101
|
Ghinnagow R, Cruz LJ, Macho-Fernandez E, Faveeuw C, Trottein F. Enhancement of Adjuvant Functions of Natural Killer T Cells Using Nanovector Delivery Systems: Application in Anticancer Immune Therapy. Front Immunol 2017; 8:879. [PMID: 28798749 PMCID: PMC5529346 DOI: 10.3389/fimmu.2017.00879] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 11/13/2022] Open
Abstract
Type I natural killer T (NKT) cells have gained considerable interest in anticancer immune therapy over the last decade. This “innate-like” T lymphocyte subset has the unique ability to recognize foreign and self-derived glycolipid antigens in association with the CD1d molecule expressed by antigen-presenting cells. An important property of these cells is to bridge innate and acquired immune responses. The adjuvant function of NKT cells might be exploited in the clinics. In this review, we discuss the approaches currently being used to target NKT cells for cancer therapy. In particular, we highlight ongoing strategies utilizing NKT cell-based nanovaccines to optimize immune therapy.
Collapse
Affiliation(s)
- Reem Ghinnagow
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elodie Macho-Fernandez
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| |
Collapse
|
102
|
|
103
|
Li X, Huang J, Kawamura A, Funakoshi R, Porcelli SA, Tsuji M. Co-localization of a CD1d-binding glycolipid with an adenovirus-based malaria vaccine for a potent adjuvant effect. Vaccine 2017; 35:3171-3177. [PMID: 28483194 PMCID: PMC5489412 DOI: 10.1016/j.vaccine.2017.04.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/26/2022]
Abstract
A CD1d-binding, invariant (i) natural killer T (NKT)-cell stimulatory glycolipid, α-Galactosylceramide (αGalCer), has been shown to act as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying a higher binding affinity for CD1d molecule and more potent adjuvant activity than αGalCer. In the present study, 7DW8-5 co-administered intramuscularly (i.m.) with a recombinant adenovirus expressing a Plasmodium yoelii circumsporozoite protein (PyCSP), AdPyCS, has led to a co-localization of 7DW8-5 and a PyCSP in draining lymph nodes (dLNs), particularly in dendritic cells (DCs). This occurrence initiates a cascade of events, such as the recruitment of DCs to dLNs and their activation and maturation, and the enhancement of the ability of DCs to prime CD8+ T cells induced by AdPyCS and ultimately leading to a potent adjuvant effect and protection against malaria.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adjuvants, Immunologic
- Animals
- Antigens, CD1d/immunology
- Antigens, CD1d/metabolism
- Antigens, Protozoan/administration & dosage
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- CD8-Positive T-Lymphocytes/immunology
- Dendritic Cells/immunology
- Galactosylceramides/chemistry
- Galactosylceramides/immunology
- Galactosylceramides/metabolism
- Immunogenicity, Vaccine
- Injections, Intramuscular
- Interferon-gamma/immunology
- Killer Cells, Natural/immunology
- Lymphocyte Activation
- Malaria/immunology
- Malaria/prevention & control
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/immunology
- Mice
- Natural Killer T-Cells/immunology
- Plasmodium yoelii/chemistry
- Plasmodium yoelii/genetics
- Plasmodium yoelii/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Xiangming Li
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA
| | - Jing Huang
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA
| | - Akira Kawamura
- Department of Chemistry, Hunter College of The City University of New York, New York, NY 10065, USA
| | - Ryota Funakoshi
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA
| | - Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA.
| |
Collapse
|
104
|
Keller CW, Freigang S, Lünemann JD. Reciprocal Crosstalk between Dendritic Cells and Natural Killer T Cells: Mechanisms and Therapeutic Potential. Front Immunol 2017; 8:570. [PMID: 28596767 PMCID: PMC5442181 DOI: 10.3389/fimmu.2017.00570] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/28/2017] [Indexed: 12/23/2022] Open
Abstract
Natural killer T cells carrying a highly conserved, semi-invariant T cell receptor (TCR) [invariant natural killer T (iNKT) cells] are a subset of unconventional T lymphocytes that recognize glycolipids presented by CD1d molecules. Although CD1d is expressed on a variety of hematopoietic and non-hematopoietic cells, dendritic cells (DCs) are key presenters of glycolipid antigen in vivo. When stimulated through their TCR, iNKT cells rapidly secrete copious amounts of cytokines and induce maturation of DCs, thereby facilitating coordinated stimulation of innate and adaptive immune responses. The bidirectional crosstalk between DCs and iNKT cells determines the functional outcome of iNKT cell-targeted responses and iNKT cell agonists are used and currently being evaluated as adjuvants to enhance the efficacy of antitumor immunotherapy. This review illustrates mechanistic underpinnings of reciprocal DCs and iNKT cell interactions and discusses how those can be harnessed for cancer therapy.
Collapse
Affiliation(s)
- Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Stefan Freigang
- Institute of Pathology, Laboratory of Immunopathology, University of Bern, Bern, Switzerland
| | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland.,Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
105
|
Terabe M, Robertson FC, Clark K, De Ravin E, Bloom A, Venzon DJ, Kato S, Mirza A, Berzofsky JA. Blockade of only TGF-β 1 and 2 is sufficient to enhance the efficacy of vaccine and PD-1 checkpoint blockade immunotherapy. Oncoimmunology 2017. [PMID: 28638730 DOI: 10.1080/2162402x.2017.1308616] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Checkpoint inhibition has established immunotherapy as a major modality of cancer treatment. However, the success of cancer immunotherapy is still limited as immune regulation of tumor immunity is very complicated and mechanisms involved may also differ among cancer types. Beside checkpoints, other good candidates for immunotherapy are immunosuppressive cytokines. TGF-β is a very potent immunosuppressive cytokine involved in suppression of tumor immunity and also necessary for the function of some regulatory cells. TGF-β has three isoforms, TGF-β 1, 2 and 3. It has been demonstrated in multiple mouse tumor models that inhibition of all three isoforms of TGF-β facilitates natural tumor immunosurveillance and tumor vaccine efficacy. However, individual isoforms of TGF-β are not well studied yet. Here, by using monoclonal antibodies (mAbs) specific for TGF-β isoforms, we asked whether it is necessary to inhibit TGF-β3 to enhance tumor immunity. We found that blockade of TGF-β1 and 2 and of all isoforms provided similar effects on tumor natural immunosurveillance and therapeutic vaccine-induced tumor immunity. The protection was CD8+ T cell-dependent. Blockade of TGF-β increased vaccine-induced Th1-type response measured by IFNγ production or T-bet expression in both tumor draining lymph nodes and tumors, although it did not increase tumor antigen-specific CD8+ T cell numbers. Therefore, protection correlated with qualitative rather than quantitative changes in T cells. Furthermore, when combined with PD-1 blockade, blockade of TGF-β1 and 2 further increased vaccine efficacy. In conclusion, blocking TGF-β1 and 2 is sufficient to enhance tumor immunity, and it can be further enhanced with PD-1 blockade.
Collapse
Affiliation(s)
- Masaki Terabe
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Faith C Robertson
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Katharine Clark
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Emma De Ravin
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Anja Bloom
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David J Venzon
- Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shingo Kato
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Jay A Berzofsky
- Vaccine Branch and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
106
|
Lopes AMM, Michelin MA, Murta EFC. Monocyte-derived dendritic cells from patients with cervical intraepithelial lesions. Oncol Lett 2017; 13:1456-1462. [PMID: 28454277 DOI: 10.3892/ol.2017.5595] [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: 04/27/2016] [Accepted: 11/17/2016] [Indexed: 01/23/2023] Open
Abstract
Immunotherapy with dendritic cells (DCs) is a great promise for the treatment of neoplasms. However, the obtainment and protocol of differentiation of these cells may depend on extrinsic factors such as the tumor itself. The aim of the present study was to verify the influence of cervical neoplasia on different protocols of differentiation of monocyte-derived DCs resulting in an increased maturation phenotype. A total of 83 women were included in the study. The patients were grouped in low-grade squamous intraepithelial lesion (LSIL) (n=30), high-grade squamous intraepithelial lesion (HSIL) (n=22), cervical cancer (n=10) and healthy patients (n=21) groups. The mononuclear cells of patients were subjected to three differentiation protocols. In protocol I (pI), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-4 and tumor necrosis factor (TNF)-α were used for the differentiation of mature DCs (pIDCs). In protocol II (pII), monocytes were stimulated with GM-CSF, IL-4, TNF-α and activated lymphocytes in the absence of non-adherent cells (pIIDCs). In protocol III (pIII), monocytes were stimulated with GM-CSF, IL-4, TNF-α and activated lymphocytes in the presence of non-adherent cells (pIIIDCs). These cells were evaluated by flow cytometry for the expression of maturation markers such as cluster of differentiation (CD)11c, CD86 and human leukocyte antigen-antigen D related (HLA-DR). The main cytokines secreted (IL-4, IL-12 and transforming growth factor-β) were measured by ELISA. Our results indicate a significantly lower mature profile of pIIDCs and a significant increase in CD11c+ pIIIDCs able to produce IL-12 (P=0.0007). Furthermore, a significant reduction in cervical cancer HLA-DR+ pIDCs (P=0.0113) was also observed. HSIL patients exhibited a higher percentage of HLA-DR+ pIIDCs (P=0.0113), while LSIL patients had a lower percentage of CD11c+ pIIIDCs (P=0.0411). These findings suggest that the extent of cervical lesions affects the process of differentiation of DCs. Furthermore, activated lymphocytes may induce a better maturation of monocyte-derived DCs, and the presence of mononuclear cells appears to contribute to the DC differentiation process.
Collapse
Affiliation(s)
- Angela Maria Moed Lopes
- Oncology Research Institute, Federal University of The Triângulo Mineiro, Uberaba, MG 38025-440, Brazil
| | - Márcia Antoniazi Michelin
- Oncology Research Institute, Federal University of The Triângulo Mineiro, Uberaba, MG 38025-440, Brazil.,Discipline of Immunology, Clinical Hospital of Federal University of The Triângulo Mineiro, Uberaba, MG 38025-440, Brazil
| | - Eddie Fernando Cândido Murta
- Oncology Research Institute, Federal University of The Triângulo Mineiro, Uberaba, MG 38025-440, Brazil.,Discipline of Gynecology and Obstetrics, Clinical Hospital of Federal University of The Triângulo Mineiro, Uberaba, MG 38025-440, Brazil
| |
Collapse
|
107
|
Eby JM, Barse L, Henning SW, Rabelink MJWE, Klarquist J, Gilbert ER, Hammer AM, Fernandez MF, Yung N, Khan S, Miller HG, Kessler ER, Garrett-Mayer E, Dilling DF, Hoeben RC, Le Poole IC. Alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 1 can support immune responses toward tumors overexpressing ganglioside D3 in mice. Cancer Immunol Immunother 2017; 66:63-75. [PMID: 27787577 PMCID: PMC11028533 DOI: 10.1007/s00262-016-1920-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
Abstract
An immunotherapeutic strategy is discussed supporting anti-tumor activity toward malignancies overexpressing ganglioside D3. GD3 can be targeted by NKT cells when derived moieties are presented in the context of CD1d. NKT cells can support anti-tumor responses by secreting inflammatory cytokines and through cytotoxicity toward CD1d+GD3+ tumors. To overexpress GD3, we generated expression vector DNA and an adenoviral vector encoding the enzyme responsible for generating GD3 from its ubiquitous precursor GM3. We show that DNA encoding α-N-acetyl-neuraminide α-2,8-sialyltransferase 1 (SIAT8) introduced by gene gun vaccination in vivo leads to overexpression of GD3 and delays tumor growth. Delayed tumor growth is dependent on CD1d expression by host immune cells, as shown in experiments engaging CD1d knockout mice. A trend toward greater NKT cell populations among tumor-infiltrating lymphocytes is associated with SIAT8 vaccination. A single adenoviral vaccination introduces anti-tumor activity similarly to repeated vaccination with naked DNA. Here, greater NKT tumor infiltrates were accompanied by marked overexpression of IL-17 in the tumor, later switching to IL-4. Our results suggest that a single intramuscular adenoviral vaccination introduces overexpression of GD3 by antigen-presenting cells at the injection site, recruiting NKT cells that provide an inflammatory anti-tumor environment. We propose adenoviral SIAT8 (AdV-SIAT8) can slow the growth of GD3 expressing tumors in patients.
Collapse
Affiliation(s)
- Jonathan M Eby
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Levi Barse
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Steven W Henning
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Martijn J W E Rabelink
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jared Klarquist
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
- Department of Immunology and Microbiology, University of Colorado Denver, Denver, CO, USA
| | - Emily R Gilbert
- Department of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Adam M Hammer
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
- Burn and Shock Trauma Institute, Loyola University Chicago, Maywood, IL, USA
| | - Manuel F Fernandez
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Nathan Yung
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Safia Khan
- Oakton Community College, Des Plaines, IL, USA
| | | | - Edward R Kessler
- Department of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel F Dilling
- Department of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Rob C Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - I Caroline Le Poole
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA.
- Departments of Pathology, Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
108
|
Abstract
Conventional vaccine adjuvants enhance peptide-specific T-cell and B-cell responses by modifying peptide stability or uptake or by binding to pattern-recognition receptors on antigen-presenting cells (APCs). This article discusses the application of a distinct mechanism of adjuvant activity: the activation of type I, or invariant, natural killer T (iNKT) cells to drive cellular and humoral immune responses. Using a semi-invariant T-cell receptor (TCR), iNKT cells recognize glycolipid antigens presented on cluster of differentiation (CD)-1d molecules. When their ligands are presented in concert with peptides, iNKT cells can provide T-cell help, 'licensing' APCs to augment peptide-specific T-cell and antibody responses. We discuss the potential benefits and limitations of exploiting iNKT cells as 'universal helpers' to enhance vaccine responses for the treatment and prevention of cancer and infectious diseases.
Collapse
Affiliation(s)
- Mary Speir
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
- School of Biological Sciences, Victoria University Wellington, PO Box 600, Wellington, 6140, New Zealand.
- Maurice Wilkins Centre, Private Bag 92019, Auckland, New Zealand.
| | - Robert Weinkove
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
- Wellington Blood and Cancer Centre, Wellington Hospital, Private Bag 7902, Wellington, 6242, New Zealand.
- Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, 6021, New Zealand.
| |
Collapse
|
109
|
Li X, Huang J, Kaneko I, Zhang M, Iwanaga S, Yuda M, Tsuji M. A potent adjuvant effect of a CD1d-binding NKT cell ligand in human immune system mice. Expert Rev Vaccines 2017; 16:73-80. [PMID: 27801602 PMCID: PMC5526659 DOI: 10.1080/14760584.2017.1256208] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/31/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVES A CD1d-binding invariant natural killer T (iNKT)-cell stimulatory glycolipid, namely 7DW8-5, is shown to enhance the efficacy of radiation-attenuated sporozoites (RAS)-based malaria vaccine in mice. In the current study, we aim to determine whether 7DW8-5 can display a potent adjuvant effect in human immune system (HIS) mice. METHODS HIS-A2/hCD1d mice, which possess both functional human iNKT cells and CD8+ T cells, were generated by the transduction of NSG mice with adeno-associated virus serotype 9 expressing genes that encode human CD1d molecules and HLA-A*0201, followed by the engraftment of human hematopoietic stem cells. The magnitudes of human iNKT-cell response against 7DW8-5 and HLA-A*0201-restricted human CD8+ T-cell response against a human malaria antigen in HIS-A2/hCD1d mice were determined by using human CD1d tetramer and human HLA-A*0201 tetramer, respectively. RESULTS We found that 7DW8-5 stimulates human iNKT cells in HIS-A2/hCD1d mice, as well as those derived from HIS-A2/hCD1d mice in vitro. We also found that 7DW8-5 significantly increases the level of a human malarial antigen-specific HLA-A*0201-restricted human CD8+ T-cell response in HIS-A2/hCD1d mice. CONCLUSIONS Our study indicates that 7DW8-5 can display a potent adjuvant effect on RAS vaccine-induced anti-malarial immunity by augmenting malaria-specific human CD8+ T-cell response.
Collapse
Affiliation(s)
- Xiangming Li
- a HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center , Affiliate of The Rockefeller University , New York , NY , USA
| | - Jing Huang
- a HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center , Affiliate of The Rockefeller University , New York , NY , USA
| | - Izumi Kaneko
- b Department of Medical Zoology , Mie University Graduate School of Medicine , Tsu , Mie , Japan
| | - Min Zhang
- a HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center , Affiliate of The Rockefeller University , New York , NY , USA
- c Department of Pathology , New York University School of Medicine , New York , NY , USA
| | - Shiroh Iwanaga
- b Department of Medical Zoology , Mie University Graduate School of Medicine , Tsu , Mie , Japan
| | - Masao Yuda
- b Department of Medical Zoology , Mie University Graduate School of Medicine , Tsu , Mie , Japan
| | - Moriya Tsuji
- a HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center , Affiliate of The Rockefeller University , New York , NY , USA
| |
Collapse
|
110
|
Saez de Guinoa J, Jimeno R, Farhadi N, Jervis PJ, Cox LR, Besra GS, Barral P. CD1d-mediated activation of group 3 innate lymphoid cells drives IL-22 production. EMBO Rep 2017; 18:39-47. [PMID: 27799287 PMCID: PMC5210076 DOI: 10.15252/embr.201642412] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/07/2016] [Accepted: 10/04/2016] [Indexed: 12/21/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous family of immune cells that play a critical role in a variety of immune processes including host defence against infection, wound healing and tissue repair. Whether these cells are involved in lipid-dependent immunity remains unexplored. Here we show that murine ILCs from a variety of tissues express the lipid-presenting molecule CD1d, with group 3 ILCs (ILC3s) showing the highest level of expression. Within the ILC3 family, natural cytotoxicity triggering receptor (NCR)-CCR6+ cells displayed the highest levels of CD1d. Expression of CD1d on ILCs is functionally relevant as ILC3s can acquire lipids in vitro and in vivo and load lipids on CD1d to mediate presentation to the T-cell receptor of invariant natural killer T (iNKT) cells. Conversely, engagement of CD1d in vitro and administration of lipid antigen in vivo induce ILC3 activation and production of IL-22. Taken together, our data expose a previously unappreciated role for ILCs in CD1d-mediated immunity, which can modulate tissue homeostasis and inflammatory responses.
Collapse
Affiliation(s)
| | - Rebeca Jimeno
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Nazanin Farhadi
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Peter J Jervis
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Liam R Cox
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, UK
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| |
Collapse
|
111
|
Gilchuk P, Knight FC, Wilson JT, Joyce S. Eliciting Epitope-Specific CD8+ T Cell Response by Immunization with Microbial Protein Antigens Formulated with α-Galactosylceramide: Theory, Practice, and Protocols. Methods Mol Biol 2017; 1494:321-352. [PMID: 27718206 DOI: 10.1007/978-1-4939-6445-1_25] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD8+ cytotoxic T lymphocytes confer protection against infectious diseases caused by viruses, bacteria, and parasites. Hence, significant efforts have been invested into devising ways to generate CD8+ T cell-targeted vaccines. Generation of microbe-free protein subunit vaccines requires a thorough knowledge of protective target antigens. Such antigens are proteolytically processed peptides presented by MHC class I molecules. To induce a robust antigen-specific CD8+ T cell response through vaccination, it is essential to formulate the antigen with an effective adjuvant. Here, we describe a versatile method for generating high-frequency antigen-specific CD8+ T cells through immunization of mice using the invariant natural killer T cell agonist α-galactosylceramide as the adjuvant.
Collapse
Affiliation(s)
- Pavlo Gilchuk
- Veterans Administration Tennessee Valley Healthcare System, US Department of Veterans Affairs, Nashville, TN, 37332, USA.,Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, A4223 Medical Centre North, 1161 21st Avenue South, Nashville, TN, 37332, USA
| | - Frances C Knight
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, 37332, USA
| | - John T Wilson
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, 37332, USA.,Department of Chemical & Biomolecular Engineering, School of Engineering, Vanderbilt University, Nashville, TN, 37332, USA
| | - Sebastian Joyce
- Veterans Administration Tennessee Valley Healthcare System, US Department of Veterans Affairs, Nashville, TN, 37332, USA. .,Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, A4223 Medical Centre North, 1161 21st Avenue South, Nashville, TN, 37332, USA.
| |
Collapse
|
112
|
Brown DL. Immunopathology of the Hepatobiliary System. MOLECULAR AND INTEGRATIVE TOXICOLOGY 2017:329-417. [DOI: 10.1007/978-3-319-47385-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
113
|
Kohlgruber AC, Donado CA, LaMarche NM, Brenner MB, Brennan PJ. Activation strategies for invariant natural killer T cells. Immunogenetics 2016; 68:649-63. [PMID: 27457886 PMCID: PMC5745583 DOI: 10.1007/s00251-016-0944-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/15/2016] [Indexed: 12/17/2022]
Abstract
Invariant natural killer T (iNKT) cells are a specialized T cell subset that plays an important role in host defense, orchestrating both innate and adaptive immune effector responses against a variety of microbes. Specific microbial lipids and mammalian self lipids displayed by the antigen-presenting molecule CD1d can activate iNKT cells through their semi-invariant αβ T cell receptors (TCRs). iNKT cells also constitutively express receptors for inflammatory cytokines typically secreted by antigen-presenting cells (APCs) after recognition of pathogen-associated molecular patterns (PAMPs), and they can be activated through these cytokine receptors either in combination with TCR signals, or in some cases even in the absence of TCR signaling. During infection, experimental evidence suggests that both TCR-driven and cytokine-driven mechanisms contribute to iNKT cell activation. While the relative contributions of these two signaling mechanisms can vary widely depending on the infectious context, both lipid antigens and PAMPs mediate reciprocal activation of iNKT cells and APCs, leading to downstream activation of multiple other immune cell types to promote pathogen clearance. In this review, we discuss the mechanisms involved in iNKT cell activation during infection, focusing on the central contributions of both lipid antigens and PAMP-induced inflammatory cytokines, and highlight in vivo examples of activation during bacterial, viral, and fungal infections.
Collapse
Affiliation(s)
- Ayano C Kohlgruber
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos A Donado
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nelson M LaMarche
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick J Brennan
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
114
|
Immunotherapeutic strategies targeting natural killer T cell responses in cancer. Immunogenetics 2016; 68:623-38. [PMID: 27393665 DOI: 10.1007/s00251-016-0928-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022]
Abstract
Natural killer T (NKT) cells are a unique subset of lymphocytes that bridge the innate and adaptive immune system. NKT cells possess a classic αβ T cell receptor (TCR) that is able to recognize self and foreign glycolipid antigens presented by the nonclassical class I major histocompatibility complex (MHC) molecule, CD1d. Type I NKT cells (referred to as invariant NKT cells) express a semi-invariant Vα14Jα18 TCR in mice and Vα24Jα18 TCR in humans. Type II NKT cells are CD1d-restricted T cells that express a more diverse set of TCR α chains. The two types of NKT cells often exert opposing effects especially in tumor immunity, where type II cells generally suppress tumor immunity while type I NKT cells can enhance anti-tumor immune responses. In this review, we focus on the role of NKT cells in cancer. We discuss their effector and suppressive functions, as well as describe preclinical and clinical studies utilizing therapeutic strategies focused on harnessing their potent anti-tumor effector functions, and conclude with a discussion on potential next steps for the utilization of NKT cell-targeted therapies for the treatment of cancer.
Collapse
|
115
|
Kharkwal SS, Arora P, Porcelli SA. Glycolipid activators of invariant NKT cells as vaccine adjuvants. Immunogenetics 2016; 68:597-610. [PMID: 27377623 DOI: 10.1007/s00251-016-0925-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/22/2016] [Indexed: 11/26/2022]
Abstract
Natural Killer T cells (NKT cells) are a subpopulation of T lymphocytes with unique phenotypic properties and a remarkably broad range of immune effector and regulatory functions. One subset of these cells, known as invariant NKT cells (iNKT cells), has become a significant focus in the search for new and better ways to enhance immunotherapies and vaccination. These unconventional T cells are characterized by their ability to be specifically activated by a range of foreign and self-derived glycolipid antigens presented by CD1d, an MHC class I-related antigen presenting molecule that has evolved to bind and present lipid antigens. The development of synthetic α-galactosylceramides as a family of powerful glycolipid agonists for iNKT cells has led to approaches for augmenting a wide variety of immune responses, including those involved in vaccination against infections and cancers. Here we review the basic background biology of iNKT cells that is relevant to their potential for improving immune responses, and summarize recent work supporting the further development of glycolipid activators of iNKT cells as a new class of vaccine adjuvants.
Collapse
Affiliation(s)
- Shalu Sharma Kharkwal
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Pooja Arora
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Steven A Porcelli
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
116
|
Shimizu K, Yamasaki S, Shinga J, Sato Y, Watanabe T, Ohara O, Kuzushima K, Yagita H, Komuro Y, Asakura M, Fujii SI. Systemic DC Activation Modulates the Tumor Microenvironment and Shapes the Long-Lived Tumor-Specific Memory Mediated by CD8+ T Cells. Cancer Res 2016; 76:3756-66. [DOI: 10.1158/0008-5472.can-15-3219] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/04/2016] [Indexed: 11/16/2022]
|
117
|
Jukes J, Gileadi U, Ghadbane H, Yu T, Shepherd D, Cox LR, Besra GS, Cerundolo V. Non-glycosidic compounds can stimulate both human and mouse iNKT cells. Eur J Immunol 2016; 46:1224-34. [PMID: 26873393 PMCID: PMC4913735 DOI: 10.1002/eji.201546114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/05/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022]
Abstract
Invariant natural killer T (iNKT) cells recognize CD1d/glycolipid complexes and upon activation with synthetic agonists display immunostimulatory properties. We have previously described that the non-glycosidic CD1d-binding lipid, threitolceramide (ThrCer) activates murine and human iNKT cells. Here, we show that incorporating the headgroup of ThrCer into a conformationally more restricted 6- or 7-membered ring results in significantly more potent non-glycosidic analogs. In particular, ThrCer 6 was found to promote strong anti-tumor responses and to induce a more prolonged stimulation of iNKT cells than does the canonical α-galactosylceramide (α-GalCer), achieving an enhanced T-cell response at lower concentrations compared with α-GalCer both in vitro, using human iNKT-cell lines and in vivo, using C57BL/6 mice. Collectively, these studies describe novel non-glycosidic ThrCer-based analogs that have improved potency in iNKT-cell activation compared with that of α-GalCer, and are clinically relevant iNKT-cell agonists.
Collapse
Affiliation(s)
- John‐Paul Jukes
- MRC Human Immunology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Hemza Ghadbane
- MRC Human Immunology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Ting‐Fong Yu
- School of Biosciences, University of BirminghamEdgbastonBirminghamUK
- School of ChemistryUniversity of BirminghamEdgbastonBirminghamUK
| | - Dawn Shepherd
- MRC Human Immunology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Liam R. Cox
- School of ChemistryUniversity of BirminghamEdgbastonBirminghamUK
| | - Gurdyal S. Besra
- School of Biosciences, University of BirminghamEdgbastonBirminghamUK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
118
|
Coelho-Dos-Reis JG, Huang J, Tsao T, Pereira FV, Funakoshi R, Nakajima H, Sugiyama H, Tsuji M. Co-administration of α-GalCer analog and TLR4 agonist induces robust CD8(+) T-cell responses to PyCS protein and WT-1 antigen and activates memory-like effector NKT cells. Clin Immunol 2016; 168:6-15. [PMID: 27132023 DOI: 10.1016/j.clim.2016.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/18/2016] [Accepted: 04/26/2016] [Indexed: 12/28/2022]
Abstract
In the present study, the combined adjuvant effect of 7DW8-5, a potent α-GalCer-analog, and monophosphoryl lipid A (MPLA), a TLR4 agonist, on the induction of vaccine-induced CD8(+) T-cell responses and protective immunity was evaluated. Mice were immunized with peptides corresponding to the CD8(+) T-cell epitopes of a malaria antigen, a circumsporozoite protein of Plasmodium yoelii, and a tumor antigen, a Wilms Tumor antigen-1 (WT-1), together with 7DW8-5 and MPLA, as an adjuvant. These immunization regimens were able to induce higher levels of CD8(+) T-cell responses and, ultimately, enhanced levels of protection against malaria and tumor challenges compared to the levels induced by immunization with peptides mixed with 7DW8-5 or MPLA alone. Co-administration of 7DW8-5 and MPLA induces activation of memory-like effector natural killer T (NKT) cells, i.e. CD44(+)CD62L(-)NKT cells. Our study indicates that 7DW8-5 greatly enhances important synergistic pathways associated to memory immune responses when co-administered with MPLA, thus rendering this combination of adjuvants a novel vaccine adjuvant formulation.
Collapse
Affiliation(s)
- Jordana G Coelho-Dos-Reis
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA; Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz - FIOCRUZ, Minas Gerais 30192, Brazil.
| | - Jing Huang
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA
| | - Tiffany Tsao
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA
| | - Felipe V Pereira
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA; Federal University of Sao Paulo, Sao Paulo 04021, Brazil
| | - Ryota Funakoshi
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA
| | - Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016, USA.
| |
Collapse
|
119
|
Veinotte L, Gebremeskel S, Johnston B. CXCL16-positive dendritic cells enhance invariant natural killer T cell-dependent IFNγ production and tumor control. Oncoimmunology 2016; 5:e1160979. [PMID: 27471636 PMCID: PMC4938370 DOI: 10.1080/2162402x.2016.1160979] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 12/16/2022] Open
Abstract
Crosstalk interactions between dendritic cells (DCs) and invariant natural killer T (iNKT) cells are important in regulating antitumor responses elicited by glycolipid antigens. iNKT cells constitutively express the chemokine receptor CXCR6, while cytokine-activated DCs upregulate the transmembrane chemokine ligand, CXCL16. This study examined the co-stimulatory role of CXCR6/CXCL16 interactions in glycolipid-dependent iNKT cell activation and tumor control. Spleen and liver DCs in wild-type mice, but not iNKT cell deficient (Jα18−/−) mice, transiently upregulated surface CXCL16 following in vivo administration of the glycolipid antigen α-galactosylceramide. Recombinant CXCL16 did not directly induce iNKT cell activation in vitro but enhanced interferon (IFN)-γ production when mouse or human iNKT cells were stimulated with plate-bound anti-CD3. Compared with glycolipid-loaded CXCL16neg DCs, CXCL16hi DCs induced higher levels of IFNγ production in iNKT cell cultures and following adoptive transfer in vivo. The number of IFNγ+ iNKT cells and expansion of T-bet+ iNKT cells were reduced in vivo when CXCL16−/− DCs were used to activate iNKT cells. Enhanced IFNγ production in vivo was not dependent on CXCR6 expression on natural killer (NK) cells. Adoptive transfer of glycolipid-loaded CXCL16hi DCs provided superior protection against tumor metastasis compared to CXCL16neg DC transfers. Similarly, wild-type DCs provided superior protection against metastasis compared with CXCL16−/− DCs. These experiments implicate an important role for CXCR6/CXCL16 interactions in regulating iNKT cell IFNγ production and tumor control. The selective use of CXCL16hi DCs in adoptive transfer immunotherapies may prove useful for enhancing T helper (Th) type 1 responses and clinical outcomes in cancer patients.
Collapse
Affiliation(s)
- Linnea Veinotte
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Simon Gebremeskel
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Brent Johnston
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada; Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
120
|
Dwivedi V, Manickam C, Dhakal S, Binjawadagi B, Ouyang K, Hiremath J, Khatri M, Hague JG, Lee CW, Renukaradhya GJ. Adjuvant effects of invariant NKT cell ligand potentiates the innate and adaptive immunity to an inactivated H1N1 swine influenza virus vaccine in pigs. Vet Microbiol 2016; 186:157-63. [PMID: 27016770 DOI: 10.1016/j.vetmic.2016.02.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 02/27/2016] [Accepted: 02/29/2016] [Indexed: 02/07/2023]
Abstract
Pigs are considered as the source of some of the emerging human flu viruses. Inactivated swine influenza virus (SwIV) vaccine has been in use in the US swine herds, but it failed to control the flu outbreaks. The main reason has been attributed to lack of induction of strong local mucosal immunity in the respiratory tract. Invariant natural killer T (iNKT) cell is a unique T cell subset, and activation of iNKT cell using its ligand α-Galactosylceramide (α-GalCer) has been shown to potentiate the cross-protective immunity to inactivated influenza virus vaccine candidates in mice. Recently, we discovered iNKT cell in pig and demonstrated its activation using α-GalCer. In this study, we evaluated the efficacy of an inactivated H1N1 SwIV coadministered with α-GalCer intranasally against a homologous viral challenge. Our results demonstrated the potent adjuvant effects of α-GalCer in potentiating both innate and adaptive immune responses to SwIV Ags in the lungs of pigs, which resulted in reduction in the lung viral load by 3 logs compared to without adjuvant. Immunologically, in the lungs of pigs vaccinated with α-GalCer an increased virus specific IgA response, IFN-α secretion and NK cell-cytotoxicity was observed. In addition, iNKT cell-stimulation enhanced the secretion of Th1 cytokines (IFN-γ and IL-12) and reduced the production of immunosuppressive cytokines (IL-10 and TGF-β) in the lungs of pigs⋅ In conclusion, we demonstrated for the first time iNKT cell adjuvant effects in pigs to SwIV Ags through augmenting the innate and adaptive immune responses in the respiratory tract.
Collapse
Affiliation(s)
- Varun Dwivedi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Cordelia Manickam
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Basavaraj Binjawadagi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Kang Ouyang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States; College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jagadish Hiremath
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States; Indian Council of Agricultural Research, National Institute of Veterinary Epidemiology and Disease Informatics, Bangalore, India
| | - Mahesh Khatri
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Jacquelyn Gervay Hague
- Department of Chemistry, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Chang Won Lee
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States.
| |
Collapse
|
121
|
Compton BJ, Tang CW, Johnston KA, Osmond TL, Hayman CM, Larsen DS, Hermans IF, Painter GF. Synthesis and Activity of 6″-Deoxy-6″-thio-α-GalCer and Peptide Conjugates. Org Lett 2015; 17:5954-7. [PMID: 26606283 DOI: 10.1021/acs.orglett.5b02836] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A major challenge in the development of highly defined synthetic vaccines is the codelivery of vaccine components (i.e., antigen and adjuvant) to secondary lymphoid tissue to induce optimal immune responses. This problem can be addressed by synthesizing vaccines that comprise peptide antigens covalently attached to glycolipid adjuvants through biologically cleavable linkers. Toward this, a strategy utilizing previously unreported 6″-deoxy-6″-thio analogues of α-GalCer that can undergo chemoselective conjugation with peptide antigens is described. Administration of these conjugate vaccines leads to enhanced priming of antigen specific T cells. This simple vaccine design is broadly applicable to multiple disease indications such as cancer and infectious disease.
Collapse
Affiliation(s)
- Benjamin J Compton
- The Ferrier Research Institute, Victoria University of Wellington , P.O. Box 600, Wellington 6140, New Zealand
| | - Ching-wen Tang
- Malaghan Institute of Medical Research , P.O. Box 7060, Wellington 6242, New Zealand
| | - Karen A Johnston
- The Ferrier Research Institute, Victoria University of Wellington , P.O. Box 600, Wellington 6140, New Zealand
| | - Taryn L Osmond
- Malaghan Institute of Medical Research , P.O. Box 7060, Wellington 6242, New Zealand
| | - Colin M Hayman
- The Ferrier Research Institute, Victoria University of Wellington , P.O. Box 600, Wellington 6140, New Zealand
| | - David S Larsen
- Department of Chemistry, University of Otago , P.O. Box 56, Dunedin 9054, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research , P.O. Box 7060, Wellington 6242, New Zealand.,School of Biological Sciences, Victoria University of Wellington , P.O. Box 600, Wellington 6140, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery , 3 Symonds Street, Auckland Central 1142, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington , P.O. Box 600, Wellington 6140, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery , 3 Symonds Street, Auckland Central 1142, New Zealand
| |
Collapse
|
122
|
Beitzen-Heineke A, Bouzani M, Schmitt AL, Kurzai O, Hünniger K, Einsele H, Loeffler J. Human Invariant Natural Killer T cells possess immune-modulating functions during Aspergillus infection. Med Mycol 2015; 54:169-76. [PMID: 26483428 DOI: 10.1093/mmy/myv074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 07/12/2015] [Indexed: 01/25/2023] Open
Abstract
Aspergillus fumigatus is the most common cause for invasive fungal infections, a disease associated with high mortality in immune-compromised patients. CD1d-restricted invariant natural killer T (iNKT) cells compose a small subset of T cells known to impact the immune response toward various infectious pathogens. To investigate the role of human iNKT cells during A. fumigatus infection, we studied their activation as determined by CD69 expression and cytokine production in response to distinct fungal morphotypes in the presence of different CD1d(+) antigen presenting cells using flow cytometry and multiplex enzyme-linked immunosorbent assay (ELISA). Among CD1d(+) subpopulations, CD1d(+)CD1c(+) mDCs showed the highest potential to activate iNKT cells on a per cell basis. The presence of A. fumigatus decreased this effect of CD1d(+)CD1c(+) mDCs on iNKT cells and led to reduced secretion of TNF-α, G-CSF and RANTES. Production of other Th1 and Th2 cytokines was not affected by the fungus, suggesting an immune-modulating function for human iNKT cells during A. fumigatus infection.
Collapse
Affiliation(s)
| | - Maria Bouzani
- Medizinische Klinik und Poliklinik II, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Anna-Lena Schmitt
- Medizinische Klinik und Poliklinik II, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Oliver Kurzai
- Septomics Research Centre, Friedrich-Schiller-University Jena and Leibniz-Institute for Natural Products Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Kerstin Hünniger
- Septomics Research Centre, Friedrich-Schiller-University Jena and Leibniz-Institute for Natural Products Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Juergen Loeffler
- Medizinische Klinik und Poliklinik II, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
123
|
Neumann S, Young K, Compton B, Anderson R, Painter G, Hook S. Synthetic TRP2 long-peptide and α-galactosylceramide formulated into cationic liposomes elicit CD8+ T-cell responses and prevent tumour progression. Vaccine 2015; 33:5838-5844. [PMID: 26363382 DOI: 10.1016/j.vaccine.2015.08.083] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/28/2015] [Accepted: 08/29/2015] [Indexed: 12/22/2022]
Abstract
The lipid antigen α-galactosylceramide (α-GalCer) is a potent activator of invariant natural killer T-cells (iNKT cells) and can stimulate cytotoxic and anti-tumour immune responses. However optimal responses appear to be induced by α-GalCer when cell-based vaccines are delivered intravenously. Here we investigated if co-delivery of protein and peptide antigens along with α-GalCer in a liposomal formulation could stimulate therapeutic anti-tumour immune responses. Cationic liposomes were inherently immune-stimulatory and induced cytotoxic immune responses when delivered both by intravenous and subcutaneous injection. However, only vaccine delivered intravenously stimulated therapeutic anti-tumour immune responses to a peptide antigen. Surface modification with polyethylene glycol (PEG) did not improve immune responses to either intravenously or subcutaneously delivered vaccines. Immune responses to short and long peptide sequences (CD8 and CD4 epitopes) of the self-antigen tyrosinase-related protein 2 (TRP2) as a vaccine antigen, co-delivered with α-GalCer in either cationic liposomes or PBS were further examined. Enhanced production of IFN-γ, increased cytotoxic T-cell responses and tumour survival were observed when a long TRP2-peptide was delivered with α-GalCer in cationic liposomes.
Collapse
Affiliation(s)
- Silke Neumann
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Katie Young
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Benji Compton
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Regan Anderson
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Gavin Painter
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
124
|
Dölen Y, Kreutz M, Gileadi U, Tel J, Vasaturo A, van Dinther EAW, van Hout-Kuijer MA, Cerundolo V, Figdor CG. Co-delivery of PLGA encapsulated invariant NKT cell agonist with antigenic protein induce strong T cell-mediated antitumor immune responses. Oncoimmunology 2015; 5:e1068493. [PMID: 26942088 PMCID: PMC4760331 DOI: 10.1080/2162402x.2015.1068493] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/26/2015] [Accepted: 06/27/2015] [Indexed: 02/02/2023] Open
Abstract
Antitumor immunity can be enhanced by the coordinated release and delivery of antigens and immune-stimulating agents to antigen-presenting cells via biodegradable vaccine carriers. So far, encapsulation of TLR ligands and tumor-associated antigens augmented cytotoxic T cell (CTLs) responses. Here, we compared the efficacy of the invariant NKT (iNKT) cell agonist α-galactosylceramide (α-GalCer) and TLR ligands (R848 and poly I:C) as an adjuvant for the full length ovalbumin (OVA) in PLGA nanoparticles. We observed that OVA+α-GalCer nanoparticles (NP) are superior over OVA+TLR-L NP in generating and stimulating antigen-specific cytotoxic T lymphocytes without the need for CD4+ T cell help. Not only a 4-fold higher induction of antigen-specific T cells was observed, but also a more profound IFN-γ secretion was obtained by the addition α-GalCer. Surprisingly, we observed that mixtures of OVA containing NP with α-GalCer were ineffective, demonstrating that co-encapsulation of both α-GalCer and antigen within the same nanoparticle is essential for the observed T cell responses. Moreover, a single immunization with OVA+α-GalCer NP provided substantial protection from tumor formation and even delayed the growth of already established tumors, which coincided with a prominent and enhanced antigen-specific CD8+ T cell infiltration. The provided evidence on the advantage of antigen and α-GalCer coencapsulation should be considered in the design of future nanoparticle vaccines for therapeutic purposes.
Collapse
Affiliation(s)
- Yusuf Dölen
- Department of Tumor Immunology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Martin Kreutz
- Department of Tumor Immunology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jurjen Tel
- Department of Tumor Immunology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Angela Vasaturo
- Department of Tumor Immunology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Eric A. W. van Dinther
- Department of Tumor Immunology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Maaike A. van Hout-Kuijer
- Department of Tumor Immunology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud University Medical Center and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
125
|
Li X, Kawamura A, Andrews CD, Miller JL, Wu D, Tsao T, Zhang M, Oren D, Padte NN, Porcelli SA, Wong CH, Kappe SHI, Ho DD, Tsuji M. Colocalization of a CD1d-Binding Glycolipid with a Radiation-Attenuated Sporozoite Vaccine in Lymph Node-Resident Dendritic Cells for a Robust Adjuvant Effect. THE JOURNAL OF IMMUNOLOGY 2015; 195:2710-21. [PMID: 26254338 DOI: 10.4049/jimmunol.1403017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 07/07/2015] [Indexed: 01/20/2023]
Abstract
A CD1d-binding glycolipid, α-Galactosylceramide (αGalCer), activates invariant NK T cells and acts as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying nearly 100-fold stronger CD1d binding affinity. In the current study, 7DW8-5 was found to exert a more potent adjuvant effect than αGalCer for a vaccine based on radiation-attenuated sporozoites of a rodent malaria parasite, Plasmodium yoelii, also referred to as irradiated P. yoelii sporozoites (IrPySpz). 7DW8-5 had a superb adjuvant effect only when the glycolipid and IrPySpz were conjointly administered i.m. Therefore, we evaluated the effect of distinctly different biodistribution patterns of αGalCer and 7DW8-5 on their respective adjuvant activities. Although both glycolipids induce a similar cytokine response in sera of mice injected i.v., after i.m. injection, αGalCer induces a systemic cytokine response, whereas 7DW8-5 is locally trapped by CD1d expressed by dendritic cells (DCs) in draining lymph nodes (dLNs). Moreover, the i.m. coadministration of 7DW8-5 with IrPySpz results in the recruitment of DCs to dLNs and the activation and maturation of DCs. These events cause the potent adjuvant effect of 7DW8-5, resulting in the enhancement of the CD8(+) T cell response induced by IrPySpz and, ultimately, improved protection against malaria. Our study is the first to show that the colocalization of a CD1d-binding invariant NK T cell-stimulatory glycolipid and a vaccine, like radiation-attenuated sporozoites, in dLN-resident DCs upon i.m. conjoint administration governs the potency of the adjuvant effect of the glycolipid.
Collapse
Affiliation(s)
- Xiangming Li
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Akira Kawamura
- Department of Chemistry, Hunter College of The City University of New York, New York, NY 10065
| | - Chasity D Andrews
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | | | - Douglass Wu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Tiffany Tsao
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Min Zhang
- Department of Pathology, New York University, New York, NY 10016
| | - Deena Oren
- Structural Biology Resource Center, The Rockefeller University, New York, NY 10065
| | - Neal N Padte
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; and
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037; Academia Sinica, Taipei 115-74, Taiwan, Republic of China
| | | | - David D Ho
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY 10016;
| |
Collapse
|
126
|
Anderson CK, Salter AI, Toussaint LE, Reilly EC, Fugère C, Srivastava N, Kerr WG, Brossay L. Role of SHIP1 in Invariant NKT Cell Development and Functions. THE JOURNAL OF IMMUNOLOGY 2015; 195:2149-2156. [PMID: 26232432 DOI: 10.4049/jimmunol.1500567] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/07/2015] [Indexed: 11/19/2022]
Abstract
SHIP1 is a 5'-inositol phosphatase known to negatively regulate the signaling product of the PI3K pathway, phosphatidylinositol (3-5)-trisphosphate. SHIP1 is recruited to a large number of inhibitory receptors expressed on invariant NK (iNKT) cells. We hypothesized that SHIP1 deletion would have major effects on iNKT cell development by altering the thresholds for positive and negative selection. Germline SHIP1 deletion has been shown to affect T cells as well as other immune cell populations. However, the role of SHIP1 on T cell function has been controversial, and its participation on iNKT cell development and function has not been examined. We evaluated the consequences of SHIP1 deletion on iNKT cells using germline-deficient mice, chimeric mice, and conditionally deficient mice. We found that T cell and iNKT cell development are impaired in germline-deficient animals. However, this phenotype can be rescued by extrinsic expression of SHIP1. In contrast, SHIP1 is required cell autonomously for optimal iNKT cell cytokine secretion. This suggests that SHIP1 calibrates the threshold of iNKT cell reactivity. These data further our understanding of how iNKT cell activation is regulated and provide insights into the biology of this unique cell lineage.
Collapse
Affiliation(s)
- Courtney K Anderson
- Department of Molecular Microbiology & Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02912
| | - Alexander I Salter
- Department of Molecular Microbiology & Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02912
| | - Leon E Toussaint
- Department of Molecular Microbiology & Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02912
| | - Emma C Reilly
- Department of Molecular Microbiology & Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02912
| | - Céline Fugère
- Department of Molecular Microbiology & Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02912
| | - Neetu Srivastava
- Departments of Pediatrics and Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - William G Kerr
- Departments of Pediatrics and Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210.,Chemistry Department, Syracuse University, Syracuse, NY 13210
| | - Laurent Brossay
- Department of Molecular Microbiology & Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02912
| |
Collapse
|
127
|
Gottschalk C, Mettke E, Kurts C. The Role of Invariant Natural Killer T Cells in Dendritic Cell Licensing, Cross-Priming, and Memory CD8(+) T Cell Generation. Front Immunol 2015; 6:379. [PMID: 26284065 PMCID: PMC4517377 DOI: 10.3389/fimmu.2015.00379] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/11/2015] [Indexed: 12/23/2022] Open
Abstract
New vaccination strategies focus on achieving CD8+ T cell (CTL) immunity rather than on induction of protective antibody responses. While the requirement of CD4+ T (Th) cell help in dendritic cell (DC) activation and licensing, and in CTL memory induction has been described in several disease models, CTL responses may occur in a Th cell help-independent manner. Invariant natural killer T cells (iNKT cells) can substitute for Th cell help and license DC as well. iNKT cells produce a broad spectrum of Th1 and Th2 cytokines, thereby inducing a similar set of costimulatory molecules and cytokines in DC. This form of licensing differs from Th cell help by inducing other chemokines, while Th cell-licensed DCs produce CCR5 ligands, iNKT cell-licensed DCs produce CCL17, which attracts CCR4+ CD8+ T cells for subsequent activation. It has recently been shown that iNKT cells do not only enhance immune responses against bacterial pathogens or parasites but also play a role in viral infections. The inclusion of iNKT cell ligands in influenza virus vaccines enhanced memory CTL generation and protective immunity in a mouse model. This review will focus on the role of iNKT cells in the cross-talk with cross-priming DC and memory CD8+ T cell formation.
Collapse
Affiliation(s)
- Catherine Gottschalk
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University of Bonn , Bonn , Germany
| | - Elisabeth Mettke
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University of Bonn , Bonn , Germany
| | - Christian Kurts
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University of Bonn , Bonn , Germany
| |
Collapse
|
128
|
Opasawatchai A, Matangkasombut P. iNKT Cells and Their Potential Lipid Ligands during Viral Infection. Front Immunol 2015; 6:378. [PMID: 26257744 PMCID: PMC4513233 DOI: 10.3389/fimmu.2015.00378] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 07/11/2015] [Indexed: 01/12/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a unique population of lipid-reactive CD1d-restricted innate-like T lymphocytes. Despite being a minor population, they serve as an early source of cytokines and promote immunological crosstalk thus bridging innate and adaptive immunity. Diseases ranging from allergy, autoimmunity, and cancer, as well as infectious diseases, including viral infection, have been reported to be influenced by iNKT cells. However, it remains unclear how iNKT cells are activated during viral infection, as virus-derived lipid antigens have not been reported. Cytokines may activate iNKT cells during infections from influenza and murine cytomegalovirus, although CD1d-dependent activation is evident in other viral infections. Several viruses, such as dengue virus, induce CD1d upregulation, which correlates with iNKT cell activation. In contrast, herpes simplex virus type 1 (HSV-1), human immunodeficiency virus (HIV), Epstein–Barr virus, and human papilloma virus promote CD1d downregulation as a strategy to evade iNKT cell recognition. These observations suggest the participation of a CD1d-dependent process in the activation of iNKT cells in response to viral infection. Endogenous lipid ligands, including phospholipids as well as glycosphingolipids, such as glucosylceramide, have been proposed to mediate iNKT cell activation. Pro-inflammatory signals produced during viral infection may stimulate iNKT cells through enhanced CD1d-dependent endogenous lipid presentation. Furthermore, viral infection may alter lipid composition and inhibit endogenous lipid degradation. Recent advances in this field are reviewed.
Collapse
Affiliation(s)
- Anunya Opasawatchai
- Department of Microbiology, Faculty of Science, Mahidol University , Bangkok , Thailand ; Faculty of Dentistry, Mahidol University , Bangkok , Thailand
| | - Ponpan Matangkasombut
- Department of Microbiology, Faculty of Science, Mahidol University , Bangkok , Thailand ; Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University , Bangkok , Thailand
| |
Collapse
|
129
|
Anderson RJ, Compton BJ, Tang CW, Authier-Hall A, Hayman CM, Swinerd GW, Kowalczyk R, Harris P, Brimble MA, Larsen DS, Gasser O, Weinkove R, Hermans IF, Painter GF. NKT cell-dependent glycolipid-peptide vaccines with potent anti-tumour activity. Chem Sci 2015; 6:5120-5127. [PMID: 28717498 PMCID: PMC5500832 DOI: 10.1039/c4sc03599b] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 06/25/2015] [Indexed: 12/22/2022] Open
Abstract
Glycolipid–peptide conjugates designed to release vaccine components within target cells ensuring potent CD1d dependent T cell responses.
It is known that T cells can eliminate tumour cells through recognition of unique or aberrantly expressed antigens presented as peptide epitopes by major histocompatibility complex (MHC) molecules on the tumour cell surface. With recent advances in defining tumour-associated antigens, it should now be possible to devise therapeutic vaccines that expand specific populations of anti-tumour T cells. However there remains a need to develop simpler efficacious synthetic vaccines that possess clinical utility. We present here the synthesis and analysis of vaccines based on conjugation of MHC-binding peptide epitopes to α-galactosylceramide, a glycolipid presented by the nonpolymorphic antigen-presenting molecule CD1d to provoke the stimulatory activity of type I natural killer T (NKT) cells. The chemical design incorporates an enzymatically cleavable linker that effects controlled release of the active components in vivo. Chemical and biological analysis of different linkages with different enzymatic targets enabled selection of a synthetic vaccine construct with potent therapeutic anti-tumour activity in mice, and marked in vitro activity in human blood.
Collapse
Affiliation(s)
- Regan J Anderson
- The Ferrier Research Institute , Victoria University of Wellington , PO Box 33436 , Lower Hutt 5046 , New Zealand .
| | - Benjamin J Compton
- The Ferrier Research Institute , Victoria University of Wellington , PO Box 33436 , Lower Hutt 5046 , New Zealand .
| | - Ching-Wen Tang
- Malaghan Institute of Medical Research , PO Box 7060 , Wellington 6242 , New Zealand .
| | - Astrid Authier-Hall
- Malaghan Institute of Medical Research , PO Box 7060 , Wellington 6242 , New Zealand .
| | - Colin M Hayman
- The Ferrier Research Institute , Victoria University of Wellington , PO Box 33436 , Lower Hutt 5046 , New Zealand .
| | - Gene W Swinerd
- Malaghan Institute of Medical Research , PO Box 7060 , Wellington 6242 , New Zealand . .,School of Biological Sciences , Victoria University of Wellington , PO Box 600 , Wellington 6140 , New Zealand
| | - Renata Kowalczyk
- School of Biological Sciences , The University of Auckland , 3 Symonds St , Auckland Central , 1142 , New Zealand
| | - Paul Harris
- School of Biological Sciences , The University of Auckland , 3 Symonds St , Auckland Central , 1142 , New Zealand
| | - Margaret A Brimble
- School of Biological Sciences , The University of Auckland , 3 Symonds St , Auckland Central , 1142 , New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery , The University of Auckland , 3 Symonds St , Auckland Central , 1142 , New Zealand
| | - David S Larsen
- Department of Chemistry , University of Otago , PO Box 56 , Dunedin 9054 , New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research , PO Box 7060 , Wellington 6242 , New Zealand .
| | - Robert Weinkove
- Malaghan Institute of Medical Research , PO Box 7060 , Wellington 6242 , New Zealand . .,Department of Pathology & Molecular Medicine , University of Otago Wellington , New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research , PO Box 7060 , Wellington 6242 , New Zealand . .,Maurice Wilkins Centre for Molecular Biodiscovery , The University of Auckland , 3 Symonds St , Auckland Central , 1142 , New Zealand.,School of Biological Sciences , Victoria University of Wellington , PO Box 600 , Wellington 6140 , New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute , Victoria University of Wellington , PO Box 33436 , Lower Hutt 5046 , New Zealand .
| |
Collapse
|
130
|
Osmond TL, Farrand KJ, Painter GF, Ruedl C, Petersen TR, Hermans IF. Activated NKT Cells Can Condition Different Splenic Dendritic Cell Subsets To Respond More Effectively to TLR Engagement and Enhance Cross-Priming. THE JOURNAL OF IMMUNOLOGY 2015; 195:821-31. [PMID: 26078270 DOI: 10.4049/jimmunol.1401751] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 05/20/2015] [Indexed: 11/19/2022]
Abstract
The function of dendritic cells (DCs) can be modulated through multiple signals, including recognition of pathogen-associated molecular patterns, as well as signals provided by rapidly activated leukocytes in the local environment, such as innate-like T cells. In this article, we addressed the possibility that the roles of different murine DC subsets in cross-priming CD8(+) T cells can change with the nature and timing of activatory stimuli. We show that CD8α(+) DCs play a critical role in cross-priming CD8(+) T cell responses to circulating proteins that enter the spleen in close temporal association with ligands for TLRs and/or compounds that activate NKT cells. However, if NKT cells are activated first, then CD8α(-) DCs become conditioned to respond more vigorously to TLR ligation, and if triggered directly, these cells can also contribute to priming of CD8(+) T cell responses. In fact, the initial activation of NKT cells can condition multiple DC subsets to respond more effectively to TLR ligation, with plasmacytoid DCs making more IFN-α and both CD8α(+) and CD8α(-) DCs manufacturing more IL-12. These results suggest that different DC subsets can contribute to T cell priming if provided appropriately phased activatory stimuli, an observation that could be factored into the design of more effective vaccines.
Collapse
Affiliation(s)
- Taryn L Osmond
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Kathryn J Farrand
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand; and
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Troels R Petersen
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand;
| |
Collapse
|
131
|
Zhu H, Zhang Y, Liu H, Zhang Y, Kang Y, Mao R, Yang F, Zhou D, Zhang J. Preserved Function of Circulating Invariant Natural Killer T Cells in Patients With Chronic Hepatitis B Virus Infection. Medicine (Baltimore) 2015; 94:e961. [PMID: 26091463 PMCID: PMC4616535 DOI: 10.1097/md.0000000000000961] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
To date, the role of invariant natural killer T (iNKT) cells in chronic hepatitis B virus (HBV) infection is not fully understood. In previous reports, iNKT cells were identified by indirect methods. However, discrepancies regarding the prevalence and function of iNKT cells during HBV infection were observed. In this study, we have devised a direct, highly specific CD1d tetramer-based methodology to test whether patients with HBV infection have associated iNKT-cell defects. In our study, a total of 93 chronic HBV-infected patients and 30 healthy individuals (as control) were enrolled. The prevalence of iNKT cells, their cytokine producing capacity, and in vitro expansion were determined by flow cytometric analysis with CD1d tetramer staining. Our observation demonstrated that there was no significant difference in circulating CD1d-tetramer positive iNKT cell numbers between HBV-infected patients and healthy controls. The capacity of iNKT cells to produce IFN-γ or IL-4 as well as their in vitro expansion was also comparable between these 2 groups. However, among chronic HBV-infected patients, a decrease in iNKT cell-number was observed in chronic hepatitis B (CHB) and cirrhosis patients in comparison to that in immune tolerant (IT) patients. These results indicated that patients with chronic HBV infection may have normal prevalence and preserved function of circulating iNKT cells. And antiviral therapy with nucleot(s)ide analogue does not alter the frequency and function of circulating iNKT cells in chronic Hepatitis B patients.
Collapse
Affiliation(s)
- Haoxiang Zhu
- From the Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China (HZ, YZ, HL, YZ, YK, RM, FY, JZ); Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA (DZ); and Key laboratory of Medical Molecular Virology of the Ministries of Education and Health (MOH&MOE), Shanghai Medical College, Fudan University, Shanghai, China (JZ)
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Abstract
The immune system can be divided into innate and adaptive components that differ in their rate and mode of cellular activation, with innate immune cells being the first responders to invading pathogens. Recent advances in the identification and characterization of innate lymphoid cells have revealed reiterative developmental programs that result in cells with effector fates that parallel those of adaptive lymphoid cells and are tailored to effectively eliminate a broad spectrum of pathogenic challenges. However, activation of these cells can also be associated with pathologies such as autoimmune disease. One major distinction between innate and adaptive immune system cells is the constitutive expression of ID proteins in the former and inducible expression in the latter. ID proteins function as antagonists of the E protein transcription factors that play critical roles in lymphoid specification as well as B- and T-lymphocyte development. In this review, we examine the transcriptional mechanisms controlling the development of innate lymphocytes, including natural killer cells and the recently identified innate lymphoid cells (ILC1, ILC2, and ILC3), and innate-like lymphocytes, including natural killer T cells, with an emphasis on the known requirements for the ID proteins.
Collapse
Affiliation(s)
- Mihalis Verykokakis
- Committee on Immunology and Department of Pathology, The University of Chicago, Chicago, IL, USA
| | | | | |
Collapse
|
133
|
Abstract
Invariant natural killer T (iNKT) cells are a unique population of T lymphocytes, which lie at the interface between the innate and adaptive immune systems, and are important mediators of immune responses and tumor surveillance. iNKT cells recognize lipid antigens in a CD1d-dependent manner; their subsequent activation results in a rapid and specific downstream response, which enhances both innate and adaptive immunity. The capacity of iNKT cells to modify the immune microenvironment influences the ability of the host to control tumor growth, making them an important population to be harnessed in the clinic for the development of anticancer therapeutics. Indeed, the identification of strong iNKT-cell agonists, such as α-galactosylceramide (α-GalCer) and its analogues, has led to the development of synthetic lipids that have shown potential in vaccination and treatment against cancers. In this Masters of Immunology article, we discuss these latest findings and summarize the major discoveries in iNKT-cell biology, which have enabled the design of potent strategies for immune-mediated tumor destruction.
Collapse
Affiliation(s)
- Rosanna M McEwen-Smith
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, United Kingdom
| | - Mariolina Salio
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, United Kingdom
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, United Kingdom.
| |
Collapse
|
134
|
Nie H, Yang Q, Zhang G, Wang A, He Q, Liu M, Li P, Yang J, Huang Y, Ding X, Yu H, Hu S. Invariant NKT cells act as an adjuvant to enhance Th2 inflammatory response in an OVA-induced mouse model of asthma. PLoS One 2015; 10:e0119901. [PMID: 25830340 PMCID: PMC4382159 DOI: 10.1371/journal.pone.0119901] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 01/06/2015] [Indexed: 11/21/2022] Open
Abstract
Background Invariant natural killer T cells (iNKT cells) are a unique subset of T lymphocytes and are considered to play an important role in the development of allergic bronchial asthma. Recently, iNKT cells were shown to play an immunoregulatory role in CD4+ and CD8+ T cell-mediated adaptive immune response. Allergen-specific Th2 inflammatory responses are an important part of the adaptive immune response in asthma. However, the regulatory functions of the Th2 inflammatory response in asthma have not been studied in detail. Method In this study, we have investigated the regulatory functions of iNKT cells on the Th2 inflammatory response in an ovalbumin (OVA)-induced murine model of asthma. Results Our results demonstrate that α-Galactosylceramide (α-GalCer) administration activated iNKT cells but could not induce the Th2 inflammatory response in wild-type (WT) mice. In the OVA-induced asthma model, α-GalCer administration and adoptive transfer of iNKT cells significantly augmented the Th2 inflammatory responses, including elevated inflammatory cell infiltration in the lung and bronchoalveolar lavage fluid (BALF); increased levels of IL-4, IL-5, and IL-13 in the BALF and splenocyte culture supernatant; and increased serum levels of OVA-specific IgE and IgG1. In addition, the Th2 inflammatory response was reduced, but not completely abrogated in CD1d-/- mice immunized and challenged with OVA, compared with WT mice. Conclusion These results suggest that iNKT cells may serve as an adjuvant to enhance Th2 inflammatory response in an OVA-induced murine model of asthma.
Collapse
Affiliation(s)
- Hanxiang Nie
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- * E-mail:
| | - Qiaoyu Yang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guqin Zhang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ailing Wang
- Wuhan University HOPE School of nursing, Wuhan, China
| | - Qing He
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Min Liu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ping Li
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiong Yang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Huang
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xuhong Ding
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongying Yu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Suping Hu
- Department of Respiratory Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
135
|
DeLyria ES, Zhou D, Lee JS, Singh S, Song W, Li F, Sun Q, Lu H, Wu J, Qiao Q, Hu Y, Zhang G, Li C, Sastry KJ, Shen H. Sublingual injection of microparticles containing glycolipid ligands for NKT cells and subunit vaccines induces antibody responses in oral cavity. Carbohydr Res 2015; 405:87-92. [PMID: 25555750 DOI: 10.1016/j.carres.2014.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 10/10/2014] [Accepted: 11/14/2014] [Indexed: 12/13/2022]
Abstract
Natural Killer T (NKT) cells are a unique type of innate immune cells which exert paradoxical roles in animal models through producing either Th1 or Th2 cytokines and activating dendritic cells. Alpha-galactosylceramide (αGalCer), a synthetic antigen for NKT cells, was found to be safe and immune stimulatory in cancer and hepatitis patients. We recently developed microparticle-formulated αGalCer, which is selectively presented by dendritic cells and macrophages, but not B cells, and thus can avoid the anergy of NKT cells. In this study, we have examined the immunogenicity of microparticles containing αGalCer and protein vaccine components through sublingual injection in mice. The results showed that sublingual injection of microparticles containing αGalCer and ovalbumin triggered IgG responses in serum (titer >1:100,000), which persisted for more than 3months. Microparticles containing ovalbumin alone also induced comparable level of IgG responses. However, immunoglobulin subclass analysis showed that sublingually injected microparticles containing αGalCer and ovalbumin induced 20 fold higher Th1 biased antibody (IgG2c) than microparticles containing OVA alone (1:20,000 as compared to 1:1000 titer). Sublingual injection of microparticles containing αGalCer and ovalbumin induced secretion of both IgG (titer >1:1000) and IgA (titer=1:80) in saliva secretion, while microparticles containing ovalbumin alone only induced secretion of IgG in saliva. Our results suggest that sublingual injection of microparticles and their subsequent trafficking to draining lymph nodes may induce adaptive immune responses in mucosal compartments. Ongoing studies are focused on the mechanism of antigen presentation and lymphocyte biology in the oral cavity, as well as the toxicity and efficacy of these candidate microparticles for future applications.
Collapse
Affiliation(s)
- Elizabeth S DeLyria
- Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Dapeng Zhou
- Shanghai Pulmonary Hospital, Tongji University, School of Medicine, Shanghai, China.
| | - Jun Soo Lee
- Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Shailbala Singh
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Wei Song
- Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States; Shanghai Public Health Clinical Center, Shanghai, China
| | - Fenge Li
- Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Qing Sun
- Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Hongzhou Lu
- Shanghai Public Health Clinical Center, Shanghai, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Qian Qiao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Guodong Zhang
- Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, TX, United States
| | - Chun Li
- Department of Experimental Diagnostic Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - K Jagannadha Sastry
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Haifa Shen
- Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, TX, United States
| |
Collapse
|
136
|
Chen JL, Dawoodji A, Tarlton A, Gnjatic S, Tajar A, Karydis I, Browning J, Pratap S, Verfaille C, Venhaus RR, Pan L, Altman DG, Cebon JS, Old LL, Nathan P, Ottensmeier C, Middleton M, Cerundolo V. NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int J Cancer 2015; 136:E590-601. [PMID: 25081390 DOI: 10.1002/ijc.29118] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/19/2014] [Accepted: 07/14/2014] [Indexed: 12/14/2022]
Abstract
Vaccination strategies based on repeated injections of NY-ESO-1 protein formulated in ISCOMATRIX particles (NY-ESO-1 ISCOMATRIX) have shown to elicit combined NY-ESO-1 specific antibody and T cell responses. However, it remains unclear whether heterologous prime-boost strategies based on the combination with NY-ESO-1 ISCOMATRIX with different NY-ESO-1 boosting reagents could be used to increase NY-ESO-1 CD8(+) or CD4(+) T cell responses. To address this question, we carried out a randomized clinical trial in 39 high-risk, resected melanoma patients vaccinated with NY-ESO-1 ISCOMATRIX, and then boosted with repeated injections of either recombinant fowlpox virus encoding full length NY-ESO-1 (rF-NY-ESO-1) (Arm A) or NY-ESO-1 ISCOMATRIX alone (Arm B). We have comprehensively analyzed NY-ESO-1 specific T cells and B cells response in all patients before and after vaccination for a total of seven time points per patient. NY-ESO-1 ISCOMATRIX alone elicited a strong NY-ESO-1 specific CD4(+) T cell and antibody response, which was maintained by both regiments at similar levels. However, CD8(+) T cell responses were significantly boosted in 3 out of 18 patients in Arm A after the first rF-NY-ESO-1 injection and such responses were maintained until the end of the trial, while no patients in Arm B showed similar CD8(+) T cell responses. In addition, our results clearly identified immunodominant regions in the NY-ESO-1 protein: NY-ESO-179-102 and NY-ESO-1115-138 for CD4+ T cells and NY-ESO-185-108 for CD8+ T cells in a large proportion of vaccinated patients. These regions of NY-ESO-1 protein should be considered in future clinical trials as immunodominant epitopes.
Collapse
Affiliation(s)
- Ji-Li Chen
- Radcliffe Department of Medicine, MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Juno JA, Stalker AT, Waruk JL, Oyugi J, Kimani M, Plummer FA, Kimani J, Fowke KR. Elevated expression of LAG-3, but not PD-1, is associated with impaired iNKT cytokine production during chronic HIV-1 infection and treatment. Retrovirology 2015; 12:17. [PMID: 25810006 PMCID: PMC4332911 DOI: 10.1186/s12977-015-0142-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/16/2015] [Indexed: 01/08/2023] Open
Abstract
Background LAG-3 is a potent negative regulator of the immune response but its impact in HIV infection in poorly understood. Unlike exhaustion markers such as PD-1, Tim-3, 2B4 and CD160, LAG-3 is poorly expressed on bulk and antigen-specific T cells during chronic HIV infection and its expression on innate lymphocyte subsets is not well understood. The aim of this study was to assess LAG-3 expression and association with cellular dysfunction on T cells, NK cells and iNKT cells among a cohort of healthy and HIV-infected female sex workers in Nairobi, Kenya. Results Ex vivo LAG-3 expression was measured by multiparametric flow cytometry, and plasma cytokine/chemokine concentrations measured by bead array. Although LAG-3 expression on bulk T cells was significantly increased among HIV-infected women, the proportion of cells expressing the marker was extremely low. In contrast, LAG-3 was more highly expressed on NK and iNKT cells and was not reduced among women treated with ART. To assess the functional impact of LAG-3 on iNKT cells, iNKT cytokine production was measured in response to lipid (αGalCer) and PMA/Io stimulation by both flow cytometry and cytokine bead array. iNKT cytokine production is profoundly altered by both HIV infection and treatment, and LAG-3, but not PD-1, expression is associated with a reduction in iNKT IFNγ production. Conclusions LAG-3 does not appear to mediate T cell exhaustion in this African population, but is instead expressed on innate lymphocyte subsets including iNKT cells. HIV infection alters iNKT cytokine production patterns and LAG-3 expression is uniquely associated with iNKT dysfunction. The continued expression of LAG-3 during treatment suggests it may contribute to the lack of innate immune reconstitution commonly observed during ART. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0142-z) contains supplementary material, which is available to authorized users.
Collapse
|
138
|
Hunn MK, Hermans IF. Exploiting invariant NKT cells to promote T-cell responses to cancer vaccines. Oncoimmunology 2014; 2:e23789. [PMID: 23734325 PMCID: PMC3654595 DOI: 10.4161/onci.23789] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 01/25/2013] [Indexed: 11/19/2022] Open
Abstract
Invariant natural killer T (iNKT) cells have the capacity to amplify adaptive immune responses by licensing antigen-presenting cells. A simple vaccine consisting of whole tumor cells pulsed with an iNKT-cell agonist efficiently delivers antigens plus adjuvants to endogenous dendritic cells and has potential for clinical applications.
Collapse
Affiliation(s)
- Martin K Hunn
- Malaghan Institute of Medical Research; Wellington, New Zealand ; School of Biological Sciences; Victoria University of Wellington; Wellington, New Zealand
| | | |
Collapse
|
139
|
Fujii SI, Shimizu K. Immunotherapy with artificial adjuvant vector cells: Harnessing both arms of the immune response. Oncoimmunology 2014; 2:e23432. [PMID: 23734303 PMCID: PMC3654573 DOI: 10.4161/onci.23432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 12/28/2012] [Indexed: 12/15/2022] Open
Abstract
Both innate and adaptive immunity underpin cancer immunosurveillance. To stimulate both these arms of the immune system, we used allogeneic cells loaded with natural killer T (NKT) cell ligands and expressing tumor-associated antigens, resulting in NKT cell activation, dendritic-cell maturation and ultimately in the elicitation of adaptive T-cell responses. This approach holds great promise for the development of novel anticancer immunotherapies.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Research Unit for Cellular Immunotherapy; The Institute of Physical and Chemical Research (RIKEN); Research Center for Allergy and Immunology (RCAI); Yokohama, Japan
| | | |
Collapse
|
140
|
Singh M, Quispe-Tintaya W, Chandra D, Jahangir A, Venkataswamy MM, Ng TW, Sharma-Kharkwal S, Carreño LJ, Porcelli SA, Gravekamp C. Direct incorporation of the NKT-cell activator α-galactosylceramide into a recombinant Listeria monocytogenes improves breast cancer vaccine efficacy. Br J Cancer 2014; 111:1945-54. [PMID: 25314062 PMCID: PMC4229631 DOI: 10.1038/bjc.2014.486] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/24/2014] [Accepted: 08/07/2014] [Indexed: 12/28/2022] Open
Abstract
Background: Immune suppression in the tumour microenvironment remains a major limitation to successful immunotherapy of cancer. In the current study, we analysed whether the natural killer T cell-activating glycolipid α-galactosylceramide could overcome immune suppression and improve vaccination against metastatic breast cancer. Methods: Mice with metastatic breast cancer (4T1 model) were therapeutically treated with a Listeria monocytogenes-based vaccine expressing tumour-associated antigen Mage-b followed by α-galactosylceramide as separate agents, or as a complex of α-galactosylceramide stably incorporated into Listeria-Mage-b. Effects on metastases, tumour weight, toxicity and immune responses were determined. Results: Sequential treatments of mice with established 4T1 breast carcinomas using Listeria-Mage-b followed by α-galactosylceramide as a separate agent was highly effective at reducing metastases, but was accompanied by severe liver toxicity. In contrast, combined therapy using Listeria-Mage-b modified by incorporation of α-galactosylceramide resulted in nearly complete elimination of metastases without toxicity. This was associated with a significant increase in the percentage of natural killer T cells in the spleen, and an increase in natural killer cell activity and in T cell responses to Mage-b. Conclusions: Our results suggest that direct incorporation of α-galactosylceramide into a live bacterial vaccine vector is a promising non-toxic new approach for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- M Singh
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - W Quispe-Tintaya
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - D Chandra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - A Jahangir
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - M M Venkataswamy
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - T W Ng
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - S Sharma-Kharkwal
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - L J Carreño
- 1] Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA [2] Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Universidad de Chile, Avenue Independencia #1027, Santiago 8380453, Chile
| | - S A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - C Gravekamp
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
141
|
Anderson RJ, Tang CW, Daniels NJ, Compton BJ, Hayman CM, Johnston KA, Knight DA, Gasser O, Poyntz HC, Ferguson PM, Larsen DS, Ronchese F, Painter GF, Hermans IF. A self-adjuvanting vaccine induces cytotoxic T lymphocytes that suppress allergy. Nat Chem Biol 2014; 10:943-9. [DOI: 10.1038/nchembio.1640] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/04/2014] [Indexed: 01/12/2023]
|
142
|
Venkataswamy MM, Ng TW, Kharkwal SS, Carreño LJ, Johnson AJ, Kunnath-Velayudhan S, Liu Z, Bittman R, Jervis PJ, Cox LR, Besra GS, Wen X, Yuan W, Tsuji M, Li X, Ho DD, Chan J, Lee S, Frothingham R, Haynes BF, Panas MW, Gillard GO, Sixsmith JD, Korioth-Schmitz B, Schmitz JE, Larsen MH, Jacobs WR, Porcelli SA. Improving Mycobacterium bovis bacillus Calmette-Guèrin as a vaccine delivery vector for viral antigens by incorporation of glycolipid activators of NKT cells. PLoS One 2014; 9:e108383. [PMID: 25255287 PMCID: PMC4177913 DOI: 10.1371/journal.pone.0108383] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/20/2014] [Indexed: 01/13/2023] Open
Abstract
Recombinant Mycobacterium bovis bacillus Calmette-Guèrin (rBCG) has been explored as a vector for vaccines against HIV because of its ability to induce long lasting humoral and cell mediated immune responses. To maximize the potential for rBCG vaccines to induce effective immunity against HIV, various strategies are being employed to improve its ability to prime CD8+ T cells, which play an important role in the control of HIV infections. In this study we adopted a previously described approach of incorporating glycolipids that activate CD1d-restricted natural killer T (NKT) cells to enhance priming of CD8+ T cells by rBCG strains expressing an SIV Gag antigen (rBCG-SIV gag). We found that the incorporation of the synthetic NKT activating glycolipid α-galactosylceramide (α-GC) into rBCG-SIV gag significantly enhanced CD8+ T cell responses against an immunodominant Gag epitope, compared to responses primed by unmodified rBCG-SIV gag. The abilities of structural analogues of α-GC to enhance CD8+ T cell responses to rBCG were compared in both wild type and partially humanized mice that express human CD1d molecules in place of mouse CD1d. These studies identified an α-GC analogue known as 7DW8-5, which has previously been used successfully as an adjuvant in non-human primates, as a promising compound for enhancing immunogenicity of antigens delivered by rBCG.vectors. Our findings support the incorporation of synthetic glycolipid activators of NKT cells as a novel approach to enhance the immunogenicity of rBCG-vectored antigens for induction of CD8+ T cell responses. The glycolipid adjuvant 7DW8-5 may be a promising candidate for advancing to non-human primate and human clinical studies for the development of HIV vaccines based on rBCG vectors.
Collapse
Affiliation(s)
- Manjunatha M. Venkataswamy
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- National Institute of Mental Health and Neuroscience, Bangalore, Karnataka, India
| | - Tony W. Ng
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Shalu S. Kharkwal
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Leandro J. Carreño
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alison J. Johnson
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Shajo Kunnath-Velayudhan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Zheng Liu
- Department of Chemistry and Biochemistry, Queens College of City University of New York, Flushing, New York, United States of America
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of City University of New York, Flushing, New York, United States of America
| | - Peter J. Jervis
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Liam R. Cox
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Gurdyal S. Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Xiangshu Wen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, New York, United States of America
| | - Xiangming Li
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, New York, United States of America
| | - David D. Ho
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, New York, United States of America
| | - John Chan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Sunhee Lee
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Richard Frothingham
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Barton F. Haynes
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael W. Panas
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Geoffrey O. Gillard
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jaimie D. Sixsmith
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Birgit Korioth-Schmitz
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joern E. Schmitz
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michelle H. Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - William R. Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Steven A. Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
143
|
An autologous leukemia cell vaccine prevents murine acute leukemia relapse after cytarabine treatment. Blood 2014; 124:2953-63. [PMID: 25237205 DOI: 10.1182/blood-2014-04-568956] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Acute leukemias with adverse prognostic features carry a high relapse rate without allogeneic stem cell transplantation (allo-SCT). Allo-SCT has a high morbidity and is precluded for many patients because of advanced age or comorbidities. Postremission therapies with reduced toxicities are urgently needed. The murine acute leukemia model C1498 was used to study the efficacy of an intravenously administered vaccine consisting of irradiated leukemia cells loaded with the natural killer T (NKT)-cell agonist α-galactosylceramide (α-GalCer). Prophylactically, the vaccine was highly effective at preventing leukemia development through the downstream activities of activated NKT cells, which were dependent on splenic langerin(+)CD8α(+) dendritic cells and which led to stimulation of antileukemia CD4(+) and CD8(+) T cells. However, hosts with established leukemia received no protective benefit from the vaccine, despite inducing NKT-cell activation. Established leukemia was associated with increases in regulatory T cells and myeloid-derived suppressor cells, and the leukemic cells themselves were highly suppressive in vitro. Although this suppressive environment impaired both effector arms of the immune response, CD4(+) T-cell responses were more severely affected. When cytarabine chemotherapy was administered prior to vaccination, all animals in remission posttherapy were protected against rechallenge with viable leukemia cells.
Collapse
|
144
|
Kim MK, Lee A, Hwang YK, Kang CY, Ha SJ. Enhancing T Cell Immune Responses by B Cell-based Therapeutic Vaccine Against Chronic Virus Infection. Immune Netw 2014; 14:207-18. [PMID: 25177253 PMCID: PMC4148491 DOI: 10.4110/in.2014.14.4.207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 07/28/2014] [Accepted: 08/05/2014] [Indexed: 12/01/2022] Open
Abstract
Chronic virus infection leads to the functional impairment of dendritic cells (DCs) as well as T cells, limiting the clinical usefulness of DC-based therapeutic vaccine against chronic virus infection. Meanwhile, B cells have been known to maintain the ability to differentiate plasma cells producing antibodies even during chronic virus infection. Previously, α-galactosylceramide (αGC) and cognate peptide-loaded B cells were comparable to DCs in priming peptide-specific CD8(+) T cells as antigen presenting cells (APCs). Here, we investigated whether B cells activated by αGC can improve virus-specific T cell immune responses instead of DCs during chronic virus infection. We found that comparable to B cells isolated from naïve mice, chronic B cells isolated from chronically infected mice with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13) after αGC-loading could activate CD1d-restricted invariant natural killer T (iNKT) cells to produce effector cytokines and upregulate co-stimulatory molecules in both naïve and chronically infected mice. Similar to naïve B cells, chronic B cells efficiently primed LCMV glycoprotein (GP) 33-41-specific P14 CD8(+) T cells in vivo, thereby allowing the proliferation of functional CD8(+) T cells. Importantly, when αGC and cognate epitope-loaded chronic B cells were transferred into chronically infected mice, the mice showed a significant increase in the population of epitope-specific CD8(+) T cells and the accelerated control of viremia. Therefore, our studies demonstrate that reciprocal activation between αGC-loaded chronic B cells and iNKT cells can strengthen virus-specific T cell immune responses, providing an effective regimen of autologous B cell-based therapeutic vaccine to treat chronic virus infection.
Collapse
Affiliation(s)
- Min Ki Kim
- System Immunology Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Ara Lee
- System Immunology Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Yu Kyeong Hwang
- Cell Therapy Team, Mogam Biotechnology Institute, Yongin 446-799, Korea
| | - Chang-Yuil Kang
- College of Pharmacy, Seoul National University, Seoul 110-799, Korea
| | - Sang-Jun Ha
- System Immunology Laboratory, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
145
|
Berzins SP, Ritchie DS. Natural killer T cells: drivers or passengers in preventing human disease? Nat Rev Immunol 2014; 14:640-6. [PMID: 25103356 DOI: 10.1038/nri3725] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Natural killer T (NKT) cells are credited with regulatory roles in immunity against cancers, autoimmune diseases, allergies, and bacterial and viral infections. Studies in mice and observational research in patient groups have suggested that NKT cell-based therapies could be used to prevent or treat these diseases, yet the translation into clinical settings has been disappointing. We support the view that NKT cells have regulatory characteristics that could be exploited in clinical settings, but there are doubts about the natural roles of NKT cells in vivo and whether NKT cell defects are fundamental drivers of disease in humans. In this Opinion article, we discuss the uncertainties and opportunities regarding NKT cells in humans, and the potential for NKT cells to be manipulated to prevent or treat disease.
Collapse
Affiliation(s)
- Stuart P Berzins
- School of Health Sciences, Federation University, Ballarat, Victoria 3350, Australia, the Fiona Elsey Cancer Research Institute, Ballarat, Victoria 3350, Australia, and the Department of Microbiology and Immunology, the Peter Doherty Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - David S Ritchie
- Department of Clinical Hematology and Bone Marrow Transplant Service, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
146
|
Payne KK, Bear HD, Manjili MH. Adoptive cellular therapy of cancer: exploring innate and adaptive cellular crosstalk to improve anti-tumor efficacy. Future Oncol 2014; 10:1779-94. [DOI: 10.2217/fon.14.97] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT The mammalian immune system has evolved to produce multi-tiered responses consisting of both innate and adaptive immune cells collaborating to elicit a functional response to a pathogen or neoplasm. Immune cells possess a shared ancestry, suggestive of a degree of coevolution that has resulted in optimal functionality as an orchestrated and highly collaborative unit. Therefore, the development of therapeutic modalities that harness the immune system should consider the crosstalk between cells of the innate and adaptive immune systems in order to elicit the most effective response. In this review, the authors will discuss the success achieved using adoptive cellular therapy in the treatment of cancer, recent trends that focus on purified T cells, T cells with genetically modified T-cell receptors and T cells modified to express chimeric antigen receptors, as well as the use of unfractionated immune cell reprogramming to achieve optimal cellular crosstalk upon infusion for adoptive cellular therapy.
Collapse
Affiliation(s)
- Kyle K Payne
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Harry D Bear
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
- Department of Surgery, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| |
Collapse
|
147
|
De Libero G, Singhal A, Lepore M, Mori L. Nonclassical T cells and their antigens in tuberculosis. Cold Spring Harb Perspect Med 2014; 4:a018473. [PMID: 25059739 DOI: 10.1101/cshperspect.a018473] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
T cells that recognize nonpeptidic antigens, and thereby are identified as nonclassical, represent important yet poorly characterized effectors of the immune response. They are present in large numbers in circulating blood and tissues and are as abundant as T cells recognizing peptide antigens. Nonclassical T cells exert multiple functions including immunoregulation, tumor control, and protection against infections. They recognize complexes of nonpeptidic antigens such as lipid and glycolipid molecules, vitamin B2 precursors, and phosphorylated metabolites of the mevalonate pathway. Each of these antigens is presented by antigen-presenting molecules other than major histocompatibility complex (MHC), including CD1, MHC class I-related molecule 1 (MR1), and butyrophilin 3A1 (BTN3A1) molecules. Here, we discuss how nonclassical T cells participate in the recognition of mycobacterial antigens and in the mycobacterial-specific immune response.
Collapse
Affiliation(s)
- Gennaro De Libero
- SIgN (Singapore Immunology Network), A*STAR (Agency for Science, Technology and Research), 138648 Singapore Experimental Immunology, Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Amit Singhal
- SIgN (Singapore Immunology Network), A*STAR (Agency for Science, Technology and Research), 138648 Singapore
| | - Marco Lepore
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Lucia Mori
- SIgN (Singapore Immunology Network), A*STAR (Agency for Science, Technology and Research), 138648 Singapore Experimental Immunology, Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| |
Collapse
|
148
|
Shekhar S, Joyee AG, Yang X. Invariant natural killer T cells: boon or bane in immunity to intracellular bacterial infections? J Innate Immun 2014; 6:575-84. [PMID: 24903638 DOI: 10.1159/000361048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/04/2014] [Indexed: 11/19/2022] Open
Abstract
Invariant natural killer T (iNKT) cells represent a specialized subset of innate lymphocytes that recognize lipid and glycolipid antigens presented to them by nonclassical MHC-I CD1d molecules and are able to rapidly secrete copious amounts of a variety of cytokines. iNKT cells possess the ability to modulate innate as well as adaptive immune responses against various pathogens. Intracellular bacteria are one of the most clinically significant human pathogens that effectively evade the immune system and cause a myriad of diseases of public health concern globally. Emerging evidence suggests that iNKT cells can confer immunity to intracellular bacteria but also inflict pathology in certain cases. We summarize the current knowledge on the contribution of iNKT cells in the host defense against intracellular bacterial infections, with a focus on the underlying mechanisms by which these cells induce protective or pathogenic reactions including the pathways of direct action (acting on infected cells) and indirect action (modulating dendritic, NK and T cells). The rational exploitation of iNKT cells for prophylactic and therapeutic purposes awaits a profound understanding of their functional biology.
Collapse
Affiliation(s)
- Sudhanshu Shekhar
- Laboratory for Infection and Immunity, Department of Medical Microbiology, University of Manitoba, Winnipeg, Man., Canada
| | | | | |
Collapse
|
149
|
Dellabona P, Abrignani S, Casorati G. iNKT-cell help to B cells: a cooperative job between innate and adaptive immune responses. Eur J Immunol 2014; 44:2230-7. [PMID: 24782127 DOI: 10.1002/eji.201344399] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/21/2014] [Accepted: 04/25/2014] [Indexed: 11/11/2022]
Abstract
T-cell help to B lymphocytes is one of the most important events in adaptive immune responses in health and disease. It is generally delivered by cognate CD4(+) T follicular helper (T(FH)) cells via both cell-to-cell contacts and soluble mediators, and it is essential for both the clonal expansion of antibody (Ab)-secreting B cells and memory B-cell formation. CD1d-restricted invariant natural killer T (iNKT) cells are a subset of innate-like T lymphocytes that rapidly respond to stimulation with specific lipid antigens (Ags) that are derived from infectious pathogens or stressed host cells. Activated iNKT cells produce a wide range of cytokines and upregulate costimulatory molecules that can promote activation of dendritic cells (DCs), natural killer (NK) cells, and T cells. A decade ago, we discovered that iNKT cells can help B cells to proliferate and to produce IgG Abs in vitro and in vivo. This adjuvant-like function of Ag-activated iNKT cells provides a flexible set of helper mechanisms that expand the current paradigm of T-cell-B-cell interaction and highlights the potential of iNKT-cell targeting vaccine formulations.
Collapse
Affiliation(s)
- Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milano, Italy
| | | | | |
Collapse
|
150
|
De Libero G, Mori L. The T-Cell Response to Lipid Antigens of Mycobacterium tuberculosis. Front Immunol 2014; 5:219. [PMID: 24904574 PMCID: PMC4033098 DOI: 10.3389/fimmu.2014.00219] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 04/30/2014] [Indexed: 11/13/2022] Open
Abstract
T-cells recognize lipid antigens presented by dedicated antigen-presenting molecules that belong to the CD1 family. This review discusses the structural properties of CD1 molecules, the nature of mycobacterial lipid antigens, and the phenotypic and functional properties of T-cells recognizing mycobacterial lipids. In humans, the five CD1 genes encode structurally similar glycoproteins that recycle in and thus survey different cellular endosomal compartments. The structure of the CD1-lipid-binding pockets, their mode of intracellular recycling and the type of CD1-expressing antigen-presenting cells all contribute to diversify lipid immunogenicity and presentation to T-cells. Mycobacteria produce a large variety of lipids, which form stable complexes with CD1 molecules and stimulate specific T-cells. The structures of antigenic lipids may be greatly different from each other and each lipid may induce unique T-cells capable of discriminating small lipid structural changes. The important functions of some lipid antigens within mycobacterial cells prevent the generation of negative mutants capable of escaping this type of immune response. T-cells specific for lipid antigens are stimulated in tuberculosis and exert protective functions. The mechanisms of antigen recognition, the type of effector functions and the mode of lipid-specific T-cell priming are discussed, emphasizing recent evidence of the roles of lipid-specific T-cells in tuberculosis.
Collapse
Affiliation(s)
- Gennaro De Libero
- Singapore Immunology Network, Agency for Science, Technology and Research (ASTAR) , Singapore , Singapore ; Experimental Immunology, Department of Biomedicine, University Hospital Basel , Basel , Switzerland
| | - Lucia Mori
- Singapore Immunology Network, Agency for Science, Technology and Research (ASTAR) , Singapore , Singapore
| |
Collapse
|