1
|
He C, Wang S, Moreews M, Pei S, Chen G, Li QZ, Del Monte Monge A, Ramiro AR, Cai C, Gaya M, Barral P, Buggert M, Batista FD, Karlsson MCI. The balance between conventional and unconventional T follicular helper cells influences autoreactive B cell responses. Cell Rep 2025; 44:115602. [PMID: 40252220 DOI: 10.1016/j.celrep.2025.115602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/26/2025] [Accepted: 03/31/2025] [Indexed: 04/21/2025] Open
Abstract
Invariant natural killer T (iNKT) cells are activated by glycolipids presented on CD1d. When iNKT cells interact with and activate B cells, they can differentiate into iNKT follicular helper (iNKTfh) cells, and here, we investigate how this, in turn, regulates conventional T follicular helper (Tfh) cells. This is done in an autoimmune model where antibodies are produced against self-antigens relevant to the autoimmune disease systemic lupus erythematosus (SLE). We find a balance between iNKTfh and Tfh cells that directs the B cell response and influences Tfh cell generation. This altered balance also affects the specificities and increases the autoantibody response. We also show that CD1d expression by B cells is essential for iNKTfh cell generation. In conclusion, our data shed light on how T cell help for B cells is divided between conventional and unconventional helper cell populations and how this has an impact on autoreactive B cell responses.
Collapse
Affiliation(s)
- Chenfei He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Tomtebodavägen 16, Solna Campus, 171 65 Stockholm, Sweden
| | - Shan Wang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Tomtebodavägen 16, Solna Campus, 171 65 Stockholm, Sweden
| | - Marion Moreews
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Tomtebodavägen 16, Solna Campus, 171 65 Stockholm, Sweden
| | - Shengduo Pei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Tomtebodavägen 16, Solna Campus, 171 65 Stockholm, Sweden
| | - Guangchun Chen
- Microarray Core, Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Quan-Zhen Li
- Microarray Core, Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Almudena R Ramiro
- Spanish Center for Cardiovascular Research, Melchor Fernandez Almagro 3, Madrid, Spain
| | - Curtis Cai
- Center for Infections Medicine, Department of Medicine Karolinska Institutet, Huddinge, Sweden
| | - Mauro Gaya
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Patricia Barral
- The Francis Crick Institute, London, UK; Center for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Marcus Buggert
- Center for Infections Medicine, Department of Medicine Karolinska Institutet, Huddinge, Sweden
| | - Facundo D Batista
- Ragon Institute of MGH, MIT, and Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Tomtebodavägen 16, Solna Campus, 171 65 Stockholm, Sweden.
| |
Collapse
|
2
|
Chatterjee P, Brahma S, Cresswell P, Bandyopadhyay S. CD1d-iNKT Axis in Infectious Diseases: Lessons Learned From the Past. Scand J Immunol 2025; 101:e70024. [PMID: 40243400 DOI: 10.1111/sji.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
CD1d is an antigen-presenting molecule that presents lipid or glycolipid antigens to iNKT cells, a distinct subset of T lymphocytes characterised by their innate-like properties and restricted use of Vα, Jα and Vβ segments. The CD1d-iNKT axis represents an interesting aspect of the immune system with significant potential for therapeutic interventions against infectious diseases. Upon recognition of lipid antigens, iNKT cells initiate rapid and potent immune responses, releasing a diverse array of cytokines such as IL-4, IL-13, IFN-γ etc. that profoundly influence immune reactions against various pathogens, including bacteria and parasites, bridging innate and adaptive immunity. We identify and describe the key features of lipidic antigens and their derivatives that determine the nature of their antigenicity. Furthermore, modulating CD1d-driven iNKT cell responses by an array of lipid and glycolipid antigens holds promise as adjunctive therapy to existing antimicrobial treatments. Understanding the complexities of the CD1d-iNKT axis and exploiting its therapeutic potential in the case of infectious diseases could lead to innovative immunotherapeutic strategies, ushering in a new era of immunotherapy against pathogenic insults.
Collapse
Affiliation(s)
- Priyajit Chatterjee
- University Science Instrument Centre, The University of Burdwan, Burdwan, West Bengal, India
| | - Shubhranil Brahma
- Department of Zoology, Iswar Chandra Vidyasagar College, Belonia, South Tripura, Tripura, India
| | - Peter Cresswell
- Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
3
|
Salou M, Paiva RA, Lantz O. Development and Functions of MAIT Cells. Annu Rev Immunol 2025; 43:253-283. [PMID: 39879553 DOI: 10.1146/annurev-immunol-082323-025943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Mucosal-associated invariant T (MAIT) cells are evolutionarily conserved T cells that recognize microbial metabolites. They are abundant in humans and conserved during mammalian evolution, which suggests that they have important nonredundant functions. In this article, we discuss the evolutionary conservation of MAIT cells and describe their original developmental process. MAIT cells exert a wide variety of effector functions, from killing infected cells and promoting inflammation to repairing tissues. We provide insights into these functions and discuss how they result from the context of stimulation encountered by MAIT cells in different tissues and pathological settings. We describe how MAIT cell numbers and features are modified in disease states, focusing mainly on in vivo models. Lastly, we discuss emerging strategies to manipulate MAIT cells for therapeutic purposes.
Collapse
Affiliation(s)
- Marion Salou
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Rafael A Paiva
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Olivier Lantz
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
- Laboratoire d'Immunologie Clinique, Institut Curie, Paris, France
- Centre d'Investigation Clinique en Biothérapie, Gustave-Roussy and Institut Curie (CIC-BT1428), Paris, France
| |
Collapse
|
4
|
Ahmadivand S, Fux R, Palić D. Role of T Follicular Helper Cells in Viral Infections and Vaccine Design. Cells 2025; 14:508. [PMID: 40214462 PMCID: PMC11987902 DOI: 10.3390/cells14070508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
T follicular helper (Tfh) cells are a specialized subset of CD4+ T lymphocytes that are essential for the development of long-lasting humoral immunity. Tfh cells facilitate B lymphocyte maturation, promote germinal center formation, and drive high-affinity antibody production. Our current knowledge of Tfh interactions with the humoral immune system effectors suggests that they have a critical role in supporting the immune response against viral infections. This review discusses the mechanisms through which Tfh cells influence anti-viral immunity, highlighting their interactions with B cells and their impact on antibody quality and quantity. We explore the role of Tfh cells in viral infections and examine how vaccine design can be improved to enhance Tfh cell responses. Innovative vaccine platforms, such as mRNA vaccines and self-assembling protein nanoplatforms (SAPNs), are promising strategies to enhance Tfh cell activation. Their integration and synergistic combination could further enhance immunity and Tfh responses (SAPN-RNA vaccines). In summary, we provide a comprehensive overview of the current insights into Tfh cells' role during viral infections, emphasizing their potential as strategic targets for innovative vaccine development.
Collapse
Affiliation(s)
- Sohrab Ahmadivand
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University Munich, 80539 Munich, Germany;
| | - Dušan Palić
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| |
Collapse
|
5
|
Palacios PA, Santibañez Á, Aguirre-Muñoz F, Gutiérrez-Vera C, Niño de Zepeda-Carrizo V, Góngora-Pimentel M, Müller M, Cáceres M, Kalergis AM, Carreño LJ. Can invariant Natural Killer T cells drive B cell fate? a look at the humoral response. Front Immunol 2025; 16:1505883. [PMID: 40040714 PMCID: PMC11876049 DOI: 10.3389/fimmu.2025.1505883] [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: 10/03/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Invariant Natural Killer T (NKT) cells represent a unique subset of innate-like T cells that express both NK cell and T cell receptors. These cells are rapidly activated by glycolipid antigens presented via CD1d molecules on antigen-presenting cells (APCs), including B cells, dendritic cells (DCs), and macrophages, or through cytokine-dependent mechanisms. Their ability to produce a wide range of cytokines and express costimulatory molecules underscores their critical role in bridging innate and adaptive immunity. B cells, traditionally recognized for their role in antibody production, also act as potent APCs due to their high expression of CD1d, enabling direct interactions with iNKT cells. This interaction has significant implications for humoral immunity, influencing B cell activation, class-switch recombination (CSR), germinal center formation, and memory B cell differentiation, thus expanding the conventional paradigm of T cell-B cell interactions. While the influence of iNKT cells on B cell biology and humoral responses is well-supported, many aspects of their interaction remain unresolved. Key questions include the roles of different iNKT cell subsets, the diversity of APCs, the spatiotemporal dynamics of these interactions, especially during early activation, and the potential for distinct glycolipid ligands to modulate immune outcomes. Understanding these factors could provide valuable insights into how iNKT cells regulate B cell-mediated immunity and offer opportunities to harness these interactions in immunotherapeutic applications, such as vaccine development. In this review, we examine these unresolved aspects and propose a novel perspective on the regulatory potential of iNKT cells in humoral immunity, emphasizing their promise as a target for innovative vaccine strategies.
Collapse
Affiliation(s)
- Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Álvaro Santibañez
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fernanda Aguirre-Muñoz
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Valentina Niño de Zepeda-Carrizo
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Martín Góngora-Pimentel
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Li K, Hu X, Tu XY, Xian MY, Huang LL, Huang T, Luo R, Jin H, Liu Z. Enhancing COVID-19 Vaccine Efficacy: Dual Adjuvant Strategies with TLR7/8 Agonists and Glycolipids. J Med Chem 2024; 67:21916-21933. [PMID: 39648985 DOI: 10.1021/acs.jmedchem.4c01801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
The controlled release of immunostimulatory agents represents a promising strategy to enhance vaccine efficacy while minimizing side effects. This study aimed to improve the efficacy of the RBD-Fc-based COVID-19 vaccine through combining of an iNKT cell agonist and a TLR7/8 agonist using covalent conjugation and temporal delivery. We hypothesized that these combinations would yield a more balanced Th1/Th2 immune response. For covalent conjugation, we employed an uncleavable linker and a self-immolative disulfide linker to conjugate α-galactosylceramide (αGC) to imidazoquinoline (IMDQ). The αGC-SS-IMDQ-Ac conjugate, designed with a prodrug strategy for controlled TLR7/8 agonist release, elicited a higher IFN-γ/IL-4 T cell response ratio than individual adjuvants or their admixture. In the temporal delivery approach, administering IMDQ followed by αGC after 2 h resulted in the highest IgG2a/IgG1 ratio, significantly surpassing other groups. A 6 h delay between glycolipid and IMDQ injections yielded balanced IgG responses, enhancing IgG, IgG1, and IgG2a levels synergistically.
Collapse
Affiliation(s)
- Ke Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, P. R. China
| | - Xing Hu
- National Key Laboratory of Green Pesticide, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, Hubei, P. R. China
| | - Xin-Yi Tu
- National Key Laboratory of Green Pesticide, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, Hubei, P. R. China
| | - Mao-Ying Xian
- National Key Laboratory of Green Pesticide, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, Hubei, P. R. China
| | - Lei-Lei Huang
- National Key Laboratory of Green Pesticide, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, Hubei, P. R. China
| | - Ting Huang
- National Key Laboratory of Green Pesticide, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, Hubei, P. R. China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, P. R. China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, P. R. China
| | - Zheng Liu
- National Key Laboratory of Green Pesticide, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, Hubei, P. R. China
| |
Collapse
|
7
|
Lebrusant-Fernandez M, Ap Rees T, Jimeno R, Angelis N, Ng JC, Fraternali F, Li VSW, Barral P. IFN-γ-dependent regulation of intestinal epithelial homeostasis by NKT cells. Cell Rep 2024; 43:114948. [PMID: 39580798 PMCID: PMC11876105 DOI: 10.1016/j.celrep.2024.114948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/23/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024] Open
Abstract
Intestinal homeostasis is maintained through the combined functions of epithelial and immune cells that collaborate to preserve the integrity of the intestinal barrier. However, the mechanisms by which immune cell populations regulate intestinal epithelial cell (IEC) homeostasis remain unclear. Here, we use a multi-omics approach to study the immune-epithelial crosstalk and identify CD1d-restricted natural killer T (NKT) cells as key regulators of IEC biology. We find that NKT cells are abundant in the proximal small intestine and show hallmarks of activation at steady state. Subsequently, NKT cells regulate the survival and the transcriptional and cellular composition landscapes of IECs in intestinal organoids, through interferon-γ (IFN-γ) and interleukin-4 secretion. In vivo, lack of NKT cells results in an increase in IEC turnover, while NKT cell activation leads to IFN-γ-dependent epithelial apoptosis. Our findings propose NKT cells as potent producers of cytokines that contribute to the regulation of IEC homeostasis.
Collapse
Affiliation(s)
- Marta Lebrusant-Fernandez
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK
| | - Tom Ap Rees
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK
| | - Rebeca Jimeno
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK
| | | | - Joseph C Ng
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK; Institute of Structural and Molecular Biology, University College London, London, UK
| | - Franca Fraternali
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK; Institute of Structural and Molecular Biology, University College London, London, UK
| | | | - Patricia Barral
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
8
|
Huang X, Yi N, Zhu P, Gao J, Lv J. Sorafenib-induced macrophage extracellular traps via ARHGDIG/IL4/PADI4 axis confer drug resistance through inhibiting ferroptosis in hepatocellular carcinoma. Biol Direct 2024; 19:110. [PMID: 39529192 PMCID: PMC11555812 DOI: 10.1186/s13062-024-00560-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common as well as leading causes of mortality worldwide, and sorafenib is the first-line treatment in HCC patients. Unfortunately, drug resistance to sorafenib often develops. However, the underlying mechanism remains unclear. Here, we reveal the important role of macrophage extracellular traps (METs)-mediated crosstalk between macrophages and tumor cells in sorafenib resistance. METHODS METs in HCC tumor tissues were detected using immunofluorescence. The concentrations of MPO-DNA, elastase and cytokines were measured using ELISA. The mRNA expression levels of genes were confirmed by qRT-PCR. The siRNAs were conducted to knock ARHGDIG in Hepa1-6 and Hep3B cells. Western Blot assay was performed to determine protein expression of Rho GDP dissociation inhibitor gamma (ARHGDIG, or RHOGDI-3), PADI2, and PADI4. Cell viability and migration were evaluated by CCK-8 assay and transwell assay, respectively. Cell ferroptosis was assessed by measurement of Fe2+ concentration, flow cytometry assay of lipid ROS, and western blot assay of GPX4. The functions of sorafenib, DNase I, IL4 neutralization antibody and GPX4 in tumor growth were explored through in vivo experiments. RESULTS Sorafenib induced MET formation in M2 macrophages rather than M1 macrophages derived from both human and mice. In Hepa1-6 HCC mice, METs clearance by DNase I improved response to sorafenib therapy, detected by tumor weight, tumor growth curve, tumor volume, and survival. By screening candidate cytokines that affect macrophage function, we found that sorafenib-promoting IL4 secretion by HCC cells plays a crucial role in sorafenib-induced MET formation. Understanding the critical role of IL4 in sorafenib-induced MET formation led us to find that IL4 neutralization significantly improved the efficiency of sorafenib in HCC models. Mechanistically, we discovered that sorafenib increased the expression of ARHGDIG in HCC cells, which led to the release of IL4. In M2 macrophages, IL4 triggered MET formation by elevating the mRNA and protein expression of peptidyl arginine deiminase 4 (PADI4) rather than PADI2. In HCC models, GSK484 inhibition of PADI4 could consistently weaken sorafenib resistance and improve sorafenib efficiency. Importantly, we discovered that METs contribute to sorafenib resistance by inhibiting the ferroptosis of HCC cells. Meanwhile, PADI4 inhibition or DNase I could reverse the sorafenib resistance caused by METs-inhibiting ferroptosis of HCC cells. CONCLUSION Our study concludes that sorafenib-induced METs inhibit the ferroptosis of tumor cells, suggesting that targeting the IL4/PADI4/METs axis in HCC could reduce or prevent sorafenib resistance.
Collapse
Affiliation(s)
- Xiangbo Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, People's Republic of China
| | - Nan Yi
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, People's Republic of China
| | - Pengfei Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, People's Republic of China
| | - Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China.
| | - Jun Lv
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
| |
Collapse
|
9
|
Rajashekar V, Stern L, Almeida CF, Slobedman B, Abendroth A. The surveillance of viral infections by the unconventional Type I NKT cell. Front Immunol 2024; 15:1472854. [PMID: 39355244 PMCID: PMC11442276 DOI: 10.3389/fimmu.2024.1472854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Type I NKT cells, also known as Invariant Natural Killer T (iNKT) cells, are a subpopulation of unconventional, innate-like T (ILT) cells which can proficiently influence downstream immune effector functions. Type I NKT cells express a semi-invariant αβ T cell receptor (TCR) that recognises lipid-based ligands specifically presented by the non-classical cluster of differentiation (CD1) protein d (CD1d) molecule. Due to their potent immunomodulatory functional capacity, type I NKT cells are being increasingly considered in prophylactic and therapeutic approaches towards various diseases, including as vaccine-adjuvants. As viruses do not encode lipid synthesis, it is surprising that many studies have shown that some viruses can directly impede type I NKT activation through downregulating CD1d expression. Therefore, in order to harness type I NKT cells for potential anti-viral therapeutic uses, it is critical that we fully appreciate how the CD1d-iNKT cell axis interacts with viral immunity. In this review, we examine clinical findings that underpin the importance of type I NKT cell function in viral infections. This review also explores how certain viruses employ immunoevasive mechanisms and directly encode functions to target CD1d expression and type I NKT cell function. Overall, we suggest that the CD1d-iNKT cell axis may hold greater gravity within viral infections than what was previously appreciated.
Collapse
Affiliation(s)
- Varshini Rajashekar
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Lauren Stern
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Catarina F. Almeida
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Barry Slobedman
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| | - Allison Abendroth
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases , University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
10
|
Melo-Silva CR, Sigal LJ. Innate and adaptive immune responses that control lymph-borne viruses in the draining lymph node. Cell Mol Immunol 2024; 21:999-1007. [PMID: 38918577 PMCID: PMC11364670 DOI: 10.1038/s41423-024-01188-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
The interstitial fluids in tissues are constantly drained into the lymph nodes (LNs) as lymph through afferent lymphatic vessels and from LNs into the blood through efferent lymphatics. LNs are strategically positioned and have the appropriate cellular composition to serve as sites of adaptive immune initiation against invading pathogens. However, for lymph-borne viruses, which disseminate from the entry site to other tissues through the lymphatic system, immune cells in the draining LN (dLN) also play critical roles in curbing systemic viral dissemination during primary and secondary infections. Lymph-borne viruses in tissues can be transported to dLNs as free virions in the lymph or within infected cells. Regardless of the entry mechanism, infected myeloid antigen-presenting cells, including various subtypes of dendritic cells, inflammatory monocytes, and macrophages, play a critical role in initiating the innate immune response within the dLN. This innate immune response involves cellular crosstalk between infected and bystander innate immune cells that ultimately produce type I interferons (IFN-Is) and other cytokines and recruit inflammatory monocytes and natural killer (NK) cells. IFN-I and NK cell cytotoxicity can restrict systemic viral spread during primary infections and prevent serious disease. Additionally, the memory CD8+ T-cells that reside or rapidly migrate to the dLN can contribute to disease prevention during secondary viral infections. This review explores the intricate innate immune responses orchestrated within dLNs that contain primary viral infections and the role of memory CD8+ T-cells following secondary infection or CD8+ T-cell vaccination.
Collapse
Affiliation(s)
- Carolina R Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Bluemle Life Sciences Building Room 709, 233 South 10th Street, Philadelphia, PA, 19107, USA.
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Bluemle Life Sciences Building Room 709, 233 South 10th Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
11
|
Choi Y, Saron WA, O'Neill A, Senanayake M, Wilder-Smith A, Rathore AP, St John AL. NKT cells promote Th1 immune bias to dengue virus that governs long-term protective antibody dynamics. J Clin Invest 2024; 134:e169251. [PMID: 39088280 PMCID: PMC11405039 DOI: 10.1172/jci169251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/23/2024] [Indexed: 08/03/2024] Open
Abstract
NKT cells are innate-like T cells, recruited to the skin during viral infection, yet their contributions to long-term immune memory to viruses are unclear. We identified granzyme K, a product made by cytotoxic cells including NKT cells, as linked to induction of Th1-associated antibodies during primary dengue virus (DENV) infection in humans. We examined the role of NKT cells in vivo using DENV-infected mice lacking CD1d-dependent (CD1ddep) NKT cells. In CD1d-KO mice, Th1-polarized immunity and infection resolution were impaired, which was dependent on intrinsic NKT cell production of IFN-γ, since it was restored by adoptive transfer of WT but not IFN-γ-KO NKT cells. Furthermore, NKT cell deficiency triggered immune bias, resulting in higher levels of Th2-associated IgG1 than Th1-associated IgG2a, which failed to protect against a homologous DENV rechallenge and promoted antibody-dependent enhanced disease during secondary heterologous infections. Similarly, Th2 immunity, typified by a higher IgG4/IgG3 ratio, was associated with worsened human disease severity during secondary infections. Thus, CD1ddep NKT cells establish Th1 polarity during the early innate response to DENV, which promotes infection resolution, memory formation, and long-term protection from secondary homologous and heterologous infections in mice, with consistent associations observed in humans. These observations illustrate how early innate immune responses during primary infections can influence secondary infection outcomes.
Collapse
Affiliation(s)
- Youngjoo Choi
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Wilfried Aa Saron
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Aled O'Neill
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
| | - Manouri Senanayake
- Department of Paediatrics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- Lady Ridgeway Children's Hospital, Colombo, Sri Lanka
| | - Annelies Wilder-Smith
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Abhay Ps Rathore
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Ashley L St John
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore
| |
Collapse
|
12
|
Gunbin KV, Kopeina GS, Zhivotovsky B, Zamaraev AV. Features of the CD1 gene family in rodents and the uniqueness of the immune system of naked mole-rat. Biol Direct 2024; 19:58. [PMID: 39075541 PMCID: PMC11285450 DOI: 10.1186/s13062-024-00503-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
Cluster of Differentiation 1 (CD1) proteins are widely expressed throughout jawed vertebrates and present lipid antigens to specific CD1-restricted T lymphocytes. CD1 molecules play an important role in immune defense with the presence or absence of particular CD1 proteins frequently associated with the functional characteristics of the immune system. Here, we show the evolution of CD1 proteins in the Rodentia family and the diversity among its members. Based on the analysis of CD1 protein-coding regions in rodent genomes and the reconstruction of protein structures, we found that Heterocephalus glaber represents a unique member of the suborder Hystricomorpha with significant changes in protein sequences and structures of the CD1 family. Multiple lines of evidence point to the absence of CD1d and CD1e and probably a dysfunctional CD1b protein in Heterocephalus glaber. In addition, the impact of CD1d loss on the CD1d/Natural killer T (NKT) cell axis in the naked mole-rat and its potential implications for immune system function are discussed in detail.
Collapse
Affiliation(s)
- Konstantin V Gunbin
- Center for Mitochondrial Functional Genomics, Immanuel Kant Baltic Federal University, Kaliningrad, 236016, Russia
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, 630090, Russia
| | - Gelina S Kopeina
- Engelhardt Institute of Molecular Biology, RAS, Moscow, 119991, Russia
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Boris Zhivotovsky
- Engelhardt Institute of Molecular Biology, RAS, Moscow, 119991, Russia.
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia.
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, Stockholm, 17177, Sweden.
| | - Alexey V Zamaraev
- Engelhardt Institute of Molecular Biology, RAS, Moscow, 119991, Russia.
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
13
|
Evans L, Barral P. CD1 molecules: Beyond antigen presentation. Mol Immunol 2024; 170:1-8. [PMID: 38579449 PMCID: PMC11481681 DOI: 10.1016/j.molimm.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
CD1 molecules are well known for their role in binding and presenting lipid antigens to mediate the activation of CD1-restricted T cells. However, much less appreciated is the fact that CD1 molecules can have additional "unconventional" roles which impact the activation and functions of CD1-expressing cells, ultimately controlling tissue homeostasis as well as the progression of inflammatory and infectious diseases. Some of these roles are mediated by so-called reverse signalling, by which crosslinking of CD1 molecules at the cell surface initiates intracellular signalling. On the other hand, CD1 molecules can also control metabolic and inflammatory pathways in CD1-expressing cells through cell-intrinsic mechanisms independent of CD1 ligation. Here, we review the evidence for "unconventional" functions of CD1 molecules and the outcomes of such roles for health and disease.
Collapse
Affiliation(s)
- Lauren Evans
- The Peter Gorer Department of Immunobiology. King's College London, London, UK; The Francis Crick Institute, London, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology. King's College London, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
14
|
Hayashizaki K, Kamii Y, Kinjo Y. Glycolipid antigen recognition by invariant natural killer T cells and its role in homeostasis and antimicrobial responses. Front Immunol 2024; 15:1402412. [PMID: 38863694 PMCID: PMC11165115 DOI: 10.3389/fimmu.2024.1402412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Due to the COVID-19 pandemic, the importance of developing effective vaccines has received more attention than ever before. To maximize the effects of vaccines, it is important to select adjuvants that induce strong and rapid innate and acquired immune responses. Invariant natural killer T (iNKT) cells, which constitute a small population among lymphocytes, bypass the innate and acquired immune systems through the rapid production of cytokines after glycolipid recognition; hence, their activation could be used as a vaccine strategy against emerging infectious diseases. Additionally, the diverse functions of iNKT cells, including enhancing antibody production, are becoming more understood in recent years. In this review, we briefly describe the functional subset of iNKT cells and introduce the glycolipid antigens recognized by them. Furthermore, we also introduce novel vaccine development taking advantages of iNKT cell activation against infectious diseases.
Collapse
Affiliation(s)
- Koji Hayashizaki
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo, Japan
- Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasuhiro Kamii
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo, Japan
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuki Kinjo
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo, Japan
- Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Hsu SC, Lin KH, Tseng YC, Cheng YY, Ma HH, Chen YC, Jan JT, Wu CY, Ma C. An Adjuvanted Vaccine-Induced Pathogenesis Following Influenza Virus Infection. Vaccines (Basel) 2024; 12:569. [PMID: 38932298 PMCID: PMC11209567 DOI: 10.3390/vaccines12060569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
An incomplete Freund's adjuvant elicited an overt pathogenesis in vaccinated mice following the intranasal challenge of A/California/07/2009 (H1N1) virus despite the induction of a higher specific antibody titer than other adjuvanted formulations. Aluminum hydroxide adjuvants have not induced any pathogenic signs in a variety of formulations with glycolipids. A glycolipid, α-galactosyl ceramide, improved a stimulatory effect of distinct adjuvanted formulations on an anti-influenza A antibody response. In contrast to α-galactosyl ceramide, its synthetic analogue C34 was antagonistic toward a stimulatory effect of an aluminum hydroxide adjuvant on a specific antibody response. The aluminum hydroxide adjuvant alone could confer complete vaccine-induced protection against mortality as well as morbidity caused by a lethal challenge of the same strain of an influenza A virus. The research results indicated that adjuvants could reshape immune responses either to improve vaccine-induced immunity or to provoke an unexpected pathogenic consequence. On the basis of these observations, this research connotes the prominence to develop a precision adjuvant for innocuous vaccination aimed at generating a protective immunity without aberrant responses.
Collapse
Affiliation(s)
- Shiou-Chih Hsu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| | - Kun-Hsien Lin
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| | - Yung-Chieh Tseng
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| | - Yang-Yu Cheng
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| | - Hsiu-Hua Ma
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| | - Ying-Chun Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan;
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| | - Chung-Yi Wu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| | - Che Ma
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115201, Taiwan; (K.-H.L.); (Y.-C.T.); (Y.-Y.C.); (H.-H.M.); (J.-T.J.); (C.-Y.W.); (C.M.)
| |
Collapse
|
16
|
Lin W, Li Q, Liu L, Wang Q, Zhang D, Wang F, Xu R, Fan Y, Xing M, Zhou C, Yuan Q. Early infiltrating NKT lymphocytes attenuate bone regeneration through secretion of CXCL2. SCIENCE ADVANCES 2024; 10:eadl6343. [PMID: 38758783 PMCID: PMC11100573 DOI: 10.1126/sciadv.adl6343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Trauma rapidly mobilizes the immune response of surrounding tissues and activates regeneration program. Manipulating immune response to promote tissue regeneration shows a broad application prospect. However, the understanding of bone healing dynamics at cellular level remains limited. Here, we characterize the landscape of immune cells after alveolar bone injury and reveal a pivotal role of infiltrating natural killer T (NKT) cells. We observe a rapid increase in NKT cells after injury, which inhibit osteogenic differentiation of mesenchymal stem cells (MSCs) and impair alveolar bone healing. Cxcl2 is up-regulated in NKT cells after injury. Systemic administration of CXCL2-neutralizing antibody or genetic deletion of Cxcl2 improves the bone healing process. In addition, we fabricate a gelatin-based porous hydrogel to deliver NK1.1 depletion antibody, which successfully promotes alveolar bone healing. In summary, our study highlights the importance of NKT cells in the early stage of bone healing and provides a potential therapeutic strategy for accelerating bone regeneration.
Collapse
Affiliation(s)
- Weimin Lin
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Qiwen Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Linfeng Liu
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Qian Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Feiyu Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Ruoshi Xu
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg R3T 2N2, Canada
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| |
Collapse
|
17
|
Steach HR, York AG, Skadow MH, Chen S, Zhao J, Williams KJ, Zhou Q, Hsieh WY, Brewer JR, Qu R, Shyer JA, Harman C, Sefik E, Mowell WK, Bailis W, Cui C, Kluger Y, Bensinger SJ, Craft J, Flavell RA. IL-4 Licenses B Cell Activation Through Cholesterol Synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593964. [PMID: 38798553 PMCID: PMC11118339 DOI: 10.1101/2024.05.13.593964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Lymphocyte activation involves a transition from quiescence and associated catabolic metabolism to a metabolic state with noted similarities to cancer cells such as heavy reliance on aerobic glycolysis for energy demands and increased nutrient requirements for biomass accumulation and cell division 1-3 . Following antigen receptor ligation, lymphocytes require spatiotemporally distinct "second signals". These include costimulatory receptor or cytokine signaling, which engage discrete programs that often involve remodeling of organelles and increased nutrient uptake or synthesis to meet changing biochemical demands 4-6 . One such signaling molecule, IL-4, is a highly pleiotropic cytokine that was first identified as a B cell co-mitogen over 30 years ago 7 . However, how IL-4 signaling mechanistically supports B cell proliferation is incompletely understood. Here, using single cell RNA sequencing we find that the cholesterol biosynthetic program is transcriptionally upregulated following IL-4 signaling during the early B cell response to influenza virus infection, and is required for B cell activation in vivo . By limiting lipid availability in vitro , we determine cholesterol to be essential for B cells to expand their endoplasmic reticulum, progress through cell cycle, and proliferate. In sum, we demonstrate that the well-known ability of IL-4 to act as a B cell growth factor is through a previously unknown rewiring of specific lipid anabolic programs, relieving sensitivity of cells to environmental nutrient availability.
Collapse
|
18
|
Cruz de Casas P, Knöpper K, Dey Sarkar R, Kastenmüller W. Same yet different - how lymph node heterogeneity affects immune responses. Nat Rev Immunol 2024; 24:358-374. [PMID: 38097778 DOI: 10.1038/s41577-023-00965-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 05/04/2024]
Abstract
Lymph nodes are secondary lymphoid organs in which immune responses of the adaptive immune system are initiated and regulated. Distributed throughout the body and embedded in the lymphatic system, local lymph nodes are continuously informed about the state of the organs owing to a constant drainage of lymph. The tissue-derived lymph carries products of cell metabolism, proteins, carbohydrates, lipids, pathogens and circulating immune cells. Notably, there is a growing body of evidence that individual lymph nodes differ from each other in their capacity to generate immune responses. Here, we review the structure and function of the lymphatic system and then focus on the factors that lead to functional heterogeneity among different lymph nodes. We will discuss how lymph node heterogeneity impacts on cellular and humoral immune responses and the implications for vaccination, tumour development and tumour control by immunotherapy.
Collapse
Affiliation(s)
- Paulina Cruz de Casas
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Konrad Knöpper
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Rupak Dey Sarkar
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Wolfgang Kastenmüller
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
19
|
Shehata L, Thouvenel CD, Hondowicz BD, Pew LA, Pritchard GH, Rawlings DJ, Choi J, Pepper M. Interleukin-4 downregulates transcription factor BCL6 to promote memory B cell selection in germinal centers. Immunity 2024; 57:843-858.e5. [PMID: 38513666 PMCID: PMC11104266 DOI: 10.1016/j.immuni.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/04/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
Germinal center (GC)-derived memory B cells (MBCs) are critical for humoral immunity as they differentiate into protective antibody-secreting cells during re-infection. GC formation and cellular interactions within the GC have been studied in detail, yet the exact signals that allow for the selection and exit of MBCs are not understood. Here, we showed that IL-4 cytokine signaling in GC B cells directly downregulated the transcription factor BCL6 via negative autoregulation to release cells from the GC program and to promote MBC formation. This selection event required additional survival cues and could therefore result in either GC exit or death. We demonstrate that both increasing IL-4 bioavailability or limiting IL-4 signaling disrupted MBC selection stringency. In this way, IL-4 control of BCL6 expression serves as a tunable switch within the GC to tightly regulate MBC selection and affinity maturation.
Collapse
Affiliation(s)
- Laila Shehata
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Christopher D Thouvenel
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Brian D Hondowicz
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Lucia A Pew
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | - David J Rawlings
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA; Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Jinyong Choi
- Department of Microbiology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
20
|
Kamii Y, Hayashizaki K, Kanno T, Chiba A, Ikegami T, Saito M, Akeda Y, Ohteki T, Kubo M, Yoshida K, Kawakami K, Oishi K, Araya J, Kuwano K, Kronenberg M, Endo Y, Kinjo Y. IL-27 regulates the differentiation of follicular helper NKT cells via metabolic adaptation of mitochondria. Proc Natl Acad Sci U S A 2024; 121:e2313964121. [PMID: 38394242 PMCID: PMC10907256 DOI: 10.1073/pnas.2313964121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/12/2024] [Indexed: 02/25/2024] Open
Abstract
Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that express an invariant T cell receptor α chain and contribute to bridging innate and acquired immunity with rapid production of large amounts of cytokines after stimulation. Among effecter subsets of iNKT cells, follicular helper NKT (NKTFH) cells are specialized to help B cells. However, the mechanisms of NKTFH cell differentiation remain to be elucidated. In this report, we studied the mechanism of NKTFH cell differentiation induced by pneumococcal surface protein A and α-galactosylceramide (P/A) vaccination. We found that Gr-1+ cells helped iNKT cell proliferation and NKTFH cell differentiation in the spleen by producing interleukin-27 (IL-27) in the early phase after vaccination. The neutralization of IL-27 impaired NKTFH cell differentiation, which resulted in compromised antibody production and diminished protection against Streptococcus pneumoniae infection by the P/A vaccine. Our data indicated that Gr-1+ cell-derived IL-27 stimulated mitochondrial metabolism, meeting the energic demand required for iNKT cells to differentiate into NKTFH cells. Interestingly, Gr-1+ cell-derived IL-27 was induced by iNKT cells via interferon-γ production. Collectively, our findings suggest that optimizing the metabolism of iNKT cells was essential for acquiring specific effector functions, and they provide beneficial knowledge on iNKT cell-mediated vaccination-mediated therapeutic strategies.
Collapse
Affiliation(s)
- Yasuhiro Kamii
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Koji Hayashizaki
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Toshio Kanno
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, Chiba292-0818, Japan
| | - Akio Chiba
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Taku Ikegami
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Yukihiro Akeda
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo162-8640, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University, Tokyo113-8510, Japan
| | - Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba278-0022, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi980-8575, Japan
| | | | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA92037
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA92093
| | - Yusuke Endo
- Department of Frontier Research and Development, Laboratory of Medical Omics Research, Kazusa DNA Research Institute, Chiba292-0818, Japan
| | - Yuki Kinjo
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| |
Collapse
|
21
|
Lee EG, Oh JE. From neglect to spotlight: the underappreciated role of B cells in cutaneous inflammatory diseases. Front Immunol 2024; 15:1328785. [PMID: 38426103 PMCID: PMC10902158 DOI: 10.3389/fimmu.2024.1328785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
The skin, covering our entire body as its largest organ, manifests enormous complexities and a profound interplay of systemic and local responses. In this heterogeneous domain, B cells were considered strangers. Yet, recent studies have highlighted their existence in the skin and their distinct role in modulating cutaneous immunity across various immune contexts. Accumulating evidence is progressively shedding light on the significance of B cells in maintaining skin health and in skin disorders. Herein, we integrate current insights on the systemic and local contributions of B cells in three prevalent inflammatory skin conditions: Pemphigus Vulgaris (PV), Systemic Lupus Erythematosus (SLE), and Atopic Dermatitis (AD), underscoring the previously underappreciated importance of B cells within skin immunity. Moreover, we address the potential adverse effects of current treatments used for skin diseases, emphasizing their unintentional consequences on B cells. These comprehensive approaches may pave the way for innovative therapeutic strategies that effectively address the intricate nature of skin disorders.
Collapse
Affiliation(s)
- Eun-Gang Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- BioMedical Research Center, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
22
|
Kwon DI, Park S, Jeong YL, Kim YM, Min J, Lee C, Choi JA, Choi YH, Kong HJ, Choi Y, Baek S, Lee KJ, Kang YW, Jeong C, You G, Oh Y, Im SK, Song M, Kim JK, Chang J, Choi D, Lee SW. Fc-fused IL-7 provides broad antiviral effects against respiratory virus infections through IL-17A-producing pulmonary innate-like T cells. Cell Rep Med 2024; 5:101362. [PMID: 38232693 PMCID: PMC10829794 DOI: 10.1016/j.xcrm.2023.101362] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/15/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024]
Abstract
Repeated pandemics caused by the influenza virus and severe acute respiratory syndrome coronavirus (SARS-CoV) have resulted in serious problems in global public health, emphasizing the need for broad-spectrum antiviral therapeutics against respiratory virus infections. Here, we show the protective effects of long-acting recombinant human interleukin-7 fused with hybrid Fc (rhIL-7-hyFc) against major respiratory viruses, including influenza virus, SARS-CoV-2, and respiratory syncytial virus. Administration of rhIL-7-hyFc in a therapeutic or prophylactic regimen induces substantial antiviral effects. During an influenza A virus (IAV) infection, rhIL-7-hyFc treatment increases pulmonary T cells composed of blood-derived interferon γ (IFNγ)+ conventional T cells and locally expanded IL-17A+ innate-like T cells. Single-cell RNA transcriptomics reveals that rhIL-7-hyFc upregulates antiviral genes in pulmonary T cells and induces clonal expansion of type 17 innate-like T cells. rhIL-7-hyFc-mediated disease prevention is dependent on IL-17A in both IAV- and SARS-CoV-2-infected mice. Collectively, we suggest that rhIL-7-hyFc can be used as a broadly active therapeutic for future respiratory virus pandemic.
Collapse
Affiliation(s)
- Dong-Il Kwon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Subin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Yujin L Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Young-Min Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jeongyong Min
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Changhyung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jung-Ah Choi
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Yoon Ha Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Hyun-Jung Kong
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngwon Choi
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seungtae Baek
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea
| | - Kun-Joo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Yeon-Woo Kang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Chaerim Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Gihoon You
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Youngsik Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Sun-Kyoung Im
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Donghoon Choi
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea.
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea.
| |
Collapse
|
23
|
Méndez Y, Vasco AV, Ebensen T, Schulze K, Yousefi M, Davari MD, Wessjohann LA, Guzmán CA, Rivera DG, Westermann B. Diversification of a Novel α-Galactosyl Ceramide Hotspot Boosts the Adjuvant Properties in Parenteral and Mucosal Vaccines. Angew Chem Int Ed Engl 2024; 63:e202310983. [PMID: 37857582 DOI: 10.1002/anie.202310983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023]
Abstract
The development of potent adjuvants is an important step for improving the performance of subunit vaccines. CD1d agonists, such as the prototypical α-galactosyl ceramide (α-GalCer), are of special interest due to their ability to activate iNKT cells and trigger rapid dendritic cell maturation and B-cell activation. Herein, we introduce a novel derivatization hotspot at the α-GalCer skeleton, namely the N-substituent at the amide bond. The multicomponent diversification of this previously unexplored glycolipid chemotype space permitted the introduction of a variety of extra functionalities that can either potentiate the adjuvant properties or serve as handles for further conjugation to antigens toward the development of self-adjuvanting vaccines. This strategy led to the discovery of compounds eliciting enhanced antigen-specific T cell stimulation and a higher antibody response when delivered by either the parenteral or the mucosal route, as compared to a known potent CD1d agonist. Notably, various functionalized α-GalCer analogues showed a more potent adjuvant effect after intranasal immunization than a PEGylated α-GalCer analogue previously optimized for this purpose. Ultimately, this work could open multiple avenues of opportunity for the use of mucosal vaccines against microbial infections.
Collapse
Affiliation(s)
- Yanira Méndez
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 6120 Halle, Saale), Germany
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata & G, Havana, 10400, Cuba
| | - Aldrin V Vasco
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 6120 Halle, Saale), Germany
| | - Thomas Ebensen
- Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Kai Schulze
- Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Mohammad Yousefi
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 6120 Halle, Saale), Germany
| | - Mehdi D Davari
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 6120 Halle, Saale), Germany
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 6120 Halle, Saale), Germany
| | - Carlos A Guzmán
- Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Daniel G Rivera
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 6120 Halle, Saale), Germany
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata & G, Havana, 10400, Cuba
- Finlay Institute of Vaccines, 200 and 21 Street, Havana, 11600, Cuba
| | - Bernhard Westermann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 6120 Halle, Saale), Germany
| |
Collapse
|
24
|
Hu X, Xian MY, Wang XF, Zou GQ, Luo R, Peng H, Liu Z. Conformationally Restricted Analogues of α-Galactosylceramide as Adjuvant in COVID-19 Subunit Vaccine. ACS Med Chem Lett 2023; 14:1647-1655. [PMID: 38116441 PMCID: PMC10726466 DOI: 10.1021/acsmedchemlett.3c00154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023] Open
Abstract
iNKT cells are a type of T lymphocyte that recognizes glycolipid antigens presented by CD1d protein. αGC is an agonistic glycolipid that activates iNKT cells and triggers immune modulatory cytokine responses, making it a promising vaccine adjuvant. To find more potent immunostimulating glycolipids, we prepared 4,6-O-galactosyl conformationally restricted analogues of αGC. Mice vaccinated with the SARS-CoV-2 RBD-Fc vaccine adjuvanted with these newly developed glycolipids produced robust anti-RBD antibody responses, comparable to those achieved with αGC. Importantly, we also found that omitting αGC, α-C-GalCer (Th1-type agonist), or C20:2 (Th2-type agonist) from the booster vaccine had negligible impact on antibody and cellular responses, potentially reducing the frequency of adjuvant use required to maintain potent immune responses.
Collapse
Affiliation(s)
- Xing Hu
- Key
Laboratory of Pesticide & Chemical Biology of Ministry of Education,
Hubei International Scientific and Technological Cooperation Base
of Pesticide and Green Synthesis, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Mao-Ying Xian
- Key
Laboratory of Pesticide & Chemical Biology of Ministry of Education,
Hubei International Scientific and Technological Cooperation Base
of Pesticide and Green Synthesis, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Xi-Feng Wang
- Key
Laboratory of Pesticide & Chemical Biology of Ministry of Education,
Hubei International Scientific and Technological Cooperation Base
of Pesticide and Green Synthesis, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Guo-Qing Zou
- Key
Laboratory of Pesticide & Chemical Biology of Ministry of Education,
Hubei International Scientific and Technological Cooperation Base
of Pesticide and Green Synthesis, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Rui Luo
- State
Key Laboratory of Agricultural Microbiology, College of Veterinary
Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P. R. China
| | - Hao Peng
- Key
Laboratory of Pesticide & Chemical Biology of Ministry of Education,
Hubei International Scientific and Technological Cooperation Base
of Pesticide and Green Synthesis, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| | - Zheng Liu
- Key
Laboratory of Pesticide & Chemical Biology of Ministry of Education,
Hubei International Scientific and Technological Cooperation Base
of Pesticide and Green Synthesis, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, P. R. China
| |
Collapse
|
25
|
Zhang H, Chen S, Zhang Y, Tian C, Pan J, Wang Y, Bai S, Wu Q, Su M, Xie D, Fu S, Li S, Zhang J, Chen Y, Zhu S, Qian Y, Bai L. Antigen Priming Induces Functional Reprogramming in iNKT Cells via Metabolic and Epigenetic Regulation: An Insight into iNKT Cell-Based Antitumor Immunotherapy. Cancer Immunol Res 2023; 11:1598-1610. [PMID: 37756568 DOI: 10.1158/2326-6066.cir-23-0448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/04/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Dysfunction of intratumoral invariant natural killer T (iNKT) cells hinders their antitumor efficacy, but the underlying mechanisms and the relationship with endogenous antigen priming remain to be explored. Here, we report that antigen priming leads to metabolic reprogramming and epigenetic remodeling, which causes functional reprogramming in iNKT cells, characterized by limited cytokine responses upon restimulation but constitutive high cytotoxicity. Mechanistically, impaired oxidative phosphorylation (OXPHOS) in antigen-primed iNKT cells inhibited T-cell receptor signaling, as well as elevation of glycolysis, upon restimulation via reducing mTORC1 activation, and thus led to impaired cytokine production. However, the metabolic reprogramming in antigen-primed iNKT cells was uncoupled with their enhanced cytotoxicity; instead, epigenetic remodeling explained their high expression of granzymes. Notably, intratumoral iNKT cells shared similar metabolic reprogramming and functional reprogramming with antigen-primed iNKT cells due to endogenous antigen priming in tumors, and thus recovery of OXPHOS in intratumoral iNKT cells by ZLN005 successfully enhanced their antitumor responses. Our study deciphers the influences of antigen priming-induced metabolic reprogramming and epigenetic remodeling on functionality of intratumoral iNKT cells, and proposes a way to enhance efficacy of iNKT cell-based antitumor immunotherapy by targeting cellular metabolism.
Collapse
Affiliation(s)
- Huimin Zhang
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sanwei Chen
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuwei Zhang
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chenxi Tian
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jun Pan
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu Wang
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shiyu Bai
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qielan Wu
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Miya Su
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Di Xie
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sicheng Fu
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuhang Li
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Zhang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Yusheng Chen
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Shasha Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
| | - Yeben Qian
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Bai
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, China
| |
Collapse
|
26
|
Shehata L, Thouvenel CD, Hondowicz BD, Pew LA, Rawlings DJ, Choi J, Pepper M. IL-4 downregulates BCL6 to promote memory B cell selection in germinal centers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525749. [PMID: 36747852 PMCID: PMC9900890 DOI: 10.1101/2023.01.26.525749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Germinal center (GC)-derived memory B cells (MBCs) are critical for humoral immunity as they differentiate into protective antibody-secreting cells during re-infection. GC formation and cellular interactions within the GC have been studied in detail, yet the exact signals that allow for the selection and exit of MBCs are not understood. Here, we show that IL-4 signaling in GC B cells directly downregulates BCL6 via negative autoregulation to release cells from the GC program and promote MBC formation. This selection event requires additional survival cues and can therefore result in either GC exit or death. We demonstrate that both increasing IL-4 bioavailability or limiting IL-4 signaling disrupt MBC selection stringency. In this way, IL-4 control of BCL6 expression serves as a tunable switch within the GC to tightly regulate MBC selection and affinity maturation.
Collapse
|
27
|
Tsuji M, Nair MS, Masuda K, Castagna C, Chong Z, Darling TL, Seehra K, Hwang Y, Ribeiro ÁL, Ferreira GM, Corredor L, Coelho-Dos-Reis JGA, Tsuji Y, Mori M, Boon ACM, Diamond MS, Huang Y, Ho DD. An immunostimulatory glycolipid that blocks SARS-CoV-2, RSV, and influenza infections in vivo. Nat Commun 2023; 14:3959. [PMID: 37402814 DOI: 10.1038/s41467-023-39738-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
Prophylactic vaccines for SARS-CoV-2 have lowered the incidence of severe COVID-19, but emergence of viral variants that are antigenically distinct from the vaccine strains are of concern and additional, broadly acting preventive approaches are desirable. Here, we report on a glycolipid termed 7DW8-5 that exploits the host innate immune system to enable rapid control of viral infections in vivo. This glycolipid binds to CD1d on antigen-presenting cells and thereby stimulates NKT cells to release a cascade of cytokines and chemokines. The intranasal administration of 7DW8-5 prior to virus exposure significantly blocked infection by three different authentic variants of SARS-CoV-2, as well as by respiratory syncytial virus and influenza virus, in mice or hamsters. We also found that this protective antiviral effect is both host-directed and mechanism-specific, requiring both the CD1d molecule and interferon-[Formula: see text]. A chemical compound like 7DW8-5 that is easy to administer and cheap to manufacture may be useful not only in slowing the spread of COVID-19 but also in responding to future pandemics long before vaccines or drugs are developed.
Collapse
Affiliation(s)
- Moriya Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Manoj S Nair
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Kazuya Masuda
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Candace Castagna
- Institute of Comparative Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zhenlu Chong
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tamarand L Darling
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kuljeet Seehra
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Youngmin Hwang
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Ágata Lopes Ribeiro
- Basic and Applied Virology Laboratory, Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geovane Marques Ferreira
- Basic and Applied Virology Laboratory, Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laura Corredor
- Institute of Comparative Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Yukiko Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Munemasa Mori
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
| |
Collapse
|
28
|
Zhang X, Zhou Z. The Mechanism of bnAb Production and Its Application in Mutable Virus Broad-Spectrum Vaccines: Inspiration from HIV-1 Broad Neutralization Research. Vaccines (Basel) 2023; 11:1143. [PMID: 37514959 PMCID: PMC10384589 DOI: 10.3390/vaccines11071143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Elite controllers among HIV-1-infected individuals have demonstrated a stronger ability to control the viral load in their bodies. Scientists have isolated antibodies with strong neutralizing ability from these individuals, which can neutralize HIV-1 variations; these are known as broadly neutralizing antibodies. The nucleic acid of some viruses will constantly mutate during replication (such as SARS-CoV-2), which will reduce the protective ability of the corresponding vaccines. The immune escape caused by this mutation is the most severe challenge faced by humans in the battle against the virus. Therefore, developing broad-spectrum vaccines that can induce broadly neutralizing antibodies against various viruses and their mutated strains is the best way to combat virus mutations. Exploring the mechanism by which the human immune system produces broadly neutralizing antibodies and its induction strategies is crucial in the design process of broad-spectrum vaccines.
Collapse
Affiliation(s)
- Xinyu Zhang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, No. 31 Huatuo Street, Daxing District, Beijing 102629, China
- College of Life Science, Jilin University, Changchun 130012, China
| | - Zehua Zhou
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
29
|
Chakma CR, Good-Jacobson KL. Requirements of IL-4 during the Generation of B Cell Memory. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1853-1860. [PMID: 37276051 DOI: 10.4049/jimmunol.2200922] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/02/2023] [Indexed: 06/07/2023]
Abstract
IL-4 has long been established as a key regulator of Th cells and for promoting effective B cell survival and isotype class switching. Yet, despite having been extensively studied, the specific role of IL-4 in generating humoral memory in vivo is unclear. In this review, we explore the recent studies that unravel the cellular sources and spatiotemporal production of IL-4, the relationship between IL-4 and IL-21 during germinal center responses and the formation of Ab-secreting cells, and the current understanding of whether IL-4 promotes or suppresses memory B cell generation in vitro and in vivo.
Collapse
Affiliation(s)
- Clarissa R Chakma
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia; and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kim L Good-Jacobson
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia; and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
30
|
Crouse B, Baehr C, Hicks D, Pravetoni M. IL-4 Predicts the Efficacy of a Candidate Antioxycodone Vaccine and Alters Vaccine-Specific Antibody-Secreting Cell Proliferation in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1272-1280. [PMID: 36939374 PMCID: PMC11321710 DOI: 10.4049/jimmunol.2200605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/27/2023] [Indexed: 03/21/2023]
Abstract
Opioid use disorders (OUDs) are a public health concern in the United States and worldwide. Current medications for OUDs may trigger side effects and are often heavily regulated. A novel treatment strategy to be used alone or in combination with existing medications is active immunization with antiopioid vaccines, which stimulate production of opioid-specific Abs that bind to the target drug and prevent its distribution to the brain. Although antiopioid vaccines have shown promising preclinical efficacy, prior clinical evaluations of vaccines targeting stimulants indicate that efficacy is limited to a subset of subjects who achieve optimal Ab responses. We have previously reported that depletion of IL-4 with a mAb increased opioid-specific IgG2a and total IgG, and it increased the number of germinal centers and germinal center T follicular helper cells in response to antiopioid vaccines via type I IL-4 signaling. The current study further investigates the mechanisms associated with IL-4-mediated increases in efficacy and whether IL-4 depletion affects specific processes involved in germinal center formation, including affinity maturation, class switching, and plasma cell differentiation in mice. Additionally, results demonstrate that preimmunization production of IL-4 after ex vivo whole blood stimulation predicted in vivo vaccine-induced Ab titers in outbred mice. Such mechanistic studies are critical for rational design of next-generation vaccine formulations, and they support the use of IL-4 as a predictive biomarker in ongoing OUD vaccine clinical studies.
Collapse
Affiliation(s)
- Bethany Crouse
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN
- School of Veterinary Population Medicine, University of Minnesota, St. Paul, MN
| | - Carly Baehr
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN
| | - Dustin Hicks
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
31
|
Alves RPDS, Wang YT, Mikulski Z, McArdle S, Shafee N, Valentine KM, Miller R, Verma SK, Batiz FAS, Maule E, Nguyen MN, Timis J, Mann C, Zandonatti M, Alarcon S, Rowe J, Kronenberg M, Weiskopf D, Sette A, Hastie K, Saphire EO, Festin S, Kim K, Shresta S. SARS-CoV-2 Omicron (B.1.1.529) shows minimal neurotropism in a double-humanized mouse model. Antiviral Res 2023; 212:105580. [PMID: 36940916 PMCID: PMC10027296 DOI: 10.1016/j.antiviral.2023.105580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/08/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023]
Abstract
Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) initially infects the respiratory tract, it also directly or indirectly affects other organs, including the brain. However, little is known about the relative neurotropism of SARS-CoV-2 variants of concern (VOCs), including Omicron (B.1.1.529), which emerged in November 2021 and has remained the dominant pathogenic lineage since then. To address this gap, we examined the relative ability of Omicron, Beta (B.1.351), and Delta (B.1.617.2) to infect the brain in the context of a functional human immune system by using human angiotensin-converting enzyme 2 (hACE2) knock-in triple-immunodeficient NGC mice with or without reconstitution with human CD34+ stem cells. Intranasal inoculation of huCD34+-hACE2-NCG mice with Beta and Delta resulted in productive infection of the nasal cavity, lungs, and brain on day 3 post-infection, but Omicron was surprisingly unique in its failure to infect either the nasal tissue or brain. Moreover, the same infection pattern was observed in hACE2-NCG mice, indicating that antiviral immunity was not responsible for the lack of Omicron neurotropism. In independent experiments, we demonstrate that nasal inoculation with Beta or with D614G, an ancestral SARS-CoV-2 with undetectable replication in huCD34+-hACE2-NCG mice, resulted in a robust response by human innate immune cells, T cells, and B cells, confirming that exposure to SARS-CoV-2, even without detectable infection, is sufficient to induce an antiviral immune response. Collectively, these results suggest that modeling of the neurologic and immunologic sequelae of SARS-CoV-2 infection requires careful selection of the appropriate SARS-CoV-2 strain in the context of a specific mouse model.
Collapse
Affiliation(s)
| | - Ying-Ting Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Zbigniew Mikulski
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Sara McArdle
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Norazizah Shafee
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kristen M Valentine
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Robyn Miller
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Shailendra Kumar Verma
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Fernanda Ana Sosa Batiz
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Erin Maule
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Michael N Nguyen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Julia Timis
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Colin Mann
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Michelle Zandonatti
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Suzie Alarcon
- Sequencing Core Facility, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jenny Rowe
- Charles River Laboratories Research Models and Services Inc., Wilmington, MA, USA
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Kathryn Hastie
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Stephen Festin
- Charles River Laboratories Research Models and Services Inc., Wilmington, MA, USA
| | - Kenneth Kim
- Histopathology Core Facility, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | - Sujan Shresta
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA.
| |
Collapse
|
32
|
Wang WB, Lin YD, Zhao L, Liao C, Zhang Y, Davila M, Sun J, Chen Y, Xiong N. Developmentally programmed early-age skin localization of iNKT cells supports local tissue development and homeostasis. Nat Immunol 2023; 24:225-238. [PMID: 36624165 DOI: 10.1038/s41590-022-01399-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 12/01/2022] [Indexed: 01/11/2023]
Abstract
Skin is exposed to various environmental assaults and undergoes morphological changes immediately after birth. Proper localization and function of immune cells in the skin is crucial for protection and establishment of skin tissue homeostasis. Here we report the discovery of a developmentally programmed process that directs preferential localization of invariant natural killer T (iNKT) cells to the skin for early local homeostatic regulation. We show that iNKT cells are programmed predominantly with a CCR10+ skin-homing phenotype during thymic development in infant and young mice. Early skin localization of iNKT cells is critical for proper commensal bacterial colonization and tissue development. Mechanistically, skin iNKT cells provide a local source of transferrin that regulates iron metabolism in hair follicle progenitor cells and helps hair follicle development. These findings provide molecular insights into the establishment and physiological functions of iNKT cells in the skin during early life.
Collapse
Affiliation(s)
- Wei-Bei Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Yang-Ding Lin
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Luming Zhao
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Chang Liao
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Yang Zhang
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Micha Davila
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Jasmine Sun
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Yidong Chen
- Department of Population Health Sciences, and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Na Xiong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
- Department of Medicine-Division of Dermatology and Cutaneous Surgery, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
33
|
Soliman H, Hogue D, Han H, Mooney B, Costa R, Lee MC, Niell B, Williams A, Chau A, Falcon S, Soyano A, Armaghani A, Khakpour N, Weinfurtner RJ, Hoover S, Kiluk J, Laronga C, Rosa M, Khong H, Czerniecki B. Oncolytic T-VEC virotherapy plus neoadjuvant chemotherapy in nonmetastatic triple-negative breast cancer: a phase 2 trial. Nat Med 2023; 29:450-457. [PMID: 36759673 DOI: 10.1038/s41591-023-02210-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 01/06/2023] [Indexed: 02/11/2023]
Abstract
Talimogene laherparepvec (T-VEC) is an oncolytic virus hypothesized to enhance triple-negative breast cancer (TNBC) responses to neoadjuvant chemotherapy (NAC). This article describes the phase 2 trial of T-VEC plus NAC (ClinicalTrials.gov ID: NCT02779855 ). Patients with stage 2-3 TNBC received five intratumoral T-VEC injections with paclitaxel followed by doxorubicin and cyclophosphamide and surgery to assess residual cancer burden index (RCB). The primary end point was RCB0 rate. Secondary end points were RCB0-1 rate, recurrence rate, toxicity and immune correlates. Thirty-seven patients were evaluated. Common T-VEC toxicities were fevers, chills, headache, fatigue and injection site pain. NAC toxicities were as expected. Four thromboembolic events occurred. The primary end point was met with an estimated RCB0 rate = 45.9% and RCB0-1 descriptive rate = 65%. The 2-year disease-free rate is equal to 89% with no recurrences in RCB0-1 patients. Immune activation during treatment correlated with response. T-VEC plus NAC in TNBC may increase RCB0-1 rates. These results support continued investigation of T-VEC plus NAC for TNBC.
Collapse
Affiliation(s)
- Hatem Soliman
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA.
| | - Deanna Hogue
- Clinical Trials Office, Moffitt Cancer Center, Tampa, FL, USA
| | - Hyo Han
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Blaise Mooney
- Department of Radiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ricardo Costa
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Marie C Lee
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Bethany Niell
- Department of Radiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Angela Williams
- Department of Radiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Alec Chau
- Department of Radiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Shannon Falcon
- Department of Radiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Aixa Soyano
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Avan Armaghani
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Nazanin Khakpour
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Susan Hoover
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - John Kiluk
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Christine Laronga
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Marilin Rosa
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
| | - Hung Khong
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Brian Czerniecki
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
34
|
Gao X, Luo K, Wang D, Wei Y, Yao Y, Deng J, Yang Y, Zeng Q, Dong X, Xiong L, Gong D, Lin L, Pohl K, Liu S, Liu Y, Liu L, Nguyen THO, Allen LF, Kedzierska K, Jin Y, Du MR, Chen W, Lu L, Shen N, Liu Z, Cockburn IA, Luo W, Yu D. T follicular helper 17 (Tfh17) cells are superior for immunological memory maintenance. eLife 2023; 12:82217. [PMID: 36655976 PMCID: PMC9891720 DOI: 10.7554/elife.82217] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
A defining feature of successful vaccination is the ability to induce long-lived antigen-specific memory cells. T follicular helper (Tfh) cells specialize in providing help to B cells in mounting protective humoral immunity in infection and after vaccination. Memory Tfh cells that retain the CXCR5 expression can confer protection through enhancing humoral response upon antigen re-exposure but how they are maintained is poorly understood. CXCR5+ memory Tfh cells in human blood are divided into Tfh1, Tfh2, and Tfh17 cells by the expression of chemokine receptors CXCR3 and CCR6 associated with Th1 and Th17, respectively. Here, we developed a new method to induce Tfh1, Tfh2, and Tfh17-like (iTfh1, iTfh2, and iTfh17) mouse cells in vitro. Although all three iTfh subsets efficiently support antibody responses in recipient mice with immediate immunization, iTfh17 cells are superior to iTfh1 and iTfh2 cells in supporting antibody response to a later immunization after extended resting in vivo to mimic memory maintenance. Notably, the counterpart human Tfh17 cells are selectively enriched in CCR7+ central memory Tfh cells with survival and proliferative advantages. Furthermore, the analysis of multiple human cohorts that received different vaccines for HBV, influenza virus, tetanus toxin or measles revealed that vaccine-specific Tfh17 cells outcompete Tfh1 or Tfh2 cells for the persistence in memory phase. Therefore, the complementary mouse and human results showing the advantage of Tfh17 cells in maintenance and memory function supports the notion that Tfh17-induced immunization might be preferable in vaccine development to confer long-term protection.
Collapse
Affiliation(s)
- Xin Gao
- Immunology and Infectious Disease Division, John Curtin School of Medical Research, The Australian National UniversityCanberraAustralia
- China-Australia Centre for Personalised Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Kaiming Luo
- China-Australia Centre for Personalised Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Diya Wang
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical UniversityXi'anChina
| | - Yunbo Wei
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology, Shandong Academy of SciencesJinanChina
| | - Yin Yao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jun Deng
- China-Australia Centre for Personalised Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yang Yang
- Frazer Institute, Faculty of Medicine, University of QueenslandBrisbaneAustralia
| | - Qunxiong Zeng
- China-Australia Centre for Personalised Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoru Dong
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical UniversityXi'anChina
| | - Le Xiong
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Dongcheng Gong
- China-Australia Centre for Personalised Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Lin Lin
- Department of Laboratory Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Kai Pohl
- Immunology and Infectious Disease Division, John Curtin School of Medical Research, The Australian National UniversityCanberraAustralia
| | - Shaoling Liu
- Shanghai Children's Medical Centre, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yu Liu
- Shanghai Children's Medical Centre, Shanghai Jiao Tong UniversityShanghaiChina
| | - Lu Liu
- Obstetrics and Gynecology Hospital of Fudan University (Shanghai Red House Obstetrics and Gynecology Hospital)ShanghaiChina
| | - Thi HO Nguyen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Lilith F Allen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Yanliang Jin
- Shanghai Children's Medical Centre, Shanghai Jiao Tong UniversityShanghaiChina
| | - Mei-Rong Du
- Obstetrics and Gynecology Hospital of Fudan University (Shanghai Red House Obstetrics and Gynecology Hospital)ShanghaiChina
| | - Wanping Chen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical UniversityXi'anChina
| | - Liangjing Lu
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Nan Shen
- China-Australia Centre for Personalised Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Department of Rheumatology, Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Ian A Cockburn
- Immunology and Infectious Disease Division, John Curtin School of Medical Research, The Australian National UniversityCanberraAustralia
| | - Wenjing Luo
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical UniversityXi'anChina
| | - Di Yu
- Immunology and Infectious Disease Division, John Curtin School of Medical Research, The Australian National UniversityCanberraAustralia
- Frazer Institute, Faculty of Medicine, University of QueenslandBrisbaneAustralia
- Ian Frazer Centre for Children’s Immunotherapy Research, Children’s Health Research Centre, Faculty of Medicine, University of QueenslandBrisbaneAustralia
| |
Collapse
|
35
|
Avraham R, Melamed S, Achdout H, Erez N, Israeli O, Barlev-Gross M, Pasmanik-Chor M, Paran N, Israely T, Vitner EB. Antiviral activity of glucosylceramide synthase inhibitors in alphavirus infection of the central nervous system. Brain Commun 2023; 5:fcad086. [PMID: 37168733 PMCID: PMC10165247 DOI: 10.1093/braincomms/fcad086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 12/23/2022] [Accepted: 03/23/2023] [Indexed: 05/13/2023] Open
Abstract
Virus-induced CNS diseases impose a considerable human health burden worldwide. For many viral CNS infections, neither antiviral drugs nor vaccines are available. In this study, we examined whether the synthesis of glycosphingolipids, major membrane lipid constituents, could be used to establish an antiviral therapeutic target. We found that neuroinvasive Sindbis virus altered the sphingolipid levels early after infection in vitro and increased the levels of gangliosides GA1 and GM1 in the sera of infected mice. The alteration in the sphingolipid levels appears to play a role in neuroinvasive Sindbis virus replication, as treating infected cells with UDP-glucose ceramide glucosyltransferase (UGCG) inhibitors reduced the replication rate. Moreover, the UGCG inhibitor GZ-161 increased the survival rates of Sindbis-infected mice, most likely by reducing the detrimental immune response activated by sphingolipids in the brains of Sindbis virus-infected mice. These findings suggest a role for glycosphingolipids in the host immune response against neuroinvasive Sindbis virus and suggest that UGCG inhibitors should be further examined as antiviral therapeutics for viral infections of the CNS.
Collapse
Affiliation(s)
- Roy Avraham
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Hagit Achdout
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Noam Erez
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Ofir Israeli
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Moria Barlev-Gross
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Science, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Einat B Vitner
- Correspondence to: Einat B. Vitner Department of Infectious Diseases Israel Institute for Biological Research P.O.B 19, 7410001 Ness-Ziona, Israel E-mail:
| |
Collapse
|
36
|
Brailey PM, Evans L, López-Rodríguez JC, Sinadinos A, Tyrrel V, Kelly G, O'Donnell V, Ghazal P, John S, Barral P. CD1d-dependent rewiring of lipid metabolism in macrophages regulates innate immune responses. Nat Commun 2022; 13:6723. [PMID: 36344546 PMCID: PMC9640663 DOI: 10.1038/s41467-022-34532-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Alterations in cellular metabolism underpin macrophage activation, yet little is known regarding how key immunological molecules regulate metabolic programs in macrophages. Here we uncover a function for the antigen presenting molecule CD1d in the control of lipid metabolism. We show that CD1d-deficient macrophages exhibit a metabolic reprogramming, with a downregulation of lipid metabolic pathways and an increase in exogenous lipid import. This metabolic rewiring primes macrophages for enhanced responses to innate signals, as CD1d-KO cells show higher signalling and cytokine secretion upon Toll-like receptor stimulation. Mechanistically, CD1d modulates lipid import by controlling the internalization of the lipid transporter CD36, while blocking lipid uptake through CD36 restores metabolic and immune responses in macrophages. Thus, our data reveal CD1d as a key regulator of an inflammatory-metabolic circuit in macrophages, independent of its function in the control of T cell responses.
Collapse
Affiliation(s)
- Phillip M Brailey
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
- The Francis Crick Institute, London, UK
| | - Lauren Evans
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
- The Francis Crick Institute, London, UK
| | - Juan Carlos López-Rodríguez
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
- The Francis Crick Institute, London, UK
| | - Anthony Sinadinos
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
- The Francis Crick Institute, London, UK
| | | | | | | | - Peter Ghazal
- School of Medicine, Cardiff University, Cardiff, UK
| | - Susan John
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology, King's College London, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
37
|
Li YQ, Yan C, Wang XF, Xian MY, Zou GQ, Gao XF, Luo R, Liu Z. A New iNKT-Cell Agonist-Adjuvanted SARS-CoV-2 Subunit Vaccine Elicits Robust Neutralizing Antibody Responses. ACS Infect Dis 2022; 8:2161-2170. [PMID: 36043698 DOI: 10.1021/acsinfecdis.2c00296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Adjuvants are essential components of vaccines. Invariant natural killer T (iNKT) cells are a distinct subset of T cells that function to bridge the innate and adaptive immunities and are capable of mediating strong and rapid responses to a range of diseases, including cancer and infectious disease. An increasing amount of evidence suggests that iNKT cells can help fight viral infection. In particular, iNKT-secreting IL-4 is a key mediator of humoral immunity and has a positive correlation with the levels of neutralizing antibodies. As iNKT cell agonists, αGC glycolipid (α-galactosylceramide, or KRN7000) and its analogues as vaccine adjuvants have begun to provide vaccinologists with a new toolset. Herein we found that a new iNKT-cell agonist αGC-CPOEt elicited a strong cytokine response with increased IL-4 production. Remarkably, after three immunizations, SARS-CoV-2 RBD-Fc adjuvanted by αGC-CPOEt evoked robust neutralizing antibody responses that were about 5.5-fold more than those induced by αGC/RBD-Fc and 25-fold greater than those induced by unadjuvanted RBD-Fc. These findings imply that αGC-CPOEt could be investigated further as a new COVID-19 vaccine adjuvant to prevent current and future infectious disease outbreaks.
Collapse
Affiliation(s)
- Ya-Qian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Cheng Yan
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xi-Feng Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Mao-Ying Xian
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Guo-Qing Zou
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| | - Xiao-Fei Gao
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, East China University of Technology, Nanchang, Jiangxi 330013, PR China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Zheng Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, PR China
| |
Collapse
|
38
|
Ataide MA, Knöpper K, Cruz de Casas P, Ugur M, Eickhoff S, Zou M, Shaikh H, Trivedi A, Grafen A, Yang T, Prinz I, Ohlsen K, Gomez de Agüero M, Beilhack A, Huehn J, Gaya M, Saliba AE, Gasteiger G, Kastenmüller W. Lymphatic migration of unconventional T cells promotes site-specific immunity in distinct lymph nodes. Immunity 2022; 55:1813-1828.e9. [PMID: 36002023 DOI: 10.1016/j.immuni.2022.07.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/06/2022] [Accepted: 07/27/2022] [Indexed: 12/31/2022]
Abstract
Lymphatic transport of molecules and migration of myeloid cells to lymph nodes (LNs) continuously inform lymphocytes on changes in drained tissues. Here, using LN transplantation, single-cell RNA-seq, spectral flow cytometry, and a transgenic mouse model for photolabeling, we showed that tissue-derived unconventional T cells (UTCs) migrate via the lymphatic route to locally draining LNs. As each tissue harbored a distinct spectrum of UTCs with locally adapted differentiation states and distinct T cell receptor repertoires, every draining LN was thus populated by a distinctive tissue-determined mix of these lymphocytes. By making use of single UTC lineage-deficient mouse models, we found that UTCs functionally cooperated in interconnected units and generated and shaped characteristic innate and adaptive immune responses that differed between LNs that drained distinct tissues. Lymphatic migration of UTCs is, therefore, a key determinant of site-specific immunity initiated in distinct LNs with potential implications for vaccination strategies and immunotherapeutic approaches.
Collapse
Affiliation(s)
- Marco A Ataide
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany.
| | - Konrad Knöpper
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Paulina Cruz de Casas
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Milas Ugur
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Sarah Eickhoff
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Haroon Shaikh
- Department of Medicine II and Pediatrics, Würzburg University Hospital, ZEMM, 97078 Würzburg, Germany
| | - Apurwa Trivedi
- Centre d'Immunologie de Marseille-Luminy (CIML), Department of Immunology, 13288 Marseille, France
| | - Anika Grafen
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Tao Yang
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Immo Prinz
- Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Knut Ohlsen
- Institute for Molecular Infection Biology (IMIB), 97078 Würzburg, Germany
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Andreas Beilhack
- Department of Medicine II and Pediatrics, Würzburg University Hospital, ZEMM, 97078 Würzburg, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Mauro Gaya
- Centre d'Immunologie de Marseille-Luminy (CIML), Department of Immunology, 13288 Marseille, France
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97078 Würzburg, Germany
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Wolfgang Kastenmüller
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany.
| |
Collapse
|
39
|
Lin Q, Kuypers M, Liu Z, Copeland JK, Chan D, Robertson SJ, Kontogiannis J, Guttman DS, Banks EK, Philpott DJ, Mallevaey T. Invariant natural killer T cells minimally influence gut microbiota composition in mice. Gut Microbes 2022; 14:2104087. [PMID: 35912530 PMCID: PMC9348128 DOI: 10.1080/19490976.2022.2104087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Invariant Natural Killer T (iNKT) cells are unconventional T cells that respond to glycolipid antigens found in microbes in a CD1d-dependent manner. iNKT cells exert innate-like functions and produce copious amounts of cytokines, chemokines and cytotoxic molecules within only minutes of activation. As such, iNKT cells can fuel or dampen inflammation in a context-dependent manner. In addition, iNKT cells provide potent immunity against bacteria, viruses, parasites and fungi. Although microbiota-iNKT cell interactions are not well-characterized, mounting evidence suggests that microbiota colonization early in life impacts iNKT cell homeostasis and functions in disease. In this study, we showed that CD1d-/- and Vα14 Tg mice, which lack and have increased numbers of iNKT cells, respectively, had no significant alterations in gut microbiota composition compared to their littermate controls. Furthermore, specific iNKT cell activation by glycolipid antigens only resulted in a transient and minimal shift in microbiota composition when compared to the natural drift found in our colony. Our findings demonstrate that iNKT cells have little to no influence in regulating commensal bacteria at steady state.Abbreviations: iNKT: invariant Natural Killer T cell; αGC: α-galactosylceramide.
Collapse
Affiliation(s)
- Qiaochu Lin
- Department of Immunology, University of Toronto, Toronto, ON, Canada,CONTACT Thierry Mallevaey University of Toronto, Department of Immunology, Medical Sciences Building, Room 7334,1 King’s College Circle, Toronto, OntarioM5S 1A8, Canada
| | - Meggie Kuypers
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Zhewei Liu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Donny Chan
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Susan J Robertson
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Jean Kontogiannis
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - E. Kate Banks
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, ON, Canada,Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Loureiro JP, Cruz MS, Cardoso AP, Oliveira MJ, Macedo MF. Human iNKT Cells Modulate Macrophage Survival and Phenotype. Biomedicines 2022; 10:1723. [PMID: 35885028 PMCID: PMC9313099 DOI: 10.3390/biomedicines10071723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
CD1d-restricted invariant Natural Killer T (iNKT) cells are unconventional innate-like T cells whose functions highly depend on the interactions they establish with other immune cells. Although extensive studies have been reported on the communication between iNKT cells and macrophages in mice, less data is available regarding the relevance of this crosstalk in humans. Here, we dove into the human macrophage-iNKT cell axis by exploring how iNKT cells impact the survival and polarization of pro-inflammatory M1-like and anti-inflammatory M2-like monocyte-derived macrophages. By performing in vitro iNKT cell-macrophage co-cultures followed by flow cytometry analysis, we demonstrated that antigen-stimulated iNKT cells induce a generalized activated state on all macrophage subsets, leading to upregulation of CD40 and CD86 expression. CD40L blocking with a specific monoclonal antibody prior to co-cultures abrogated CD40 and CD86 upregulation, thus indicating that iNKT cells required CD40-CD40L co-stimulation to trigger macrophage activation. In addition, activated iNKT cells were cytotoxic towards macrophages in a CD1d-dependent manner, killing M1-like macrophages more efficiently than their naïve M0 or anti-inflammatory M2-like counterparts. Hence, this work highlighted the role of human iNKT cells as modulators of macrophage survival and phenotype, untangling key features of the human macrophage-iNKT cell axis and opening perspectives for future therapeutic modulation.
Collapse
Affiliation(s)
- J. Pedro Loureiro
- Cell Activation and Gene Expression Group, Institute for Molecular and Cell Biology (IBMC), Institute for Research and Innovation in Health (i3S), University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (J.P.L.); (M.S.C.)
- Experimental Immunology Group, Department of Biomedicine (DBM), University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Mariana S. Cruz
- Cell Activation and Gene Expression Group, Institute for Molecular and Cell Biology (IBMC), Institute for Research and Innovation in Health (i3S), University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (J.P.L.); (M.S.C.)
- Department of Medical Sciences, University of Aveiro (UA), 3810-193 Aveiro, Portugal
| | - Ana P. Cardoso
- Tumour and Microenvironment Interactions Group, Institute of Biomedical Engineering (INEB), Institute for Research and Innovation in Health (i3S), University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.C.); (M.J.O.)
| | - Maria J. Oliveira
- Tumour and Microenvironment Interactions Group, Institute of Biomedical Engineering (INEB), Institute for Research and Innovation in Health (i3S), University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.P.C.); (M.J.O.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - M. Fátima Macedo
- Cell Activation and Gene Expression Group, Institute for Molecular and Cell Biology (IBMC), Institute for Research and Innovation in Health (i3S), University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (J.P.L.); (M.S.C.)
- Department of Medical Sciences, University of Aveiro (UA), 3810-193 Aveiro, Portugal
| |
Collapse
|
41
|
Porcine reproductive and respiratory syndrome virus reinfection causes the distribution of porcine interleukin-4 in close proximity to B lymphocytes within lymphoid follicles and a reduction in B and T lymphocytes. Vet Microbiol 2022; 272:109498. [PMID: 35793585 DOI: 10.1016/j.vetmic.2022.109498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/15/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022]
Abstract
Interleukin 4 (IL-4) plays a major role in T-lymphocyte development and is thought to be a central regulator as a cofactor in resting B-lymphocyte proliferation. Primary infection with porcine reproductive and respiratory syndrome virus (PRRSV) induces minimal IL-4 production, whereas an IL-4 response occurs in the peripheral blood of piglets reinfected by PRRSV. The locations and interaction partners for the massive volume of IL-4 triggered by PRRSV reinfection remain unclear. This study aimed to investigate the characteristics of IL-4 secretion and location changes in peripheral immune organs induced by PRRSV infection and reinfection. Our results show that PRRSV reinfection induced higher levels of IL-4 mRNA and protein expression in the peripheral immune organs (e.g., lymph node and spleen) and peripheral blood compared with PRRSV primary infection. Importantly, we found that, following PRRSV reinfection, an obvious large-scale migration of IL-4 occurred in the lymph nodes. During PRRSV primary infection, IL-4 was mainly concentrated around the lymphoid follicles and paracortical regions of the lymph node and also located in the marginal area and periarterial lymphatic sheath region of the spleen. During PRRSV reinfection, the now abundant IL-4 gathered into the lymphoid follicles of the lymph node and spleen. Notably, IL-4 changed its location state from scattered and sparse during primary infection to clinging to B lymphocytes in the lymphoid follicles during reinfection. During reinfection, IL-4 was often co-localized with T and B lymphocytes; furthermore, the percentages of several T lymphocyte subsets, N protein-specific antibody levels, and viral load in the peripheral blood or lymph tissues underwent remarkable variation. Another important finding of this study was that the numbers of B lymphocytes and T lymphocytes in the lymphoid nodes were significantly reduced after PRRSV infection or reinfection, presumably due to PRRSV-induced acute bone marrow failure and autophagy in thymic epithelial cells. This study revealed the characteristics of IL-4 migration and distribution in the peripheral lymph organs induced by PRRSV reinfection and provides valuable clues for further exploration of the interactions between IL-4, B lymphocytes, and T lymphocytes during PRRSV infection and reinfection.
Collapse
|
42
|
Serrán MG, Vernengo FF, Almada L, Beccaria CG, Gazzoni Y, Canete PF, Roco JA, Boari JT, Ramello MC, Wehrens E, Cai Y, Zuniga EI, Montes CL, Cockburn IA, Rodriguez EVA, Vinuesa CG, Gruppi A. Extrafollicular Plasmablasts Present in the Acute Phase of Infections Express High Levels of PD-L1 and Are Able to Limit T Cell Response. Front Immunol 2022; 13:828734. [PMID: 35651611 PMCID: PMC9149371 DOI: 10.3389/fimmu.2022.828734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/11/2022] [Indexed: 11/20/2022] Open
Abstract
During infections with protozoan parasites or some viruses, T cell immunosuppression is generated simultaneously with a high B cell activation. It has been described that, as well as producing antibodies, plasmablasts, the differentiation product of activated B cells, can condition the development of protective immunity in infections. Here, we show that, in T. cruzi infection, all the plasmablasts detected during the acute phase of the infection had higher surface expression of PD-L1 than other mononuclear cells. PD-L1hi plasmablasts were induced in vivo in a BCR-specific manner and required help from Bcl-6+CD4+T cells. PD-L1hi expression was not a characteristic of all antibody-secreting cells since plasma cells found during the chronic phase of infection expressed PD-L1 but at lower levels. PD-L1hi plasmablasts were also present in mice infected with Plasmodium or with lymphocytic choriomeningitis virus, but not in mice with autoimmune disorders or immunized with T cell-dependent antigens. In vitro experiments showed that PD-L1hi plasmablasts suppressed the T cell response, partially via PD-L1. Thus, this study reveals that extrafollicular PD-L1hi plasmablasts, whose peaks of response precede the peak of germinal center response, may have a modulatory function in infections, thus influencing T cell response.
Collapse
Affiliation(s)
- Melisa Gorosito Serrán
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Facundo Fiocca Vernengo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Laura Almada
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cristian G Beccaria
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Yamila Gazzoni
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Pablo F Canete
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jonathan A Roco
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jimena Tosello Boari
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Cecilia Ramello
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ellen Wehrens
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| | - Yeping Cai
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Elina I Zuniga
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| | - Carolina L Montes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Eva V Acosta Rodriguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Carola G Vinuesa
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,China-Australia Centre for Personalised Immunology, Shanghai Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
43
|
Baranek T, de Amat Herbozo C, Mallevaey T, Paget C. Deconstructing iNKT cell development at single-cell resolution. Trends Immunol 2022; 43:503-512. [PMID: 35654639 DOI: 10.1016/j.it.2022.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 12/22/2022]
Abstract
Invariant natural killer T (iNKT) cells are increasingly regarded as disease biomarkers and immunotherapeutic targets. However, a greater understanding of their biology is necessary to effectively target these cells in the clinic. The discovery of iNKT1/2/17 cell effector subsets was a milestone in our understanding of iNKT cell development and function. Recent transcriptomic studies have uncovered an even greater heterogeneity and challenge our understanding of iNKT cell ontogeny and effector differentiation. We propose a refined model whereby iNKT cells differentiate through a dynamic and continuous instructive process that requires the accumulation and integration of various signals within the thymus or peripheral tissues. Within this framework, we question the existence of true iNKT2 cells and discuss the parallels between mouse and human iNKT cells.
Collapse
Affiliation(s)
- Thomas Baranek
- Centre d'Étude des Pathologies Respiratoires (CEPR), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 1100, Faculté de Médecine, Université de Tours, Tours, France
| | - Carolina de Amat Herbozo
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Thierry Mallevaey
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
| | - Christophe Paget
- Centre d'Étude des Pathologies Respiratoires (CEPR), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche 1100, Faculté de Médecine, Université de Tours, Tours, France.
| |
Collapse
|
44
|
Takano T, Morikawa M, Adachi Y, Kabasawa K, Sax N, Moriyama S, Sun L, Isogawa M, Nishiyama A, Onodera T, Terahara K, Tonouchi K, Nishimura M, Tomii K, Yamashita K, Matsumura T, Shinkai M, Takahashi Y. Distinct immune cell dynamics correlate with the immunogenicity and reactogenicity of SARS-CoV-2 mRNA vaccine. Cell Rep Med 2022; 3:100631. [PMID: 35545084 PMCID: PMC9023335 DOI: 10.1016/j.xcrm.2022.100631] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/28/2022] [Accepted: 04/18/2022] [Indexed: 12/14/2022]
Abstract
Two doses of Pfizer/BioNTech BNT162b2 mRNA vaccine elicit robust severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-neutralizing antibodies with frequent adverse events. Here, by applying a high-dimensional immune profiling on 92 vaccinees, we identify six vaccine-induced immune dynamics that correlate with the amounts of neutralizing antibodies, the severity of adverse events, or both. The early dynamics of natural killer (NK)/monocyte subsets (CD16+ NK cells, CD56high NK cells, and non-classical monocytes), dendritic cell (DC) subsets (DC3s and CD11c- Axl+ Siglec-6+ [AS]-DCs), and NKT-like cells are revealed as the distinct cell correlates for neutralizing-antibody titers, severity of adverse events, and both, respectively. The cell correlates for neutralizing antibodies or adverse events are consistently associated with elevation of interferon gamma (IFN-γ)-inducible chemokines, but the chemokine receptors CCR2 and CXCR3 are expressed in distinct manners between the two correlates: vaccine-induced expression on the neutralizing-antibody correlate and constitutive expression on the adverse-event correlate. The finding may guide vaccine strategies that balance immunogenicity and reactogenicity.
Collapse
Affiliation(s)
- Tomohiro Takano
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | - Nicolas Sax
- KOTAI Biotechnologies, Inc., Osaka 565-0871, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Lin Sun
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Masanori Isogawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Ayae Nishiyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kazutaka Terahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Keisuke Tonouchi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | - Kentaro Tomii
- Artificial Intelligence Research Center (AIRC), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo 135-0064, Japan; AIST-Tokyo Tech Real World Big-Data Computation Open Innovation Laboratory (RWBC-OIL), Tokyo 152-8550, Japan
| | | | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.
| | | | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.
| |
Collapse
|
45
|
Johnson DN, Ruan Z, Petley EV, Devi S, Holz LE, Uldrich AP, Mak JYW, Hor JL, Mueller SN, McCluskey J, Fairlie DP, Darcy PK, Beavis PA, Heath WR, Godfrey DI. Differential location of NKT and MAIT cells within lymphoid tissue. Sci Rep 2022; 12:4034. [PMID: 35260653 PMCID: PMC8904549 DOI: 10.1038/s41598-022-07704-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Natural Killer T (NKT) cells and Mucosal-Associated Invariant T (MAIT) cells are innate-like T cells that express semi-invariant αβ T cell receptors (TCRs) through which they recognise CD1d and MR1 molecules, respectively, in complex with specific ligands. These cells play important roles in health and disease in many organs, but their precise intra-organ location is not well established. Here, using CD1d and MR1 tetramer staining techniques, we describe the precise location of NKT and MAIT cells in lymphoid and peripheral organs. Within the thymus, NKT cells were concentrated in the medullary side of the corticomedullary junction. In spleen and lymph nodes, NKT cells were mainly localised within T cell zones, although following in vivo activation with the potent NKT-cell ligand α-GalCer, they expanded throughout the spleen. MAIT cells were clearly detectable in Vα19 TCR transgenic mice and were rare but detectable in lymphoid tissue of non-transgenic mice. In contrast to NKT cells, MAIT cells were more closely associated with the B cell zone and red pulp of the spleen. Accordingly, we have provided an extensive analysis of the in situ localisation of NKT and MAIT cells and suggest differences between the intra-organ location of these two cell types.
Collapse
Affiliation(s)
- Darryl N Johnson
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Zheng Ruan
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Emma V Petley
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sapna Devi
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lauren E Holz
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jeffrey Y W Mak
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jyh Liang Hor
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - James McCluskey
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - David P Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - William R Heath
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia. .,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
46
|
Bond NG, Fahlberg MD, Yu S, Rout N, Tran D, Fitzpatrick-Schmidt T, Sprehe LM, Scheef EA, Mudd JC, Schaub R, Kaur A. Immunomodulatory potential of in vivo natural killer T (NKT) activation by NKTT320 in Mauritian-origin cynomolgus macaques. iScience 2022; 25:103889. [PMID: 35243248 PMCID: PMC8866157 DOI: 10.1016/j.isci.2022.103889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/22/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Invariant natural killer T-lymphocytes (iNKT) are unique immunomodulatory innate T cells with an invariant TCRα recognizing glycolipids presented on MHC class-I-like CD1d molecules. Activated iNKT rapidly secrete pro-and anti-inflammatory cytokines, potentiate immunity, and modulate inflammation. Here, we report the effects of in vivo iNKT activation in Mauritian-origin cynomolgus macaques by a humanized monoclonal antibody, NKTT320, that binds to the invariant region of the iNKT TCR. NKTT320 led to rapid iNKT activation, increased polyfunctionality, and elevation of multiple plasma analytes within 24 hours. Flow cytometry and RNA-Seq confirmed downstream activation of multiple immune subsets, enrichment of JAK/STAT and PI3K/AKT pathway genes, and upregulation of inflammation-modulating genes. NKTT320 also increased iNKT frequency in adipose tissue and did not cause iNKT anergy. Our data indicate that NKTT320 has a sustained effect on in vivo iNKT activation, potentiation of innate and adaptive immunity, and resolution of inflammation, which supports its future use as an immunotherapeutic. NKTT320 rapidly activates iNKT in vivo, modulating downstream immune function In vivo NKTT320 treatment modulates pro- and anti-inflammatory genes NKTT320 treatment results in activation of innate and adaptive immune subsets NKTT320 has promise as an immunotherapeutic with translational potential
Collapse
|
47
|
Abstract
Germinal centers (GCs) are microanatomical sites of B cell clonal expansion and antibody affinity maturation. Therein, B cells undergo the Darwinian process of somatic diversification and affinity-driven selection of immunoglobulins that produces the high-affinity antibodies essential for effective humoral immunity. Here, we review recent developments in the field of GC biology, primarily as it pertains to GCs induced by infection or immunization. First, we summarize the phenotype and function of the different cell types that compose the GC, focusing on GC B cells. Then, we review the cellular and molecular bases of affinity-dependent selection within the GC and the export of memory and plasma cells. Finally, we present an overview of the emerging field of GC clonal dynamics, focusing on how GC and post-GC selection shapes the diversity of antibodies secreted into serum. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA;
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology and Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA;
| |
Collapse
|
48
|
Cruz MS, Loureiro JP, Oliveira MJ, Macedo MF. The iNKT Cell-Macrophage Axis in Homeostasis and Disease. Int J Mol Sci 2022; 23:ijms23031640. [PMID: 35163561 PMCID: PMC8835952 DOI: 10.3390/ijms23031640] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are CD1d-restricted, lipid-reactive T cells that exhibit preponderant immunomodulatory properties. The ultimate protective or deleterious functions displayed by iNKT cells in tissues are known to be partially shaped by the interactions they establish with other immune cells. In particular, the iNKT cell–macrophage crosstalk has gained growing interest over the past two decades. Accumulating evidence has highlighted that this immune axis plays central roles not only in maintaining homeostasis but also during the development of several pathologies. Hence, this review summarizes the reported features of the iNKT cell–macrophage axis in health and disease. We discuss the pathophysiological significance of this interplay and provide an overview of how both cells communicate with each other to regulate disease onset and progression in the context of infection, obesity, sterile inflammation, cancer and autoimmunity.
Collapse
Affiliation(s)
- Mariana S. Cruz
- Cell Activation and Gene Expression Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.S.C.); (J.P.L.)
- Department of Medical Sciences, University of Aveiro (UA), 3810-193 Aveiro, Portugal
| | - José Pedro Loureiro
- Cell Activation and Gene Expression Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.S.C.); (J.P.L.)
- Experimental Immunology Group, Department of Biomedicine (DBM), University of Basel and University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Maria J. Oliveira
- Tumour and Microenvironment Interactions Group, Instituto Nacional de Engenharia Biomédica (INEB), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal;
- Department of Molecular Biology, ICBAS-Institute of Biomedical Sciences Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria Fatima Macedo
- Cell Activation and Gene Expression Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.S.C.); (J.P.L.)
- Department of Medical Sciences, University of Aveiro (UA), 3810-193 Aveiro, Portugal
- Correspondence:
| |
Collapse
|
49
|
Courey-Ghaouzi A, Gaya M. Identification of Non-classical Follicular T Cells. Methods Mol Biol 2022; 2380:77-84. [PMID: 34802123 DOI: 10.1007/978-1-0716-1736-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nonclassical T cells are a heterogeneous group of T lymphocytes that are activated during the early stages of infection and act as a bridge between the innate and adaptive immune system. Among them, Natural Killer T (NKT) cells have been extensively studied in the last two decades due to their unique ability to recognize foreign/self-lipid antigens in the context of CD1d, a nonclassical major histocompatibility complex molecule. In this chapter, we describe our protocols to track murine NKT cells in lymph nodes by flow cytometry and confocal microscopy.
Collapse
Affiliation(s)
- Alan Courey-Ghaouzi
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, INSERM, CNRS, Marseille, France
| | - Mauro Gaya
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, INSERM, CNRS, Marseille, France.
| |
Collapse
|
50
|
Preferential and persistent impact of acute HIV-1 infection on CD4 + iNKT cells in colonic mucosa. Proc Natl Acad Sci U S A 2021; 118:2104721118. [PMID: 34753817 PMCID: PMC8609642 DOI: 10.1073/pnas.2104721118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
Evidence suggests that HIV-1 disease progression is determined in the early stages of infection. Here, preinfection invariant natural killer T (iNKT) cell levels were predictive of the peak viral load during acute HIV-1 infection (AHI). Furthermore, iNKT cells were preferentially lost in AHI. This was particularly striking in the colonic mucosa, where iNKT cells were depleted more profoundly than conventional CD4+ T cells. The initiation of antiretroviral therapy during AHI-prevented iNKT cell dysregulation in peripheral blood but not in the colonic mucosa. Overall, our results support a model in which iNKT cells are early and preferential targets for HIV-1 infection during AHI. Acute HIV-1 infection (AHI) results in the widespread depletion of CD4+ T cells in peripheral blood and gut mucosal tissue. However, the impact on the predominantly CD4+ immunoregulatory invariant natural killer T (iNKT) cells during AHI remains unknown. Here, iNKT cells from peripheral blood and colonic mucosa were investigated during treated and untreated AHI. iNKT cells in blood were activated and rapidly depleted in untreated AHI. At the time of peak HIV-1 viral load, these cells showed the elevated expression of cell death–associated transcripts compared to preinfection. Residual peripheral iNKT cells suffered a diminished responsiveness to in vitro stimulation early into chronic infection. Additionally, HIV-1 DNA, as well as spliced and unspliced viral RNA, were detected in iNKT cells isolated from blood, indicating the active infection of these cells in vivo. The loss of iNKT cells occurred from Fiebig stage III in the colonic mucosa, and these cells were not restored to normal levels after initiation of ART during AHI. CD4+ iNKT cells were depleted faster and more profoundly than conventional CD4+ T cells, and the preferential infection of CD4+ iNKT cells over conventional CD4+ T cells was confirmed by in vitro infection experiments. In vitro data also provided evidence of latent infection in iNKT cells. Strikingly, preinfection levels of peripheral blood CD4+ iNKT cells correlated directly with the peak HIV-1 load. These findings support a model in which iNKT cells are early targets for HIV-1 infection, driving their rapid loss from circulation and colonic mucosa.
Collapse
|