1
|
Almeida CF, Gully BS, Jones CM, Kedzierski L, Gunasinghe SD, Rice MT, Berry R, Gherardin NA, Nguyen TT, Mok YF, Reijneveld JF, Moody DB, Van Rhijn I, La Gruta NL, Uldrich AP, Rossjohn J, Godfrey DI. Direct recognition of an intact foreign protein by an αβ T cell receptor. Nat Commun 2024; 15:8816. [PMID: 39394178 PMCID: PMC11470135 DOI: 10.1038/s41467-024-51897-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 08/21/2024] [Indexed: 10/13/2024] Open
Abstract
αβ T cell receptors (αβTCRs) co-recognise antigens when bound to Major Histocompatibility Complex (MHC) or MHC class I-like molecules. Additionally, some αβTCRs can bind non-MHC molecules, but how much intact antigen reactivities are achieved remains unknown. Here, we identify an αβ T cell clone that directly recognises the intact foreign protein, R-phycoerythrin (PE), a multimeric (αβ)6γ protein complex. This direct αβTCR-PE interaction occurs in an MHC-independent manner, yet triggers T cell activation and bound PE with an affinity comparable to αβTCR-peptide-MHC interactions. The crystal structure reveals how six αβTCR molecules simultaneously engage the PE hexamer, mediated by the complementarity-determining regions (CDRs) of the αβTCR. Here, the αβTCR mainly binds to two α-helices of the globin fold in the PE α-subunit, which is analogous to the antigen-binding platform of the MHC molecule. Using retrogenic mice expressing this TCR, we show that it supports intrathymic T cell development, maturation, and exit into the periphery as mature CD4/CD8 double negative (DN) T cells with TCR-mediated functional capacity. Accordingly, we show how an αβTCR can recognise an intact foreign protein in an antibody-like manner.
Collapse
MESH Headings
- Animals
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Mice
- Phycoerythrin/metabolism
- Phycoerythrin/chemistry
- Lymphocyte Activation/immunology
- Protein Binding
- Crystallography, X-Ray
- Mice, Inbred C57BL
- Humans
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/metabolism
- Models, Molecular
Collapse
Affiliation(s)
- Catarina F Almeida
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin S Gully
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Claerwen M Jones
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lukasz Kedzierski
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Sachith D Gunasinghe
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- European Molecular Biology Laboratory (EMBL) Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, New South Wales, Australia
| | - Michael T Rice
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Richard Berry
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Nicholas A Gherardin
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Trang T Nguyen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Yee-Foong Mok
- Melbourne Protein Characterisation Platform, Bio21 Molecular Science and Biotechnology Institute, Melbourne, VIC, Australia
| | - Josephine F Reijneveld
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - D Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Nicole L La Gruta
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Adam P Uldrich
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK.
| | - Dale I Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
2
|
Taheri MM, Javan F, Poudineh M, Athari SS. CAR-NKT Cells in Asthma: Use of NKT as a Promising Cell for CAR Therapy. Clin Rev Allergy Immunol 2024; 66:328-362. [PMID: 38995478 DOI: 10.1007/s12016-024-08998-0] [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] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
NKT cells, unique lymphocytes bridging innate and adaptive immunity, offer significant potential for managing inflammatory disorders like asthma. Activating iNKT induces increasing IFN-γ, TGF-β, IL-2, and IL-10 potentially suppressing allergic asthma. However, their immunomodulatory effects, including granzyme-perforin-mediated cytotoxicity, and expression of TIM-3 and TRAIL warrant careful consideration and targeted approaches. Although CAR-T cell therapy has achieved remarkable success in treating certain cancers, its limitations necessitate exploring alternative approaches. In this context, CAR-NKT cells emerge as a promising approach for overcoming these challenges, potentially achieving safer and more effective immunotherapies. Strategies involve targeting distinct IgE-receptors and their interactions with CAR-NKT cells, potentially disrupting allergen-mast cell/basophil interactions and preventing inflammatory cytokine release. Additionally, targeting immune checkpoints like PDL-2, inducible ICOS, FASL, CTLA-4, and CD137 or dectin-1 for fungal asthma could further modulate immune responses. Furthermore, artificial intelligence and machine learning hold immense promise for revolutionizing NKT cell-based asthma therapy. AI can optimize CAR-NKT cell functionalities, design personalized treatment strategies, and unlock a future of precise and effective care. This review discusses various approaches to enhancing CAR-NKT cell efficacy and longevity, along with the challenges and opportunities they present in the treatment of allergic asthma.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyyed Shamsadin Athari
- Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
3
|
Tomás-Cortázar J, Quinn C, Corcoran N, Blanco A, Christensen D, McClean S. BpOmpW antigen administered with CAF01 adjuvant stimulates comparable T cell responses to Sigma adjuvant system. Vaccine X 2024; 17:100438. [PMID: 38303776 PMCID: PMC10831100 DOI: 10.1016/j.jvacx.2024.100438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
There are no licensed vaccines to protect vulnerable populations from the potentially fatal tropical infection, melioidosis, despite its causative agent, Burkholderia pseudomallei, being endemic in tropical and subtropical regions. A promising vaccine candidate, BpOmpW protected mice from melioidosis infection for up to 81 days and stimulated robust interferon gamma responses in CD4+, CD8+, NK and NKT cells. In order to progress to human studies, selection of an adjuvant with an acceptable human safety profile that stimulates appropriate correlates of protection is essential. Here we demonstrate that the CAF01 vaccine adjuvant elicits optimal immune correlates of protection when administered with our BpOmpW vaccine. Specifically, we demonstrate that CAF01 administered with BpOmpW elicits robust Th1 responses, with potent IFN-γ responses in CD4+ and CD8+ T cells and NKT cells, in addition to Th17 and Th2 responses. This formulation will be particularly effective in protecting susceptible populations including people with type 2 diabetes from melioidosis.
Collapse
Affiliation(s)
- Julen Tomás-Cortázar
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Conor Quinn
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Niamh Corcoran
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Alfonso Blanco
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen S, Denmark
| | - Siobhán McClean
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Eckhardt E, Schinköthe J, Gischke M, Sehl-Ewert J, Corleis B, Dorhoi A, Teifke J, Albrecht D, Geluk A, Gilleron M, Bastian M. Phosphatidylinositolmannoside vaccination induces lipid-specific Th1-responses and partially protects guinea pigs from Mycobacterium tuberculosis challenge. Sci Rep 2023; 13:18613. [PMID: 37903877 PMCID: PMC10616071 DOI: 10.1038/s41598-023-45898-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/25/2023] [Indexed: 11/01/2023] Open
Abstract
The concept of donor-unrestricted T cells (DURTs) comprises a heterogeneity of lymphoid cells that respond to an abundance of unconventional epitopes in a non-MHC-restricted manner. Vaccinologists strive to harness this so far underexplored branch of the immune system for new vaccines against tuberculosis. A particular division of DURTs are T cells that recognize their cognate lipid antigen in the context of CD1-molecules. Mycobacteria are characterized by a particular lipid-rich cell wall. Several of these lipids have been shown to be presented to T cells via CD1b-molecules. Guinea pigs functionally express CD1b and are hence an appropriate small animal model to study the role of CD1b-restricted, lipid-specific immune responses. In the current study, guinea pigs were vaccinated with BCG or highly-purified, liposome-formulated phosphatidylinositol-hexa-mannoside (PIM6) to assess the effect of CD1-restricted DURTs on the course of infection after virulent Mycobacterium tuberculosis (Mtb) challenge. Robust PIM6-specific T cell-responses were observed both after BCG- and PIM6-vaccination. The cellular response was significantly reduced in the presence of monoclonal, CD1b-blocking antibodies, indicating that a predominant part of this reactivity was CD1b-restricted. When animals were challenged with Mtb, BCG- and PIM6-vaccinated animals showed significantly reduced pathology, smaller necrotic granulomas in lymph node and spleen and reduced bacterial loads. While BCG conferred an almost sterile protection in this setting, compared to control animals' lesions were reduced roughly by two thirds in PIM6-vaccinated. Comprehensive histological and transcriptional analyses in the draining lymph node revealed that protected animals showed reduced transcription-levels of inflammatory cyto- and chemokines and higher levels of CD1b-expression on professional antigen cells compared to controls. Although BCG as a comparator induced by far stronger effects, our observations in the guinea pig model suggest that CD1b-restricted, PIM6-reactive DURTs contribute to immune-mediated containment of virulent Mtb.
Collapse
Affiliation(s)
- Emmelie Eckhardt
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald - Isle of Riems, Germany
| | - Jan Schinköthe
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Marcel Gischke
- Institute of Microbiology, Greifswald University, Greifswald, Germany
| | - Julia Sehl-Ewert
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald - Isle of Riems, Germany
| | - Björn Corleis
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald - Isle of Riems, Germany
| | - Anca Dorhoi
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald - Isle of Riems, Germany
| | - Jens Teifke
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald - Isle of Riems, Germany
| | - Dirk Albrecht
- Institute of Microbiology, Greifswald University, Greifswald, Germany
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Martine Gilleron
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | - Max Bastian
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald - Isle of Riems, Germany.
| |
Collapse
|
5
|
Lee MS, Webb TJ. Novel lipid antigens for NKT cells in cancer. Front Immunol 2023; 14:1173375. [PMID: 37908366 PMCID: PMC10613688 DOI: 10.3389/fimmu.2023.1173375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Cancer immunotherapy aims to unleash the power of the immune system against tumors without the side effects of traditional chemotherapy. Immunotherapeutic methods vary widely, but all follow the same basic principle: overcome the barriers utilized by cancers to avoid immune destruction. These approaches often revolve around classical T cells, such as with CAR T cells and neoantigen vaccines; however, the utility of the innate-like iNKT cell in cancer immunotherapy has gained significant recognition. iNKT cells parallel classic T cell recognition of peptide antigens presented on MHC through their recognition of lipid antigens presented on the MHC I-like molecule CD1d. Altered metabolism and a lipogenic phenotype are essential properties of tumor cells, representing a unique feature that may be exploited by iNKT cells. In this review, we will cover properties of iNKT cells, CD1d, and lipid antigen presentation. Next, we will discuss the cancer lipidome and how it may be exploited by iNKT cells through a window of opportunity. Finally, we will review, in detail, novel lipid antigens for iNKT cells in cancer.
Collapse
Affiliation(s)
- Michael S. Lee
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tonya J. Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
6
|
Hackstein CP, Klenerman P. MAITs and their mates: "Innate-like" behaviors in conventional and unconventional T cells. Clin Exp Immunol 2023; 213:1-9. [PMID: 37256718 PMCID: PMC10324555 DOI: 10.1093/cei/uxad058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/01/2023] [Accepted: 05/30/2023] [Indexed: 06/02/2023] Open
Abstract
Most CD4 and CD8 T cells are restricted by conventional major histocompatibility complex (MHC) molecules and mount TCR-dependent adaptive immune responses. In contrast, MAIT, iNKT, and certain γδ TCR bearing cells are characterized by their abilities to recognize antigens presented by unconventional antigen-presenting molecules and to mount cytokine-mediated TCR-independent responses in an "innate-like" manner. In addition, several more diverse T-cell subsets have been described that in a similar manner are restricted by unconventional antigen-presenting molecules but mainly depend on their TCRs for activation. Vice versa, innate-like behaviour was reported in defined subpopulations of conventional T cells, particularly in barrier sites, showing that these two features are not necessarily linked. The abilities to recognize antigens presented by unconventional antigen-presenting molecules or to mount TCR-independent responses creates unique niches for these T cells and is linked to wide range of functional capabilities. This is especially exemplified by unconventional and innate-like T cells present at barrier sites where they are involved in pathogen defense, tissue homeostasis as well as in pathologic processes.
Collapse
Affiliation(s)
- Carl-Philipp Hackstein
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Abstract
Metabolites produced by commensal gut microbes impact host health through their recognition by the immune system and their influence on numerous metabolic pathways. Notably, the gut microbiota can both transform and synthesize lipids as well as break down dietary lipids to generate secondary metabolites with host modulatory properties. Although lipids have largely been consigned to structural roles, particularly in cell membranes, recent research has led to an increased appreciation of their signaling activities, with potential impacts on host health and physiology. This review focuses on studies that highlight the functions of bioactive lipids in mammalian physiology, with a special emphasis on immunity and metabolism.
Collapse
Affiliation(s)
- Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute, Boston, MA 02115, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
8
|
Monnot GC, Wegrecki M, Cheng TY, Chen YL, Sallee BN, Chakravarthy R, Karantza IM, Tin SY, Khaleel AE, Monga I, Uwakwe LN, Tillman A, Cheng B, Youssef S, Ng SW, Shahine A, Garcia-Vilas JA, Uhlemann AC, Bordone LA, Han A, Rohde CH, Ogg G, Moody DB, Rossjohn J, de Jong A. Staphylococcal phosphatidylglycerol antigens activate human T cells via CD1a. Nat Immunol 2023; 24:110-122. [PMID: 36550321 PMCID: PMC10389259 DOI: 10.1038/s41590-022-01375-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
Expressed on epidermal Langerhans cells, CD1a presents a range of self-lipid antigens found within the skin; however, the extent to which CD1a presents microbial ligands from bacteria colonizing the skin is unclear. Here we identified CD1a-dependent T cell responses to phosphatidylglycerol (PG), a ubiquitous bacterial membrane phospholipid, as well as to lysylPG, a modified PG, present in several Gram-positive bacteria and highly abundant in Staphylococcus aureus. The crystal structure of the CD1a-PG complex showed that the acyl chains were buried within the A'- and F'-pockets of CD1a, while the phosphoglycerol headgroup remained solvent exposed in the F'-portal and was available for T cell receptor contact. Using lysylPG and PG-loaded CD1a tetramers, we identified T cells in peripheral blood and in skin that respond to these lipids in a dose-dependent manner. Tetramer+CD4+ T cell lines secreted type 2 helper T cell cytokines in response to phosphatidylglycerols as well as to co-cultures of CD1a+ dendritic cells and Staphylococcus bacteria. The expansion in patients with atopic dermatitis of CD4+ CD1a-(lysyl)PG tetramer+ T cells suggests a response to lipids made by bacteria associated with atopic dermatitis and provides a link supporting involvement of PG-based lipid-activated T cells in atopic dermatitis pathogenesis.
Collapse
Affiliation(s)
- Gwennaëlle C Monnot
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Marcin Wegrecki
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Tan-Yun Cheng
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yi-Ling Chen
- Medical Research Council Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Brigitte N Sallee
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Reka Chakravarthy
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioanna Maria Karantza
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Shin Yi Tin
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Alexandra E Khaleel
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Isha Monga
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Laura N Uwakwe
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Alice Tillman
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Microbiome and Pathogen Genomics Core, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Bin Cheng
- Department of Biostatistics, Columbia University Irving Medical Center, New York, NY, USA
| | - Soundos Youssef
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Soo Weei Ng
- Medical Research Council Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Javier A Garcia-Vilas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Microbiome and Pathogen Genomics Core, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lindsey A Bordone
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA
| | - Arnold Han
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Christine H Rohde
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Graham Ogg
- Medical Research Council Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - D Branch Moody
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Annemieke de Jong
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Li T, Lin X, Shen B, Zhang W, Liu Y, Liu H, Wang Y, Zheng L, Zhi F. Akkermansia muciniphila suppressing nonalcoholic steatohepatitis associated tumorigenesis through CXCR6 + natural killer T cells. Front Immunol 2022; 13:1047570. [PMID: 36531991 PMCID: PMC9755844 DOI: 10.3389/fimmu.2022.1047570] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Gut microbiota plays a crucial role in the development and progression of nonalcoholic steatohepatitis (NASH) and associated hepatocellular carcinoma (HCC). Akkermansia muciniphila was reported to inhibit inflammation-associated cancer in the intestine. The anti-NASH ability of A. muciniphila has recently been found. Thus, we were to investigate whether supplementation of A. muciniphila could prevent NASH-associated HCC. Methods In a model we called STAM, male C57BL/6J mice were subcutaneously injected with 200 µg streptozotocin at 4 days after birth, and fed with high-fat diet at 4 weeks of age to induce NASH-associated HCC. Faeces from mice and patients with NASH-related HCC were collected for 16S rRNA sequencing. STAM mice were orally administered either saline or A. muciniphila twice a day starting at 4 or 10 weeks of age. The effects of A. muciniphila on the immune responses were also evaluated. Results Patients and mice with NASH-related HCC showed significantly reduced gut A. muciniphila in comparison to healthy controls. Administration of breast milk-isolated A. muciniphila (AM06) but not feces-isolated A. muciniphila (AM02) could improve NASH severity. Interestingly, breast milk-isolated A. muciniphila treatment suppressed the progression of NASH to HCC, accompanied with an increased hepatic CXCR6+ natural killer T (NKT) cell and decreased macrophage infiltration. The antitumor ability of A. muciniphila was not evident in NKT cell-deficient mice (CD1d-/- and CXCR6-/-). In vitro, A. muciniphila promoted the killing of hepG2 cells by NKT cells. Discussion Our study will provide the rationale for the application of A. muciniphila to treat NASH and for the prevention of its progression to HCC.
Collapse
Affiliation(s)
- Tao Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinlong Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Binhai Shen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wujian Zhang
- Department of General Surgery of the First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Haerbin, China
| | - Yangyang Liu
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Hongbin Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Wang
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Lijun Zheng
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Zheng Q, Xue C, Gu X, Shan D, Chu Q, Wang J, Zhu H, Chen Z. Multi-Omics Characterizes the Effects and Mechanisms of CD1d in Nonalcoholic Fatty Liver Disease Development. Front Cell Dev Biol 2022; 10:830702. [PMID: 35465315 PMCID: PMC9024148 DOI: 10.3389/fcell.2022.830702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a class of metabolic-associated liver diseases. Aberrant lipid consumption plays an important role in NAFLD pathogenesis. It has been shown CD1d can bind to multiple different lysophospholipids and associated with NAFLD progression. However, the mechanism of CD1d regulation in NAFLD is not completely understood. In this study, we established a NAFLD mouse model by feeding C57/BL6J mice a high-fat diet (HFD) for 24 weeks. Subsequently, we performed integrated transcriptomics and metabolomics analyses to thoroughly probe the role of CD1d in NAFLD progression. In the present study, we demonstrate that CD1d expression was significantly decreased in our murine model of NAFLD. Additionally, we show CD1d knockdown (CD1d KO) in HFD-fed wild-type (WT) mice induced NAFLD, which resulted in weight gain, exaggerated liver injury, and hepatic steatosis. We uncover the crucial roles of CD1d deficiency results in accumulated lipid accumulation. We further explored the CD1d deficiency in NAFLD regarding the transcriptional landscapes, microbiota environment, metabolomics change, and transcriptomics differences. In conclusion, our data demonstrate CD1d plays an important role in NAFLD pathogenesis and may represent a potential therapeutic target for the further therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhi Chen
- *Correspondence: Zhi Chen, ; Haihong Zhu,
| |
Collapse
|
11
|
Zhao L, Yang X. Cross Talk Between Natural Killer T and Dendritic Cells and Its Impact on T Cell Responses in Infections. Front Immunol 2022; 13:837767. [PMID: 35185930 PMCID: PMC8850912 DOI: 10.3389/fimmu.2022.837767] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 11/16/2022] Open
Abstract
Both innate and adaptive immunity is vital for host defense against infections. Dendritic cells (DCs) are critical for initiating and modulating adaptive immunity, especially for T-cell responses. Natural killer T (NKT) cells are a small population of innate-like T cells distributed in multiple organs. Many studies have suggested that the cross-talk between these two immune cells is critical for immunobiology and host defense mechanisms. Not only can DCs influence the activation/function of NKT cells, but NKT cells can feedback on DCs also, thus modulating the phenotype and function of DCs and DC subsets. This functional feedback of NKT cells on DCs, especially the preferential promoting effect on CD8α+ and CD103+ DC subsets in lymphoid and non-lymphoid tissues, significantly impacts the systemic and local adaptive CD4 and CD8 T cell responses in infections. This review focuses on the two-way interaction between NKT cells and DCs, emphasizing the importance of NKT cell feedback on DCs in bridging innate and adaptive immune responses for host defense purposes.
Collapse
Affiliation(s)
- Lei Zhao
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xi Yang
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
12
|
Gwynne PJ, Clendenen LH, Turk SP, Marques AR, Hu LT. Antiphospholipid autoantibodies in Lyme disease arise after scavenging of host phospholipids by Borrelia burgdorferi. J Clin Invest 2022; 132:152506. [PMID: 35289310 PMCID: PMC8920326 DOI: 10.1172/jci152506] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 02/02/2022] [Indexed: 12/13/2022] Open
Abstract
A close association with its vertebrate and tick hosts allows Borrelia burgdorferi, the bacterium responsible for Lyme disease, to eliminate many metabolic pathways and instead scavenge key nutrients from the host. A lipid-defined culture medium was developed to demonstrate that exogenous lipids are an essential nutrient of B. burgdorferi, which can accumulate intact phospholipids from its environment to support growth. Antibody responses to host phospholipids were studied in mice and humans using an antiphospholipid ELISA. Several of these environmentally acquired phospholipids including phosphatidylserine and phosphatidic acid, as well as borrelial phosphatidylcholine, are the targets of antibodies that arose early in infection in the mouse model. Patients with acute infections demonstrated antibody responses to the same lipids. The elevation of antiphospholipid antibodies predicted early infection with better sensitivity than did the standardized 2-tier tests currently used in diagnosis. Sera obtained from patients with Lyme disease before and after antibiotic therapy showed declining antiphospholipid titers after treatment. Further study will be required to determine whether these antibodies have utility in early diagnosis of Lyme disease, tracking of the response to therapy, and diagnosis of reinfection, areas in which current standardized tests are inadequate.
Collapse
Affiliation(s)
- Peter J Gwynne
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Luke H Clendenen
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Siu-Ping Turk
- Laboratory of Clinical Microbiology and Immunology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Adriana R Marques
- Laboratory of Clinical Microbiology and Immunology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Li Y, Sharma A, Maciaczyk J, Schmidt-Wolf IGH. Recent Development in NKT-Based Immunotherapy of Glioblastoma: From Bench to Bedside. Int J Mol Sci 2022; 23:ijms23031311. [PMID: 35163235 PMCID: PMC8835986 DOI: 10.3390/ijms23031311] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive and dismal disease with a median overall survival of around 15 months and a 5-year survival rate of 7.2%. Owing to genetic mutations, drug resistance, disruption to the blood–brain barrier (BBB)/blood–brain tumor barrier (BBTB), and the complexity of the immunosuppressive environment, the therapeutic approaches to GBM represent still major challenges. Conventional therapies, including surgery, radiotherapy, and standard chemotherapy with temozolomide, have not resulted in satisfactory improvements in the overall survival of GBM patients. Among cancer immunotherapeutic approaches, we propose that adjuvant NKT immunotherapy with invariant NKT (iNKT) and cytokine-induced killer (CIK) cells may improve the clinical scenario of this devastating disease. Considering this, herein, we discuss the current strategies of NKT therapy for GBM based primarily on in vitro/in vivo experiments, clinical trials, and the combinatorial approaches with future therapeutic potential.
Collapse
Affiliation(s)
- Yutao Li
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Amit Sharma
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
| | - Jarek Maciaczyk
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
- Department of Surgical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ingo G. H. Schmidt-Wolf
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-2871-7050
| |
Collapse
|
14
|
Burchill L, Williams SJ. From the banal to the bizarre: unravelling immune recognition and response to microbial lipids. Chem Commun (Camb) 2022; 58:925-940. [PMID: 34989357 DOI: 10.1039/d1cc06003a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microbes produce a rich array of lipidic species that through their location in the cell wall and ability to mingle with host lipids represent a privileged class of immune-active molecules. Lipid-sensing immunity recognizes microbial lipids from pathogens and commensals causing immune responses. Yet microbial lipids are often heterogeneous, in limited supply and in some cases their structures are incompletely defined. Total synthesis can assist in structural determination, overcome supply issues, and provide access to high-purity, homogeneous samples and analogues. This account highlights synthetic approaches to lipidic species from pathogenic and commensal bacteria and fungi that have supported immunological studies involving lipid sensing through the pattern recognition receptor Mincle and cell-mediated immunity through the CD1-T cell axis.
Collapse
Affiliation(s)
- Laura Burchill
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Spencer J Williams
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
15
|
Vogt S, Mattner J. NKT Cells Contribute to the Control of Microbial Infections. Front Cell Infect Microbiol 2021; 11:718350. [PMID: 34595131 PMCID: PMC8477047 DOI: 10.3389/fcimb.2021.718350] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022] Open
Abstract
Innate (-like) T lymphocytes such as natural killer T (NKT) cells play a pivotal role in the recognition of microbial infections and their subsequent elimination. They frequently localize to potential sites of pathogen entry at which they survey extracellular and intracellular tissue spaces for microbial antigens. Engagement of their T cell receptors (TCRs) induces an explosive release of different cytokines and chemokines, which often pre-exist as constitutively expressed gene transcripts in NKT cells and underlie their poised effector state. Thus, NKT cells regulate immune cell migration and activation and subsequently, bridge innate and adaptive immune responses. In contrast to conventional T cells, which react to peptide antigens, NKT cells recognize lipids presented by the MHC class I like CD1d molecule on antigen presenting cells (APCs). Furthermore, each NKT cell TCR can recognize various antigen specificities, whereas a conventional T lymphocyte TCR reacts mostly only to one single antigen. These lipid antigens are either intermediates of the intracellular APC`s-own metabolism or originate from the cell wall of different bacteria, fungi or protozoan parasites. The best-characterized subset, the type 1 NKT cell subset expresses a semi-invariant TCR. In contrast, the TCR repertoire of type 2 NKT cells is diverse. Furthermore, NKT cells express a panoply of inhibitory and activating NK cell receptors (NKRs) that contribute to their primarily TCR-mediated rapid, innate like immune activation and even allow an adaption of their immune response in an adoptive like manner. Dueto their primary localization at host-environment interfaces, NKT cells are one of the first immune cells that interact with signals from different microbial pathogens. Vice versa, the mutual exchange with local commensal microbiota shapes also the biology of NKT cells, predominantly in the gastrointestinal tract. Following infection, two main signals drive the activation of NKT cells: first, cognate activation upon TCR ligation by microbial or endogenous lipid antigens; and second, bystander activation due to cytokines. Here we will discuss the role of NKT cells in the control of different microbial infections comparing pathogens expressing lipid ligands in their cell walls to infectious agents inducing endogenous lipid antigen presentation by APCs.
Collapse
Affiliation(s)
- Stefan Vogt
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
16
|
Almeida CF, Smith DGM, Cheng TY, Harpur CM, Batleska E, Nguyen-Robertson CV, Nguyen T, Thelemann T, Reddiex SJJ, Li S, Eckle SBG, Van Rhijn I, Rossjohn J, Uldrich AP, Moody DB, Williams SJ, Pellicci DG, Godfrey DI. Benzofuran sulfonates and small self-lipid antigens activate type II NKT cells via CD1d. Proc Natl Acad Sci U S A 2021; 118:e2104420118. [PMID: 34417291 PMCID: PMC8403964 DOI: 10.1073/pnas.2104420118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Natural killer T (NKT) cells detect lipids presented by CD1d. Most studies focus on type I NKT cells that express semi-invariant αβ T cell receptors (TCR) and recognize α-galactosylceramides. However, CD1d also presents structurally distinct lipids to NKT cells expressing diverse TCRs (type II NKT cells), but our knowledge of the antigens for type II NKT cells is limited. An early study identified a nonlipidic NKT cell agonist, phenyl pentamethyldihydrobenzofuransulfonate (PPBF), which is notable for its similarity to common sulfa drugs, but its mechanism of NKT cell activation remained unknown. Here, we demonstrate that a range of pentamethylbenzofuransulfonates (PBFs), including PPBF, activate polyclonal type II NKT cells from human donors. Whereas these sulfa drug-like molecules might have acted pharmacologically on cells, here we demonstrate direct contact between TCRs and PBF-treated CD1d complexes. Further, PBF-treated CD1d tetramers identified type II NKT cell populations expressing αβTCRs and γδTCRs, including those with variable and joining region gene usage (TRAV12-1-TRAJ6) that was conserved across donors. By trapping a CD1d-type II NKT TCR complex for direct mass-spectrometric analysis, we detected molecules that allow the binding of CD1d to TCRs, finding that both selected PBF family members and short-chain sphingomyelin lipids are present in these complexes. Furthermore, the combination of PPBF and short-chain sphingomyelin enhances CD1d tetramer staining of PPBF-reactive T cell lines over either molecule alone. This study demonstrates that nonlipidic small molecules, which resemble sulfa drugs implicated in systemic hypersensitivity and drug allergy reactions, are targeted by a polyclonal population of type II NKT cells in a CD1d-restricted manner.
Collapse
Affiliation(s)
- Catarina F Almeida
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Dylan G M Smith
- School of Chemistry, The University of Melbourne, Melbourne, VIC 3052, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Tan-Yun Cheng
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Boston, MA 02115
| | - Chris M Harpur
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Elena Batleska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Catriona V Nguyen-Robertson
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Tram Nguyen
- School of Chemistry, The University of Melbourne, Melbourne, VIC 3052, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Tamara Thelemann
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Scott J J Reddiex
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Shihan Li
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Sidonia B G Eckle
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Ildiko Van Rhijn
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Boston, MA 02115
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, University Utrecht, 3584CL Utrecht, Netherlands
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Adam P Uldrich
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - D Branch Moody
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Boston, MA 02115;
| | - Spencer J Williams
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia;
- School of Chemistry, The University of Melbourne, Melbourne, VIC 3052, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Daniel G Pellicci
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
17
|
Holzheimer M, Buter J, Minnaard AJ. Chemical Synthesis of Cell Wall Constituents of Mycobacterium tuberculosis. Chem Rev 2021; 121:9554-9643. [PMID: 34190544 PMCID: PMC8361437 DOI: 10.1021/acs.chemrev.1c00043] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The pathogen Mycobacterium tuberculosis (Mtb), causing
tuberculosis disease, features an extraordinary
thick cell envelope, rich in Mtb-specific lipids,
glycolipids, and glycans. These cell wall components are often directly
involved in host–pathogen interaction and recognition, intracellular
survival, and virulence. For decades, these mycobacterial natural
products have been of great interest for immunology and synthetic
chemistry alike, due to their complex molecular structure and the
biological functions arising from it. The synthesis of many of these
constituents has been achieved and aided the elucidation of their
function by utilizing the synthetic material to study Mtb immunology. This review summarizes the synthetic efforts of a quarter
century of total synthesis and highlights how the synthesis layed
the foundation for immunological studies as well as drove the field
of organic synthesis and catalysis to efficiently access these complex
natural products.
Collapse
Affiliation(s)
- Mira Holzheimer
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Jeffrey Buter
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| |
Collapse
|
18
|
Ruibal P, Voogd L, Joosten SA, Ottenhoff THM. The role of donor-unrestricted T-cells, innate lymphoid cells, and NK cells in anti-mycobacterial immunity. Immunol Rev 2021; 301:30-47. [PMID: 33529407 PMCID: PMC8154655 DOI: 10.1111/imr.12948] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/15/2022]
Abstract
Vaccination strategies against mycobacteria, focusing mostly on classical T‐ and B‐cells, have shown limited success, encouraging the addition of alternative targets. Classically restricted T‐cells recognize antigens presented via highly polymorphic HLA class Ia and class II molecules, while donor‐unrestricted T‐cells (DURTs), with few exceptions, recognize ligands via genetically conserved antigen presentation molecules. Consequently, DURTs can respond to the same ligands across diverse human populations. DURTs can be activated either through cognate TCR ligation or via bystander cytokine signaling. TCR‐driven antigen‐specific activation of DURTs occurs upon antigen presentation via non‐polymorphic molecules such as HLA‐E, CD1, MR1, and butyrophilin, leading to the activation of HLA‐E–restricted T‐cells, CD1‐restricted T‐cells, mucosal‐associated invariant T‐cells (MAITs), and TCRγδ T‐cells, respectively. NK cells and innate lymphoid cells (ILCs), which lack rearranged TCRs, are activated through other receptor‐triggering pathways, or can be engaged through bystander cytokines, produced, for example, by activated antigen‐specific T‐cells or phagocytes. NK cells can also develop trained immune memory and thus could represent cells of interest to mobilize by novel vaccines. In this review, we summarize the latest findings regarding the contributions of DURTs, NK cells, and ILCs in anti–M tuberculosis, M leprae, and non‐tuberculous mycobacterial immunity and explore possible ways in which they could be harnessed through vaccines and immunotherapies to improve protection against Mtb.
Collapse
Affiliation(s)
- Paula Ruibal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Linda Voogd
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
19
|
The thick waxy coat of mycobacteria, a protective layer against antibiotics and the host's immune system. Biochem J 2020; 477:1983-2006. [PMID: 32470138 PMCID: PMC7261415 DOI: 10.1042/bcj20200194] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/22/2022]
Abstract
Tuberculosis, caused by the pathogenic bacterium Mycobacterium tuberculosis (Mtb), is the leading cause of death from an infectious disease, with a mortality rate of over a million people per year. This pathogen's remarkable resilience and infectivity is largely due to its unique waxy cell envelope, 40% of which comprises complex lipids. Therefore, an understanding of the structure and function of the cell wall lipids is of huge indirect clinical significance. This review provides a synopsis of the cell envelope and the major lipids contained within, including structure, biosynthesis and roles in pathogenesis.
Collapse
|
20
|
Genardi S, Visvabharathy L, Cao L, Morgun E, Cui Y, Qi C, Chen YH, Gapin L, Berdyshev E, Wang CR. Type II Natural Killer T Cells Contribute to Protection Against Systemic Methicillin-Resistant Staphylococcus aureus Infection. Front Immunol 2020; 11:610010. [PMID: 33312179 PMCID: PMC7708336 DOI: 10.3389/fimmu.2020.610010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/22/2020] [Indexed: 12/31/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (SA) bacteremia is responsible for over 10,000 deaths in the hospital setting each year. Both conventional CD4+ T cells and γδ T cells play protective roles in SA infection through secretion of IFN-γ and IL-17. However, the role of other unconventional T cells in SA infection is largely unknown. Natural killer T (NKT) cells, a subset of innate-like T cells, are activated rapidly in response to a wide range of self and microbial lipid antigens presented by MHC I-like molecule CD1d. NKT cells are divided into two groups, invariant NKT (iNKT) and type II NKT cells, based on TCR usage. Using mice lacking either iNKT cells or both types of NKT cells, we show that both NKT cell subsets are activated after systemic SA infection and produce IFN-γ in response to SA antigen, however type II NKT cells are sufficient to control bacterial burden and inflammatory infiltrate in infected organs. This protective capacity was specific for NKT cells, as mice lacking mucosal associated invariant T (MAIT) cells, another innate-like T cell subset, had no increased susceptibility to SA systemic infection. We identify polar lipid species from SA that induce IFN-γ production from type II NKT cells, which requires both CD1d-TCR engagement and IL-12 production by antigen presenting cells. We also demonstrate that a population of T cells enriched for type II NKT cells are increased in PBMC of SA bacteremic patients compared to healthy controls. Therefore, type II NKT cells perform effector functions that enhance control of SA infection prior to conventional T cell activation and recognize SA-derived lipid antigens. As CD1d is highly conserved in humans, these CD1d-restricted SA lipid antigens could be used in the design of next generation SA vaccines targeting cell-mediated immunity.
Collapse
Affiliation(s)
- Samantha Genardi
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lavanya Visvabharathy
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Liang Cao
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Eva Morgun
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yongyong Cui
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Chao Qi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yi-Hua Chen
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Evgeny Berdyshev
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
21
|
Valestrand L, Berntsen NL, Zheng F, Schrumpf E, Hansen SH, Karlsen TH, Blumberg RS, Hov JR, Jiang X, Melum E. Lipid antigens in bile from patients with chronic liver diseases activate natural killer T cells. Clin Exp Immunol 2020; 203:304-314. [PMID: 33089489 DOI: 10.1111/cei.13541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
Natural killer T (NKT) cells are an abundant subset of liver lymphocytes activated by lipid antigens presented on CD1d molecules that are expressed by cholangiocytes. We aimed to determine if bile from patients with chronic liver diseases contains antigenic lipids that can activate NKT cells. Using murine invariant (24.7, 24.8 and DN32.D3) and non-invariant (14S.6, 14S.7 and 14S.10) NKT hybridomas we investigated the presence of lipid antigens in bile collected from the gallbladder of patients undergoing liver transplantation due to end-stage liver disease. Biliary microbiota profiles were generated using 16S rRNA amplicon sequencing. We found that the patient bile samples contain antigens that activate both invariant and non-invariant NKT hybridomas (24.7, 24.8, DN32.D3, 14S.6, 14S.7 and 14S.10), as demonstrated by activation of at least one hybridoma by eight of 10 bile samples. Activation at high dilutions suggests that some antigens are highly potent. We used the non-invariant NKT hybridoma 14S.6 to screen 21 additional patient bile samples for NKT-reactivity and demonstrated that 12 of 21 bile samples resulted in activation, three of which gave a strong activation. Four of 12 activating bile samples contained microbial DNA. Our results reveal an immunological pathway that could be of critical importance in biliary immunology.
Collapse
Affiliation(s)
- L Valestrand
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Section for Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - N L Berntsen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - F Zheng
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - E Schrumpf
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - S H Hansen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - T H Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Section for Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - R S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - J R Hov
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Section for Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - X Jiang
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - E Melum
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Section for Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Le Naour J, Liu P, Zhao L, Adjemian S, Sztupinszki Z, Taieb J, Mulot C, Silvin A, Dutertre CA, Ginhoux F, Sauvat A, Cerrato G, Castoldi F, Martins I, Stoll G, Paillet J, Mangane K, Richter C, Kepp O, Maiuri MC, Pietrocola F, Vandenabeele P, André F, Delaloge S, Szallasi Z, Laurent-Puig P, Zucman-Rossi J, Zitvogel L, Pol JG, Vacchelli E, Kroemer G. A TLR3 Ligand Reestablishes Chemotherapeutic Responses in the Context of FPR1 Deficiency. Cancer Discov 2020; 11:408-423. [PMID: 33046534 DOI: 10.1158/2159-8290.cd-20-0465] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/12/2020] [Accepted: 10/06/2020] [Indexed: 11/16/2022]
Abstract
For anthracycline-based chemotherapy to be immunogenic, dying cancer cells must release annexin A1 (ANXA1) that subsequently interacts with the pattern recognition receptor, formyl peptide receptor 1 (FPR1), on the surface of dendritic cells (DC). Approximately 30% of individuals bear loss-of-function alleles of FPR1, calling for strategies to ameliorate their anticancer immune response. Here, we show that immunotherapy with a ligand of Toll-like receptor-3, polyinosinic:polycytidylic acid (pIC), restores the deficient response to chemotherapy of tumors lacking ANXA1 developing in immunocompetent mice or those of normal cancers growing in FPR1-deficient mice. This effect was accompanied by improved DC- and T-lymphocyte-mediated anticancer immunity. Of note, carcinogen-induced breast cancers precociously developed in FPR1-deficient mice as compared with wild-type controls. A similar tendency for earlier cancer development was found in patients carrying the loss-of-function allele of FPR1. These findings have potential implications for the clinical management of FPR1-deficient patients. SIGNIFICANCE: The loss-of-function variant rs867228 in FPR1, harbored by approximately 30% of the world population, is associated with the precocious manifestation of breast, colorectal, esophageal, and head and neck carcinomas. pIC restores deficient chemotherapeutic responses in mice lacking Fpr1, suggesting a personalized strategy for compensating for the FPR1 defect.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Julie Le Naour
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, Le Kremlin Bicêtre, France
| | - Peng Liu
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sandy Adjemian
- Department of Biomedical Molecular Biology, Gent University, Ghent, Belgium.,VIB Center for Inflammation Research, Ghent, Belgium
| | - Zsofia Sztupinszki
- Computational Health Informatics Program (CHIP), Boston Children's Hospital, Boston, MA.,Harvard Medical School, Boston, MA.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Julien Taieb
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Siric CARPEM, Université de Paris, Assistance Publique-Hôpitaux de Paris, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,CNRS SNC 5096, Paris, France
| | - Aymeric Silvin
- INSERM U1015, Equipe labellisée par la Ligue contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Center of Clinical Investigations BIOTHERIS (CICBT) 1428, Villejuif, France
| | - Charles-Antoine Dutertre
- INSERM U1015, Equipe labellisée par la Ligue contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Center of Clinical Investigations BIOTHERIS (CICBT) 1428, Villejuif, France
| | | | - Allan Sauvat
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Giulia Cerrato
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, Le Kremlin Bicêtre, France
| | - Francesca Castoldi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Gautier Stoll
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Juliette Paillet
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, Le Kremlin Bicêtre, France
| | - Khady Mangane
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Cornelia Richter
- Department of Pediatrics, University Clinic Carl Gustav Carus, Technische Universitaet Dresden, Dresden, Germany
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Federico Pietrocola
- Karolinska Institute, Department of Bioscience and Nutrition, Huddinge, Sweden
| | - Peter Vandenabeele
- Department of Biomedical Molecular Biology, Gent University, Ghent, Belgium.,VIB Center for Inflammation Research, Ghent, Belgium.,Methusalem program, Ghent University, Ghent, Belgium
| | - Fabrice André
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, Le Kremlin Bicêtre, France.,Department of Medical Oncology, INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Suzette Delaloge
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Zoltan Szallasi
- Computational Health Informatics Program (CHIP), Boston Children's Hospital, Boston, MA.,Harvard Medical School, Boston, MA.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,CNRS SNC 5096, Paris, France.,Institut du Cancer Paris Carpem, AP-HP, BIOPHYGEN Department, Hôpital European Georges Pompidou, Paris, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Functional Genomics of Solid Tumors laboratory, équipe labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, Paris, France
| | - Laurence Zitvogel
- INSERM U1015, Equipe labellisée par la Ligue contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Center of Clinical Investigations BIOTHERIS (CICBT) 1428, Villejuif, France
| | - Jonathan G Pol
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France.,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Erika Vacchelli
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France. .,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris, France. .,Team "Metabolism, Cancer & Immunity," Equipe labellisée par la Ligue contre le cancer, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Institut Universitaire de France, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
Shepherd FR, McLaren JE. T Cell Immunity to Bacterial Pathogens: Mechanisms of Immune Control and Bacterial Evasion. Int J Mol Sci 2020; 21:E6144. [PMID: 32858901 PMCID: PMC7504484 DOI: 10.3390/ijms21176144] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
The human body frequently encounters harmful bacterial pathogens and employs immune defense mechanisms designed to counteract such pathogenic assault. In the adaptive immune system, major histocompatibility complex (MHC)-restricted αβ T cells, along with unconventional αβ or γδ T cells, respond to bacterial antigens to orchestrate persisting protective immune responses and generate immunological memory. Research in the past ten years accelerated our knowledge of how T cells recognize bacterial antigens and how many bacterial species have evolved mechanisms to evade host antimicrobial immune responses. Such escape mechanisms act to corrupt the crosstalk between innate and adaptive immunity, potentially tipping the balance of host immune responses toward pathological rather than protective. This review examines the latest developments in our knowledge of how T cell immunity responds to bacterial pathogens and evaluates some of the mechanisms that pathogenic bacteria use to evade such T cell immunosurveillance, to promote virulence and survival in the host.
Collapse
Affiliation(s)
| | - James E. McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
| |
Collapse
|
24
|
Tezera LB, Mansour S, Elkington P. Reconsidering the Optimal Immune Response to Mycobacterium tuberculosis. Am J Respir Crit Care Med 2020; 201:407-413. [PMID: 31657633 DOI: 10.1164/rccm.201908-1506pp] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Liku B Tezera
- National Institute for Health Research Biomedical Research Centre, School of Clinical and Experimental Sciences and
| | - Salah Mansour
- National Institute for Health Research Biomedical Research Centre, School of Clinical and Experimental Sciences and.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Paul Elkington
- National Institute for Health Research Biomedical Research Centre, School of Clinical and Experimental Sciences and.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
25
|
Sphingosine Kinase Blockade Leads to Increased Natural Killer T Cell Responses to Mantle Cell Lymphoma. Cells 2020; 9:cells9041030. [PMID: 32326225 PMCID: PMC7226300 DOI: 10.3390/cells9041030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 12/22/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive subtype of non-Hodgkin’s lymphoma. Despite being responsive to combination chemotherapy, median survival remains around 5 years due to high rates of relapse. Sphingolipid metabolism regulates MCL survival and proliferation and we found that sphingosine-1-phosphate (S1P) is upregulated in MCL cells. Therapeutic targeting of the S1P1 receptor or knockdown of sphingosine kinase 1 (SK1), the enzyme responsible for generating S1P, in human MCL cells results in a significant increase in Natural Killer T (NKT) cell activation. NKT cells recognize glycolipid antigens presented on CD1d and can reduce MCL tumor burden in vivo. Lipidomic studies identified cardiolipin, which has been reported to bind to CD1d molecules, as being upregulated in SK1 knockdown cells. We found that the pretreatment of antigen presenting cells with cardiolipin leads to increased cytokine production by NKT cell hybridomas. Furthermore, the ability of cardiolipin to activate NKT cells was dependent on the structure of its acyl chains. Collectively, these studies delineate novel pathways important for immune recognition of malignant cells and could lead to the development of new treatments for lymphoma.
Collapse
|
26
|
Navigating the Role of CD1d/Invariant Natural Killer T-cell/Glycolipid Immune Axis in Multiple Myeloma Evolution: Therapeutic Implications. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:358-365. [PMID: 32234294 DOI: 10.1016/j.clml.2020.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 12/28/2022]
Abstract
Multiple myeloma (MM) is an incurable B-cell malignancy. The immunotherapeutic approach for MM therapy is evolving. The Cd1d/invariant natural killer T-cell/glycolipid immune axis belongs to the innate immunity, and we have highlighted role in myeloma pathogenesis in the present study. The recent development of the chimeric antigen receptor (CAR19)-invariant natural killer T-cells resulted in our renewed interest in this immune system and offer new perspectives for future anti-MM immunotherapies.
Collapse
|
27
|
Visvabharathy L, Genardi S, Cao L, He Y, Alonzo F, Berdyshev E, Wang CR. Group 1 CD1-restricted T cells contribute to control of systemic Staphylococcus aureus infection. PLoS Pathog 2020; 16:e1008443. [PMID: 32343740 PMCID: PMC7188215 DOI: 10.1371/journal.ppat.1008443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/28/2020] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus (SA) is the causative agent of both skin/soft tissue infections as well as invasive bloodstream infections. Though vaccines have been developed to target both humoral and T cell-mediated immune responses against SA, they have largely failed due to lack of protective efficacy. Group 1 CD1-restricted T cells recognize lipid rather than peptide antigens. Previously found to recognize lipids derived from cell wall of Mycobacterium tuberculosis (Mtb), these cells were associated with protection against Mtb infection in humans. Using a transgenic mouse model expressing human group 1 CD1 molecules (hCD1Tg), we demonstrate that group 1 CD1-restricted T cells can recognize SA-derived lipids in both immunization and infection settings. Systemic infection of hCD1Tg mice showed that SA-specific group 1 CD1-restricted T cell response peaked at 10 days post-infection, and hCD1Tg mice displayed significantly decreased kidney pathology at this time point compared with WT control mice. Immunodominant SA lipid antigens recognized by group 1 CD1-restricted T cells were comprised mainly of cardiolipin and phosphatidyl glycerol, with little contribution from lysyl-phosphatidyl glycerol which is a unique bacterial lipid not present in mammals. Group 1 CD1-restricted T cell lines specific for SA lipids also conferred protection against SA infection in the kidney after adoptive transfer. They were further able to effectively control SA replication in vitro through direct antigen presentation by group 1 CD1-expressing BMDCs. Together, our data demonstrate a previously unknown role for group 1 CD1-restricted SA lipid-specific T cells in the control of systemic MRSA infection.
Collapse
Affiliation(s)
- Lavanya Visvabharathy
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Samantha Genardi
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Liang Cao
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Ying He
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Francis Alonzo
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, United States of America
| | - Evgeny Berdyshev
- Department of Medicine, National Jewish Health, Denver, United States of America
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| |
Collapse
|
28
|
Salminen A. Activation of immunosuppressive network in the aging process. Ageing Res Rev 2020; 57:100998. [PMID: 31838128 DOI: 10.1016/j.arr.2019.100998] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
Chronic low-grade inflammation has a key role in the aging process, a state called inflammaging. It is known that the chronic inflammatory condition generates counteracting immunosuppressive state in many diseases. Inflammaging is also associated with an immune deficiency; generally termed as immunosenescence, although it is not known whether it represents the senescence of immune cells or the active remodeling of immune system. Evidence has accumulated since the 1970's indicating that immunosenescence might be caused by an increased activity of immunosuppressive cells rather than cellular senescence. Immune cells display remarkable plasticity; many of these cells can express both proinflammatory and immunosuppressive phenotypes in a context-dependent manner. The immunosuppressive network involves the regulatory subtypes of T (Treg) and B (Breg) cells as well as regulatory phenotypes of macrophages (Mreg), dendritic (DCreg), natural killer (NKreg), and type II natural killer T (NKT) cells. The immunosuppressive network also includes monocytic (M-MDSC) and polymorphonuclear (PMN-MDSC) myeloid-derived suppressor cells which are immature myeloid cells induced by inflammatory mediators. This co-operative network is stimulated in chronic inflammatory conditions preventing excessive inflammatory responses but at the same time they exert harmful effects on the immune system and tissue homeostasis. Recent studies have revealed that the aging process is associated with the activation of immunosuppressive network, especially the functions of MDSCs, Tregs, and Mregs are increased. I will briefly review the properties of the regulatory phenotypes of immune cells and examine in detail the evidences for an activation of immunosuppressive network with aging.
Collapse
|
29
|
Kawashima M, Tokiwa M, Nishimura T, Kawata Y, Sugimoto M, Kataoka TR, Sakurai T, Iwaisako K, Suzuki E, Hagiwara M, Harris AL, Toi M. High-resolution imaging mass spectrometry combined with transcriptomic analysis identified a link between fatty acid composition of phosphatidylinositols and the immune checkpoint pathway at the primary tumour site of breast cancer. Br J Cancer 2020; 122:245-257. [PMID: 31819188 PMCID: PMC7051979 DOI: 10.1038/s41416-019-0662-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The fatty acid (FA) composition of phosphatidylinositols (PIs) is tightly regulated in mammalian tissue since its disruption impairs normal cellular functions. We previously found its significant alteration in breast cancer by using matrix-assisted laser desorption and ionisation imaging mass spectrometry (MALDI-IMS). METHODS We visualised the histological distribution of PIs containing different FAs in 65 primary breast cancer tissues using MALDI-IMS and investigated its association with clinicopathological features and gene expression profiles. RESULTS Normal ductal cells (n = 7) predominantly accumulated a PI containing polyunsaturated FA (PI-PUFA), PI(18:0/20:4). PI(18:0/20:4) was replaced by PIs containing monounsaturated FA (PIs-MUFA) in all non-invasive cancer cells (n = 12). While 54% of invasive cancer cells (n = 27) also accumulated PIs-MUFA, 46% of invasive cancer cells (n = 23) accumulated the PIs-PUFA, PI(18:0/20:3) and PI(18:0/20:4). The accumulation of PI(18:0/20:3) was associated with higher incidence of lymph node metastasis and activation of the PD-1-related immune checkpoint pathway. Fatty acid-binding protein 7 was identified as a putative molecule controlling PI composition. CONCLUSIONS MALDI-IMS identified PI composition associated with invasion and nodal metastasis of breast cancer. The accumulation of PI(18:0/20:3) could affect the PD-1-related immune checkpoint pathway, although its precise mechanism should be further validated.
Collapse
Affiliation(s)
- Masahiro Kawashima
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan.
- Molecular Oncology Laboratories, Wheaterall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - Mariko Tokiwa
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Tomomi Nishimura
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Yukiko Kawata
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Masahiro Sugimoto
- Health Promotion and Preemptive Medicine, Research and Development Center for Minimally Invasive Therapies, Tokyo Medical University, Sinjuku-ku, Tokyo, 160-8402, Japan
| | - Tatsuki R Kataoka
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takaki Sakurai
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Keiko Iwaisako
- Department of Target Therapy Oncology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eiji Suzuki
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Adrian L Harris
- Molecular Oncology Laboratories, Wheaterall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Masakazu Toi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606 8507, Japan
| |
Collapse
|
30
|
Almeida CF, Sundararaj S, Le Nours J, Praveena T, Cao B, Burugupalli S, Smith DGM, Patel O, Brigl M, Pellicci DG, Williams SJ, Uldrich AP, Godfrey DI, Rossjohn J. Distinct CD1d docking strategies exhibited by diverse Type II NKT cell receptors. Nat Commun 2019; 10:5242. [PMID: 31748533 PMCID: PMC6868179 DOI: 10.1038/s41467-019-12941-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 10/11/2019] [Indexed: 12/20/2022] Open
Abstract
Type I and type II natural killer T (NKT) cells are restricted to the lipid antigen-presenting molecule CD1d. While we have an understanding of the antigen reactivity and function of type I NKT cells, our knowledge of type II NKT cells in health and disease remains unclear. Here we describe a population of type II NKT cells that recognise and respond to the microbial antigen, α-glucuronosyl-diacylglycerol (α-GlcADAG) presented by CD1d, but not the prototypical type I NKT cell agonist, α-galactosylceramide. Surprisingly, the crystal structure of a type II NKT TCR-CD1d-α-GlcADAG complex reveals a CD1d F’-pocket-docking mode that contrasts sharply with the previously determined A’-roof positioning of a sulfatide-reactive type II NKT TCR. Our data also suggest that diverse type II NKT TCRs directed against distinct microbial or mammalian lipid antigens adopt multiple recognition strategies on CD1d, thereby maximising the potential for type II NKT cells to detect different lipid antigens. Natural killer T (NKT) cells include type I that express semi-invariant T cell receptor (TCR), and type II that cover a broader repertoire. Here the authors describe the crystal structure of a type II NKT TCR complexed with CD1d/antigen to propose that type II NKT TCRs may adapt multiple CD1d docking modes to maximise antigen recognition efficacy.
Collapse
Affiliation(s)
- Catarina F Almeida
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Srinivasan Sundararaj
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia
| | - T Praveena
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia
| | - Benjamin Cao
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Satvika Burugupalli
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dylan G M Smith
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Onisha Patel
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Manfred Brigl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Daniel G Pellicci
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Spencer J Williams
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Adam P Uldrich
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Dale I Godfrey
- Department of Microbiology & Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3010, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia. .,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
31
|
Tiwary S, Berzofsky JA, Terabe M. Altered Lipid Tumor Environment and Its Potential Effects on NKT Cell Function in Tumor Immunity. Front Immunol 2019; 10:2187. [PMID: 31620124 PMCID: PMC6759687 DOI: 10.3389/fimmu.2019.02187] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/30/2019] [Indexed: 01/26/2023] Open
Abstract
Natural killer T (NKT) cells are CD1d restricted T cells that mostly recognize lipid antigens. These cells share characteristics with both adaptive and innate immune cells and have multiple immunoregulatory roles. In a manner similar to innate immune cells, they respond quickly to stimuli and secrete large amounts of cytokines, amplifying and modulating the immune response. As T cells, they express T cell receptors (TCRs) and respond in an antigen-specific manner like conventional T cells. There are at least two subtypes of NKT cells, type I and type II, that differ in the nature of their TCR, either semi-invariant (type I) or diverse (type II). The two sub-types generally have opposing functions in tumor immunity, with type I promoting and type II suppressing tumor immunity, and they cross-regulate each other, forming an immunoregulatory axis. The tumor has multiple mechanisms by which it can evade immune-surveillance. One such mechanism involves alteration in tumor lipid repertoire and accumulation of lipids and fatty acids that favor tumor growth and evade anti-tumor immunity. Since NKT cells mostly recognize lipid antigens, an altered tumor lipid metabolic profile will also alter the repertoire of lipid antigens that can potentially affect their immune-modulatory function. In this review, we will explore the effects of alterations in the lipid metabolites on tumor growth, antigen cross-presentation, and overall effect on anti-tumor immunity, especially in the context of NKT cells.
Collapse
Affiliation(s)
- Shweta Tiwary
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Masaki Terabe
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
32
|
Ying G, Wang J, Mallevaey T, Van Calenbergh S, Zajonc DM. Structural basis of NKT cell inhibition using the T-cell receptor-blocking anti-CD1d antibody 1B1. J Biol Chem 2019; 294:12947-12956. [PMID: 31296659 PMCID: PMC6721955 DOI: 10.1074/jbc.ra119.009403] [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: 05/16/2019] [Revised: 07/05/2019] [Indexed: 01/07/2023] Open
Abstract
Natural killer T (NKT) cells are a subset of T lymphocytes that recognize glycolipid antigens presented by the CD1d molecule (CD1d). They rapidly respond to antigen challenge and can activate both innate and adaptive immune cells. To study the role of antigen presentation in NKT cell activation, previous studies have developed several anti-CD1d antibodies that block CD1d binding to T-cell receptors (TCRs). Antibodies that are specific to both CD1d and the presented antigen can only be used to study the function of only a limited number of antigens. In contrast, antibodies that bind CD1d and block TCR binding regardless of the presented antigen can be widely used to assess the role of TCR-mediated NKT cell activation in various disease models. Here, we report the crystal structure of the widely used anti-mouse CD1d antibody 1B1 bound to CD1d at a resolution of 2.45 Å and characterized its binding to CD1d-presented glycolipids. We observed that 1B1 uses a long hydrophobic H3 loop that is inserted deep into the binding groove of CD1d where it makes intimate nonpolar contacts with the lipid backbone of an incorporated spacer lipid. Using an NKT cell agonist that has a modified sphingosine moiety, we further demonstrate that 1B1 in its monovalent form cannot block TCR-mediated NKT cell activation, because 1B1 fails to bind with high affinity to mCD1d. Our results suggest potential limitations of using 1B1 to assess antigen recognition by NKT cells, especially when investigating antigens that do not follow the canonical two alkyl-chain rule.
Collapse
Affiliation(s)
- Ge Ying
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, California 92037
| | - Jing Wang
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, California 92037
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (FFW), Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Dirk M. Zajonc
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, California 92037,Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium, To whom correspondence should be addressed:
Cancer Immunology Discovery, Pfizer, San Diego, CA 92121. E-mail:
| |
Collapse
|
33
|
Sebode M, Wigger J, Filpe P, Fischer L, Weidemann S, Krech T, Weiler-Normann C, Peiseler M, Hartl J, Tolosa E, Herkel J, Schramm C, Lohse AW, Arrenberg P. Inflammatory Phenotype of Intrahepatic Sulfatide-Reactive Type II NKT Cells in Humans With Autoimmune Hepatitis. Front Immunol 2019; 10:1065. [PMID: 31191516 PMCID: PMC6546815 DOI: 10.3389/fimmu.2019.01065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/25/2019] [Indexed: 01/06/2023] Open
Abstract
Background: Natural Killer T (NKT) cells are CD1d-restricted innate-like T cells that can rapidly release stored cytokines upon recognition of lipid antigens. In mice, type I NKT cells seem to promote liver inflammation, whereas type II NKT cells seem to restrict hepatitis. Here, we aimed at characterizing the role of human type I and type II NKT in patients with autoimmune hepatitis (AIH). Methods: NKT cells were analyzed by flow cytometry in peripheral blood and liver of AIH patients and control groups. α-galactosylceramide-loaded or sulfatide-loaded tetramers were used to detect type I or II NKT cells, respectively. Hepatic CD1d was stained by in situ-hybridization of liver biopsies. Results and Conclusions: Type II NKT cells were more prevalent in human peripheral blood and liver than type I NKT cells. In AIH patients, the frequency of sulfatide-reactive type II NKT cells was significantly increased in peripheral blood (0.11% of peripheral blood leukocytes) and liver (3.78% of intrahepatic leukocytes) compared to healthy individuals (0.05% and 1.82%) and patients with drug-induced liver injury (0.06% and 2.03%; p < 0.05). Intrahepatic type II NKT cells of AIH patients had a different cytokine profile than healthy subjects with an increased frequency of TNFα (77.8% vs. 59.1%, p < 0.05), decreased IFNγ (32.7% vs. 63.0%, p < 0.05) and a complete lack of IL-4 expressing cells (0% vs. 2.1%, p < 0.05). T cells in portal tracts expressed significantly more CD1d-RNA in AIH livers compared to controls. This study supports that in contrast to their assumed protective role in mice, human intrahepatic, sulfatide-reactive type II NKT cells displayed a proinflammatory cytokine profile in patients with AIH. Infiltrating T cells in portal areas of AIH patients overexpressed CD1d and could thereby activate type II NKT cells.
Collapse
Affiliation(s)
- Marcial Sebode
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Jennifer Wigger
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pamela Filpe
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lutz Fischer
- Department of Hepatobiliary Surgery and Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sören Weidemann
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Krech
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Weiler-Normann
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Moritz Peiseler
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Johannes Hartl
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Eva Tolosa
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Herkel
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany.,Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Philomena Arrenberg
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
Fu S, Zhu S, Tian C, Bai S, Zhang J, Zhan C, Xie D, Wang L, Li Z, Li J, Zhang H, Zhou R, Tian Z, Xu T, Bai L. Immunometabolism regulates TCR recycling and iNKT cell functions. Sci Signal 2019; 12:12/570/eaau1788. [PMID: 30808817 DOI: 10.1126/scisignal.aau1788] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Invariant natural killer T (iNKT) cells are innate-like T lymphocytes that express an invariant T cell receptor (TCR), which recognizes glycolipid antigens presented on CD1d molecules. These cells are phenotypically and functionally distinct from conventional T cells. When we characterized the metabolic activity of iNKT cells, consistent with their activated phenotype, we found that they had much less mitochondrial respiratory capacity but increased glycolytic activity in comparison to naïve conventional CD4+ T cells. After TCR engagement, iNKT cells further increased aerobic glycolysis, which was important for the expression of interferon-γ (IFN-γ). Glycolytic metabolism promoted the translocation of hexokinase-II to mitochondria and the activation of mammalian target of rapamycin complex 2 (mTORC2). Inhibiting glycolysis reduced the activity of Akt and PKCθ, which inhibited TCR recycling and accumulation within the immune synapse. Diminished TCR accumulation in the immune synapse reduced the activation of proximal and distal TCR signaling pathways and IFN-γ production in activated iNKT cells. Our studies demonstrate that glycolytic metabolism augments TCR signaling duration and IFN-γ production in iNKT cells by increasing TCR recycling.
Collapse
Affiliation(s)
- Sicheng Fu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Shasha Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei 230022, China
| | - Chenxi Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Shiyu Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Jiqian Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230022, China
| | - Chonglun Zhan
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Di Xie
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Lu Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Zonghong Li
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jie Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Huimin Zhang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Rongbin Zhou
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Signaling Network, Hefei 230027, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Tao Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Li Bai
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China. .,Innovation Center for Cell Signaling Network, Hefei 230027, China
| |
Collapse
|
35
|
Halder RC, Tran C, Prasad P, Wang J, Nallapothula D, Ishikawa T, Wang M, Zajonc DM, Singh RR. Self-glycerophospholipids activate murine phospholipid-reactive T cells and inhibit iNKT cell activation by competing with ligands for CD1d loading. Eur J Immunol 2018; 49:242-254. [PMID: 30508304 DOI: 10.1002/eji.201847717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/15/2018] [Accepted: 11/27/2018] [Indexed: 01/12/2023]
Abstract
Glycosphingolipids and glycerophospholipids bind CD1d. Glycosphingolipid-reactive invariant NKT-cells (iNKT) exhibit myriad immune effects, however, little is known about the functions of phospholipid-reactive T cells (PLT). We report that the normal mouse immune repertoire contains αβ T cells, which recognize self-glycerophospholipids such as phosphatidic acid (PA) in a CD1d-restricted manner and don't cross-react with iNKT-cell ligands. PA bound to CD1d in the absence of lipid transfer proteins. Upon in vivo priming, PA induced an expansion and activation of T cells in Ag-specific manner. Crystal structure of the CD1d:PA complex revealed that the ligand is centrally located in the CD1d-binding groove opening for TCR recognition. Moreover, the increased flexibility of the two acyl chains in diacylglycerol ligands and a less stringent-binding orientation for glycerophospholipids as compared with the bindings of glycosphingolipids may allow glycerophospholipids to readily occupy CD1d. Indeed, PA competed with α-galactosylceramide to load onto CD1d, leading to reduced expression of CD1d:α-galactosylceramide complexes on the surface of dendritic cells. Consistently, glycerophospholipids reduced iNKT-cell proliferation, expansion, and cytokine production in vitro and in vivo. Such superior ability of self-glycerophospholipids to compete with iNKT-cell ligands to occupy CD1d may help maintain homeostasis between the diverse subsets of lipid-reactive T cells, with important pathogenetic and therapeutic implications.
Collapse
Affiliation(s)
- Ramesh Chandra Halder
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Cynthia Tran
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Priti Prasad
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA.,Molecular Toxicology Interdepartmental Program, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Jing Wang
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Dhiraj Nallapothula
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Tatsuya Ishikawa
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Meiying Wang
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Dirk M Zajonc
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, CA, USA.,Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA.,Molecular Toxicology Interdepartmental Program, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
36
|
Unappreciated diversity within the pool of CD1d-restricted T cells. Semin Cell Dev Biol 2018; 84:42-47. [DOI: 10.1016/j.semcdb.2017.11.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022]
|
37
|
Collins JM, Walker DI, Jones DP, Tukvadze N, Liu KH, Tran VT, Uppal K, Frediani JK, Easley KA, Shenvi N, Khadka M, Ortlund EA, Kempker RR, Blumberg HM, Ziegler TR. High-resolution plasma metabolomics analysis to detect Mycobacterium tuberculosis-associated metabolites that distinguish active pulmonary tuberculosis in humans. PLoS One 2018; 13:e0205398. [PMID: 30308073 PMCID: PMC6181350 DOI: 10.1371/journal.pone.0205398] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 09/25/2018] [Indexed: 12/01/2022] Open
Abstract
Introduction Pulmonary tuberculosis (TB) is a major worldwide health problem that lacks robust blood-based biomarkers for detection of active disease. High-resolution metabolomics (HRM) is an innovative method to discover low-abundance metabolites as putative blood biomarkers to detect TB disease, including those known to be produced by the causative organism, Mycobacterium tuberculosis (Mtb). Methods We used HRM profiling to measure the plasma metabolome for 17 adults with active pulmonary TB disease and 16 of their household contacts without active TB. We used a suspect screening approach to identify metabolites previously described in cell culture studies of Mtb based on retention time and accurate mass matches. Results The association of relative metabolite abundance in active TB disease subjects compared to their household contacts predicted three Mtb-associated metabolites that were significantly increased in the active TB patients based on accurate mass matches: phosphatidylglycerol (PG) (16:0_18:1), lysophosphatidylinositol (Lyso-PI) (18:0) and acylphosphatidylinositol mannoside (Ac1PIM1) (56:1) (p<0.001 for all). These three metabolites provided excellent classification accuracy for active TB disease (AUC = 0.97). Ion dissociation spectra (tandem MS/MS) supported the identification of PG (16:0_18:1) and Lyso-PI (18:0) in the plasma of patients with active TB disease, though the identity of Ac1PIM1 could not be definitively confirmed. Conclusions Presence of the Mtb-associated lipid metabolites PG (16:0_18:1) and Lyso-PI (18:0) in plasma accurately identified patients with active TB disease. Consistency of in vitro and in vivo data suggests suitability for exploring these in future studies for possible development as TB disease biomarkers.
Collapse
Affiliation(s)
- Jeffrey M. Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| | - Douglas I. Walker
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Dean P. Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nestani Tukvadze
- National Center for Tuberculosis and Lung Disease, Tbilisi, Georgia
| | - Ken H. Liu
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - ViLinh T. Tran
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Karan Uppal
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jennifer K. Frediani
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, Georgia, United States of America
| | - Kirk A. Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Neeta Shenvi
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
| | - Manoj Khadka
- Emory Integrated Lipidomics Core, Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Eric A. Ortlund
- Emory Integrated Lipidomics Core, Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Russell R. Kempker
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Henry M. Blumberg
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Thomas R. Ziegler
- Division of Endocrinology, Metabolism and Lipids and Center for Clinical and Molecular Nutrition, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Section of Endocrinology, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, United States of America
| |
Collapse
|
38
|
Singh AK, Tripathi P, Cardell SL. Type II NKT Cells: An Elusive Population With Immunoregulatory Properties. Front Immunol 2018; 9:1969. [PMID: 30210505 PMCID: PMC6120993 DOI: 10.3389/fimmu.2018.01969] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022] Open
Abstract
Natural killer T (NKT) cells are unique unconventional T cells that are reactive to lipid antigens presented on the non-polymorphic major histocompatibility class (MHC) I-like molecule CD1d. They have characteristics of both innate and adaptive immune cells, and have potent immunoregulatory roles in tumor immunity, autoimmunity, and infectious diseases. Based on their T cell receptor (TCR) expression, NKT cells are divided into two subsets, type I NKT cells with an invariant TCRα-chain (Vα24 in humans, Vα14 in mice) and type II NKT cells with diverse TCRs. While type I NKT cells are well-studied, knowledge about type II NKT cells is still limited, and it is to date only possible to identify subsets of this population. However, recent advances have shown that both type I and type II NKT cells play important roles in many inflammatory situations, and can sometimes regulate the functions of each other. Type II NKT cells can be both protective and pathogenic. Here, we review current knowledge on type II NKT cells and their functions in different disease settings and how these cells can influence immunological outcomes.
Collapse
Affiliation(s)
- Avadhesh Kumar Singh
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Prabhanshu Tripathi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanna L Cardell
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
39
|
Trottein F, Paget C. Natural Killer T Cells and Mucosal-Associated Invariant T Cells in Lung Infections. Front Immunol 2018; 9:1750. [PMID: 30116242 PMCID: PMC6082944 DOI: 10.3389/fimmu.2018.01750] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
The immune system has been traditionally divided into two arms called innate and adaptive immunity. Typically, innate immunity refers to rapid defense mechanisms that set in motion within minutes to hours following an insult. Conversely, the adaptive immune response emerges after several days and relies on the innate immune response for its initiation and subsequent outcome. However, the recent discovery of immune cells displaying merged properties indicates that this distinction is not mutually exclusive. These populations that span the innate-adaptive border of immunity comprise, among others, CD1d-restricted natural killer T cells and MR1-restricted mucosal-associated invariant T cells. These cells have the unique ability to swiftly activate in response to non-peptidic antigens through their T cell receptor and/or to activating cytokines in order to modulate many aspects of the immune response. Despite they recirculate all through the body via the bloodstream, these cells mainly establish residency at barrier sites including lungs. Here, we discuss the current knowledge into the biology of these cells during lung (viral and bacterial) infections including activation mechanisms and functions. We also discuss future strategies targeting these cell types to optimize immune responses against respiratory pathogens.
Collapse
Affiliation(s)
- François Trottein
- Univ. Lille, U1019 – UMR 8204 – CIIL – Centre d’Infection et d’Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Christophe Paget
- Institut National de la Santé et de la Recherche Médicale U1100, Centre d’Etude des Pathologies Respiratoires (CEPR), Tours, France
- Université de Tours, Tours, France
| |
Collapse
|
40
|
Doherty DG, Melo AM, Moreno-Olivera A, Solomos AC. Activation and Regulation of B Cell Responses by Invariant Natural Killer T Cells. Front Immunol 2018; 9:1360. [PMID: 29967611 PMCID: PMC6015876 DOI: 10.3389/fimmu.2018.01360] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/31/2018] [Indexed: 12/16/2022] Open
Abstract
CD1d-restricted invariant natural killer T (iNKT) cells play central roles in the activation and regulation of innate and adaptive immunity. Cytokine-mediated and CD1d-dependent interactions between iNKT cells and myeloid and lymphoid cells enable iNKT cells to contribute to the activation of multiple cell types, with important impacts on host immunity to infection and tumors and on the prevention of autoimmunity. Here, we review the mechanisms by which iNKT cells contribute to B cell maturation, antibody and cytokine production, and antigen presentation. Cognate interactions with B cells contribute to the rapid production of antibodies directed against conserved non-protein antigens resulting in rapid but short-lived innate humoral immunity. iNKT cells can also provide non-cognate help for the generation of antibodies directed against protein antigens, by promoting the activation of follicular helper T cells, resulting in long-lasting adaptive humoral immunity and B cell memory. iNKT cells can also regulate humoral immunity by promoting the development of autoreactive B cells into regulatory B cells. Depletions and functional impairments of iNKT cells are found in patients with infectious, autoimmune and malignant diseases associated with altered B cell function and in murine models of these conditions. The adjuvant and regulatory activities that iNKT cells have for B cells makes them attractive therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Derek G Doherty
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Ashanty M Melo
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Ana Moreno-Olivera
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Andreas C Solomos
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
41
|
DeWitt WS, Yu KKQ, Wilburn DB, Sherwood A, Vignali M, Day CL, Scriba TJ, Robins HS, Swanson WJ, Emerson RO, Bradley PH, Seshadri C. A Diverse Lipid Antigen-Specific TCR Repertoire Is Clonally Expanded during Active Tuberculosis. THE JOURNAL OF IMMUNOLOGY 2018; 201:888-896. [PMID: 29914888 DOI: 10.4049/jimmunol.1800186] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/21/2018] [Indexed: 01/06/2023]
Abstract
Human T cells that recognize lipid Ags presented by highly conserved CD1 proteins often express semi-invariant TCRs, but the true diversity of lipid Ag-specific TCRs remains unknown. We use CD1b tetramers and high-throughput immunosequencing to analyze thousands of TCRs from ex vivo-sorted or in vitro-expanded T cells specific for the mycobacterial lipid Ag, glucose monomycolate. Our results reveal a surprisingly diverse repertoire resulting from editing of germline-encoded gene rearrangements analogous to MHC-restricted TCRs. We used a distance-based metric (TCRDist) to show how this diverse TCR repertoire builds upon previously reported conserved motifs by including subject-specific TCRs. In a South African cohort, we show that TCRDist can identify clonal expansion of diverse glucose monomycolate-specific TCRs and accurately distinguish patients with active tuberculosis from control subjects. These data suggest that similar mechanisms govern the selection and expansion of peptide and lipid Ag-specific T cells despite the nonpolymorphic nature of CD1.
Collapse
Affiliation(s)
- William S DeWitt
- Adaptive Biotechnologies, Seattle, WA 98102.,Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109.,Department of Genome Sciences, University of Washington, Seattle, WA 98195
| | - Krystle K Q Yu
- Department of Medicine, University of Washington, Seattle, WA 98195
| | - Damien B Wilburn
- Department of Genome Sciences, University of Washington, Seattle, WA 98195
| | | | | | - Cheryl L Day
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322.,Emory Vaccine Center, Atlanta, GA 30329
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.,Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Harlan S Robins
- Adaptive Biotechnologies, Seattle, WA 98102.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Willie J Swanson
- Department of Genome Sciences, University of Washington, Seattle, WA 98195
| | | | - Philip H Bradley
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and.,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Chetan Seshadri
- Department of Medicine, University of Washington, Seattle, WA 98195;
| |
Collapse
|
42
|
Shahine A, Van Rhijn I, Cheng TY, Iwany S, Gras S, Moody DB, Rossjohn J. A molecular basis of human T cell receptor autoreactivity toward self-phospholipids. Sci Immunol 2018; 2:2/16/eaao1384. [PMID: 29054999 PMCID: PMC6649662 DOI: 10.1126/sciimmunol.aao1384] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 08/25/2017] [Indexed: 12/14/2022]
Abstract
Human T cell autoreactivity toward lipid antigens presented by CD1 proteins can manifest in numerous diseases, including psoriasis, contact hypersensitivities, and allergies. However, the molecular mechanisms for regulating T cell autoreactivity toward lipid antigens remain unclear. We determined the basis for T cell receptor (TCR) autoreactivity toward CD1b bound to self-phospholipids. The spectrum of self-antigens captured by CD1b skews toward abundant membrane phospholipids such as phosphatidylcholine and phosphatidylethanolamine. However, TCRs can specifically recognize rare phospholipids, including phosphatidylglycerol (PG). The structure of an autoreactive TCR bound to CD1b-PG shows that discrimination occurs through a marked induced fit movement of PG so that its polar head group fits snugly into the cationic cup of the TCR. Conversely, TCR binding toward ubiquitous self-phospholipids was sterically or electrostatically repelled. Accordingly, we describe a mechanism of TCR autoreactivity toward rare phospholipids and avoidance of autoreactivity to the most abundant self-phospholipids.
Collapse
Affiliation(s)
- Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Ildiko Van Rhijn
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584CL Utrecht, Netherlands
| | - Tan-Yun Cheng
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sarah Iwany
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - D Branch Moody
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia. .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
43
|
Gras S, Van Rhijn I, Shahine A, Le Nours J. Molecular recognition of microbial lipid-based antigens by T cells. Cell Mol Life Sci 2018; 75:1623-1639. [PMID: 29340708 PMCID: PMC6328055 DOI: 10.1007/s00018-018-2749-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/17/2017] [Accepted: 01/08/2018] [Indexed: 02/06/2023]
Abstract
The immune system has evolved to protect hosts from pathogens. T cells represent a critical component of the immune system by their engagement in host defence mechanisms against microbial infections. Our knowledge of the molecular recognition by T cells of pathogen-derived peptidic antigens that are presented by the major histocompatibility complex glycoproteins is now well established. However, lipids represent an additional, distinct chemical class of molecules that when presented by the family of CD1 antigen-presenting molecules can serve as antigens, and be recognized by specialized subsets of T cells leading to antigen-specific activation. Over the past decades, numerous CD1-presented self- and bacterial lipid-based antigens have been isolated and characterized. However, our understanding at the molecular level of T cell immunity to CD1 molecules presenting microbial lipid-based antigens is still largely unexplored. Here, we review the insights and the molecular basis underpinning the recognition of microbial lipid-based antigens by T cells.
Collapse
Affiliation(s)
- Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia
| | - Ildiko Van Rhijn
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital/Harvard Medical School, Boston, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, University Utrecht, Utrecht, The Netherlands
| | - Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
44
|
Burugupalli S, Richardson MB, Williams SJ. Total synthesis and mass spectrometric analysis of a Mycobacterium tuberculosis phosphatidylglycerol featuring a two-step synthesis of (R)-tuberculostearic acid. Org Biomol Chem 2018; 15:7422-7429. [PMID: 28831486 DOI: 10.1039/c7ob01786c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We report the total synthesis of (R)-tuberculostearic acid-containing Mycobacterium tuberculosis phosphatidylglycerol (PG). The approach features a two-step synthesis of (R)-tuberculostearic acid, involving an (S)-citronellyl bromide linchpin, and the phosphoramidite-assisted assembly of the full PG structure. Collision-induced dissociation mass spectrometry of two chemically-synthesized PG acyl regioisomers revealed diagnostic product ions formed by preferential loss of carboxylate at the secondary (sn-2) position.
Collapse
Affiliation(s)
- Satvika Burugupalli
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia 3010.
| | - Mark B Richardson
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia 3010.
| | - Spencer J Williams
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia 3010.
| |
Collapse
|
45
|
O'Donnell VB, Rossjohn J, Wakelam MJ. Phospholipid signaling in innate immune cells. J Clin Invest 2018; 128:2670-2679. [PMID: 29683435 DOI: 10.1172/jci97944] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Phospholipids comprise a large body of lipids that define cells and organelles by forming membrane structures. Importantly, their complex metabolism represents a highly controlled cellular signaling network that is essential for mounting an effective innate immune response. Phospholipids in innate cells are subject to dynamic regulation by enzymes, whose activities are highly responsive to activation status. Along with their metabolic products, they regulate multiple aspects of innate immune cell biology, including shape change, aggregation, blood clotting, and degranulation. Phospholipid hydrolysis provides substrates for cell-cell communication, enables regulation of hemostasis, immunity, thrombosis, and vascular inflammation, and is centrally important in cardiovascular disease and associated comorbidities. Phospholipids themselves are also recognized by innate-like T cells, which are considered essential for recognition of infection or cancer, as well as self-antigens. This Review describes the major phospholipid metabolic pathways present in innate immune cells and summarizes the formation and metabolism of phospholipids as well as their emerging roles in cell biology and disease.
Collapse
Affiliation(s)
- Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jamie Rossjohn
- Systems Immunity Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, and.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
46
|
Migalovich Sheikhet H, Villacorta Hidalgo J, Fisch P, Balbir-Gurman A, Braun-Moscovici Y, Bank I. Dysregulated CD25 and Cytokine Expression by γδ T Cells of Systemic Sclerosis Patients Stimulated With Cardiolipin and Zoledronate. Front Immunol 2018; 9:753. [PMID: 29706966 PMCID: PMC5909681 DOI: 10.3389/fimmu.2018.00753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/26/2018] [Indexed: 11/13/2022] Open
Abstract
Objectives γδ T cells, a non-conventional innate lymphocyte subset containing cells that can be activated by lipids and phosphoantigens, are abnormally regulated in systemic sclerosis (SSc). To further evaluate the significance of this dysregulation, we compared how exposure to an autoantigenic lipid, cardiolipin (CL), during co-stimulation with an amino-bisphosphonate (zoledronate, zol), affects the activation and cytokine production of SSc and healthy control (HC) γδ T cells. Methods Expression of CD25 on Vγ9+, Vδ1+, and total CD3+ T cells in cultured peripheral blood mononuclear cells (PBMCs), their binding of CD1d tetramers, and the effect of monoclonal antibody (mAb) blockade of CD1d were monitored by flow cytometry after 4 days of in vitro culture. Intracellular production of IFNγ and IL-4 was assessed after overnight culture. Results Percentages of CD25+ among CD3+ and Vδ1+ T cells were elevated significantly in short-term cultured SSc PBMC compared to HC. In SSc but not HC, CL and zol, respectively, suppressed %CD25+ Vγ9+ and Vδ1+ T cells but, when combined, CL + zol significantly activated both subsets in HC and partially reversed inhibition by the individual reagents in SSc. Importantly, Vδ1+ T cells in both SSc and HC were highly reactive with lipid presenting CD1d tetramers, and a CD1d-blocking mAb decreased CL-induced enhancement of %SSc CD25+ Vδ1+ T cells in the presence of zol. %IFNγ+ cells among Vγ9+ T cells of SSc was lower than HC cultured in medium, CL, zol, or CL + zol, whereas %IFNγ+ Vδ1+ T cells was lower only in the presence of CL or CL + zol. %IL-4+ T cells were similar in SSc and HC in all conditions, with the exception of being increased in SSc Vγ9+ T cells in the presence of CL. Conclusion Abnormal functional responses of γδ T cell subsets to stimulation by CL and phosphoantigens in SSc may contribute to fibrosis and immunosuppression, characteristics of this disease.
Collapse
Affiliation(s)
| | | | - Paul Fisch
- Department of Clinical Pathology, University of Freiburg Medical Center, Freiburg, Germany
| | - Alexandra Balbir-Gurman
- B. Shine Rheumatology Unit, Rambam Health Care Campus and The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yolanda Braun-Moscovici
- B. Shine Rheumatology Unit, Rambam Health Care Campus and The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ilan Bank
- Laboratory of Immune-Regulation, Sheba Medical Center, Ramat Gan, Israel.,Rheumatology Unit, Tel Aviv University, Tel Aviv, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
47
|
Nishioka Y, Masuda S, Tomaru U, Ishizu A. CD1d-Restricted Type II NKT Cells Reactive With Endogenous Hydrophobic Peptides. Front Immunol 2018; 9:548. [PMID: 29599785 PMCID: PMC5862807 DOI: 10.3389/fimmu.2018.00548] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 03/05/2018] [Indexed: 11/13/2022] Open
Abstract
NKT cells belong to a distinct subset of T cells that recognize hydrophobic antigens presented by major histocompatibility complex class I-like molecules, such as CD1d. Because NKT cells stimulated by antigens can activate or suppress other immunocompetent cells through an immediate production of a large amount of cytokines, they are regarded as immunological modulators. CD1d-restricted NKT cells are classified into two subsets, namely, type I and type II. CD1d-restricted type I NKT cells express invariant T cell receptors (TCRs) and react with lipid antigens, including the marine sponge-derived glycolipid α-galactosylceramide. On the contrary, CD1d-restricted type II NKT cells recognize a wide variety of antigens, including glycolipids, phospholipids, and hydrophobic peptides, by their diverse TCRs. In this review, we focus particularly on CD1d-restricted type II NKT cells that recognize endogenous hydrophobic peptides presented by CD1d. Previous studies have demonstrated that CD1d-restricted type I NKT cells usually act as pro-inflammatory cells but sometimes behave as anti-inflammatory cells. It has been also demonstrated that CD1d-restricted type II NKT cells play opposite roles to CD1d-restricted type I NKT cells; thus, they function as anti-inflammatory or pro-inflammatory cells depending on the situation. In line with this, CD1d-restricted type II NKT cells that recognize type II collagen peptide have been demonstrated to act as anti-inflammatory cells in diverse inflammation-induction models in mice, whereas pro-inflammatory CD1d-restricted type II NKT cells reactive with sterol carrier protein 2 peptide have been demonstrated to be involved in the development of small vessel vasculitis in rats.
Collapse
Affiliation(s)
- Yusuke Nishioka
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Ishizu
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
48
|
Krijgsman D, Hokland M, Kuppen PJK. The Role of Natural Killer T Cells in Cancer-A Phenotypical and Functional Approach. Front Immunol 2018. [PMID: 29535734 PMCID: PMC5835336 DOI: 10.3389/fimmu.2018.00367] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Natural killer T (NKT) cells are a subset of CD1d-restricted T cells at the interface between the innate and adaptive immune system. NKT cells can be subdivided into functional subsets that respond rapidly to a wide variety of glycolipids and stress-related proteins using T- or natural killer (NK) cell-like effector mechanisms. Because of their major modulating effects on immune responses via secretion of cytokines, NKT cells are also considered important players in tumor immunosurveillance. During early tumor development, T helper (TH)1-like NKT cell subsets have the potential to rapidly stimulate tumor-specific T cells and effector NK cells that can eliminate tumor cells. In case of tumor progression, NKT cells may become overstimulated and anergic leading to deletion of a part of the NKT cell population in patients via activation-induced cell death. In addition, the remaining NKT cells become hyporesponsive, or switch to immunosuppressive TH2-/T regulatory-like NKT cell subsets, thereby facilitating tumor progression and immune escape. In this review, we discuss this important role of NKT cells in tumor development and we conclude that there should be three important focuses of future research in cancer patients in relation with NKT cells: (1) expansion of the NKT cell population, (2) prevention and breaking of NKT cell anergy, and (3) skewing of NKT cells toward TH1-like subsets with antitumor activity.
Collapse
Affiliation(s)
- Daniëlle Krijgsman
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
49
|
Kato S, Berzofsky JA, Terabe M. Possible Therapeutic Application of Targeting Type II Natural Killer T Cell-Mediated Suppression of Tumor Immunity. Front Immunol 2018; 9:314. [PMID: 29520281 PMCID: PMC5827362 DOI: 10.3389/fimmu.2018.00314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/05/2018] [Indexed: 12/17/2022] Open
Abstract
Natural killer T (NKT) cells are a unique T cell subset that exhibits characteristics from both the innate immune cells and T cells. There are at least two subsets of NKT cells, type I and type II. These two subsets of NKT cells have opposite functions in antitumor immunity. Type I NKT cells usually enhance and type II NKT cells suppress antitumor immunity. In addition, these two subsets of NKT cells cross-regulate each other. In this review, we mainly focus on immunosuppressive NKT cells, type II NKT cells. After summarizing their definition, experimental tools to study them, and subsets of them, we will discuss possible therapeutic applications of type II NKT cell pathway targeted therapies.
Collapse
Affiliation(s)
- Shingo Kato
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Jay A. Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Masaki Terabe
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
50
|
Torina A, Guggino G, La Manna MP, Sireci G. The Janus Face of NKT Cell Function in Autoimmunity and Infectious Diseases. Int J Mol Sci 2018; 19:ijms19020440. [PMID: 29389901 PMCID: PMC5855662 DOI: 10.3390/ijms19020440] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/19/2022] Open
Abstract
Natural killer T cells (NKT) are a subset of T lymphocytes bridging innate and adaptive immunity. These cells recognize self and microbial glycolipids bound to non-polymorphic and highly conserved CD1d molecules. Three NKT cell subsets, type I, II, and NKT-like expressing different antigen receptors (TCR) were described and TCR activation promotes intracellular events leading to specific functional activities. NKT can exhibit different functions depending on the secretion of soluble molecules and the interaction with other cell types. NKT cells act as regulatory cells in the defense against infections but, on the other hand, their effector functions can be involved in the pathogenesis of several inflammatory disorders due to their exposure to different microbial or self-antigens, respectively. A deep understanding of the biology and functions of type I, II, and NKT-like cells as well as their interplay with cell types acting in innate (neuthrophils, innate lymphoid cells, machrophages, and dendritic cells) and adaptive immunity (CD4⁺,CD8⁺, and double negative T cells) should be important to design potential immunotherapies for infectious and autoimmune diseases.
Collapse
Affiliation(s)
- Alessandra Torina
- Experimental Zooprophylactic Institute of Sicily, Via Marinuzzi 3, 90100 Palermo, Italy.
| | - Giuliana Guggino
- Biomedical Department of Internal and Specialized Medicine, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90100 Palermo, Italy.
| | - Marco Pio La Manna
- Department of Biopathology and Medical Biotechnology, Section of General Pathology, University of Palermo, Via del Vespro 129, 90100 Palermo, Italy.
- Central Laboratory Advanced Diagnostic and Biological Research, University Hospital, Via del Vespro 129, 90100 Palermo, Italy.
| | - Guido Sireci
- Department of Biopathology and Medical Biotechnology, Section of General Pathology, University of Palermo, Via del Vespro 129, 90100 Palermo, Italy.
- Central Laboratory Advanced Diagnostic and Biological Research, University Hospital, Via del Vespro 129, 90100 Palermo, Italy.
| |
Collapse
|