1
|
Jaiswal S, Fatima S, Velarde de la Cruz E, Kumar S. Unraveling the role of the immune landscape in tuberculosis granuloma. Tuberculosis (Edinb) 2025; 152:102615. [PMID: 40020281 DOI: 10.1016/j.tube.2025.102615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Despite significant advances in research over the past century, Tuberculosis (TB) remains a formidable global health concern. TB granulomas are organized structures composed of immune cells, that serve as the body's primary defense against the spread of Mycobacterium tuberculosis (Mtb). The immune landscape of TB granulomas involves a complex array of immune cells, including CD4+ and CD8+ T cells, B cells, NK cells, and others, which collectively influence the fate of the granuloma. B cells contribute to the formation of the granuloma's germinal center, while the functional state of T cells-particularly their ability to control infection-dictates whether the granuloma is controlling or proliferative. The intricate interplay between T cells and the dynamic microenvironment of the granuloma plays a pivotal role in determining the outcome of the infection. However, several aspects of the immunological basis of tuberculosis are still unknown. This review delves into the immunological landscape of TB granuloma, focusing on the dynamic cellular interplay within the granuloma and its profound influence on disease pathogenesis.
Collapse
|
2
|
Castaño AR, Porcelli SA. Editorial: Cognate recognition, functional properties and immunotherapeutic applications of iNKT cells: leveling the road to the clinic. Front Immunol 2025; 16:1552225. [PMID: 39917293 PMCID: PMC11798922 DOI: 10.3389/fimmu.2025.1552225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Affiliation(s)
- A. Raul Castaño
- Department of Cellular Biology, Physiology and Immunology, Autonomous University of Barcelona, Barcelona, Spain
| | - Steven Anthony Porcelli
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
3
|
Sakai Y, Asa M, Hirose M, Kusuhara W, Fujiwara N, Tamashima H, Ikazaki T, Oka S, Kuraba K, Tanaka K, Yoshiyama T, Nagae M, Hoshino Y, Motooka D, Van Rhijn I, Lu X, Ishikawa E, Moody DB, Kato T, Inuki S, Hirai G, Yamasaki S. A conserved human CD4+ T cell subset recognizing the mycobacterial adjuvant trehalose monomycolate. J Clin Invest 2024; 135:e185443. [PMID: 39718834 PMCID: PMC11910211 DOI: 10.1172/jci185443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Mycobacterium tuberculosis causes human tuberculosis (TB). As mycobacteria are protected by a thick lipid cell wall, humans have developed immune responses against diverse mycobacterial lipids. Most of these immunostimulatory lipids are known as adjuvants acting through innate immune receptors, such as C-type lectin receptors. Although a few mycobacterial lipid antigens activate unconventional T cells, the antigenicity of most adjuvantic lipids is unknown. Here, we identified that trehalose monomycolate (TMM), an abundant mycobacterial adjuvant, activated human T cells bearing a unique αβ T cell receptor (αβTCR). This recognition was restricted by CD1b, a monomorphic antigen-presenting molecule conserved in primates but not mice. Single-cell TCR-RNA-Seq using newly established CD1b-TMM tetramers revealed that TMM-specific T cells were present as CD4+ effector memory T cells in the periphery of uninfected donors but expressed IFN-γ, TNF, and anti-mycobacterial effectors upon TMM stimulation. TMM-specific T cells were detected in cord blood and PBMCs of donors without bacillus Calmette-Guérin vaccination but were expanded in patients with active TB. A cryo-electron microscopy study of CD1b-TMM-TCR complexes revealed unique antigen recognition by conserved features of TCRs, positively charged CDR3α, and long CDR3β regions. These results indicate that humans have a commonly shared and preformed CD4+ T cell subset recognizing a typical mycobacterial adjuvant as an antigen. Furthermore, the dual role of TMM justifies reconsideration of the mechanism of action of adjuvants.
Collapse
Affiliation(s)
- Yuki Sakai
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Minori Asa
- Department of Molecular Immunology, Research Institute for Microbial Diseases
| | - Mika Hirose
- Laboratory for CryoEM Structural Biology, Institute for Protein Research, Osaka University, Suita, Japan
| | - Wakana Kusuhara
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Nagatoshi Fujiwara
- Department of Food and Nutrition, Faculty of Contemporary Human Life Science, Tezukayama University, Nara, Japan
| | - Hiroto Tamashima
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Ikazaki
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shiori Oka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kota Kuraba
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kentaro Tanaka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Takashi Yoshiyama
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Yoshihiko Hoshino
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Ildiko Van Rhijn
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, University Utrecht, Utrecht, Netherlands
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Xiuyuan Lu
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - D. Branch Moody
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takayuki Kato
- Laboratory for CryoEM Structural Biology, Institute for Protein Research, Osaka University, Suita, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Go Hirai
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
- Center for Advanced Modalities and Drug Delivery Systems (CAMaD), Osaka University, Suita, Japan
| |
Collapse
|
4
|
Laudadio E, Mangano L, Minnelli C. Chemical Scaffolds for the Clinical Development of Mutant-Selective and Reversible Fourth-Generation EGFR-TKIs in NSCLC. ACS Chem Biol 2024; 19:839-854. [PMID: 38552205 DOI: 10.1021/acschembio.4c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
In nonsmall cell lung cancer (NSCLC), as well as in other tumors, the targeted therapy is mainly represented by tyrosine kinase inhibitors (TKIs), small molecules able to target oncogenic driver alterations affecting the gene encoding the epidermal growth factor receptor (EGFR). Up to now, several different TKIs have been developed. However, cancer cells showed an incredible adaptive tumor response to the inhibition of the sequentially mutated EGFR (EGFRM+), triggering the need to explore novel pharmacochemical strategies. This Review summarizes the recent efforts in the development of new reversible next-generation EGFR TKIs to fight the resistance against T790M and C797S mutations. Specifically, after giving an overview of the role of the EGFR's signaling pathways in cancer progression, we are going to discuss the most relevant approved drugs and drug candidates in terms of chemical structure, binding modalities, and their potency and selectivity against the mutated EGFR over the wild-type form. This could provide important guidelines and rationale for the discovery and iterative development of new drugs.
Collapse
Affiliation(s)
- Emiliano Laudadio
- Department of Science and Engineering of Matter, Environment and Urban Planning, Marche Polytechnic University, 60131 Ancona, Italy
| | - Luca Mangano
- Roche Pharma Research and Early Development, Oncology Discovery, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Cristina Minnelli
- Department of Life and Environmental Sciences, Marche Polytechnic University, 60131 Ancona, Italy
| |
Collapse
|
5
|
Li X, Ma Y, Li G, Jin G, Xu L, Li Y, Wei P, Zhang L. Leprosy: treatment, prevention, immune response and gene function. Front Immunol 2024; 15:1298749. [PMID: 38440733 PMCID: PMC10909994 DOI: 10.3389/fimmu.2024.1298749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Since the leprosy cases have fallen dramatically, the incidence of leprosy has remained stable over the past years, indicating that multidrug therapy seems unable to eradicate leprosy. More seriously, the emergence of rifampicin-resistant strains also affects the effectiveness of treatment. Immunoprophylaxis was mainly carried out through vaccination with the BCG but also included vaccines such as LepVax and MiP. Meanwhile, it is well known that the infection and pathogenesis largely depend on the host's genetic background and immunity, with the onset of the disease being genetically regulated. The immune process heavily influences the clinical course of the disease. However, the impact of immune processes and genetic regulation of leprosy on pathogenesis and immunological levels is largely unknown. Therefore, we summarize the latest research progress in leprosy treatment, prevention, immunity and gene function. The comprehensive research in these areas will help elucidate the pathogenesis of leprosy and provide a basis for developing leprosy elimination strategies.
Collapse
Affiliation(s)
- Xiang Li
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Yun Ma
- Chronic Infectious Disease Control Section, Nantong Center for Disease Control and Prevention, Nantong, China
| | - Guoli Li
- Department of Chronic Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Guangjie Jin
- Department of Chronic Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Li Xu
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Yunhui Li
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Pingmin Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Lianhua Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
- Department of Chronic Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| |
Collapse
|
6
|
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
|
7
|
Torrelles JB, Chatterjee D. Collected Thoughts on Mycobacterial Lipoarabinomannan, a Cell Envelope Lipoglycan. Pathogens 2023; 12:1281. [PMID: 38003746 PMCID: PMC10675199 DOI: 10.3390/pathogens12111281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
The presence of lipoarabinomannan (LAM) in the Mycobacterium tuberculosis (Mtb) cell envelope was first reported close to 100 years ago. Since then, numerous studies have been dedicated to the isolation, purification, structural definition, and elucidation of the biological properties of Mtb LAM. In this review, we present a brief historical perspective on the discovery of Mtb LAM and the herculean efforts devoted to structurally characterizing the molecule because of its unique structural and biological features. The significance of LAM remains high to this date, mainly due to its distinct immunological properties in conjunction with its role as a biomarker for diagnostic tests due to its identification in urine, and thus can serve as a point-of-care diagnostic test for tuberculosis (TB). In recent decades, LAM has been thoroughly studied and massive amounts of information on this intriguing molecule are now available. In this review, we give the readers a historical perspective and an update on the current knowledge of LAM with information on the inherent carbohydrate composition, which is unique due to the often puzzling sugar residues that are specifically found on LAM. We then guide the readers through the complex and myriad immunological outcomes, which are strictly dependent on LAM's chemical structure. Furthermore, we present issues that remain unresolved and represent the immediate future of LAM research. Addressing the chemistry, functions, and roles of LAM will lead to innovative ways to manipulate the processes that involve this controversial and fascinating biomolecule.
Collapse
Affiliation(s)
- Jordi B. Torrelles
- International Center for the Advancement of Research and Education (I • Care), Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Delphi Chatterjee
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
8
|
Walker EM, Merino KM, Slisarenko N, Grasperge BF, Mehra S, Roy CJ, Kaushal D, Rout N. Impact of SIV infection on mycobacterial lipid-reactive T cell responses in Bacillus Calmette-Guérin (BCG) inoculated macaques. Front Immunol 2023; 13:1085786. [PMID: 36726992 PMCID: PMC9885173 DOI: 10.3389/fimmu.2022.1085786] [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: 10/31/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Background Although BCG vaccine protects infants from tuberculosis (TB), it has limited efficacy in adults against pulmonary TB. Further, HIV coinfection significantly increases the risk of developing active TB. In the lack of defined correlates of protection in TB disease, it is essential to explore immune responses beyond conventional CD4 T cells to gain a better understanding of the mechanisms of TB immunity. Methods Here, we evaluated unconventional lipid-reactive T cell responses in cynomolgus macaques following aerosol BCG inoculation and examined the impact of subsequent SIV infection on these responses. Immune responses to cellular lipids of M. bovis and M. tuberculosis were examined ex vivo in peripheral blood and bronchioalveolar lavage (BAL). Results Prior to BCG inoculation, innate-like IFN-γ responses to mycobacterial lipids were observed in T cells. Aerosol BCG exposure induced an early increase in frequencies of BAL γδT cells, a dominant subset of lipid-reactive T cells, along with enhanced IL-7R and CXCR3 expression. Further, BCG exposure stimulated greater IFN-γ responses to mycobacterial lipids in peripheral blood and BAL, suggesting the induction of systemic and local Th1-type response in lipid-reactive T cells. Subsequent SIV infection resulted in a significant loss of IL-7R expression on blood and BAL γδT cells. Additionally, IFN-γ responses of mycobacterial lipid-reactive T cells in BAL fluid were significantly lower in SIV-infected macaques, while perforin production was maintained through chronic SIV infection. Conclusions Overall, these data suggest that despite SIV-induced decline in IL-7R expression and IFN-γ production by mycobacterial lipid-reactive T cells, their cytolytic potential is maintained. A deeper understanding of anti-mycobacterial lipid-reactive T cell functions may inform novel approaches to enhance TB control in individuals with or without HIV infection.
Collapse
Affiliation(s)
- Edith M. Walker
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Kristen M. Merino
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Nadia Slisarenko
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Brooke F. Grasperge
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Chad J. Roy
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Namita Rout
- Division of Microbiology at Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
9
|
Correia-Neves M, Nigou J, Mousavian Z, Sundling C, Källenius G. Immunological hyporesponsiveness in tuberculosis: The role of mycobacterial glycolipids. Front Immunol 2022; 13:1035122. [PMID: 36544778 PMCID: PMC9761185 DOI: 10.3389/fimmu.2022.1035122] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 12/09/2022] Open
Abstract
Glycolipids constitute a major part of the cell envelope of Mycobacterium tuberculosis (Mtb). They are potent immunomodulatory molecules recognized by several immune receptors like pattern recognition receptors such as TLR2, DC-SIGN and Dectin-2 on antigen-presenting cells and by T cell receptors on T lymphocytes. The Mtb glycolipids lipoarabinomannan (LAM) and its biosynthetic relatives, phosphatidylinositol mannosides (PIMs) and lipomannan (LM), as well as other Mtb glycolipids, such as phenolic glycolipids and sulfoglycolipids have the ability to modulate the immune response, stimulating or inhibiting a pro-inflammatory response. We explore here the downmodulating effect of Mtb glycolipids. A great proportion of the studies used in vitro approaches although in vivo infection with Mtb might also lead to a dampening of myeloid cell and T cell responses to Mtb glycolipids. This dampened response has been explored ex vivo with immune cells from peripheral blood from Mtb-infected individuals and in mouse models of infection. In addition to the dampening of the immune response caused by Mtb glycolipids, we discuss the hyporesponse to Mtb glycolipids caused by prolonged Mtb infection and/or exposure to Mtb antigens. Hyporesponse to LAM has been observed in myeloid cells from individuals with active and latent tuberculosis (TB). For some myeloid subsets, this effect is stronger in latent versus active TB. Since the immune response in individuals with latent TB represents a more protective profile compared to the one in patients with active TB, this suggests that downmodulation of myeloid cell functions by Mtb glycolipids may be beneficial for the host and protect against active TB disease. The mechanisms of this downmodulation, including tolerance through epigenetic modifications, are only partly explored.
Collapse
Affiliation(s)
- Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal,Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B's), Portuguese (PT) Government Associate Laboratory, Braga, Portugal,Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France
| | - Zaynab Mousavian
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,School of Mathematics, Statistics, and Computer Science, College of Science, University of Tehran, Tehran, Iran,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,*Correspondence: Gunilla Källenius,
| |
Collapse
|
10
|
Kim H, Shin SJ. Pathological and protective roles of dendritic cells in Mycobacterium tuberculosis infection: Interaction between host immune responses and pathogen evasion. Front Cell Infect Microbiol 2022; 12:891878. [PMID: 35967869 PMCID: PMC9366614 DOI: 10.3389/fcimb.2022.891878] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) are principal defense components that play multifactorial roles in translating innate immune responses to adaptive immunity in Mycobacterium tuberculosis (Mtb) infections. The heterogeneous nature of DC subsets follows their altered functions by interacting with other immune cells, Mtb, and its products, enhancing host defense mechanisms or facilitating pathogen evasion. Thus, a better understanding of the immune responses initiated, promoted, and amplified or inhibited by DCs in Mtb infection is an essential step in developing anti-tuberculosis (TB) control measures, such as host-directed adjunctive therapy and anti-TB vaccines. This review summarizes the recent advances in salient DC subsets, including their phenotypic classification, cytokine profiles, functional alterations according to disease stages and environments, and consequent TB outcomes. A comprehensive overview of the role of DCs from various perspectives enables a deeper understanding of TB pathogenesis and could be useful in developing DC-based vaccines and immunotherapies.
Collapse
|
11
|
Gramegna A, Lombardi A, Lorè NI, Amati F, Barone I, Azzarà C, Cirillo D, Aliberti S, Gori A, Blasi F. Innate and Adaptive Lymphocytes in Non-Tuberculous Mycobacteria Lung Disease: A Review. Front Immunol 2022; 13:927049. [PMID: 35837393 PMCID: PMC9273994 DOI: 10.3389/fimmu.2022.927049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Non-tuberculous mycobacteria (NTM) are ubiquitous environmental microorganisms capable of a wide range of infections that primarily involve the lymphatic system and the lower respiratory tract. In recent years, cases of lung infection sustained by NTM have been steadily increasing, due mainly to the ageing of the population with underlying lung disease, the enlargement of the cohort of patients undergoing immunosuppressive medications and the improvement in microbiologic diagnostic techniques. However, only a small proportion of individuals at risk ultimately develop the disease due to reasons that are not fully understood. A better understanding of the pathophysiology of NTM pulmonary disease is the key to the development of better diagnostic tools and therapeutic targets for anti-mycobacterial therapy. In this review, we cover the various types of interactions between NTM and lymphoid effectors of innate and adaptive immunity. We also give a brief look into the mechanism of immune exhaustion, a phenomenon of immune dysfunction originally reported for chronic viral infections and cancer, but recently also observed in the setting of mycobacterial diseases. We try to set the scene to postulate that a better knowledge of immune exhaustion can play a crucial role in establishing prognostic/predictive factors and enabling a broader investigation of immune-modulatory drugs in the experimental treatment of NTM pulmonary disease.
Collapse
Affiliation(s)
- Andrea Gramegna
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Internal Medicine Department, Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- *Correspondence: Andrea Gramegna,
| | - Andrea Lombardi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Internal Medicine Department, Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Nicola I. Lorè
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Amati
- IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Ivan Barone
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Internal Medicine Department, Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Azzarà
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Internal Medicine Department, Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Aliberti
- IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Andrea Gori
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Internal Medicine Department, Infectious Diseases Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Internal Medicine Department, Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
12
|
Gairola A, Benjamin A, Weatherston JD, Cirillo JD, Wu HJ. Recent Developments in Drug Delivery for Treatment of Tuberculosis by Targeting Macrophages. ADVANCED THERAPEUTICS 2022; 5:2100193. [PMID: 36203881 PMCID: PMC9531895 DOI: 10.1002/adtp.202100193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/10/2022]
Abstract
Tuberculosis (TB) is among the greatest public health and safety concerns in the 21st century, Mycobacterium tuberculosis, which causes TB, infects alveolar macrophages and uses these cells as one of its primary sites of replication. The current TB treatment regimen, which consist of chemotherapy involving a combination of 3-4 antimicrobials for a duration of 6-12 months, is marked with significant side effects, toxicity, and poor compliance. Targeted drug delivery offers a strategy that could overcome many of the problems of current TB treatment by specifically targeting infected macrophages. Recent advances in nanotechnology and material science have opened an avenue to explore drug carriers that actively and passively target macrophages. This approach can increase the drug penetration into macrophages by using ligands on the nanocarrier that interact with specific receptors for macrophages. This review encompasses the recent development of drug carriers specifically targeting macrophages actively and passively. Future directions and challenges associated with development of effective TB treatment is also discussed.
Collapse
Affiliation(s)
- Anirudh Gairola
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Aaron Benjamin
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Joshua D Weatherston
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jeffrey D Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Hung-Jen Wu
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
13
|
Tsubata T. Role of inhibitory B cell co-receptors in B cell self-tolerance to non-protein antigens. Immunol Rev 2022; 307:53-65. [PMID: 34989000 DOI: 10.1111/imr.13059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022]
Abstract
Antibodies to non-protein antigens such as nucleic acids, polysaccharides, and glycolipids play important roles in both host defense against microbes and development of autoimmune diseases. Although non-protein antigens are not recognized by T cells, antibody production to non-protein antigens involve T cell-independent mechanisms such as signaling through TLR7 and TLR9 in antibody production to nucleic acids. Although self-reactive B cells are tolerized by various mechanisms including deletion, anergy, and receptor editing, T cell tolerance is also crucial in self-tolerance of B cells to protein self-antigen because self-reactive T cells induce autoantibody production to these self-antigens. However, presence of T cell-independent mechanism suggests that T cell tolerance is not able to maintain B cell tolerance to non-protein self-antigens. Lines of evidence suggest that B cell response to non-protein self-antigens such as nucleic acids and gangliosides, sialic acid-containing glycolipids, are suppressed by inhibitory B cell co-receptors CD72 and Siglec-G, respectively. These inhibitory co-receptors recognize non-protein self-antigens and suppress BCR signaling induced by these antigens, thereby inhibiting B cell response to these self-antigens. Inhibitory B cell co-receptors appear to be involved in B cell self-tolerance to non-protein self-antigens that can activate B cells by T cell-independent mechanisms.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
14
|
Silva CS, Sundling C, Folkesson E, Fröberg G, Nobrega C, Canto-Gomes J, Chambers BJ, Lakshmikanth T, Brodin P, Bruchfeld J, Nigou J, Correia-Neves M, Källenius G. High Dimensional Immune Profiling Reveals Different Response Patterns in Active and Latent Tuberculosis Following Stimulation With Mycobacterial Glycolipids. Front Immunol 2021; 12:727300. [PMID: 34887849 PMCID: PMC8650708 DOI: 10.3389/fimmu.2021.727300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Upon infection with Mycobacterium tuberculosis (Mtb) the host immune response might clear the bacteria, control its growth leading to latent tuberculosis (LTB), or fail to control its growth resulting in active TB (ATB). There is however no clear understanding of the features underlying a more or less effective response. Mtb glycolipids are abundant in the bacterial cell envelope and modulate the immune response to Mtb, but the patterns of response to glycolipids are still underexplored. To identify the CD45+ leukocyte activation landscape induced by Mtb glycolipids in peripheral blood of ATB and LTB, we performed a detailed assessment of the immune response of PBMCs to the Mtb glycolipids lipoarabinomannan (LAM) and its biosynthetic precursor phosphatidyl-inositol mannoside (PIM), and purified-protein derivate (PPD). At 24 h of stimulation, cell profiling and secretome analysis was done using mass cytometry and high-multiplex immunoassay. PIM induced a diverse cytokine response, mainly affecting antigen-presenting cells to produce both pro-inflammatory and anti-inflammatory cytokines, but not IFN-γ, contrasting with PPD that was a strong inducer of IFN-γ. The effect of PIM on the antigen-presenting cells was partly TLR2-dependent. Expansion of monocyte subsets in response to PIM or LAM was reduced primarily in LTB as compared to healthy controls, suggesting a hyporesponsive/tolerance pattern derived from Mtb infection.
Collapse
Affiliation(s)
- Carolina S Silva
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Elin Folkesson
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gabrielle Fröberg
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Claudia Nobrega
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - João Canto-Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - Benedict J Chambers
- Center for Infectious Medicine, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Tadepally Lakshmikanth
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Judith Bruchfeld
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal.,Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Keane JT, Posey AD. Chimeric Antigen Receptors Expand the Repertoire of Antigenic Macromolecules for Cellular Immunity. Cells 2021; 10:cells10123356. [PMID: 34943864 PMCID: PMC8699116 DOI: 10.3390/cells10123356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/16/2022] Open
Abstract
T-cell therapies have made significant improvements in cancer treatment over the last decade. One cellular therapy utilizing T-cells involves the use of a chimeric MHC-independent antigen-recognition receptor, typically referred to as a chimeric antigen receptor (CAR). CAR molecules, while mostly limited to the recognition of antigens on the surface of tumor cells, can also be utilized to exploit the diverse repertoire of macromolecules targetable by antibodies, which are incorporated into the CAR design. Leaning into this expansion of target macromolecules will enhance the diversity of antigens T-cells can target and may improve the tumor-specificity of CAR T-cell therapy. This review explores the types of macromolecules targetable by T-cells through endogenous and synthetic antigen-specific receptors.
Collapse
Affiliation(s)
- John T. Keane
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Avery D. Posey
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
16
|
Korenfeld D, Roussak K, Dinkel S, Vogel TP, Pollack H, Levy J, Leiding JW, Milner J, Cooper M, Klechevsky E. STAT3 Gain-of-Function Mutations Underlie Deficiency in Human Nonclassical CD16 + Monocytes and CD141 + Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:2423-2432. [PMID: 34654687 DOI: 10.4049/jimmunol.2000841] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/14/2021] [Indexed: 12/24/2022]
Abstract
Genetic analysis of human inborn errors of immunity has defined the contribution of specific cell populations and molecular pathways in the host defense against infection. The STAT family of transcription factors orchestrate hematopoietic cell differentiation. Patients with de novo activating mutations of STAT3 present with multiorgan autoimmunity, lymphoproliferation, and recurrent infections. We conducted a detailed characterization of the blood monocyte and dendritic cell (DC) subsets in patients with gain-of-function (GOF) mutations across the gene. We found a selective deficiency in circulating nonclassical CD16+ and intermediate CD16+CD14+ monocytes and a significant increase in the percentage of classical CD14+ monocytes. This suggests a role for STAT3 in the transition of classical CD14+ monocytes into the CD16+ nonclassical subset. Developmentally, ex vivo-isolated STAT3GOF CD14+ monocytes fail to differentiate into CD1a+ monocyte-derived DCs. Moreover, patients with STAT3GOF mutations display reduced circulating CD34+ hematopoietic progenitors and frequency of myeloid DCs. Specifically, we observed a reduction in the CD141+ DC population, with no difference in the frequencies of CD1c+ and plasmacytoid DCs. CD34+ hematopoietic progenitor cells from patients were found to differentiate into CD1c+ DCs, but failed to differentiate into CD141+ DCs indicating an intrinsic role for STAT3 in this process. STAT3GOF-differentiated DCs produced lower amounts of CCL22 than healthy DCs, which could further explain some of the patient pathological phenotypes. Thus, our findings provide evidence that, in humans, STAT3 serves to regulate development and differentiation of nonclassical CD16+ monocytes and a subset of myeloid DCs.
Collapse
Affiliation(s)
- Daniel Korenfeld
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Kate Roussak
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Sabrina Dinkel
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO
| | - Tiphanie P Vogel
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University School of Medicine, St. Louis, MO
| | - Henry Pollack
- Department of Pediatrics, New York University School of Medicine, New York, NY
| | - Joseph Levy
- Department of Pediatrics, New York University School of Medicine, New York, NY
| | - Jennifer W Leiding
- Department of Pediatrics, Division of Allergy and Immunology, University of South Florida, Tampa, FL; and
| | - Joshua Milner
- Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Megan Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University School of Medicine, St. Louis, MO
| | - Eynav Klechevsky
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, MO;
| |
Collapse
|
17
|
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
|
18
|
Reijneveld JF, Holzheimer M, Young DC, Lopez K, Suliman S, Jimenez J, Calderon R, Lecca L, Murray MB, Ishikawa E, Yamasaki S, Minnaard AJ, Moody DB, Van Rhijn I. Synthetic mycobacterial diacyl trehaloses reveal differential recognition by human T cell receptors and the C-type lectin Mincle. Sci Rep 2021; 11:2010. [PMID: 33479373 PMCID: PMC7820438 DOI: 10.1038/s41598-021-81474-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/04/2021] [Indexed: 11/10/2022] Open
Abstract
The cell wall of Mycobacterium tuberculosis is composed of diverse glycolipids which potentially interact with the human immune system. To overcome difficulties in obtaining pure compounds from bacterial extracts, we recently synthesized three forms of mycobacterial diacyltrehalose (DAT) that differ in their fatty acid composition, DAT1, DAT2, and DAT3. To study the potential recognition of DATs by human T cells, we treated the lipid-binding antigen presenting molecule CD1b with synthetic DATs and looked for T cells that bound the complex. DAT1- and DAT2-treated CD1b tetramers were recognized by T cells, but DAT3-treated CD1b tetramers were not. A T cell line derived using CD1b-DAT2 tetramers showed that there is no cross-reactivity between DATs in an IFN-γ release assay, suggesting that the chemical structure of the fatty acid at the 3-position determines recognition by T cells. In contrast with the lack of recognition of DAT3 by human T cells, DAT3, but not DAT1 or DAT2, activates Mincle. Thus, we show that the mycobacterial lipid DAT can be both an antigen for T cells and an agonist for the innate Mincle receptor, and that small chemical differences determine recognition by different parts of the immune system.
Collapse
Affiliation(s)
- Josephine F Reijneveld
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, 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
| | - Mira Holzheimer
- Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - David C Young
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Kattya Lopez
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA.,Socios En Salud, Lima, Peru
| | - Sara Suliman
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | | | | | | | - Megan B Murray
- Division of Global Health Equity, Department of Global Health and Social Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - D Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA. .,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
19
|
Animal models for human group 1 CD1 protein function. Mol Immunol 2020; 130:159-163. [PMID: 33384157 DOI: 10.1016/j.molimm.2020.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/09/2020] [Indexed: 11/21/2022]
Abstract
The CD1 antigen presenting system is evolutionary conserved and found in mammals, birds and reptiles. Humans express five isoforms, of which CD1a, CD1b and CD1c represent the group 1 CD1-molecules. They are recognized by T cells that express diverse αβ-T cell receptors. Investigation of the role of group 1 CD1 function has been hampered by the fact that CD1a, CD1b and CD1c are not expressed by mice. However, other animals, such as guinea pigs or cattle, serve as alternative models and have established basic aspects of CD1-dependent, antimicrobial immune functions. Group 1 CD1 transgenic mouse models became available about ten years ago. In a series of seminal studies these mouse models coined the mechanistical understanding of the role of the corresponding CD1 restricted T cell responses. This review gives a short overview of available animal studies and the lessons that have been and still can be learned.
Collapse
|
20
|
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: 88] [Impact Index Per Article: 17.6] [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
|
21
|
Benedictus L, Steinbach S, Holder T, Bakker D, Vrettou C, Morrison WI, Vordermeier M, Connelley T. Hydrophobic Mycobacterial Antigens Elicit Polyfunctional T Cells in Mycobacterium bovis Immunized Cattle: Association With Protection Against Challenge? Front Immunol 2020; 11:588180. [PMID: 33281817 PMCID: PMC7688591 DOI: 10.3389/fimmu.2020.588180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/20/2020] [Indexed: 01/09/2023] Open
Abstract
Bovine tuberculosis (bTB), caused by Mycobacterium bovis, is a chronic disease of cattle with a detrimental impact on food quality and production. Research on bTB vaccines has predominantly been focused on proteinaceous antigens. However, mycobacteria have a thick and intricate lipid outer layer and lipids as well as lipopeptides are important for immune-evasion and virulence. In humans, lipid extracts of M. tuberculosis have been shown to elicit immune responses effective against M. tuberculosis in vitro. Chloroform-methanol extraction (CME) was applied to M. bovis BCG to obtain a hydrophobic antigen extract (CMEbcg) containing lipids and lipopeptides. CMEbcg stimulated IFN-γ+IL-2+ and IL-17A+IL-22+ polyfunctional T cells and elicited T cell responses with a Th1 and Th17 cytokine release profile in both M. bovis BCG vaccinated and M. bovis challenged calves. Lipopeptides were shown to be the immunodominant antigens in CMEbcg, stimulating CD4 T cells via MHC class II. CMEbcg expanded T cells killed CMEbcg loaded monocytes and the CMEbcg-specific CD3 T cell proliferative response following M. bovis BCG vaccination was the best predictor for reduced pathology following challenge with M. bovis. Although the high predictive value of CMEbcg-specific immune responses does not confirm a causal relationship with protection against M. bovis challenge, when taking into account the in vitro antimycobacterial phenotype of CMEbcg-specific T cells (e.g. Th1/Th17 cytokine profile), it is indicative that CMEbcg-specific immune responses could play a functional role in immunity against M. bovis. Based on these findings we conclude that lipopeptides of M. bovis are potential novel subunit vaccine candidates and that further studies into the functional characterization of lipopeptide-specific immune responses together with their role in protection against bovine tuberculosis are warranted.
Collapse
Affiliation(s)
- Lindert Benedictus
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - Sabine Steinbach
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Thomas Holder
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Douwe Bakker
- Independent Researcher and Technical Consultant, Lelystad, Netherlands
| | - Christina Vrettou
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - W Ivan Morrison
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - Martin Vordermeier
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom.,Centre for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, University of Aberystwyth, Aberystwyth, United Kingdom
| | - Timothy Connelley
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| |
Collapse
|
22
|
Deshpande D, Grieshober M, Wondany F, Gerbl F, Noschka R, Michaelis J, Stenger S. Super-Resolution Microscopy Reveals a Direct Interaction of Intracellular Mycobacterium tuberculosis with the Antimicrobial Peptide LL-37. Int J Mol Sci 2020; 21:ijms21186741. [PMID: 32937921 PMCID: PMC7555347 DOI: 10.3390/ijms21186741] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/31/2020] [Accepted: 09/10/2020] [Indexed: 12/26/2022] Open
Abstract
The antimicrobial peptide LL-37 inhibits the growth of the major human pathogen Mycobacterium tuberculosis (Mtb), but the mechanism of the peptide–pathogen interaction inside human macrophages remains unclear. Super-resolution imaging techniques provide a novel opportunity to visualize these interactions on a molecular level. Here, we adapt the super-resolution technique of stimulated emission depletion (STED) microscopy to study the uptake, intracellular localization and interaction of LL-37 with macrophages and virulent Mtb. We demonstrate that LL-37 is internalized by both uninfected and Mtb infected primary human macrophages. The peptide localizes in the membrane of early endosomes and lysosomes, the compartment in which mycobacteria reside. Functionally, LL-37 disrupts the cell wall of intra- and extracellular Mtb, resulting in the killing of the pathogen. In conclusion, we introduce STED microscopy as an innovative and informative tool for studying host–pathogen–peptide interactions, clearly extending the possibilities of conventional confocal microscopy.
Collapse
Affiliation(s)
- Dhruva Deshpande
- Institute of Biophysics, Ulm University, 89081 Ulm, Germany; (D.D.); (F.W.)
| | - Mark Grieshober
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, 89081 Ulm, Germany; (M.G.); (F.G.); (R.N.)
| | - Fanny Wondany
- Institute of Biophysics, Ulm University, 89081 Ulm, Germany; (D.D.); (F.W.)
| | - Fabian Gerbl
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, 89081 Ulm, Germany; (M.G.); (F.G.); (R.N.)
| | - Reiner Noschka
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, 89081 Ulm, Germany; (M.G.); (F.G.); (R.N.)
| | - Jens Michaelis
- Institute of Biophysics, Ulm University, 89081 Ulm, Germany; (D.D.); (F.W.)
- Correspondence: (J.M.); (S.S.)
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, 89081 Ulm, Germany; (M.G.); (F.G.); (R.N.)
- Correspondence: (J.M.); (S.S.)
| |
Collapse
|
23
|
La Manna MP, Orlando V, Tamburini B, Badami GD, Dieli F, Caccamo N. Harnessing Unconventional T Cells for Immunotherapy of Tuberculosis. Front Immunol 2020; 11:2107. [PMID: 33013888 PMCID: PMC7497315 DOI: 10.3389/fimmu.2020.02107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Even if the incidence of tuberculosis (TB) has been decreasing over the last years, the number of patients with TB is increasing worldwide. The emergence of multidrug-resistant and extensively drug-resistant TB is making control of TB more difficult. Mycobacterium bovis bacillus Calmette–Guérin vaccine fails to prevent pulmonary TB in adults, and there is an urgent need for a vaccine that is also effective in patients with human immunodeficiency virus (HIV) coinfection. Therefore, TB control may benefit on novel therapeutic options beyond antimicrobial treatment. Host-directed immunotherapies could offer therapeutic strategies for patients with drug-resistant TB or with HIV and TB coinfection. In the last years, the use of donor lymphocytes after hematopoietic stem cell transplantation has emerged as a new strategy in the cure of hematologic malignancies in order to induce graft-versus leukemia and graft-versus-infection effects. Moreover, adoptive therapy has proven to be effective in controlling cytomegalovirus and Epstein-Barr virus reactivation in immunocompromised patients with ex vivo expanded viral antigen-specific T cells. Unconventional T cells are a heterogeneous group of T lymphocytes with limited diversity. One of their characteristics is that antigen recognition is not restricted by the classical major histocompatibility complex (MHC). They include CD1 (cluster of differentiation 1)–restricted T cells, MHC-related protein-1–restricted mucosal-associated invariant T (MAIT) cells, MHC class Ib–reactive T cells, and γδ T cells. Because these T cells are genotype-independent, they are also termed “donor unrestricted” T cells. The combined features of low donor diversity and the lack of genetic restriction make these cells suitable candidates for T cell–based immunotherapy of TB.
Collapse
Affiliation(s)
- Marco P La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giusto D Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
24
|
Smith AA, Villarreal-Ramos B, Mendum TA, Williams KJ, Jones GJ, Wu H, McFadden J, Vordermeier HM, Stewart GR. Genetic screening for the protective antigenic targets of BCG vaccination. Tuberculosis (Edinb) 2020; 124:101979. [PMID: 32814303 DOI: 10.1016/j.tube.2020.101979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 11/18/2022]
Abstract
Bovine tuberculosis is an important animal health problem and the predominant cause of zoonotic tuberculosis worldwide. It results in serious economic burden due to losses in productivity and the cost of control programmes. Control could be greatly improved by the introduction of an efficacious cattle vaccine but the most likely candidate, BCG, has several limitations including variable efficacy. Augmentation of BCG with a subunit vaccine booster has been shown to increase protection but the selection of antigens has hitherto been left largely to serendipity. In the present study, we take a rational approach to identify the protective antigens of BCG, selecting a BCG transposon mutant library in naïve and BCG-vaccinated cattle. Ten mutants had increased relative survival in vaccinated compared to naïve cattle, consistent with loss of protective antigen targets making the mutants less visible to the BCG immune response. The immunogenicity of three putative protective antigens, BCG_0116, BCG_0205 (YrbE1B) and BCG_1448 (PPE20) was investigated using peptide pools and PBMCs from BCG vaccinated cattle. BCG vaccination induced PBMC to release elevated levels of IP10, IL-17a and IL-10 in response to all three antigens. Taken together, the data supports the further study of these antigens for use in subunit vaccines.
Collapse
MESH Headings
- Animals
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- BCG Vaccine/administration & dosage
- BCG Vaccine/immunology
- Cattle
- Cytokines/immunology
- Cytokines/metabolism
- DNA Transposable Elements
- Immunogenicity, Vaccine
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/microbiology
- Mutation
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Tuberculosis, Bovine/immunology
- Tuberculosis, Bovine/metabolism
- Tuberculosis, Bovine/microbiology
- Tuberculosis, Bovine/prevention & control
- Vaccination/veterinary
Collapse
Affiliation(s)
- Alex A Smith
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Bernardo Villarreal-Ramos
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, KT15 3NB, UK; Centre of Excellence for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Wales, SY23 3DA, UK.
| | - Tom A Mendum
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Kerstin J Williams
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Gareth J Jones
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, KT15 3NB, UK
| | - Huihai Wu
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Johnjoe McFadden
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - H Martin Vordermeier
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, KT15 3NB, UK; Centre of Excellence for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Wales, SY23 3DA, UK.
| | - Graham R Stewart
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
25
|
Lopez K, Iwany SK, Suliman S, Reijneveld JF, Ocampo TA, Jimenez J, Calderon R, Lecca L, Murray MB, Moody DB, Van Rhijn I. CD1b Tetramers Broadly Detect T Cells That Correlate With Mycobacterial Exposure but Not Tuberculosis Disease State. Front Immunol 2020; 11:199. [PMID: 32117314 PMCID: PMC7033476 DOI: 10.3389/fimmu.2020.00199] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/27/2020] [Indexed: 12/29/2022] Open
Abstract
The non-polymorphic nature of CD1 proteins creates a situation in which T cells with invariant T cell receptors (TCRs), like CD1d-specific NKT cells, are present in all humans. CD1b is an abundant protein on human dendritic cells that presents M. tuberculosis (Mtb) lipid antigens to T cells. Analysis of T cell clones suggested that semi-invariant TCRs exist in the CD1b system, but their prevalence in humans is not known. Here we used CD1b tetramers loaded with mycolic acid or glucose monomycolate to study polyclonal T cells from 150 Peruvian subjects. We found that CD1b tetramers loaded with mycolic acid or glucose monomycolate antigens stained TRAV1-2+ GEM T cells or TRBV4-1+ LDN5-like T cells in the majority of subjects tested, at rates ~10-fold lower than NKT cells. Thus, GEM T cells and LDN5-like T cells are a normal part of the human immune system. Unlike prior studies measuring MHC- or CD1b-mediated activation, this large-scale tetramer study found no significant differences in rates of CD1b tetramer-mycobacterial lipid staining of T cells among subjects with Mtb exposure, latent Mtb infection or active tuberculosis (TB) disease. In all subjects, including “uninfected” subjects, CD1b tetramer+ T cells expressed memory markers at high levels. However, among controls with lower mycobacterial antigen exposure in Boston, we found significantly lower frequencies of T cells staining with CD1b tetramers loaded with mycobacterial lipids. These data link CD1b-specific T cell detection to mycobacterial exposure, but not TB disease status, which potentially explains differences in outcomes among CD1-based clinical studies, which used control subjects with low Mtb exposure.
Collapse
Affiliation(s)
- Kattya Lopez
- Socios En Salud, Lima, Peru.,Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Sarah K Iwany
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Sara Suliman
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Josephine F Reijneveld
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Tonatiuh A Ocampo
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | | | | | | | - Megan B Murray
- Division of Global Health Equity, Department of Global Health and Social Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - D Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
26
|
James CA, Seshadri C. T Cell Responses to Mycobacterial Glycolipids: On the Spectrum of "Innateness". Front Immunol 2020; 11:170. [PMID: 32117300 PMCID: PMC7026021 DOI: 10.3389/fimmu.2020.00170] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/22/2020] [Indexed: 12/12/2022] Open
Abstract
Diseases due to mycobacteria, including tuberculosis, leprosy, and Buruli ulcer, rank among the top causes of death and disability worldwide. Animal studies have revealed the importance of T cells in controlling these infections. However, the specific antigens recognized by T cells that confer protective immunity and their associated functions remain to be definitively established. T cells that respond to mycobacterial peptide antigens exhibit classical features of adaptive immunity and have been well-studied in humans and animal models. Recently, innate-like T cells that recognize lipid and metabolite antigens have also been implicated. Specifically, T cells that recognize mycobacterial glycolipid antigens (mycolipids) have been shown to confer protection to tuberculosis in animal models and share some biological characteristics with adaptive and innate-like T cells. Here, we review the existing data suggesting that mycolipid-specific T cells exist on a spectrum of “innateness,” which will influence how they can be leveraged to develop new diagnostics and vaccines for mycobacterial diseases.
Collapse
Affiliation(s)
- Charlotte A James
- Molecular Medicine and Mechanisms of Disease (M3D) PhD Program, Department of Pathology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Chetan Seshadri
- Department of Medicine, School of Medicine, University of Washington, Seattle, WA, United States.,Tuberculosis Research and Training Center, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
27
|
Garcia-Vilanova A, Chan J, Torrelles JB. Underestimated Manipulative Roles of Mycobacterium tuberculosis Cell Envelope Glycolipids During Infection. Front Immunol 2019; 10:2909. [PMID: 31921168 PMCID: PMC6930167 DOI: 10.3389/fimmu.2019.02909] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
The Mycobacterium tuberculosis cell envelope has been evolving over time to make the bacterium transmissible and adaptable to the human host. In this context, the M. tuberculosis cell envelope contains a peripheral barrier full of lipids, some of them unique, which confer M. tuberculosis with a unique shield against the different host environments that the bacterium will encounter at the different stages of infection. This lipid barrier is mainly composed of glycolipids that can be characterized by three different subsets: trehalose-containing, mannose-containing, and 6-deoxy-pyranose-containing glycolipids. In this review, we explore the roles of these cell envelope glycolipids in M. tuberculosis virulence and pathogenesis, drug resistance, and further, how these glycolipids may dictate the M. tuberculosis cell envelope evolution from ancient to modern strains. Finally, we address how these M. tuberculosis cell envelope glycolipids are impacted by the host lung alveolar environment, their role in vaccination and masking host immunity, and subsequently the impact of these glycolipids in shaping how M. tuberculosis interacts with host cells, manipulating their immune response to favor the establishment of an infection.
Collapse
Affiliation(s)
- Andreu Garcia-Vilanova
- Population Health Program, TB Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - John Chan
- Department of Medicine (Infectious Diseases), Albert Einstein College of Medicine & Montefiore Medical Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine & Montefiore Medical Center, Bronx, NY, United States
| | - Jordi B. Torrelles
- Population Health Program, TB Group, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
28
|
Koets AP, van den Esker MH, Riepema K, Bakker D. The Role of Phosphatidylinositol Mannosides in the Serological Diagnosis of Mycobacterial Infections. Vet Sci 2019; 6:E91. [PMID: 31766256 PMCID: PMC6958488 DOI: 10.3390/vetsci6040091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 11/16/2022] Open
Abstract
Accurate diagnosis of mycobacterial infections, such as bovine tuberculosis and paratuberculosis, remains challenging. Available direct diagnostic tests aimed at detecting the pathogen are highly specific but lack sensitivity, depending on the stage of infection and the prevalence of infection in a population. The sensitivity of indirect diagnostic assays that measure the host immune response to infection is similarly affected by disease characteristics. The choice of antigen used to detect a host response to infection has a critical impact on test sensitivity and specificity. Many indirect tests rely on crude antigen preparations and cell-free extracts, of which the production is poorly standardized. Moreover, these preparations contain ample uncharacterized cross-reactive compounds. To enhance serological test specificity, existing assays depend on the pre-treatment of samples and a relatively high cut-off value, that in turn influences test sensitivity. Research therefore focuses on the identification of more specific, defined antigens to improve diagnostics. In the current study, we extracted phosphatidylinositol mannosides (PIMs) and investigated their potential use in antibody-based tests. Our results demonstrate that specific IgG class antibodies are generated against PIMs in cows, but this is unrelated to tuberculosis or paratuberculosis infection status, making these antigens unsuitable for diagnostic applications. In addition, we demonstrate that PIMs are widely present in crude antigen preparations and in serum pre-absorption buffer. Our results indicate that PIMs are cross-reactive compounds with immunodominant B cell epitopes that could impair serological test specificity.
Collapse
Affiliation(s)
- Ad P. Koets
- Department of Bacteriology and Epidemiology, Wageningen Bioveterinary Research, Houtribweg 398221 RA Lelystad, The Netherlands; (M.H.v.d.E.); (K.R.)
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 73584 CL Utrecht, The Netherlands
| | - Marielle H. van den Esker
- Department of Bacteriology and Epidemiology, Wageningen Bioveterinary Research, Houtribweg 398221 RA Lelystad, The Netherlands; (M.H.v.d.E.); (K.R.)
| | - Karel Riepema
- Department of Bacteriology and Epidemiology, Wageningen Bioveterinary Research, Houtribweg 398221 RA Lelystad, The Netherlands; (M.H.v.d.E.); (K.R.)
| | - Douwe Bakker
- Independent Researcher, 8212 AM Lelystad, The Netherlands;
| |
Collapse
|
29
|
Correia-Neves M, Sundling C, Cooper A, Källenius G. Lipoarabinomannan in Active and Passive Protection Against Tuberculosis. Front Immunol 2019; 10:1968. [PMID: 31572351 PMCID: PMC6749014 DOI: 10.3389/fimmu.2019.01968] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/05/2019] [Indexed: 12/14/2022] Open
Abstract
Glycolipids of the cell wall of Mycobacterium tuberculosis (Mtb) are important immunomodulators in tuberculosis. In particular, lipoarabinomannan (LAM) has a profound effect on the innate immune response. LAM and its structural variants can be recognized by and activate human CD1b-restricted T cells, and emerging evidence indicates that B cells and antibodies against LAM can modulate the immune response to Mtb. Anti-LAM antibodies are induced during Mtb infection and after bacille Calmette-Guerin (BCG) vaccination, and monoclonal antibodies against LAM have been shown to confer protection by passive administration in mice and guinea pigs. In this review, we describe the immune response against LAM and the potential use of the mannose-capped arabinan moiety of LAM in the construction of vaccine candidates against tuberculosis.
Collapse
Affiliation(s)
- Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Cooper
- Leicester Tuberculosis Research Group (LTBRG), Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Muir FGW, Samadi-Bahrami Z, Moore GRW, Quandt JA. Expression of CD1d by astrocytes corresponds with relative activity in multiple sclerosis lesions. Brain Pathol 2019; 30:26-35. [PMID: 31050367 PMCID: PMC6916356 DOI: 10.1111/bpa.12733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/23/2019] [Indexed: 12/23/2022] Open
Abstract
The CD1 protein family present lipid antigens to the immune system. CD1d has been observed in the CNS of MS patients, yet no studies have quantitatively characterized this expression and related it to inflammatory demyelinative activity in MS plaques. In this study, we set out to localize and quantify the presence of CD1d expression by astrocytes in MS brain tissue lesions. Formalin‐fixed, paraffin‐embedded MS and control brain tissues were examined. Lesions were classified as active, chronic active or chronic silent. Using immunofluorescence, the density of CD1d‐positive cells was determined in active lesions, chronic active lesion edges and chronic active lesion centers. The percentage of CD1d‐positive cells that were GFAP‐positive was also determined in each of these regions. CD1d immunoreactivity was significantly increased in MS compared to control tissue, was significantly more prevalent in areas of active demyelination, and colocalized with GFAP‐positive reactive astrocytes. Increases of CD1d immunoreactivity in the CNS of MS patients being greatest in areas of active demyelination and localized to GFAP‐positive astrocytes lend support to the hypothesis of a lipid‐targeted autoimmune process contributing to the pathogenesis of MS.
Collapse
Affiliation(s)
- Fraser G W Muir
- Department of Pathology & Laboratory Medicine, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, Canada
| | - Zahra Samadi-Bahrami
- Department of Pathology & Laboratory Medicine, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, Canada
| | - George R Wayne Moore
- Department of Pathology & Laboratory Medicine, Department of Medicine (Neurology), International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, Canada
| | - Jacqueline A Quandt
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
31
|
Dang AT, Teles RM, Liu PT, Choi A, Legaspi A, Sarno EN, Ochoa MT, Parvatiyar K, Cheng G, Gilliet M, Bloom BR, Modlin RL. Autophagy links antimicrobial activity with antigen presentation in Langerhans cells. JCI Insight 2019; 4:126955. [PMID: 30996142 PMCID: PMC6538337 DOI: 10.1172/jci.insight.126955] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
DC, through the uptake, processing, and presentation of antigen, are responsible for activation of T cell responses to defend the host against infection, yet it is not known if they can directly kill invading bacteria. Here, we studied in human leprosy, how Langerhans cells (LC), specialized DC, contribute to host defense against bacterial infection. IFN-γ treatment of LC isolated from human epidermis and infected with Mycobacterium leprae (M. leprae) activated an antimicrobial activity, which was dependent on the upregulation of the antimicrobial peptide cathelicidin and induction of autophagy. IFN-γ induction of autophagy promoted fusion of phagosomes containing M. leprae with lysosomes and the delivery of cathelicidin to the intracellular compartment containing the pathogen. Autophagy enhanced the ability of M. leprae-infected LC to present antigen to CD1a-restricted T cells. The frequency of IFN-γ labeling and LC containing both cathelicidin and autophagic vesicles was greater in the self-healing lesions vs. progressive lesions, thus correlating with the effectiveness of host defense against the pathogen. These data indicate that autophagy links the ability of DC to kill and degrade an invading pathogen, ensuring cell survival from the infection while facilitating presentation of microbial antigens to resident T cells.
Collapse
Affiliation(s)
- Angeline Tilly Dang
- Division of Dermatology, Department of Medicine, and
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, UCLA, Los Angeles, California, USA
| | | | - Phillip T. Liu
- Division of Dermatology, Department of Medicine, and
- UCLA and Orthopaedic Hospital, Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California, USA
| | - Aaron Choi
- Division of Dermatology, Department of Medicine, and
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, UCLA, Los Angeles, California, USA
| | | | - Euzenir N. Sarno
- Leprosy Laboratory, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Maria T. Ochoa
- Department of Dermatology, University of Southern California School of Medicine, Los Angeles, California, USA
| | - Kislay Parvatiyar
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, UCLA, Los Angeles, California, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, UCLA, Los Angeles, California, USA
| | - Michel Gilliet
- Department of Medicine, Dermatology Service, Lausanne University Hospital of Lausanne, Lausanne, Switzerland
| | - Barry R. Bloom
- Harvard School of Public Health, Boston, Massachusetts, USA
| | - Robert L. Modlin
- Division of Dermatology, Department of Medicine, and
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, UCLA, Los Angeles, California, USA
| |
Collapse
|
32
|
Ban Y, Dong W, Zhang L, Zhou T, Altiti AS, Ali K, Mootoo DR, Blaho VA, Hla T, Ren Y, Ma X. Abrogation of Endogenous Glycolipid Antigen Presentation on Myelin-Laden Macrophages by D-Sphingosine Ameliorates the Pathogenesis of Experimental Autoimmune Encephalomyelitis. Front Immunol 2019; 10:404. [PMID: 30941120 PMCID: PMC6433838 DOI: 10.3389/fimmu.2019.00404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/15/2019] [Indexed: 01/20/2023] Open
Abstract
Background: Although myelin is composed of mostly lipids, the pathological role of myelin lipids in demyelinating diseases remains elusive. The principal lipid of the myelin sheath is β-galactosylceramide (β-Galcer). Its α-anomer (α-Galcer) has been demonstrated to be antigenically presented by macrophages via CD1d, a MHC class I-like molecule. Myelin, which is mostly composed of β-Galcer, has been long considered as an immunologically-inert neuron insulator, because the antigen-binding cleft of CD1d is highly α-form-restricted. Results: Here, we report that CD1d-mediated antigenic presentation of myelin-derived galactosylceramide (Mye-GalCer) by macrophages contributed significantly to the progression of experimental autoimmune encephalomyelitis (EAE). Surprisingly, this presentation was recognizable by α-Galcer:CD1d-specific antibody (clone L363), but incapable of triggering expansion of iNKT cells and production of iNKT signature cytokines (IFNγ and IL-4). Likewise, a synthesized analog of Mye-Galcer, fluorinated α-C-GalCer (AA2), while being efficiently presented via CD1d on macrophages, failed to stimulate production of IFNγ and IL-4. However, AA2 significantly exacerbated EAE progression. Further analyses revealed that the antigenic presentations of both Mye-GalCer and its analog (AA2) in α-form via CD1d promoted IL-17 production from T cells, leading to elevated levels of IL-17 in EAE spinal cords and sera. The IL-17 neutralizing antibody significantly reduced the severity of EAE symptoms in AA2-treated mice. Furthermore, D-sphingosine, a lipid possessing the same hydrophobic base as ceramide but without a carbohydrate residue, efficiently blocked this glycolipid antigen presentation both in vitro and in spinal cords of EAE mice, and significantly decreased IL-17 and ameliorated the pathological symptoms. Conclusion: Our findings reveal a novel pathway from the presentation of Mye-GalCer to IL-17 production, and highlight the promising therapeutic potential of D-sphingosine for the human disorder of multiple sclerosis.
Collapse
Affiliation(s)
- Yi Ban
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States
| | - Wenjuan Dong
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States
| | - Lixing Zhang
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology, Shanghai Jiaotong University, Shanghai, China
| | - Tian Zhou
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, United States.,Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ahmad S Altiti
- Department of Chemistry, Hunter College, City University of New York, New York, NY, United States
| | - Khaleel Ali
- Department of Chemistry, Hunter College, City University of New York, New York, NY, United States
| | - David R Mootoo
- Department of Chemistry, Hunter College, City University of New York, New York, NY, United States
| | - Victoria A Blaho
- Department of Pathology and Laboratory Medicine, Center for Vascular Biology, Weill Medical Medicine, New York, NY, United States.,Sanford Burnham Prepys Medical Discovery Institute, La Jolla, CA, United States
| | - Timothy Hla
- Department of Pathology and Laboratory Medicine, Center for Vascular Biology, Weill Medical Medicine, New York, NY, United States
| | - Yi Ren
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States.,State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
33
|
A T-cell receptor escape channel allows broad T-cell response to CD1b and membrane phospholipids. Nat Commun 2019; 10:56. [PMID: 30610190 PMCID: PMC6320368 DOI: 10.1038/s41467-018-07898-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 12/03/2018] [Indexed: 01/22/2023] Open
Abstract
CD1 proteins are expressed on dendritic cells, where they display lipid antigens to T-cell receptors (TCRs). Here we describe T-cell autoreactivity towards ubiquitous human membrane phospholipids presented by CD1b. These T-cells discriminate between two major types of lipids, sphingolipids and phospholipids, but were broadly cross-reactive towards diverse phospholipids including phosphatidylcholine, phosphatidylinositol and phosphatidylethanolamine. The crystal structure of a representative TCR bound to CD1b-phosphatidylcholine provides a molecular mechanism for this promiscuous recognition. We observe a lateral escape channel in the TCR, which shunted phospholipid head groups sideways along the CD1b-TCR interface, without contacting the TCR. Instead the TCR recognition site involved the neck region phosphate that is common to all major self-phospholipids but absent in sphingolipids. Whereas prior studies have focused on foreign lipids or rare self-lipids, we define a new molecular mechanism of promiscuous recognition of common self-phospholipids including those that are known targets in human autoimmune disease.
Collapse
|
34
|
Shang S, Kats D, Cao L, Morgun E, Velluto D, He Y, Xu Q, Wang CR, Scott EA. Induction of Mycobacterium Tuberculosis Lipid-Specific T Cell Responses by Pulmonary Delivery of Mycolic Acid-Loaded Polymeric Micellar Nanocarriers. Front Immunol 2018; 9:2709. [PMID: 30538700 PMCID: PMC6277542 DOI: 10.3389/fimmu.2018.02709] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/02/2018] [Indexed: 12/23/2022] Open
Abstract
Mycolic acid (MA), a major lipid component of Mycobacterium tuberculosis (Mtb) cell wall, can be presented by the non-polymorphic antigen presenting molecule CD1b to T cells isolated from Mtb-infected individuals. These MA-specific CD1b-restricted T cells are cytotoxic, produce Th1 cytokines, and form memory populations, suggesting that MA can be explored as a potential subunit vaccine candidate for TB. However, the controlled elicitation of MA-specific T cell responses has been challenging due to difficulties in the targeted delivery of lipid antigens and a lack of suitable animal models. In this study, we generated MA-loaded micellar nanocarriers (MA-Mc) comprised of self-assembled poly(ethylene glycol)-bl-poly(propylene sulfide; PEG-PPS) copolymers conjugated to an acid sensitive fluorophore to enhance intracellular delivery of MA to phagocytic immune cells. Using humanized CD1 transgenic (hCD1Tg) mice, we found these nanobiomaterials to be endocytosed by bone marrow-derived dendritic cells (DCs) and localized to lysosomal compartments. Additionally, MA-Mc demonstrated superior efficacy over free MA in activating MA-specific TCR transgenic (DN1) T cells in vitro. Following intranasal immunization, MA-Mc were primarily taken up by alveolar macrophages and DCs in the lung and induced activation and proliferation of adoptively transferred DN1 T cells. Furthermore, intranasal immunization with MA-Mc induced MA-specific T cell responses in the lungs of hCD1Tg mice. Collectively, our data demonstrates that pulmonary delivery of MA via PEG-PPS micelles to DCs can elicit potent CD1b-restricted T cell responses both in vitro and in vivo and MA-Mc could be explored as subunit vaccines against Mtb infection.
Collapse
Affiliation(s)
- Shaobin Shang
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, United States
| | - Dina Kats
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, United States
| | - Liang Cao
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, United States
| | - Eva Morgun
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, United States
| | - Diana Velluto
- Diabetes Research Institute and Cell Transplant Center, University of Miami School of Medicine, Miami, FL, United States
| | - Ying He
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, United States
| | - Qichen Xu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, United States
| | - Evan A Scott
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, United States.,Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States.,Simpson Querrey Institute, Northwestern University, Chicago, IL, United States.,Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, United States
| |
Collapse
|
35
|
Lepore M, Mori L, De Libero G. The Conventional Nature of Non-MHC-Restricted T Cells. Front Immunol 2018; 9:1365. [PMID: 29963057 PMCID: PMC6010553 DOI: 10.3389/fimmu.2018.01365] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/01/2018] [Indexed: 12/17/2022] Open
Abstract
The definition “unconventional T cells” identifies T lymphocytes that recognize non-peptide antigens presented by monomorphic antigen-presenting molecules. Two cell populations recognize lipid antigens and small metabolites presented by CD1 and MR1 molecules, respectively. A third cell population expressing the TCR Vγ9Vδ2 is stimulated by small phosphorylated metabolites. In the recent past, we have learnt a lot about the selection, tissue distribution, gene transcription programs, mode of expansion after antigen recognition, and persistence of these cells. These studies depict their functions in immune homeostasis and diseases. Current investigations are revealing that unconventional T cells include distinct sub-populations, which display unexpected similarities to classical MHC-restricted T cells in terms of TCR repertoire diversity, antigen specificity variety, functional heterogeneity, and naïve-to-memory differentiation dynamic. This review discusses the latest findings with a particular emphasis on these T cells, which appear to be more conventional than previously appreciated, and with the perspective of using CD1 and MR1-restricted T cells in vaccination and immunotherapy.
Collapse
Affiliation(s)
- Marco Lepore
- Experimental Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| |
Collapse
|
36
|
Singh P, Rameshwaram NR, Ghosh S, Mukhopadhyay S. Cell envelope lipids in the pathophysiology of Mycobacterium tuberculosis. Future Microbiol 2018; 13:689-710. [PMID: 29771143 DOI: 10.2217/fmb-2017-0135] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Mycobacterium tuberculosis is an intracellular bacterium that persists and replicates inside macrophages. The bacterium possesses an unusual lipid-rich cell envelope that provides a hydrophobic impermeable barrier against many environmental stressors and allows it to survive extremely hostile intracellular surroundings. Since the lipid-rich envelope is crucial for M. tuberculosis virulence, the components of the cell wall lipid biogenesis pathways constitute an attractive target for the development of vaccines and antimycobacterial chemotherapeutics. In this review, we provide a detailed description of the mycobacterial cell envelope lipid components and their contributions to the physiology and pathogenicity of mycobacteria. We also discussed the current status of the antimycobacterial drugs that target biosynthesis, export and regulation of cell envelope lipids.
Collapse
Affiliation(s)
- Parul Singh
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500 039, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, Karnataka, 576 104, India
| | - Nagender Rao Rameshwaram
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500 039, India
| | - Sudip Ghosh
- Molecular Biology Division, National Institute of Nutrition (ICMR), Jamai-Osmania PO, Hyderabad, 500 007, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500 039, India
| |
Collapse
|
37
|
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
|
38
|
Sadhu S, Mitra DK. Emerging Concepts of Adaptive Immunity in Leprosy. Front Immunol 2018; 9:604. [PMID: 29686668 PMCID: PMC5900054 DOI: 10.3389/fimmu.2018.00604] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 03/09/2018] [Indexed: 12/11/2022] Open
Abstract
Leprosy is a chronic intracellular infection caused by the acid-fast bacillus, Mycobacterium leprae. The disease chiefly affects the skin, peripheral nerves, mucosa of the upper respiratory tract, and the eyes. The damage to peripheral nerves results in sensory and motor impairment with characteristic deformities and disability. Presently, the disease remains concentrated in resource-poor countries in tropical and warm temperate regions with the largest number of cases reported from India. Even though innate immunity influences the clinical manifestation of the disease, it is the components of adaptive immune system which seem to tightly correlate with the characteristic spectrum of leprosy. M. leprae-specific T cell anergy with bacillary dissemination is the defining feature of lepromatous leprosy (LL) patients in contrast to tuberculoid leprosy (TT) patients, which is characterized by strong Th1-type cell response with localized lesions. Generation of Th1/Th2-like effector cells, however, cannot wholly explain the polarized state of immunity in leprosy. A comprehensive understanding of the role of various regulatory T cells, such as Treg and natural killer T cells, in deciding the polarized state of T cell immunity is crucial. Interaction of these T cell subsets with effector T cells like Th1 (IFN-γ dominant), Th2 (interluekin-4 dominant), and Th17 (IL-17+) cells through various regulatory cytokines and molecules (programmed death-1/programmed death ligand-1) may constitute key events in dictating the state of immune polarization, thus controlling the clinical manifestation. Studying these important components of the adaptive immune system in leprosy patients is essential for better understanding of immune function, correlate(s) the immunity and mechanism(s) of its containment.
Collapse
Affiliation(s)
- Soumi Sadhu
- Department of Transplant Immunology, All India Institute of Medical Sciences (AIIMS), New Delhi, India.,Department of Immunogenetics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Dipendra Kumar Mitra
- Department of Transplant Immunology, All India Institute of Medical Sciences (AIIMS), New Delhi, India.,Department of Immunogenetics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
39
|
Chancellor A, Gadola SD, Mansour S. The versatility of the CD1 lipid antigen presentation pathway. Immunology 2018; 154:196-203. [PMID: 29460282 DOI: 10.1111/imm.12912] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/12/2018] [Accepted: 02/14/2018] [Indexed: 12/19/2022] Open
Abstract
The family of non-classical major histocompatibility complex (MHC) class-I like CD1 molecules has an emerging role in human disease. Group 1 CD1 includes CD1a, CD1b and CD1c, which function to display lipids on the cell surface of antigen-presenting cells for direct recognition by T-cells. The recent advent of CD1 tetramers and the identification of novel lipid ligands has contributed towards the increasing number of CD1-restricted T-cell clones captured. These advances have helped to identify novel donor unrestricted and semi-invariant T-cell populations in humans and new mechanisms of T-cell recognition. However, although there is an opportunity to design broadly acting lipids and harness the therapeutic potential of conserved T-cells, knowledge of their role in health and disease is lacking. We briefly summarize the current evidence implicating group 1 CD1 molecules in infection, cancer and autoimmunity and show that although CD1 are not as diverse as MHC, recent discoveries highlight their versatility as they exhibit intricate mechanisms of antigen presentation.
Collapse
Affiliation(s)
- Andrew Chancellor
- Faculty of Medicine, Academic Unit of Clinical and Experimental Sciences, Southampton, UK
| | - Stephan D Gadola
- Faculty of Medicine, Academic Unit of Clinical and Experimental Sciences, Southampton, UK.,F.Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Salah Mansour
- Faculty of Medicine, Academic Unit of Clinical and Experimental Sciences, Southampton, UK
| |
Collapse
|
40
|
Teyton L. Role of lipid transfer proteins in loading CD1 antigen-presenting molecules. J Lipid Res 2018; 59:1367-1373. [PMID: 29559523 PMCID: PMC6071766 DOI: 10.1194/jlr.r083212] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/18/2018] [Indexed: 11/20/2022] Open
Abstract
Research to connect lipids with immunology is growing, but details about the specific roles of lipid transfer proteins (LTPs) in antigen presentation remain unclear. A single class of major histocompatibility class-like molecules, called CD1 molecules, can present lipids and glycolipids to the immune system. These molecules all have a common hydrophobic antigen-binding groove. The loading of this groove with various lipids throughout the life of a CD1 molecule defines the immune recognition of lipids by T cells. At each location of residence, CD1 molecules are exposed to particular physicochemical conditions, particular collections of lipids, and unique combinations of LTPs that will define which lipids bind to CD1 and which do not. The lipid transfer machinery that is used by CD1 molecules is entirely hijacked from the normal synthetic and catalytic pathways of lipids. The precise determinants that regulate the presentation of certain lipids over others with respect to chemistry, solubility, and abundance are still poorly defined and require investigation to allow the use of lipids as regular antigenic targets of immunotherapy and vaccine.
Collapse
Affiliation(s)
- Luc Teyton
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
41
|
James CA, Yu KKQ, Gilleron M, Prandi J, Yedulla VR, Moleda ZZ, Diamanti E, Khan M, Aggarwal VK, Reijneveld JF, Reinink P, Lenz S, Emerson RO, Scriba TJ, Souter MNT, Godfrey DI, Pellicci DG, Moody DB, Minnaard AJ, Seshadri C, Van Rhijn I. CD1b Tetramers Identify T Cells that Recognize Natural and Synthetic Diacylated Sulfoglycolipids from Mycobacterium tuberculosis. Cell Chem Biol 2018; 25:392-402.e14. [PMID: 29398561 DOI: 10.1016/j.chembiol.2018.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/05/2017] [Accepted: 01/04/2018] [Indexed: 12/23/2022]
Abstract
Mycobacterial cell wall lipids bind the conserved CD1 family of antigen-presenting molecules and activate T cells via their T cell receptors (TCRs). Sulfoglycolipids (SGLs) are uniquely synthesized by Mycobacterium tuberculosis, but tools to study SGL-specific T cells in humans are lacking. We designed a novel hybrid synthesis of a naturally occurring SGL, generated CD1b tetramers loaded with natural or synthetic SGL analogs, and studied the molecular requirements for TCR binding and T cell activation. Two T cell lines derived using natural SGLs are activated by synthetic analogs independently of lipid chain length and hydroxylation, but differentially by saturation status. By contrast, two T cell lines derived using an unsaturated SGL synthetic analog were not activated by the natural antigen. Our data provide a bioequivalence hierarchy of synthetic SGL analogs and SGL-loaded CD1b tetramers. These reagents can now be applied to large-scale translational studies investigating the diagnostic potential of SGL-specific T cell responses or SGL-based vaccines.
Collapse
Affiliation(s)
- Charlotte A James
- Department of Medicine, University of Washington Medical Center, 750 Republican Street, Suite E663, Seattle, WA 98115, USA; Department of Pathology, Molecular Medicine and Mechanisms of Disease Program, University of Washington, Seattle, WA 98195, USA
| | - Krystle K Q Yu
- Department of Medicine, University of Washington Medical Center, 750 Republican Street, Suite E663, Seattle, WA 98115, USA
| | - Martine Gilleron
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jacques Prandi
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Vijayendar R Yedulla
- Stratingh Institute for Chemistry, University of Groningen, 9747AG Groningen, the Netherlands
| | - Zuzanna Z Moleda
- Stratingh Institute for Chemistry, University of Groningen, 9747AG Groningen, the Netherlands
| | | | - Momin Khan
- School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | | | - Josephine F Reijneveld
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584CL Utrecht, the Netherlands
| | - Peter Reinink
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584CL Utrecht, the Netherlands
| | - Stefanie Lenz
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584CL Utrecht, the Netherlands
| | | | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative and Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7935, South Africa
| | - Michael N T Souter
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Daniel G Pellicci
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC 3010, Australia
| | - D Branch Moody
- Department of Rheumatology, Allergy & Immunology, Brigham and Women's Hospital, 60 Fenwood Road, Room 6006V, Boston, MA 02115, USA
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen, 9747AG Groningen, the Netherlands
| | - Chetan Seshadri
- Department of Medicine, University of Washington Medical Center, 750 Republican Street, Suite E663, Seattle, WA 98115, USA.
| | - Ildiko Van Rhijn
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584CL Utrecht, the Netherlands; Department of Rheumatology, Allergy & Immunology, Brigham and Women's Hospital, 60 Fenwood Road, Room 6006V, Boston, MA 02115, USA.
| |
Collapse
|
42
|
McMurtrey C, Harriff MJ, Swarbrick GM, Duncan A, Cansler M, Null M, Bardet W, Jackson KW, Lewinsohn DA, Hildebrand W, Lewinsohn DM. T cell recognition of Mycobacterium tuberculosis peptides presented by HLA-E derived from infected human cells. PLoS One 2017; 12:e0188288. [PMID: 29176828 PMCID: PMC5703486 DOI: 10.1371/journal.pone.0188288] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 11/03/2017] [Indexed: 12/28/2022] Open
Abstract
HLA-E is a non-conventional MHC Class I molecule that has been recently demonstrated to present pathogen-derived ligands, resulting in the TCR-dependent activation of αβ CD8+ T cells. The goal of this study was to characterize the ligandome displayed by HLA-E following infection with Mycobacterium tuberculosis (Mtb) using an in-depth mass spectrometry approach. Here we identified 28 Mtb ligands derived from 13 different source proteins, including the Esx family of proteins. When tested for activity with CD8+ T cells isolated from sixteen donors, nine of the ligands elicited an IFN-γ response from at least one donor, with fourteen of 16 donors responding to the Rv0634A19-29 peptide. Further evaluation of this immunodominant peptide response confirmed HLA-E restriction and the presence of Rv0634A19-29-reactive CD8+ T cells in the peripheral blood of human donors. The identification of an Mtb HLA-E ligand that is commonly recognized may provide a target for a non-traditional vaccine strategy.
Collapse
Affiliation(s)
- Curtis McMurtrey
- Department of Microbiology and Immunology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States of America
| | - Melanie J. Harriff
- VA Portland Health Care System, Portland, OR, United States of America
- Department of Pulmonary and Critical Care Medicine, Oregon Health and Sciences University, Portland, OR, United States of America
| | - Gwendolyn M. Swarbrick
- Department of Pediatric Medicine, Oregon Health and Sciences University, Portland, OR, United States of America
| | - Amanda Duncan
- Department of Pediatric Medicine, Oregon Health and Sciences University, Portland, OR, United States of America
| | - Meghan Cansler
- Department of Pediatric Medicine, Oregon Health and Sciences University, Portland, OR, United States of America
| | - Megan Null
- Department of Pediatric Medicine, Oregon Health and Sciences University, Portland, OR, United States of America
| | - Wilfried Bardet
- Department of Microbiology and Immunology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States of America
| | - Kenneth W. Jackson
- Department of Microbiology and Immunology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States of America
| | - Deborah A. Lewinsohn
- Department of Pediatric Medicine, Oregon Health and Sciences University, Portland, OR, United States of America
| | - William Hildebrand
- Department of Microbiology and Immunology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States of America
| | - David M. Lewinsohn
- VA Portland Health Care System, Portland, OR, United States of America
- Department of Pulmonary and Critical Care Medicine, Oregon Health and Sciences University, Portland, OR, United States of America
| |
Collapse
|
43
|
Le Nours J, Shahine A, Gras S. Molecular features of lipid-based antigen presentation by group 1 CD1 molecules. Semin Cell Dev Biol 2017; 84:48-57. [PMID: 29113870 DOI: 10.1016/j.semcdb.2017.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 09/12/2017] [Accepted: 11/02/2017] [Indexed: 11/18/2022]
Abstract
Lipids are now widely considered to play a variety of important roles in T-cell mediated immunity, including serving as antigens. Lipid-based antigens are presented by a specialised group of glycoproteins termed CD1. In humans, three classes of CD1 molecules exist: group 1 (CD1a, CD1b, CD1c), group 2 (CD1d), and group 3 (CD1e). While CD1d-mediated T-cell immunity has been extensively investigated, we have only recently gained insights into the structure and function of group 1 CD1 molecules. Structural studies have revealed how lipid-based antigens are presented by group 1 CD1 molecules, as well as shedding light on the molecular requirements for T-cell recognition. Here, we provide an overview of our current understanding of lipid presentation by group 1 CD1 molecules in humans and their recognition by T-cells, as well as examining the potential differences in lipid presentation that may occur across different species.
Collapse
Affiliation(s)
- Jérôme Le Nours
- 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 for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Adam Shahine
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - 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 for Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
44
|
Harnessing the CD1 restricted T cell response for leukemia adoptive immunotherapy. Cytokine Growth Factor Rev 2017; 36:117-123. [PMID: 28712863 DOI: 10.1016/j.cytogfr.2017.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/15/2017] [Indexed: 01/03/2023]
Abstract
Disease recurrence following chemotherapy and allogeneic hematopoietic cell transplantation is the major unmet clinical need of acute leukemia. Adoptive cell therapy (ACT) with allogeneic T lymphocytes can control recurrences at the cost of inducing detrimental GVHD. Targeting T cell recognition on leukemia cells is therefore needed to overcome the problem and ensure safe and durable disease remission. In this review, we discuss adoptive cells therapy based on CD1-restricted T cells specific for tumor associated self-lipid antigens. CD1 molecules are identical in every individual and expressed essentially on mature hematopoietic cells and leukemia blasts, but not by parenchymatous cells, while lipid antigens are enriched in malignant cells and unlike to mutate upon immune-mediated selective pressure. Redirecting T cells against self-lipids presented by CD1 molecules can thus provide an appealing cell therapy strategy for acute leukemia that is patient-unrestricted and can minimize risks for GVHD, implying potential prognostic improvement for this cancer.
Collapse
|
45
|
Busch M, Herzmann C, Kallert S, Zimmermann A, Höfer C, Mayer D, Zenk SF, Muche R, Lange C, Bloom BR, Modlin RL, Stenger S. Lipoarabinomannan-Responsive Polycytotoxic T Cells Are Associated with Protection in Human Tuberculosis. Am J Respir Crit Care Med 2017; 194:345-55. [PMID: 26882070 DOI: 10.1164/rccm.201509-1746oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The development of host-targeted, prophylactic, and therapeutic interventions against tuberculosis requires a better understanding of the immune mechanisms that determine the outcome of infection with Mycobacterium tuberculosis. OBJECTIVES To identify T-cell-dependent mechanisms that are protective in tuberculosis. METHODS Multicolor flow cytometry, cell sorting and growth inhibition assays were employed to compare the frequency, phenotype and function of T lymphocytes from bronchoalveolar lavage or the peripheral blood. MEASUREMENTS AND MAIN RESULTS At two independent study sites, bronchoalveolar lavage cells from donors with latent tuberculosis infection limited the growth of virulent Mycobacterium tuberculosis more efficiently than those in patients who developed disease. Unconventional, glycolipid-responsive T cells contributed to reduced mycobacterial growth because antibodies to CD1b inhibited this effect by 55%. Lipoarabinomannan was the most potent mycobacterial lipid antigen (activation of 1.3% T lymphocytes) and activated CD1b-restricted T cells that limited bacterial growth. A subset of IFN-γ-producing lipoarabinomannan-responsive T cells coexpressed the cytotoxic molecules perforin, granulysin, and granzyme B, which we termed polycytotoxic T cells. Taking advantage of two well-defined cohorts of subjects latently infected with Mycobacterium tuberculosis or patients who developed active disease after infection, we found a correlation between the frequency of polycytotoxic T cells and the ability to control infection (latent tuberculosis infection, 62%; posttuberculosis patients, 26%). CONCLUSIONS Our data define an unconventional CD8(+) T-cell subset (polycytotoxic T cells) that is based on antigen recognition and function. The results link clinical and mechanistic evidence that glycolipid-responsive, polycytotoxic T cells contribute to protection against tuberculosis.
Collapse
Affiliation(s)
- Martin Busch
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Christian Herzmann
- 2 Division of Clinical Infectious Diseases, German Center for Infection Research, Borstel, Germany
| | - Stephanie Kallert
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Andreas Zimmermann
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Christoph Höfer
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Daniel Mayer
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Sebastian F Zenk
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Rainer Muche
- 3 Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Christoph Lange
- 2 Division of Clinical Infectious Diseases, German Center for Infection Research, Borstel, Germany
| | - Barry R Bloom
- 4 Harvard School of Public Health, Boston, Massachusetts; and
| | - Robert L Modlin
- 5 Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Steffen Stenger
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | | |
Collapse
|
46
|
Kaczmarek R, Pasciak M, Szymczak-Kulus K, Czerwinski M. CD1: A Singed Cat of the Three Antigen Presentation Systems. Arch Immunol Ther Exp (Warsz) 2017; 65:201-214. [PMID: 28386696 PMCID: PMC5434122 DOI: 10.1007/s00005-017-0461-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/20/2017] [Indexed: 02/07/2023]
Abstract
Contrary to general view that the MHC Class I and II are the kapellmeisters of recognition and response to antigens, there is another big player in that part of immunity, represented by CD1 glycoproteins. In contrast to MHC Class I or II, which present peptides, CD1 molecules present lipids. Humans express five CD1 proteins (CD1a-e), four of which (CD1a-d) are trafficked to the cell surface, where they may display lipid antigens to T-cell receptors. This interaction may lead to both non-cognate and cognate T cell help to B cells, the latter eliciting anti-lipid antibody response. All CD1 proteins can bind a broad range of structurally different exogenous and endogenous lipids, but each shows a preference to one or more lipid classes. This unorthodox binding behavior is the result of elaborate architectures of CD1 binding clefts and distinct intracellular trafficking routes. Together, these features make CD1 system a versatile player in immune response, sitting at the crossroads of innate and adaptive immunity. While CD1 system may be involved in numerous infectious, inflammatory, and autoimmune diseases, its involvement may lead to opposite outcomes depending on different pathologies. Despite these ambiguities and complexity, CD1 system draws growing attention and continues to show glimmers of therapeutic potential. In this review, we summarize the current knowledge about CD1 proteins, their structures, lipid-binding profiles, and roles in immunity, and evaluate the role of CD1 proteins in eliciting humoral immune response.
Collapse
Affiliation(s)
- Radoslaw Kaczmarek
- Laboratory of Glycoconjugate Immunochemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Mariola Pasciak
- Laboratory of Medical Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Katarzyna Szymczak-Kulus
- Laboratory of Glycoconjugate Immunochemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Marcin Czerwinski
- Laboratory of Glycoconjugate Immunochemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland. .,Faculty of Physiotherapy and Physical Education, Opole University of Technology, Opole, Poland.
| |
Collapse
|
47
|
Protective efficacy of a lipid antigen vaccine in a guinea pig model of tuberculosis. Vaccine 2017; 35:1395-1402. [DOI: 10.1016/j.vaccine.2017.01.079] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/12/2017] [Accepted: 01/30/2017] [Indexed: 01/25/2023]
|
48
|
Ishikawa E, Mori D, Yamasaki S. Recognition of Mycobacterial Lipids by Immune Receptors. Trends Immunol 2017; 38:66-76. [DOI: 10.1016/j.it.2016.10.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/24/2016] [Accepted: 10/28/2016] [Indexed: 01/03/2023]
|
49
|
Abstract
Peptide-specific conventional T cells have been major targets for designing most antimycobacterial vaccines. Immune responses mediated by conventional T cells exhibit a delayed onset upon primary infection and are highly variable in different human populations. In contrast, innate-like T cells quickly respond to pathogens and display effector functions without undergoing extensive clonal expansion. Specifically, the activation of innate-like T cells depends on the promiscuous interaction of highly conserved antigen-presenting molecules, non-peptidic antigens, and likely semi-invariant T cell receptors. In antimicrobial immune responses, mucosal-associated invariant T cells are activated by riboflavin precursor metabolites presented by major histocompatibility complex-related protein I, while lipid-specific T cells including natural killer T cells are activated by lipid metabolites presented by CD1 proteins. Multiple innate-like T cell subsets have been shown to be protective or responsive in mycobacterial infections. Through rapid cytokine secretion, innate-like T cells function in early defense and memory response, offering novel advantages over conventional T cells in the design of anti-tuberculosis strategies.
Collapse
Affiliation(s)
- Shouxiong Huang
- Department of Environmental Health, University of Cincinnati College of Medicine , Cincinnati, OH , USA
| |
Collapse
|
50
|
CD1-Restricted T Cells at the Crossroad of Innate and Adaptive Immunity. J Immunol Res 2016; 2016:2876275. [PMID: 28070524 PMCID: PMC5192300 DOI: 10.1155/2016/2876275] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/13/2016] [Indexed: 11/17/2022] Open
Abstract
Lipid-specific T cells comprise a group of T cells that recognize lipids bound to the MHC class I-like CD1 molecules. There are four isoforms of CD1 that are expressed at the surface of antigen presenting cells and therefore capable of presenting lipid antigens: CD1a, CD1b, CD1c, and CD1d. Each one of these isoforms has distinct structural features and cellular localizations, which promotes binding to a broad range of different types of lipids. Lipid antigens originate from either self-tissues or foreign sources, such as bacteria, fungus, or plants and their recognition by CD1-restricted T cells has important implications in infection but also in cancer and autoimmunity. In this review, we describe the characteristics of CD1 molecules and CD1-restricted lipid-specific T cells, highlighting the innate-like and adaptive-like features of different CD1-restricted T cell subtypes.
Collapse
|