells in the treatment of autoimmune arthritis, the inhibition of the CD40-CD154 axis has two advantages, namely, attenuating CD154-mediated T cell costimulation and suppressing CD40-mediated B-cell stimulation. Furthermore, blockade of the CD40-CD154 interaction drives the conversion of CD4+ T cells to regulatory T cells that mediate immunosuppression. Currently, several biological products targeting the CD40-CD154 axis have been developed and are undergoing early phase clinical trials with encouraging success in several autoimmune disorders, including autoimmune arthritis. This review addresses the roles of the CD40-CD154 axis in the pathogenesis of autoimmune arthritis and its potential as a therapeutic target.
Collapse
|
86
|
Roco JA, Mesin L, Binder SC, Nefzger C, Gonzalez-Figueroa P, Canete PF, Ellyard J, Shen Q, Robert PA, Cappello J, Vohra H, Zhang Y, Nowosad CR, Schiepers A, Corcoran LM, Toellner KM, Polo JM, Meyer-Hermann M, Victora GD, Vinuesa CG. Class-Switch Recombination Occurs Infrequently in Germinal Centers. Immunity 2019; 51:337-350.e7. [PMID: 31375460 DOI: 10.1016/j.immuni.2019.07.001] [Citation(s) in RCA: 315] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/26/2019] [Accepted: 07/09/2019] [Indexed: 01/06/2023]
Abstract
Class-switch recombination (CSR) is a DNA recombination process that replaces the immunoglobulin (Ig) constant region for the isotype that can best protect against the pathogen. Dysregulation of CSR can cause self-reactive BCRs and B cell lymphomas; understanding the timing and location of CSR is therefore important. Although CSR commences upon T cell priming, it is generally considered a hallmark of germinal centers (GCs). Here, we have used multiple approaches to show that CSR is triggered prior to differentiation into GC B cells or plasmablasts and is greatly diminished in GCs. Despite finding a small percentage of GC B cells expressing germline transcripts, phylogenetic trees of GC BCRs from secondary lymphoid organs revealed that the vast majority of CSR events occurred prior to the onset of somatic hypermutation. As such, we have demonstrated the existence of IgM-dominated GCs, which are unlikely to occur under the assumption of ongoing switching.
Collapse
Affiliation(s)
- Jonathan A Roco
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, 10065, USA
| | - Sebastian C Binder
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Rebenring 56, 38106 Braunschweig, Germany
| | - Christian Nefzger
- Department of Anatomy and Developmental Biology and Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton VIC 3800, Australia
| | - Paula Gonzalez-Figueroa
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Pablo F Canete
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Julia Ellyard
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Qian Shen
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Philippe A Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Rebenring 56, 38106 Braunschweig, Germany
| | - Jean Cappello
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Harpreet Vohra
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia
| | - Yang Zhang
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Carla R Nowosad
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, 10065, USA
| | - Arien Schiepers
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, 10065, USA
| | - Lynn M Corcoran
- Molecular Immunology Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville VIC 3052, Australia
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Jose M Polo
- Department of Anatomy and Developmental Biology and Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton VIC 3800, Australia
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Rebenring 56, 38106 Braunschweig, Germany; Institute for Biochemistry, Biotechnology, and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, Rockefeller University, New York, NY, 10065, USA
| | - Carola G Vinuesa
- Department of Immunology and Infectious Disease and Centre for Personalised Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 0200, Australia; China-Australia Centre for Personalised Immunology, Department of Rheumatology, Shanghai Renji Hospital, Shanghai JiaoTong University, Shanghai, China.
| |
Collapse
|
87
|
Abstract
The marginal zone (MZ) is largely composed of a unique subpopulation of B cells, the so-called MZ-B cells. At a molecular level, memory B cells are characterized by the presence of somatically mutated IGV genes. The earliest studies in the rat have documented the presence of hapten-specific MZ-B cells after immunization in the MZ. This work later received experimental support demonstrating that the IGHV-Cµ transcripts expressed by phenotypically defined splenic MZ-B cells (defined as CD90negIgMhighIgDlow B cells) can carry somatic hypermutation. However, only a minor fraction (< 10%-20%) of these MZ-B cells is mutated and is considered to represent memory B cells. Memory B cells can either be class-switched (IgG, IgA, IgE), or non-class-switched (IgM) B cells. B cells in the MZ are a heterogeneous population of cells and both naïve MZ-B cells; class switched and unswitched memory MZ-B cells are present at this unique site in the spleen. Naïve MZ-B cells carry unmutated Ig genes, produce low-affinity IgM molecules and constitute a first line of defense against invading pathogens. Memory MZ-B cells express high-affinity Ig molecules, directed to (microbial) antigens that have been encountered. In this review, we report on the memory compartment of splenic MZ-B cells in the rat to provide insights into the origin and function of these memory MZ-B cells.
Collapse
Affiliation(s)
- Jacobus Hendricks
- Discipline of Human Physiology, Westville Campus, University of KwaZulu-Natal, Durban, South Africa.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nicolaas A Bos
- Discipline of Human Physiology, Westville Campus, University of KwaZulu-Natal, Durban, South Africa
| | - Frans G M Kroese
- Discipline of Human Physiology, Westville Campus, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
88
|
Smets I, Fiddes B, Garcia-Perez JE, He D, Mallants K, Liao W, Dooley J, Wang G, Humblet-Baron S, Dubois B, Compston A, Jones J, Coles A, Liston A, Ban M, Goris A, Sawcer S. Multiple sclerosis risk variants alter expression of co-stimulatory genes in B cells. Brain 2019; 141:786-796. [PMID: 29361022 PMCID: PMC5837558 DOI: 10.1093/brain/awx372] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022] Open
Abstract
The increasing evidence supporting a role for B cells in the pathogenesis of multiple sclerosis prompted us to investigate the influence of known susceptibility variants on the surface expression of co-stimulatory molecules in these cells. Using flow cytometry we measured surface expression of CD40 and CD86 in B cells from 68 patients and 162 healthy controls that were genotyped for the multiple sclerosis associated single nucleotide polymorphisms (SNPs) rs4810485, which maps within the CD40 gene, and rs9282641, which maps within the CD86 gene. We found that carrying the risk allele rs4810485*T lowered the cell-surface expression of CD40 in all tested B cell subtypes (in total B cells P ≤ 5.10 × 10−5 in patients and ≤4.09 × 10−6 in controls), while carrying the risk allele rs9282641*G increased the expression of CD86, with this effect primarily seen in the naïve B cell subset (P = 0.048 in patients and 5.38 × 10−5 in controls). In concordance with these results, analysis of RNA expression demonstrated that the risk allele rs4810485*T resulted in lower total CD40 expression (P = 0.057) but with an increased proportion of alternative splice-forms leading to decoy receptors (P = 4.00 × 10−7). Finally, we also observed that the risk allele rs4810485*T was associated with decreased levels of interleukin-10 (P = 0.020), which is considered to have an immunoregulatory function downstream of CD40. Given the importance of these co-stimulatory molecules in determining the immune reaction that appears in response to antigen our data suggest that B cells might have an important antigen presentation and immunoregulatory role in the pathogenesis of multiple sclerosis.
Collapse
Affiliation(s)
- Ide Smets
- Laboratory for Neuroimmunology, Department of Neurosciences, KU Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Barnaby Fiddes
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Josselyn E Garcia-Perez
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Laboratory for Translational Immunology, Department of Immunology and Microbiology, KU Leuven, Belgium
| | - Di He
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Klara Mallants
- Laboratory for Neuroimmunology, Department of Neurosciences, KU Leuven, Belgium
| | - Wenjia Liao
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - James Dooley
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Laboratory for Translational Immunology, Department of Immunology and Microbiology, KU Leuven, Belgium
| | - George Wang
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Stephanie Humblet-Baron
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Laboratory for Translational Immunology, Department of Immunology and Microbiology, KU Leuven, Belgium
| | - Bénédicte Dubois
- Laboratory for Neuroimmunology, Department of Neurosciences, KU Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Alastair Compston
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Joanne Jones
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Alasdair Coles
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Adrian Liston
- VIB Center for Brain and Disease Research, Leuven, Belgium.,Laboratory for Translational Immunology, Department of Immunology and Microbiology, KU Leuven, Belgium
| | - Maria Ban
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - An Goris
- Laboratory for Neuroimmunology, Department of Neurosciences, KU Leuven, Belgium
| | - Stephen Sawcer
- University of Cambridge, Department of Clinical Neurosciences, Box 165, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| |
Collapse
|
89
|
Remer M, White A, Glennie M, Al-Shamkhani A, Johnson P. The Use of Anti-CD40 mAb in Cancer. Curr Top Microbiol Immunol 2019; 405:165-207. [PMID: 25651948 DOI: 10.1007/82_2014_427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Immunomodulatory monoclonal antibody (mAb) therapy is at the forefront of developing cancer therapeutics with numerous targeted agents proving highly effective in selective patients at stimulating protective host immunity, capable of eradicating established tumours and leading to long-term disease-free states. The cell surface marker CD40 is expressed on a range of immune cells and transformed cells in malignant states whose signalling plays a critical role in modulating adaptive immune responses. Anti-CD40 mAb therapy acts via multiple mechanisms to stimulate anti-tumour immunity across a broad range of lymphoid and solid malignancies. A wealth of preclinical research in this field has led to the successful development of multiple anti-CD40 mAb agents that have shown promise in early-phase clinical trials. Significant progress has been made to enhance the engagement of antibodies with immune effectors through their interactions with Fcγ receptors (FcγRs) by the process of Fc engineering. As more is understood about how to best optimise these agents, principally through the fine-tuning of mAb structure and choice of synergistic partnerships, our ability to generate robust, clinically beneficial anti-tumour activity will form the foundation for the next generation of cancer therapeutics.
Collapse
Affiliation(s)
- Marcus Remer
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.
| | - Ann White
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Martin Glennie
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Aymen Al-Shamkhani
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Peter Johnson
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| |
Collapse
|
90
|
Yamano T, Dobeš J, Vobořil M, Steinert M, Brabec T, Ziętara N, Dobešová M, Ohnmacht C, Laan M, Peterson P, Benes V, Sedláček R, Hanayama R, Kolář M, Klein L, Filipp D. Aire-expressing ILC3-like cells in the lymph node display potent APC features. J Exp Med 2019; 216:1027-1037. [PMID: 30918005 PMCID: PMC6504225 DOI: 10.1084/jem.20181430] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 01/17/2019] [Accepted: 02/27/2019] [Indexed: 01/08/2023] Open
Abstract
The autoimmune regulator (Aire) serves an essential function for T cell tolerance by promoting the "promiscuous" expression of tissue antigens in thymic epithelial cells. Aire is also detected in rare cells in peripheral lymphoid organs, but the identity of these cells is poorly understood. Here, we report that Aire protein-expressing cells in lymph nodes exhibit typical group 3 innate lymphoid cell (ILC3) characteristics such as lymphoid morphology, absence of "classical" hematopoietic lineage markers, and dependence on RORγt. Aire+ cells are more frequent among lineage-negative RORγt+ cells of peripheral lymph nodes as compared with mucosa-draining lymph nodes, display a unique Aire-dependent transcriptional signature, express high surface levels of MHCII and costimulatory molecules, and efficiently present an endogenously expressed model antigen to CD4+ T cells. These findings define a novel type of ILC3-like cells with potent APC features, suggesting that these cells serve a function in the control of T cell responses.
Collapse
Affiliation(s)
- Tomoyoshi Yamano
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilans-Universität, Munich, Germany
| | - Jan Dobeš
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Matouš Vobořil
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Madlen Steinert
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilans-Universität, Munich, Germany
| | - Tomáš Brabec
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Natalia Ziętara
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilans-Universität, Munich, Germany
| | - Martina Dobešová
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Caspar Ohnmacht
- Helmholtz Zentrum München, Institut für Allergieforschung, Neuherberg, Germany
| | - Martti Laan
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Part Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Services and Technology Unit, Heidelberg, Germany
| | - Radislav Sedláček
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Rikinari Hanayama
- Department of Immunology, Kanazawa University Graduate School of Medical Sciences, and World Premier International Research Center Initiative Nano Life Science Institute, Kanazawa University, Ishikawa, Japan
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ludger Klein
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilans-Universität, Munich, Germany
| | - Dominik Filipp
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
91
|
Polonskaya Z, Savage PB, Finn MG, Teyton L. High-affinity anti-glycan antibodies: challenges and strategies. Curr Opin Immunol 2019; 59:65-71. [PMID: 31029911 DOI: 10.1016/j.coi.2019.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/18/2019] [Indexed: 12/19/2022]
Abstract
High-affinity binding of antibodies provides for increased specificity and usually higher effector functions in vivo. This goal, well documented in cancer immunotherapy, is very relevant to vaccines as well, and has particularly significant application toward glycan antigens. The inability to elicit high-affinity antibodies has limited potential applications of glycan-based immunogens, giving rise to insufficient population coverage due to low titers and short duration of protection. That such vaccines have achieved widespread use in spite of these shortcomings highlights the surpassing importance of glycans as prophylactic immunological targets. New advances in the combination of synthetic chemistry, bioconjugation, and mechanistic immunology offer the possibility to vastly expand the number of potential molecular targets in cancer and infectious diseases by opening a wider world of carbohydrate structures to immunological recognition and high-affinity response.
Collapse
Affiliation(s)
- Zinaida Polonskaya
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Paul B Savage
- Department of Chemistry and Biochemistry, Brigham Young University, UT, USA
| | - M G Finn
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Luc Teyton
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
92
|
Huse K, Wogsland CE, Polikowsky HG, Diggins KE, Smeland EB, Myklebust JH, Irish JM. Human Germinal Center B Cells Differ from Naïve and Memory B Cells in CD40 Expression and CD40L-Induced Signaling Response. Cytometry A 2019; 95:442-449. [PMID: 30838773 DOI: 10.1002/cyto.a.23737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 11/20/2018] [Accepted: 02/08/2019] [Indexed: 12/21/2022]
Abstract
CD40 expression is required for germinal center (GC) formation and function, but the kinetics and magnitude of signaling following CD40 engagement remain poorly characterized in human B cells undergoing GC reactions. Here, differences in CD40 expression and signaling responses were compared across differentiation stages of mature human tonsillar B cells. A combination of mass cytometry and phospho-specific flow cytometry was used to quantify protein expression and CD40L-induced signaling in primary human naïve, GC, and memory B cells. Protein expression signatures of cell subsets were quantified using viSNE and Marker Enrichment Modeling (MEM). This approach revealed enriched expression of CD40 protein in GC B cells, compared to naïve and memory B cells. Despite this, GC B cells responded to CD40L engagement with lower phosphorylation of NFκB p65 during the first 30 min following CD40L activation. Before CD40L stimulation, GC B cells expressed higher levels of suppressor protein IκBα than naïve and memory B cells. Following CD40 activation, IκBα was rapidly degraded and reached equivalently low levels in naïve, GC, and memory B cells at 30 min following CD40L. Quantifying CD40 signaling responses as a function of bound ligand revealed a correlation between bound CD40L and degree of induced NFκB p65 phosphorylation, whereas comparable IκBα degradation occurred at all measured levels of CD40L binding. These results characterize cell-intrinsic signaling differences that exist in mature human B cells undergoing GC reactions. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Centre for B cell malignancies, University of Oslo, Oslo, Norway
| | - Cara E Wogsland
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hannah G Polikowsky
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Kirsten E Diggins
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Erlend B Smeland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Centre for B cell malignancies, University of Oslo, Oslo, Norway
| | - June H Myklebust
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Centre for B cell malignancies, University of Oslo, Oslo, Norway
| | - Jonathan M Irish
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
93
|
Karnell JL, Rieder SA, Ettinger R, Kolbeck R. Targeting the CD40-CD40L pathway in autoimmune diseases: Humoral immunity and beyond. Adv Drug Deliv Rev 2019; 141:92-103. [PMID: 30552917 DOI: 10.1016/j.addr.2018.12.005] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/12/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022]
Abstract
CD40 is a TNF receptor superfamily member expressed on both immune and non-immune cells. Interactions between B cell-expressed CD40 and its binding partner, CD40L, predominantly expressed on activated CD4+ T cells, play a critical role in promoting germinal center formation and the production of class-switched antibodies. Non-hematopoietic cells expressing CD40 can also engage CD40L and trigger a pro-inflammatory response. This article will highlight what is known about the biology of the CD40-CD40L axis in humans and describe the potential contribution of CD40 signaling on both hematopoietic and non-hematopoietic cells to autoimmune disease pathogenesis. Additionally, novel therapeutic approaches to target this pathway, currently being evaluated in clinical trials, are discussed.
Collapse
|
94
|
Abstract
Class switch recombination (CSR) generates isotype-switched antibodies with distinct effector functions essential for mediating effective humoral immunity. CSR is catalyzed by activation-induced deaminase (AID) that initiates DNA lesions in the evolutionarily conserved switch (S) regions at the immunoglobulin heavy chain (Igh) locus. AID-initiated DNA lesions are subsequently converted into DNA double stranded breaks (DSBs) in the S regions of Igh locus, repaired by non-homologous end-joining to effect CSR in mammalian B lymphocytes. While molecular mechanisms of CSR are well characterized, it remains less well understood how upstream signaling pathways regulate AID expression and CSR. B lymphocytes express multiple receptors including the B cell antigen receptor (BCR) and co-receptors (e.g., CD40). These receptors may share common signaling pathways or may use distinct signaling elements to regulate CSR. Here, we discuss how signals emanating from different receptors positively or negatively regulate AID expression and CSR.
Collapse
Affiliation(s)
- Zhangguo Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| | - Jing H Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
95
|
Wennhold K, Shimabukuro-Vornhagen A, von Bergwelt-Baildon M. B Cell-Based Cancer Immunotherapy. Transfus Med Hemother 2019; 46:36-46. [PMID: 31244580 PMCID: PMC6558332 DOI: 10.1159/000496166] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
B cells are not only producers of antibodies, but also contribute to immune regulation or act as potent antigen-presenting cells. The potential of B cells for cellular therapy is still largely underestimated, despite their multiple diverse effector functions. The CD40L/CD40 signaling pathway is the most potent activator of antigen presentation capacity in B lymphocytes. CD40-activated B cells are potent antigen-presenting cells that induce specific T-cell responses in vitro and in vivo. In preclinical cancer models in mice and dogs, CD40-activated B cell-based cancer immunotherapy was able to induce effective antitumor immunity. So far, there have been only few early-stage clinical studies involving B cell-based cancer vaccines. These trials indicate that B cell-based immunotherapy is generally safe and associated with little toxicity. Furthermore, these studies suggest that B-cell immunotherapy can elicit antitumor T-cell responses. Alongside the recent advances in cellular therapies in general, major obstacles for generation of good manufacturing practice-manufactured B-cell immunotherapies have been overcome. Thus, a first clinical trial involving CD40-activated B cells might be in reach.
Collapse
Affiliation(s)
- Kerstin Wennhold
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | - Michael von Bergwelt-Baildon
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Gene Center Munich, LMU Munich, Munich, Germany
| |
Collapse
|
96
|
Dostert C, Grusdat M, Letellier E, Brenner D. The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond. Physiol Rev 2019; 99:115-160. [DOI: 10.1152/physrev.00045.2017] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The tumor necrosis factor (TNF) and TNF receptor (TNFR) superfamilies (TNFSF/TNFRSF) include 19 ligands and 29 receptors that play important roles in the modulation of cellular functions. The communication pathways mediated by TNFSF/TNFRSF are essential for numerous developmental, homeostatic, and stimulus-responsive processes in vivo. TNFSF/TNFRSF members regulate cellular differentiation, survival, and programmed death, but their most critical functions pertain to the immune system. Both innate and adaptive immune cells are controlled by TNFSF/TNFRSF members in a manner that is crucial for the coordination of various mechanisms driving either co-stimulation or co-inhibition of the immune response. Dysregulation of these same signaling pathways has been implicated in inflammatory and autoimmune diseases, highlighting the importance of their tight regulation. Investigation of the control of TNFSF/TNFRSF activities has led to the development of therapeutics with the potential to reduce chronic inflammation or promote anti-tumor immunity. The study of TNFSF/TNFRSF proteins has exploded over the last 30 yr, but there remains a need to better understand the fundamental mechanisms underlying the molecular pathways they mediate to design more effective anti-inflammatory and anti-cancer therapies.
Collapse
Affiliation(s)
- Catherine Dostert
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Melanie Grusdat
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Elisabeth Letellier
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
97
|
Tripathi P, Sedimbi SK, Singh AK, Löfbom L, Issazadeh-Navikas S, Weiss S, Förster I, Karlsson MCI, Yrlid U, Kadri N, Cardell SL. Innate and adaptive stimulation of murine diverse NKT cells result in distinct cellular responses. Eur J Immunol 2018; 49:443-453. [PMID: 30427069 PMCID: PMC6587840 DOI: 10.1002/eji.201847647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 10/24/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023]
Abstract
Natural killer T (NKT) cells recognize glycolipids presented on CD1d. They share features of adaptive T lymphocytes and innate NK cells, and mediate immunoregulatory functions via rapid production of cytokines. Invariant (iNKT) and diverse (dNKT) NKT cell subsets are defined by their TCR. The immunological role of dNKT cells, that do not express the invariant TCRα‐chain used by iNKT cells, is less well explored than that of iNKT cells. Here, we investigated signals driving Toll‐like receptor (TLR) ligand activation of TCR‐transgenic murine dNKT cells. IFN‐γ production by dNKT cells required dendritic cells (DC), cell‐to‐cell contact and presence of TLR ligands. TLR‐stimulated DC activated dNKT cells to secrete IFN‐γ in a CD1d‐, CD80/86‐ and type I IFN‐independent manner. In contrast, a requirement for IL‐12p40, and a TLR ligand‐selective dependence on IL‐18 or IL‐15 was observed. TLR ligand/DC stimulation provoked early secretion of pro‐inflammatory cytokines by both CD62L+ and CD62L− dNKT cells. However, proliferation was limited. In contrast, TCR/co‐receptor‐mediated activation resulted in proliferation and delayed production of a broader cytokine spectrum preferentially in CD62L− dNKT cells. Thus, innate (TLR ligand/DC) and adaptive (TCR/co‐receptor) stimulation of dNKT cells resulted in distinct cellular responses that may contribute differently to the formation of immune memory.
Collapse
Affiliation(s)
- Prabhanshu Tripathi
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Saikiran K Sedimbi
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Avadhesh Kumar Singh
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Linda Löfbom
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, Copenhagen, Denmark
| | - Siegfried Weiss
- Institute of Immunology, Medical School Hannover, Hannover, Germany
| | - Irmgard Förster
- Immunology and Environment, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Nadir Kadri
- Center of Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Susanna L Cardell
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
98
|
Ristov J, Espie P, Ulrich P, Sickert D, Flandre T, Dimitrova M, Müller-Ristig D, Weider D, Robert G, Schmutz P, Greutmann B, Cordoba-Castro F, Schneider MA, Warncke M, Kolbinger F, Cote S, Heusser C, Bruns C, Rush JS. Characterization of the in vitro and in vivo properties of CFZ533, a blocking and non-depleting anti-CD40 monoclonal antibody. Am J Transplant 2018; 18:2895-2904. [PMID: 29665205 DOI: 10.1111/ajt.14872] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/23/2018] [Accepted: 04/04/2018] [Indexed: 01/25/2023]
Abstract
The CD40-CD154 costimulatory pathway is essential for T cell-dependent immune responses, development of humoral memory, and antigen presenting cell function. These immune functions have been implicated in the pathology of multiple autoimmune diseases as well as allograft rejection. We have generated CFZ533, a fully human, pathway blocking anti-CD40 monoclonal antibody that has been modified with a N297A mutation to render it unable to mediate Fcγ-dependent effector functions. CFZ533 inhibited CD154-induced activation of human leukocytes in vitro, but failed to induce human leukocyte activation. Additionally, CFZ533 was unable to mediate depletion of human CD40 expressing B cells. In vivo, CFZ533 blocked primary and recall T cell-dependent antibody responses in nonhuman primates and abrogated germinal formation without depleting peripheral blood B cells. We also established a relationship between plasma concentrations of CFZ533 and CD40 pathway-relevant pharmacodynamic effects in tissue. Collectively these data support the scientific rationale and posology for clinical utility of this antibody in select autoimmune diseases and solid organ transplantation.
Collapse
Affiliation(s)
- Jacinda Ristov
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Pascal Espie
- Translational Medicine, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Peter Ulrich
- Translational Medicine, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Denise Sickert
- Translational Medicine, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Thierry Flandre
- Translational Medicine, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Mirela Dimitrova
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Dorothee Müller-Ristig
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Doris Weider
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Gautier Robert
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Patrick Schmutz
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Barbara Greutmann
- Translational Medicine, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | | | - Martin A Schneider
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Max Warncke
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Frank Kolbinger
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Serge Cote
- Translational Medicine, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Christoph Heusser
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Christian Bruns
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - James S Rush
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes of Biomedical Research, Basel, Switzerland
| |
Collapse
|
99
|
Ko KH, Lee IK, Kim G, Gu MJ, Kim HY, Park BC, Park TS, Han SH, Yun CH. Changes in bursal B cells in chicken during embryonic development and early life after hatching. Sci Rep 2018; 8:16905. [PMID: 30442912 PMCID: PMC6238004 DOI: 10.1038/s41598-018-34897-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/01/2018] [Indexed: 11/25/2022] Open
Abstract
The bursa of Fabricius, the primary lymphoid organ for B cell development found only in birds, offers novel approaches to study B cell differentiation at various developmental stages. Here, we explored the changes and mechanism involved in the developmental stages of bursal B cells. The bursal B cells rapidly increased in the late embryonic stage and around hatching, which coincided with changes in specific cell surface markers. Moreover, the cells in the bursa were divided by size into small (low forward- and side-scatter) or large (high forward- and side-scatter) via flow cytometry. It is intriguing that the proportion of small and large B cells was reversed during this period. Because little is known about this phenomenon, we hypothesized that size-based B cell population could be used as an indicator to distinguish their status and stage during B cell development in chicken. The results demonstrated that large B cells are actively proliferating cells than small B cells. Additionally, large B cells showed higher mRNA expression of both proliferation- and differentiation-associated genes compared to small B cells. Taken together, these data show that large bursal B cells are the main source of proliferation and differentiation during B cell development in chickens.
Collapse
Affiliation(s)
- Kwang Hyun Ko
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.,Biomodulation Major and Center for Food Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
| | - In Kyu Lee
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Girak Kim
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min Jeong Gu
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.,Biomodulation Major and Center for Food Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyun Young Kim
- Department of Oral Microbiology and Immunology, Dental Research Institute, and Brain Korea 21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Chul Park
- Institute of Green Bio Science Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Tae Sub Park
- Institute of Green Bio Science Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, Dental Research Institute, and Brain Korea 21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea. .,Biomodulation Major and Center for Food Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea. .,Institute of Green Bio Science Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea.
| |
Collapse
|
100
|
Mener A, Patel SR, Arthur CM, Chonat S, Wieland A, Santhanakrishnan M, Liu J, Maier CL, Jajosky RP, Girard-Pierce K, Bennett A, Zerra PE, Smith NH, Hendrickson JE, Stowell SR. Complement serves as a switch between CD4+ T cell-independent and -dependent RBC antibody responses. JCI Insight 2018; 3:121631. [PMID: 30429364 DOI: 10.1172/jci.insight.121631] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/10/2018] [Indexed: 01/11/2023] Open
Abstract
RBC alloimmunization represents a significant immunological challenge for patients requiring lifelong transfusion support. The majority of clinically relevant non-ABO(H) blood group antigens have been thought to drive antibody formation through T cell-dependent immune pathways. Thus, we initially sought to define the role of CD4+ T cells in formation of alloantibodies to KEL, one of the leading causes of hemolytic transfusion reactions. Unexpectedly, our findings demonstrated that KEL RBCs actually possess the ability to induce antibody formation independent of CD4+ T cells or complement component 3 (C3), two common regulators of antibody formation. However, despite the ability of KEL RBCs to induce anti-KEL antibodies in the absence of complement, removal of C3 or complement receptors 1 and 2 (CR1/2) rendered recipients completely reliant on CD4+ T cells for IgG anti-KEL antibody formation. Together, these findings suggest that C3 may serve as a novel molecular switch that regulates the type of immunological pathway engaged following RBC transfusion.
Collapse
Affiliation(s)
- Amanda Mener
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Seema R Patel
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Connie M Arthur
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Satheesh Chonat
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, and
| | - Andreas Wieland
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Jingchun Liu
- Yale School of Medicine, Department of Laboratory Medicine, New Haven, Connecticut, USA
| | - Cheryl L Maier
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Ryan P Jajosky
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Kathryn Girard-Pierce
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Ashley Bennett
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Patricia E Zerra
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Nicole H Smith
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| | - Jeanne E Hendrickson
- Yale School of Medicine, Department of Laboratory Medicine, New Haven, Connecticut, USA
| | - Sean R Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology
| |
Collapse
|