251
|
Adjuvants: Classification, Modus Operandi, and Licensing. J Immunol Res 2016; 2016:1459394. [PMID: 27274998 PMCID: PMC4870346 DOI: 10.1155/2016/1459394] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/02/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
Vaccination is one of the most efficient strategies for the prevention of infectious diseases. Although safer, subunit vaccines are poorly immunogenic and for this reason the use of adjuvants is strongly recommended. Since their discovery in the beginning of the 20th century, adjuvants have been used to improve immune responses that ultimately lead to protection against disease. The choice of the adjuvant is of utmost importance as it can stimulate protective immunity. Their mechanisms of action have now been revealed. Our increasing understanding of the immune system, and of correlates of protection, is helping in the development of new vaccine formulations for global infections. Nevertheless, few adjuvants are licensed for human vaccines and several formulations are now being evaluated in clinical trials. In this review, we briefly describe the most well known adjuvants used in experimental and clinical settings based on their main mechanisms of action and also highlight the requirements for licensing new vaccine formulations.
Collapse
|
252
|
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
|
253
|
Loi M, Müller A, Steinbach K, Niven J, Barreira da Silva R, Paul P, Ligeon LA, Caruso A, Albrecht RA, Becker AC, Annaheim N, Nowag H, Dengjel J, García-Sastre A, Merkler D, Münz C, Gannagé M. Macroautophagy Proteins Control MHC Class I Levels on Dendritic Cells and Shape Anti-viral CD8(+) T Cell Responses. Cell Rep 2016; 15:1076-1087. [PMID: 27117419 DOI: 10.1016/j.celrep.2016.04.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/10/2016] [Accepted: 03/27/2016] [Indexed: 01/08/2023] Open
Abstract
The macroautophagy machinery has been implicated in MHC class II restricted antigen presentation. Here, we report that this machinery assists in the internalization of MHC class I molecules. In the absence of the autophagy factors Atg5 and Atg7, MHC class I surface levels are elevated due to decreased endocytosis and degradation. Internalization of MHC class I molecules occurs less efficiently if AAK1 cannot be recruited via Atg8/LC3B. In the absence of Atg-dependent MHC class I internalization, dendritic cells stimulate CD8(+) T cell responses more efficiently in vitro and in vivo. During viral infections, lack of Atg5 results in enhanced influenza- and LCMV-specific CD8(+) T cell responses in vivo. Elevated influenza-specific CD8(+) T cell responses are associated with better immune control of this infection. Thus, the macroautophagy machinery orchestrates T cell immunity by supporting MHC class II but compromises MHC class I restricted antigen presentation.
Collapse
Affiliation(s)
- Monica Loi
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Anne Müller
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Karin Steinbach
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jennifer Niven
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva, Switzerland; Division of Rheumatology, Department of Internal Medicine, University Hospital, Geneva 1205, Switzerland
| | - Rosa Barreira da Silva
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Petra Paul
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Laure-Anne Ligeon
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Assunta Caruso
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva, Switzerland; Division of Rheumatology, Department of Internal Medicine, University Hospital, Geneva 1205, Switzerland
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrea C Becker
- Department of Dermatology, Medical Center; ZBSA Center for Biological Systems Analysis; BIOSS Centre for Biological Signalling Studies; and FRIAS Freiburg Institute for Advanced Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Nicolas Annaheim
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Heike Nowag
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Jörn Dengjel
- Department of Dermatology, Medical Center; ZBSA Center for Biological Systems Analysis; BIOSS Centre for Biological Signalling Studies; and FRIAS Freiburg Institute for Advanced Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Doron Merkler
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva, Switzerland; Department of Neuropathology, University Medical Center, 37099 Göttingen, Germany
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland.
| | - Monique Gannagé
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland; Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva, Switzerland; Division of Rheumatology, Department of Internal Medicine, University Hospital, Geneva 1205, Switzerland.
| |
Collapse
|
254
|
Mitogen-activated Tasmanian devil blood mononuclear cells kill devil facial tumour disease cells. Immunol Cell Biol 2016; 94:673-9. [PMID: 27089941 DOI: 10.1038/icb.2016.38] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/25/2016] [Accepted: 02/26/2016] [Indexed: 12/11/2022]
Abstract
Devil facial tumour disease (DFTD) is a transmissible cancer that has brought the host species, the Tasmanian devil, to the brink of extinction. The cancer cells avoid allogeneic immune recognition by downregulating cell surface major histocompatibility complex (MHC) I expression. This should prevent CD8(+) T cell, but not natural killer (NK) cell, cytotoxicity. The reason why NK cells, normally reactive to MHC-negative cells, are not activated to kill DFTD cells has not been determined. The immune response of wild devils to DFTD, if it occurs, is uncharacterised. To investigate this, we tested 12 wild devils with DFTD, and found suggestive evidence of low levels of antibodies against DFTD cells in one devil. Eight of these devils were also analysed for cytotoxicity, however, none showed evidence for cytotoxicity against cultured DFTD cells. To establish whether mimicking activation of antitumour responses could induce cytotoxic activity against DFTD, Tasmanian devil peripheral blood mononuclear cells (PBMCs) were treated with either the mitogen Concanavalin A, the Toll-like receptor agonist polyinosinic:polycytidylic acid or recombinant Tasmanian devil IL-2. All induced the PBMC cells to kill cultured DFTD cells, suggesting that activation does not occur after encounter with DFTD cells in vivo, but can be induced. The identification of agents that activate cytotoxicity against DFTD target cells is critical for developing strategies to protect against DFTD. Such agents could function as adjuvants to induce functional immune responses capable of targeting DFTD cells and tumours in vivo.
Collapse
|
255
|
Dye JM, Warfield KL, Wells JB, Unfer RC, Shulenin S, Vu H, Nichols DK, Aman MJ, Bavari S. Virus-Like Particle Vaccination Protects Nonhuman Primates from Lethal Aerosol Exposure with Marburgvirus (VLP Vaccination Protects Macaques against Aerosol Challenges). Viruses 2016; 8:94. [PMID: 27070636 PMCID: PMC4848589 DOI: 10.3390/v8040094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 01/21/2023] Open
Abstract
Marburg virus (MARV) was the first filovirus to be identified following an outbreak of viral hemorrhagic fever disease in Marburg, Germany in 1967. Due to several factors inherent to filoviruses, they are considered a potential bioweapon that could be disseminated via an aerosol route. Previous studies demonstrated that MARV virus-like particles (VLPs) containing the glycoprotein (GP), matrix protein VP40 and nucleoprotein (NP) generated using a baculovirus/insect cell expression system could protect macaques from subcutaneous (SQ) challenge with multiple species of marburgviruses. In the current study, the protective efficacy of the MARV VLPs in conjunction with two different adjuvants: QS-21, a saponin derivative, and poly I:C against homologous aerosol challenge was assessed in cynomolgus macaques. Antibody responses against the GP antigen were equivalent in all groups receiving MARV VLPs irrespective of the adjuvant; adjuvant only-vaccinated macaques did not demonstrate appreciable antibody responses. All macaques were subsequently challenged with lethal doses of MARV via aerosol or SQ as a positive control. All MARV VLP-vaccinated macaques survived either aerosol or SQ challenge while animals administered adjuvant only exhibited clinical signs and lesions consistent with MARV disease and were euthanized after meeting the predetermined criteria. Therefore, MARV VLPs induce IgG antibodies recognizing MARV GP and VP40 and protect cynomolgus macaques from an otherwise lethal aerosol exposure with MARV.
Collapse
Affiliation(s)
- John M Dye
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | | | - Jay B Wells
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Robert C Unfer
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Sergey Shulenin
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Hong Vu
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Donald K Nichols
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - M Javad Aman
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| |
Collapse
|
256
|
Sultan H, Fesenkova VI, Addis D, Fan AE, Kumai T, Wu J, Salazar AM, Celis E. Designing therapeutic cancer vaccines by mimicking viral infections. Cancer Immunol Immunother 2016; 66:203-213. [PMID: 27052572 DOI: 10.1007/s00262-016-1834-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/23/2016] [Indexed: 12/25/2022]
Abstract
The design of efficacious and cost-effective therapeutic vaccines against cancer remains both a research priority and a challenge. For more than a decade, our laboratory has been involved in the development of synthetic peptide-based anti-cancer therapeutic vaccines. We first dedicated our efforts in the identification and validation of peptide epitopes for both CD8 and CD4 T cells from tumor-associated antigens (TAAs). Because of suboptimal immune responses and lack of therapeutic benefit of peptide vaccines containing these epitopes, we have focused our recent efforts in optimizing peptide vaccinations in mouse tumor models using numerous TAA epitopes. In this focused research review, we describe how after taking lessons from the immune system's way of dealing with acute viral infections, we have designed peptide vaccination strategies capable of generating very high numbers of therapeutically effective CD8 T cells. We also discuss some of the remaining challenges to translate these findings into the clinical setting.
Collapse
Affiliation(s)
- Hussein Sultan
- Augusta University GRU Cancer Center, CN-4121, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
- Microbiology and Immunology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Valentyna I Fesenkova
- Augusta University GRU Cancer Center, CN-4121, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
| | - Diane Addis
- Augusta University GRU Cancer Center, CN-4121, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
| | - Aaron E Fan
- Augusta University GRU Cancer Center, CN-4121, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
| | - Takumi Kumai
- Augusta University GRU Cancer Center, CN-4121, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Juan Wu
- Augusta University GRU Cancer Center, CN-4121, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
| | | | - Esteban Celis
- Augusta University GRU Cancer Center, CN-4121, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA.
- Departments of Medicine and Biochemistry, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
257
|
Zhou J, Jin JO, Du J, Yu Q. Innate Immune Signaling Induces IL-7 Production, Early Inflammatory Responses, and Sjögren's-Like Dacryoadenitis in C57BL/6 Mice. Invest Ophthalmol Vis Sci 2016; 56:7831-8. [PMID: 26658504 DOI: 10.1167/iovs.15-17368] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Innate immune signaling elicited by polyinosinic-polycytidylic acid (poly I:C) induces IL-7 production and early inflammatory responses in the salivary gland and accelerates the development of Sjögren's syndrome (SS)-like sialadenitis. Whether poly I:C can induce similar responses in the lacrimal gland (LAC) has not been characterized. In this study, we examined the early responses and pathologic changes of the LAC tissue in response to poly I:C treatment. METHODS Poly I:C or recombinant human IL-7 was injected intraperitoneally into C57BL/6 mice, and the LAC was harvested at different time points. Expression of chemokines and cytokines in the LAC was measured by RT-PCR, immunofluorescence staining, and immunohistochemistry. Leukocytic infiltration and caspase-3 activation were analyzed by hematoxylin and eosin staining and immunohistochemistry. Serum antinuclear antibody levels were also determined. Tear secretion was measured by phenol red cotton threads. RESULTS Administration of poly I:C induced IL-7 gene expression and protein production in the LAC. Poly I:C also induced the expression of CXCR3 ligands, monocyte chemoattractant protein-1, IL-23p19, and TNF-α in the LAC in an IL-7-dependent fashion. Similarly to poly I:C, administration of exogenous IL-7 also up-regulated these proinflammatory mediators. Furthermore, repeated administration of poly I:C to C57BL/6 mice over an 8-day period caused leukocytic infiltration and caspase-3 activation in the LAC, antinuclear antibody production, and impaired tear secretion. CONCLUSIONS Poly I:C induces IL-7 production, early inflammatory responses, and characteristic pathologies of SS-like dacryoadenitis in non-autoimmune-prone C57BL/6 mice. These findings provide new evidence that viral infection-elicited innate immune signaling may be one of the early triggers of SS-like dacryoadenitis.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts, United States 2Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, United States
| | - Jun-O Jin
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts, United States
| | - Juan Du
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts, United States
| | - Qing Yu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts, United States 2Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, United States
| |
Collapse
|
258
|
Direct Delivery of Antigens to Dendritic Cells via Antibodies Specific for Endocytic Receptors as a Promising Strategy for Future Therapies. Vaccines (Basel) 2016; 4:vaccines4020008. [PMID: 27043640 PMCID: PMC4931625 DOI: 10.3390/vaccines4020008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/15/2016] [Accepted: 03/18/2016] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are the most potent professional antigen presenting cells and are therefore indispensable for the control of immunity. The technique of antibody mediated antigen targeting to DC subsets has been the basis of intense research for more than a decade. Many murine studies have utilized this approach of antigen delivery to various kinds of endocytic receptors of DCs both in vitro and in vivo. Today, it is widely accepted that different DC subsets are important for the induction of select immune responses. Nevertheless, many questions still remain to be answered, such as the actual influence of the targeted receptor on the initiation of the immune response to the delivered antigen. Further efforts to better understand the induction of antigen-specific immune responses will support the transfer of this knowledge into novel treatment strategies for human diseases. In this review, we will discuss the state-of-the-art aspects of the basic principles of antibody mediated antigen targeting approaches. A table will also provide a broad overview of the latest studies using antigen targeting including addressed DC subset, targeted receptors, outcome, and applied coupling techniques.
Collapse
|
259
|
Tai N, Wong FS, Wen L. The role of the innate immune system in destruction of pancreatic beta cells in NOD mice and humans with type I diabetes. J Autoimmun 2016; 71:26-34. [PMID: 27021275 DOI: 10.1016/j.jaut.2016.03.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 03/12/2016] [Indexed: 02/08/2023]
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease characterized by T cell-mediated destruction of the insulin-producing pancreatic β cells. A combination of genetic and environmental factors eventually leads to the loss of functional β cell mass and hyperglycemia. Both innate and adaptive immunity are involved in the development of T1D. In this review, we have highlighted the most recent findings on the role of innate immunity, especially the pattern recognition receptors (PRRs), in disease development. In murine models and human studies, different PRRs, such as toll-like receptors (TLRs) and nucleotide-binding domain, leucine-rich repeat-containing (or Nod-like) receptors (NLRs), have different roles in the pathogenesis of T1D. These PRRs play a critical role in defending against infection by sensing specific ligands derived from exogenous microorganisms to induce innate immune responses and shape adaptive immunity. Animal studies have shown that TLR7, TLR9, MyD88 and NLPR3 play a disease-predisposing role in T1D, while controversial results have been found with other PRRs, such as TLR2, TLR3, TLR4, TLR5 and others. Human studies also shown that TLR2, TLR3 and TLR4 are expressed in either islet β cells or infiltrated immune cells, indicating the innate immunity plays a role in β cell autoimmunity. Furthermore, some human genetic studies showed a possible association of TLR3, TLR7, TLR8 or NLRP3 genes, at single nucleotide polymorphism (SNP) level, with human T1D. Increasing evidence suggest that the innate immunity modulates β cell autoimmunity. Thus, targeting pathways of innate immunity may provide novel therapeutic strategies to fight this disease.
Collapse
Affiliation(s)
- Ningwen Tai
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, USA
| | - F Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
260
|
Chan J, Babb R, David SC, McColl SR, Alsharifi M. Vaccine-Induced Antibody Responses Prevent the Induction of Interferon Type I Responses Upon a Homotypic Live Virus Challenge. Scand J Immunol 2016; 83:165-73. [DOI: 10.1111/sji.12410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/22/2015] [Indexed: 12/31/2022]
Affiliation(s)
- J. Chan
- Vaccine Research Group; Centre for Molecular Pathology; School of Biological Sciences; The University of Adelaide; Adelaide SA Australia
| | - R. Babb
- Vaccine Research Group; Centre for Molecular Pathology; School of Biological Sciences; The University of Adelaide; Adelaide SA Australia
| | - S. C. David
- Vaccine Research Group; Centre for Molecular Pathology; School of Biological Sciences; The University of Adelaide; Adelaide SA Australia
| | - S. R. McColl
- Vaccine Research Group; Centre for Molecular Pathology; School of Biological Sciences; The University of Adelaide; Adelaide SA Australia
| | - M. Alsharifi
- Vaccine Research Group; Centre for Molecular Pathology; School of Biological Sciences; The University of Adelaide; Adelaide SA Australia
| |
Collapse
|
261
|
The Vaccine Adjuvant Chitosan Promotes Cellular Immunity via DNA Sensor cGAS-STING-Dependent Induction of Type I Interferons. Immunity 2016; 44:597-608. [PMID: 26944200 DOI: 10.1016/j.immuni.2016.02.004] [Citation(s) in RCA: 444] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/03/2015] [Accepted: 12/03/2015] [Indexed: 01/17/2023]
Abstract
The cationic polysaccharide chitosan is an attractive candidate adjuvant capable of driving potent cell-mediated immunity, but the mechanism by which it acts is not clear. We show that chitosan promotes dendritic cell maturation by inducing type I interferons (IFNs) and enhances antigen-specific T helper 1 (Th1) responses in a type I IFN receptor-dependent manner. The induction of type I IFNs, IFN-stimulated genes and dendritic cell maturation by chitosan required the cytoplasmic DNA sensor cGAS and STING, implicating this pathway in dendritic cell activation. Additionally, this process was dependent on mitochondrial reactive oxygen species and the presence of cytoplasmic DNA. Chitosan-mediated enhancement of antigen specific Th1 and immunoglobulin G2c responses following vaccination was dependent on both cGAS and STING. These findings demonstrate that a cationic polymer can engage the STING-cGAS pathway to trigger innate and adaptive immune responses.
Collapse
|
262
|
Lutz MB. Induction of CD4(+) Regulatory and Polarized Effector/helper T Cells by Dendritic Cells. Immune Netw 2016; 16:13-25. [PMID: 26937228 PMCID: PMC4770096 DOI: 10.4110/in.2016.16.1.13] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/15/2016] [Accepted: 01/18/2016] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are considered to play major roles during the induction of T cell immune responses as well as the maintenance of T cell tolerance. Naive CD4(+) T cells have been shown to respond with high plasticity to signals inducing their polarization into effector/helper or regulatory T cells. Data obtained from in vitro generated bone-marrow (BM)-derived DCs as well as genetic mouse models revealed an important but not exclusive role of DCs in shaping CD4(+) T cell responses. Besides the specialization of some conventional DC subsets for the induction of polarized immunity, also the maturation stage, activation of specialized transcription factors and the cytokine production of DCs have major impact on CD4(+) T cells. Since in vitro generated BM-DCs show a high diversity to shape CD4(+) T cells and their high similarity to monocyte-derived DCs in vivo, this review reports data mainly on BM-DCs in this process and only touches the roles of transcription factors or of DC subsets, which have been discussed elsewhere. Here, recent findings on 1) the conversion of naive into anergic and further into Foxp3(-) regulatory T cells (Treg) by immature DCs, 2) the role of RelB in steady state migratory DCs (ssmDCs) for conversion of naive T cells into Foxp3(+) Treg, 3) the DC maturation signature for polarized Th2 cell induction and 4) the DC source of IL-12 for Th1 induction are discussed.
Collapse
Affiliation(s)
- Manfred B Lutz
- Institute of Virology and Immunobiology, University of Würzburg, 97078 Würzburg, Germany
| |
Collapse
|
263
|
Pathogen-Associated Molecular Patterns Induced Crosstalk between Dendritic Cells, T Helper Cells, and Natural Killer Helper Cells Can Improve Dendritic Cell Vaccination. Mediators Inflamm 2016; 2016:5740373. [PMID: 26980946 PMCID: PMC4766350 DOI: 10.1155/2016/5740373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/30/2015] [Indexed: 12/29/2022] Open
Abstract
A coordinated cellular interplay is of crucial importance in both host defense against pathogens and malignantly transformed cells. The various interactions of Dendritic Cells (DC), Natural Killer (NK) cells, and T helper (Th) cells can be influenced by a variety of pathogen-associated molecular patterns (PAMPs) and will lead to enhanced CD8+ effector T cell responses. Specific Pattern Recognition Receptor (PRR) triggering during maturation enables DC to enhance Th1 as well as NK helper cell responses. This effect is correlated with the amount of IL-12p70 released by DC. Activated NK cells are able to amplify the proinflammatory cytokine profile of DC via the release of IFN-γ. The knowledge on how PAMP recognition can modulate the DC is of importance for the design and definition of appropriate therapeutic cancer vaccines. In this review we will discuss the potential role of specific PAMP-matured DC in optimizing therapeutic DC-based vaccines, as some of these DC are efficiently activating Th1, NK cells, and cytotoxic T cells. Moreover, to optimize these vaccines, also the inhibitory effects of tumor-derived suppressive factors, for example, on the NK-DC crosstalk, should be taken into account. Finally, the suppressive role of the tumor microenvironment in vaccination efficacy and some proposals to overcome this by using combination therapies will be described.
Collapse
|
264
|
A Comparative Study of the T Cell Stimulatory and Polarizing Capacity of Human Primary Blood Dendritic Cell Subsets. Mediators Inflamm 2016; 2016:3605643. [PMID: 27057096 PMCID: PMC4761397 DOI: 10.1155/2016/3605643] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/21/2015] [Accepted: 01/04/2016] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) are central players of immune responses; they become activated upon infection or inflammation and migrate to lymph nodes, where they can initiate an antigen-specific immune response by activating naive T cells. Two major types of naturally occurring DCs circulate in peripheral blood, namely, myeloid and plasmacytoid DCs (pDCs). Myeloid DCs (mDCs) can be subdivided based on the expression of either CD1c or CD141. These human DC subsets differ in surface marker expression, Toll-like receptor (TLR) repertoire, and transcriptional profile, suggesting functional differences between them. Here, we directly compared the capacity of human blood mDCs and pDCs to activate and polarize CD4(+) T cells. CD141(+) mDCs show an overall more mature phenotype over CD1c(+) mDC and pDCs; they produce less IL-10 and more IL-12 than CD1c(+) mDCs. Despite these differences, all subsets can induce the production of IFN-γ in naive CD4(+) T cells. CD1c(+) and CD141(+) mDCs especially induce a strong T helper 1 profile. Importantly, naive CD4(+) T cells are not polarized towards regulatory T cells by any subset. These findings further establish all three human blood DCs-despite their differences-as promising candidates for immunostimulatory effectors in cancer immunotherapy.
Collapse
|
265
|
Gupta S, Termini JM, Issac B, Guirado E, Stone GW. Constitutively Active MAVS Inhibits HIV-1 Replication via Type I Interferon Secretion and Induction of HIV-1 Restriction Factors. PLoS One 2016; 11:e0148929. [PMID: 26849062 PMCID: PMC4743994 DOI: 10.1371/journal.pone.0148929] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/15/2015] [Indexed: 12/24/2022] Open
Abstract
Type I interferon is known to inhibit HIV-1 replication through the induction of interferon stimulated genes (ISG), including a number of HIV-1 restriction factors. To better understand interferon-mediated HIV-1 restriction, we constructed a constitutively active form of the RIG-I adapter protein MAVS. Constitutive MAVS was generated by fusion of full length MAVS to a truncated form of the Epstein Barr virus protein LMP1 (ΔLMP1). Supernatant from ΔLMP1-MAVS-transfected 293T cells contained high levels of type I interferons and inhibited HIV replication in both TZM-bl and primary human CD4+ T cells. Supernatant from ΔLMP1-MAVS-transfected 293T cells also inhibited replication of VSV-G pseudotyped single cycle SIV in TZM-bl cells, suggesting restriction was post-entry and common to both HIV and SIV. Gene array analysis of ΔLMP1-MAVS-transfected 293T cells and trans-activated CD4+ T cells showed significant upregulation of ISG, including previously characterized HIV restriction factors Viperin, Tetherin, MxB, and ISG56. Interferon blockade studies implicated interferon-beta in this response. In addition to direct viral inhibition, ΔLMP1-MAVS markedly enhanced secretion of IFN-β and IL-12p70 by dendritic cells and the activation and maturation of dendritic cells. Based on this immunostimulatory activity, an adenoviral vector (Ad5) expressing ΔLMP1-MAVS was tested as a molecular adjuvant in an HIV vaccine mouse model. Ad5-Gag antigen combined with Ad5-ΔLMP1-MAVS enhanced control of vaccinia-gag replication in a mouse challenge model, with 4/5 animals showing undetectable virus following challenge. Overall, ΔLMP1-MAVS is a promising reagent to inhibit HIV-1 replication in infected tissues and enhance vaccine-mediated immune responses, while avoiding toxicity associated with systemic type I interferon administration.
Collapse
Affiliation(s)
- Sachin Gupta
- Department of Microbiology and Immunology, Miami Center for AIDS Research and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - James M. Termini
- Department of Microbiology and Immunology, Miami Center for AIDS Research and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Biju Issac
- Division of Bioinformatics, Biostatistics and Bioinformatics Core, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Elizabeth Guirado
- Department of Microbiology and Immunology, Miami Center for AIDS Research and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Geoffrey W. Stone
- Department of Microbiology and Immunology, Miami Center for AIDS Research and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
266
|
Elayeb R, Tamagne M, Bierling P, Noizat-Pirenne F, Vingert B. Red blood cell alloimmunization is influenced by the delay between Toll-like receptor agonist injection and transfusion. Haematologica 2016; 101:209-18. [PMID: 26430173 PMCID: PMC4938341 DOI: 10.3324/haematol.2015.134171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/01/2015] [Indexed: 11/09/2022] Open
Abstract
Murine models of red blood cell transfusion show that inflammation associated with viruses or methylated DNA promotes red blood cell alloimmunization. In vaccination studies, the intensity of antigen-specific responses depends on the delay between antigen and adjuvant administration, with a short delay limiting immune responses. In mouse models of alloimmunization, the delay between the injection of Toll-like receptor agonists and transfusion is usually short. In this study, we hypothesized that the timing of Toll-like receptor 3 agonist administration affects red blood cell alloimmunization. Poly(I:C), a Toll-like receptor 3 agonist, was administered to B10BR mice at various time points before the transfusion of HEL-expressing red blood cells. For each time point, we measured the activation of splenic HEL-presenting dendritic cells, HEL-specific CD4(+) T cells and anti-HEL antibodies in serum. The phenotype of activated immune cells depended on the delay between transfusion and Toll-like receptor-dependent inflammation. The production of anti-HEL antibodies was highest when transfusion occurred 7 days after agonist injection. The proportion of HEL-presenting CD8α(+) dendritic cells producing interleukin-12 was highest in mice injected with poly(I:C) 3 days before transfusion. Although the number of early-induced HEL-specific CD4(+) T cells was similar between groups, a high proportion of these cells expressed CD134, CD40 and CD44 in mice injected with poly(I:C) 7 days before transfusion. This study clearly shows that the delay between transfusion and Toll-like receptor-induced inflammation influences the immune response to transfused red blood cells.
Collapse
Affiliation(s)
- Rahma Elayeb
- Établissement Français du Sang, Créteil, France Institut Mondor de Recherche Biomédicale, lnserm U955, Equipe 2, Créteil, France Laboratory of Excellence GR-Ex, Paris, France
| | - Marie Tamagne
- Établissement Français du Sang, Créteil, France Institut Mondor de Recherche Biomédicale, lnserm U955, Equipe 2, Créteil, France Laboratory of Excellence GR-Ex, Paris, France
| | - Philippe Bierling
- Établissement Français du Sang, Créteil, France Institut Mondor de Recherche Biomédicale, lnserm U955, Equipe 2, Créteil, France Université Paris Est, Faculté de Médecine, Créteil, France Laboratory of Excellence GR-Ex, Paris, France
| | - France Noizat-Pirenne
- Établissement Français du Sang, Créteil, France Institut Mondor de Recherche Biomédicale, lnserm U955, Equipe 2, Créteil, France Université Paris Est, Faculté de Médecine, Créteil, France Laboratory of Excellence GR-Ex, Paris, France
| | - Benoît Vingert
- Établissement Français du Sang, Créteil, France Institut Mondor de Recherche Biomédicale, lnserm U955, Equipe 2, Créteil, France Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
267
|
Ruiz J, Kanagavelu S, Flores C, Romero L, Riveron R, Shih DQ, Fukata M. Systemic Activation of TLR3-Dependent TRIF Signaling Confers Host Defense against Gram-Negative Bacteria in the Intestine. Front Cell Infect Microbiol 2016; 5:105. [PMID: 26793623 PMCID: PMC4710052 DOI: 10.3389/fcimb.2015.00105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/24/2015] [Indexed: 12/11/2022] Open
Abstract
Recognition of Gram-negative bacteria by toll-like receptor (TLR)4 induces MyD88 and TRIF mediated responses. We have shown that TRIF-dependent responses play an important role in intestinal defense against Gram-negative enteropathogens. In the current study, we examined underlying mechanisms of how systemic TRIF activation enhances intestinal immune defense against Gram-negative bacteria. First we confirmed that the protective effect of poly I:C against enteric infection of mice with Yersinia enterocolitica was dependent on TLR3-mediated TRIF signaling by using TLR3-deficient mice. This protection was unique in TRIF-dependent TLR signaling because systemic stimulation of mice with agonists for TLR2 (Pam3CSK4) or TLR5 (flagellin) did not reduce mortality on Y. enterocolitica infection. Systemic administration of poly I:C mobilized CD11c+, F4/80+, and Gr−1hi cells from lamina propria and activated NK cells in the mesenteric lymph nodes (MLN) within 24 h. This innate immune cell rearrangement was type I IFN dependent and mediated through upregulation of TLR4 followed by CCR7 expression in these innate immune cells found in the intestinal mucosa. Poly I:C induced IFN-γ expression by NK cells in the MLN, which was mediated through type I IFNs and IL-12p40 from antigen presenting cells and consequent activation of STAT1 and STAT4 in NK cells. This formation of innate immunity significantly contributed to the elimination of bacteria in the MLN. Our results demonstrated an innate immune network in the intestine that can be established by systemic stimulation of TRIF, which provides a strong host defense against Gram-negative pathogens. The mechanism underlying TRIF-mediated protective immunity may be useful to develop novel therapies for enteric bacterial infection.
Collapse
Affiliation(s)
- Jose Ruiz
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine Miami, FL, USA
| | - Saravana Kanagavelu
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of MedicineMiami, FL, USA; Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, F. Widjaja Foundation, Inflammatory Bowel and Immunology Research InstituteLos Angeles, CA, USA
| | - Claudia Flores
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, F. Widjaja Foundation, Inflammatory Bowel and Immunology Research Institute Los Angeles, CA, USA
| | - Laura Romero
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine Miami, FL, USA
| | - Reldy Riveron
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine Miami, FL, USA
| | - David Q Shih
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, F. Widjaja Foundation, Inflammatory Bowel and Immunology Research InstituteLos Angeles, CA, USA; Department of Medicine, David Geffen School of Medicine, University of CaliforniaLos Angeles, CA, USA
| | - Masayuki Fukata
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of MedicineMiami, FL, USA; Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, F. Widjaja Foundation, Inflammatory Bowel and Immunology Research InstituteLos Angeles, CA, USA; Department of Medicine, David Geffen School of Medicine, University of CaliforniaLos Angeles, CA, USA; Department of Cell Biology, University of Miami Miller School of MedicineMiami, FL, USA; Department of Biomedical Science, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| |
Collapse
|
268
|
Thompson LJ, Lai JF, Valladao AC, Thelen TD, Urry ZL, Ziegler SF. Conditioning of naive CD4(+) T cells for enhanced peripheral Foxp3 induction by nonspecific bystander inflammation. Nat Immunol 2016; 17:297-303. [PMID: 26752376 PMCID: PMC4757503 DOI: 10.1038/ni.3329] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/21/2015] [Indexed: 12/13/2022]
Abstract
Inflammation induced during infection can both promote and suppress immunity. This contradiction suggests that inflammatory cytokines impact the immune system in a context-dependent manner. Here we show that non-specific bystander inflammation conditioned naïve CD4+ T cells for enhanced peripheral Foxp3 induction and reduced effector differentiation. This resulted in inhibition of immune responses in vivo via Foxp3-dependent effect on antigen-specific naïve CD4+ T cell precursors. Such conditioning may have evolved to allow immunity to infection while limiting subsequent autoimmunity caused by release of self-antigens in the wake of infection. Furthermore, this phenomenon suggests a mechanistic explanation for the concept that early tuning of the immune system by infection impacts the long-term quality of immune regulation.
Collapse
Affiliation(s)
- Lucas J Thompson
- Benaroya Research Institute, Immunology Research Program, Seattle, Washington, USA
| | - Jen-Feng Lai
- Benaroya Research Institute, Immunology Research Program, Seattle, Washington, USA
| | - Andrea C Valladao
- Benaroya Research Institute, Immunology Research Program, Seattle, Washington, USA.,Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Tennille D Thelen
- Benaroya Research Institute, Immunology Research Program, Seattle, Washington, USA.,Department of Immunology, University of Washington, Seattle, Washington, USA
| | | | - Steven F Ziegler
- Benaroya Research Institute, Immunology Research Program, Seattle, Washington, USA
| |
Collapse
|
269
|
Cauwelaert ND, Desbien AL, Hudson TE, Pine SO, Reed SG, Coler RN, Orr MT. The TLR4 Agonist Vaccine Adjuvant, GLA-SE, Requires Canonical and Atypical Mechanisms of Action for TH1 Induction. PLoS One 2016; 11:e0146372. [PMID: 26731269 PMCID: PMC4701231 DOI: 10.1371/journal.pone.0146372] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
The Toll-like receptor 4 agonist glucopyranosyl lipid adjuvant formulated in a stable emulsion (GLA-SE) promotes strong TH1 and balanced IgG1/IgG2 responses to protein vaccine antigens. This enhanced immunity is sufficient to provide protection against many diseases including tuberculosis and leishmaniasis. To better characterize the adjuvant action it is important to understand how the different cytokines and transcription factors contribute to the initiation of immunity. In the present study using T-bet-/- and IL-12-/- mice and a blocking anti-IFNαR1 monoclonal antibody, we define mechanisms of adjuvant activity of GLA-SE. In accordance with previous studies of TLR4 agonist based adjuvants, we found that TH1 induction via GLA-SE was completely dependent upon T-bet, a key transcription factor for IFNγ production and TH1 differentiation. Consistent with this, deficiency of IL-12, a cytokine canonical to TH1 induction, ablated TH1 induction via GLA-SE. Finally we demonstrate that the innate immune response to GLA-SE, including rapid IFNγ production by memory CD8+ T cells and NK cells, was contingent on type I interferon, a cytokine group whose association with TH1 induction is contextual, and that they contributed to the adjuvant activity of GLA-SE.
Collapse
Affiliation(s)
| | - Anthony L. Desbien
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Thomas E. Hudson
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Samuel O. Pine
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Rhea N. Coler
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Mark T. Orr
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
270
|
Martins KAO, Cooper CL, Stronsky SM, Norris SLW, Kwilas SA, Steffens JT, Benko JG, van Tongeren SA, Bavari S. Adjuvant-enhanced CD4 T Cell Responses are Critical to Durable Vaccine Immunity. EBioMedicine 2015; 3:67-78. [PMID: 26870818 PMCID: PMC4739439 DOI: 10.1016/j.ebiom.2015.11.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/08/2023] Open
Abstract
Protein-based vaccines offer a safer alternative to live-attenuated or inactivated vaccines but have limited immunogenicity. The identification of adjuvants that augment immunogenicity, specifically in a manner that is durable and antigen-specific, is therefore critical for advanced development. In this study, we use the filovirus virus-like particle (VLP) as a model protein-based vaccine in order to evaluate the impact of four candidate vaccine adjuvants on enhancing long term protection from Ebola virus challenge. Adjuvants tested include poly-ICLC (Hiltonol), MPLA, CpG 2395, and alhydrogel. We compared and contrasted antibody responses, neutralizing antibody responses, effector T cell responses, and T follicular helper (Tfh) cell frequencies with each adjuvant's impact on durable protection. We demonstrate that in this system, the most effective adjuvant elicits a Th1-skewed antibody response and strong CD4 T cell responses, including an increase in Tfh frequency. Using immune-deficient animals and adoptive transfer of serum and cells from vaccinated animals into naïve animals, we further demonstrate that serum and CD4 T cells play a critical role in conferring protection within effective vaccination regimens. These studies inform on the requirements of long term immune protection, which can potentially be used to guide screening of clinical-grade adjuvants for vaccine clinical development. Adjuvants can prolong the protection afforded by protein-based vaccines and impact adaptive immune responses Enhanced CD4 T cell responses, helper and effector, correlate with duration of protection Durable protection from ma-EBOV is associated with Tfh frequency, Th1 antibody titers, and effector CD4 T cells
Protein-based vaccines are extremely safe, but they sometimes require the addition of adjuvants to enhance immunogenicity. In this study, we compared the impact of multiple adjuvants on immunogenicity, focusing on the duration of vaccine-mediated protection in mice. We then looked at how each adjuvant impacted the immune response in order to identify correlates of that long lasting immunity. The most effective adjuvant/vaccine combinations elicited multifunctional CD4 T cell responses and a Th1-skewed antibody response. By transferring antigen-experienced CD4 T cells and serum into naïve animals, we demonstrated that both CD4 T cells and serum were critical for durable vaccine-mediated protection.
Collapse
Key Words
- Adjuvant
- BME, beta mercaptoethanol
- CD, cluster of differentiation
- DSCF, Dwass, Steel, Critchlow-Fligner
- Durable protection
- ELISA, Enzyme linked immunosorbent assay
- ELISPOT, enzyme-linked immunospot assay
- Ebola virus
- FACS, fluorescence activated cell sorting
- FBS, fetal bovine serum
- GP, glycoprotein
- IACUC, Institutional Animal Care and Use Committee
- IM, intramuscular
- IP, intraperitoneal
- IQR, interquartile range
- Immune correlates
- LN, lymph node
- MPLA, monophosphoryl lipid A
- NAb, neutralizing antibody
- Ns, not significant
- PBS, phosphate buffered saline
- PRR, pattern recognition receptor
- Pfu, plaque forming unit
- PsVNA, pseudovirion neutralization assay
- TLR, Toll-like receptor
- USAMRIID, United States Army Medical Research Institute of Infectious Diseases
- VLP, virus-like particle
- Vaccine
- ma-EBOV, mouse-adapted Ebola virus
Collapse
Affiliation(s)
- Karen A O Martins
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Christopher L Cooper
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sabrina M Stronsky
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sarah L W Norris
- Research Support Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Steven A Kwilas
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Jesse T Steffens
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Jacqueline G Benko
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sean A van Tongeren
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sina Bavari
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA.
| |
Collapse
|
271
|
Dong H, Franklin NA, Ritchea SB, Yagita H, Glennie MJ, Bullock TNJ. CD70 and IFN-1 selectively induce eomesodermin or T-bet and synergize to promote CD8+ T-cell responses. Eur J Immunol 2015; 45:3289-301. [PMID: 26461455 DOI: 10.1002/eji.201445291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 08/21/2015] [Accepted: 09/24/2015] [Indexed: 12/17/2022]
Abstract
CD70-mediated stimulation of CD27 is an important cofactor of CD4(+) T-cell licensed dendritic cells (DCs). However, it is unclear how CD70-mediated stimulation of T cells is integrated with signals that emanate from signal 3 pathways, such as type-1 interferon (IFN-1) and IL-12. We find that while stimulation of CD27 in isolation drives weak Eomesodermin(hi) T-bet(lo) CD8(+) T-cell responses to OVA immunization, profound synergistic expansion is achieved by cotargeting TLR. This cooperativity can substantially boost antiviral CD8(+) T-cell responses during acute infection. Concomitant stimulation of TLR significantly increases per cell IFN-γ production and the proportion of the population with characteristics of short-lived effector cells, yet also promotes the ability to form long-lived memory. Notably, while IFN-1 contributes to the expression of CD70 on DCs, the synergy between CD27 and TLR stimulation is dependent upon IFN-1's effect directly on CD8(+) T cells, and is associated with the increased expression of T-bet in T cells. Surprisingly, we find that IL-12 fails to synergize with CD27 stimulation to promote CD8(+) T-cell expansion, despite its capacity to drive effector CD8(+) T-cell differentiation. Together, these data identify complex interactions between signal 3 and costimulatory pathways, and identify opportunities to influence the differentiation of CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Han Dong
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Nathan A Franklin
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Shane B Ritchea
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Martin J Glennie
- Cancer Sciences Division, Southampton University School of Medicine, General Hospital, Southampton, UK
| | | |
Collapse
|
272
|
Differential outcome of TRIF-mediated signaling in TLR4 and TLR3 induced DC maturation. Proc Natl Acad Sci U S A 2015; 112:13994-9. [PMID: 26508631 DOI: 10.1073/pnas.1510760112] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Recognition of pathogen-associated molecular patterns by Toll-like receptors (TLRs) on dendritic cells (DCs) leads to DC maturation, a process involving up-regulation of MHC and costimulatory molecules and secretion of proinflammatory cytokines. All TLRs except TLR3 achieve these outcomes by using the signaling adaptor myeloid differentiation factor 88. TLR4 and TLR3 can both use the Toll-IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF)-dependent signaling pathway leading to IFN regulatory factor 3 (IRF3) activation and induction of IFN-β and -α4. The TRIF signaling pathway, downstream of both of these TLRs, also leads to DC maturation, and it has been proposed that the type I IFNs act in cis to induce DC maturation and subsequent effects on adaptive immunity. The present study was designed to understand the molecular mechanisms of TRIF-mediated DC maturation. We have discovered that TLR4-TRIF-induced DC maturation was independent of both IRF3 and type I IFNs. In contrast, TLR3-mediated DC maturation was completely dependent on type I IFN feedback. We found that differential activation of mitogen-activated protein kinases by the TLR4- and TLR3-TRIF axes determined the type I IFN dependency for DC maturation. In addition, we found that the adjuvanticity of LPS to induce T-cell activation is completely independent of type I IFNs. The important distinction between the TRIF-mediated signaling pathways of TLR4 and TLR3 discovered here could have a major impact in the design of future adjuvants that target this pathway.
Collapse
|
273
|
Beljanski V, Chiang C, Kirchenbaum GA, Olagnier D, Bloom CE, Wong T, Haddad EK, Trautmann L, Ross TM, Hiscott J. Enhanced Influenza Virus-Like Particle Vaccination with a Structurally Optimized RIG-I Agonist as Adjuvant. J Virol 2015; 89:10612-24. [PMID: 26269188 PMCID: PMC4580177 DOI: 10.1128/jvi.01526-15] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The molecular interaction between viral RNA and the cytosolic sensor RIG-I represents the initial trigger in the development of an effective immune response against infection with RNA viruses, resulting in innate immune activation and subsequent induction of adaptive responses. In the present study, the adjuvant properties of a sequence-optimized 5'-triphosphate-containing RNA (5'pppRNA) RIG-I agonist (termed M8) were examined in combination with influenza virus-like particles (VLP) (M8-VLP) expressing H5N1 influenza virus hemagglutinin (HA) and neuraminidase (NA) as immunogens. In combination with VLP, M8 increased the antibody response to VLP immunization, provided VLP antigen sparing, and protected mice from a lethal challenge with H5N1 influenza virus. M8-VLP immunization also led to long-term protective responses against influenza virus infection in mice. M8 adjuvantation of VLP increased endpoint and antibody titers and inhibited influenza virus replication in lungs compared with approved or experimental adjuvants alum, AddaVax, and poly(I·C). Uniquely, immunization with M8-VLP stimulated a TH1-biased CD4 T cell response, as determined by increased TH1 cytokine levels in CD4 T cells and increased IgG2 levels in sera. Collectively, these data demonstrate that a sequence-optimized, RIG-I-specific agonist is a potent adjuvant that can be utilized to increase the efficacy of influenza VLP vaccination and dramatically improve humoral and cellular mediated protective responses against influenza virus challenge. IMPORTANCE The development of novel adjuvants to increase vaccine immunogenicity is an important goal that seeks to improve vaccine efficacy and ultimately prevent infections that endanger human health. This proof-of-principle study investigated the adjuvant properties of a sequence-optimized 5'pppRNA agonist (M8) with enhanced capacity to stimulate antiviral and inflammatory gene networks using influenza virus-like particles (VLP) expressing HA and NA as immunogens. Vaccination with VLP in combination with M8 increased anti-influenza virus antibody titers and protected animals from lethal influenza virus challenge, highlighting the potential clinical use of M8 as an adjuvant in vaccine development. Altogether, the results describe a novel immunostimulatory agonist targeted to the cytosolic RIG-I sensor as an attractive vaccine adjuvant candidate that can be used to increase vaccine efficacy, a pressing issue in children and the elderly population.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Animals
- Antibodies, Viral/biosynthesis
- DEAD Box Protein 58
- DEAD-box RNA Helicases/chemistry
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/immunology
- Dendritic Cells/immunology
- Dendritic Cells/virology
- Female
- HEK293 Cells
- Hemagglutinins, Viral/chemistry
- Hemagglutinins, Viral/genetics
- Hemagglutinins, Viral/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunization
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice
- Mice, Inbred BALB C
- Neuraminidase/chemistry
- Neuraminidase/genetics
- Neuraminidase/immunology
- Oligoribonucleotides/administration & dosage
- Oligoribonucleotides/genetics
- Oligoribonucleotides/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/mortality
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Primary Cell Culture
- Receptors, Immunologic
- Survival Analysis
- Th1-Th2 Balance/drug effects
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Vladimir Beljanski
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Cindy Chiang
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Greg A Kirchenbaum
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - David Olagnier
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Chalise E Bloom
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Terianne Wong
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Elias K Haddad
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Lydie Trautmann
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Ted M Ross
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - John Hiscott
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| |
Collapse
|
274
|
da Costa TA, Di Gangi R, Martins P, Longhini ALF, Zanucoli F, de Oliveira ALR, Stach-Machado DR, Burger E, Verinaud L, Thomé R. Protection against Paracoccidioides brasiliensis infection in mice treated with modulated dendritic cells relies on inhibition of interleukin-10 production by CD8+ T cells. Immunology 2015; 146:486-95. [PMID: 26302057 DOI: 10.1111/imm.12526] [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: 04/22/2015] [Revised: 08/08/2015] [Accepted: 08/18/2015] [Indexed: 11/27/2022] Open
Abstract
Paracoccidioidomycosis is a systemic infection prevalent in Latin American countries. Disease develops after inhalation of Paracoccidioides brasiliensis conidia followed by an improper immune activation by the host leucocytes. Dendritic cells (DCs) are antigen-presenting cells with the unique ability to direct the adaptive immune response by the time of activation of naive T cells. This study was conducted to test whether extracts of P. brasiliensis would induce maturation of DCs. We found that DCs treated with extracts acquired an inflammatory phenotype and upon adoptive transfer conferred protection to infection. Interestingly, interleukin-10 production by CD8(+) T cells was ablated following DC transfer. Further analyses showed that lymphocytes from infected mice were high producers of interleukin-10, with CD8(+) T cells being the main source. Blockage of cross-presentation to CD8(+) T cells by modulated DCs abolished the protective effect of adoptive transfer. Collectively, our data show that adoptive transfer of P. brasiliensis-modulated DCs is an interesting approach for the control of infection in paracoccidioidomycosis.
Collapse
Affiliation(s)
- Thiago Alves da Costa
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Rosária Di Gangi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Paula Martins
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | - Fábio Zanucoli
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | - Dagmar Ruth Stach-Machado
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Eva Burger
- Instituto de Biociências, Universidade Federal de Alfenas, Alfenas, MG, Brazil
| | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
275
|
Sehgal M, Zeremski M, Talal AH, Ginwala R, Elrod E, Grakoui A, Li QG, Philip R, Khan ZK, Jain P. IFN-α-Induced Downregulation of miR-221 in Dendritic Cells: Implications for HCV Pathogenesis and Treatment. J Interferon Cytokine Res 2015; 35:698-709. [PMID: 26090579 PMCID: PMC4560851 DOI: 10.1089/jir.2014.0211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/19/2015] [Indexed: 12/19/2022] Open
Abstract
Although interferon (IFN)-α is known to exert immunomodulatory and antiproliferative effects on dendritic cells (DCs) through induction of protein-coding IFN-stimulated genes (ISGs), little is known about IFN-α-regulated miRNAs in DCs. Since several miRNAs are involved in regulating DC functions, it is important to investigate whether IFN-α's effects on DCs are mediated through miRNAs as well. In this study, we examined miRNA expression patterns in myeloid DCs (mDCs) and plasmacytoid DCs after exposing them to IFN-α. We report that IFN-α downregulates miR-221 in both DC subsets via inhibition of STAT3. We validated proapoptotic proteins BCL2L11 and CDKN1C as miR-221 targets suggesting that IFN-α can induce DC apoptosis via miR-221 downregulation. In addition, we validated another miR-221 target, SOCS1, which is known to be a negative regulator of JAK/STAT signaling. Consistent with this, miR-221 overexpression in mDCs enhanced the secretion of proinflammatory cytokines IL-6 and TNF-α. In peripheral blood mononuclear cells (PBMCs) of HIV-1/HCV co-infected individuals undergoing IFN-α-based treatment the baseline miR-221 expression was lower in non-responders compared with responders; and miR-221 expression directly correlated with DC frequency and IL-6/TNF-α secretion. In addition to PBMCs, we isolated total liver cells and kupffer cells from HCV-infected individuals and individuals with alcoholic cirrhosis. We found that both total liver cells and kupffer cells from HCV-infected individuals had significantly higher miR-221 levels compared with individuals with cirrhosis. Overall, we demonstrate that IFN-α exerts both antiproliferative and immunomodulatory effects on mDCs via miR-221 downregulation; and IFN-miR-221 axis can play important role in HCV pathogenesis and treatment.
Collapse
Affiliation(s)
- Mohit Sehgal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | | | - Andrew H. Talal
- School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Rashida Ginwala
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | | | | | - Qi-Ging Li
- Duke University Medical Center, Durham, North Carolina
| | - Ramila Philip
- Immunotope, Inc., Pennsylvania Biotechnology Center, Doylestown, Pennsylvania
| | - Zafar K. Khan
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
276
|
Kerbrat S, Vingert B, Junier MP, Castellano F, Renault-Mihara F, Dos Reis Tavares S, Surenaud M, Noizat-Pirenne F, Boczkowski J, Guellaën G, Chneiweiss H, Le Gouvello S. Absence of the Adaptor Protein PEA-15 Is Associated with Altered Pattern of Th Cytokines Production by Activated CD4+ T Lymphocytes In Vitro, and Defective Red Blood Cell Alloimmune Response In Vivo. PLoS One 2015; 10:e0136885. [PMID: 26317969 PMCID: PMC4552951 DOI: 10.1371/journal.pone.0136885] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/10/2015] [Indexed: 01/08/2023] Open
Abstract
TCR-dependent and costimulation signaling, cell division, and cytokine environment are major factors driving cytokines expression induced by CD4+ T cell activation. PEA-15 15 (Protein Enriched in Astrocyte / 15kDa) is an adaptor protein that regulates death receptor-induced apoptosis and proliferation signaling by binding to FADD and relocating ERK1/2 to the cytosol, respectively. By using PEA-15-deficient mice, we examined the role of PEA-15 in TCR-dependent cytokine production in CD4+ T cells. TCR-stimulated PEA-15-deficient CD4+ T cells exhibited defective progression through the cell cycle associated with impaired expression of cyclin E and phosphoRb, two ERK1/2-dependent proteins of the cell cycle. Accordingly, expression of the division cycle-dependent cytokines IL-2 and IFNγ, a Th1 cytokine, was reduced in stimulated PEA-15-deficient CD4+ T cells. This was associated with abnormal subcellular compartmentalization of activated ERK1/2 in PEA-15-deficient T cells. Furthermore, in vitro TCR-dependent differentiation of naive CD4+ CD62L+ PEA-15-deficient T cells was associated with a lower production of the Th2 cytokine, IL-4, whereas expression of the Th17-associated molecule IL4I1 was enhanced. Finally, a defective humoral response was shown in PEA-15-deficient mice in a model of red blood cell alloimmunization performed with Poly IC, a classical adjuvant of Th1 response in vivo. Collectively, our data suggest that PEA-15 contributes to the specification of the cytokine pattern of activated Th cells, thus highlighting a potential new target to interfere with T cell functional polarization and subsequent immune response.
Collapse
Affiliation(s)
- Stéphane Kerbrat
- Université Paris-Est, Créteil, France
- Inserm U955, Créteil, France
| | - Benoit Vingert
- Inserm U955, Créteil, France
- Etablissement Français du Sang, Créteil, France
| | - Marie-Pierre Junier
- Inserm, U1130, Neuroscience Paris Seine, IBPS, Paris, France
- Université Pierre et Marie Curie, UM119, Neuroscience Paris Seine, IBPS, Paris, France
- CNRS, UMR8246, Neuroscience Paris Seine, IBPS, Paris, France
| | - Flavia Castellano
- Université Paris-Est, Créteil, France
- Inserm U955, Créteil, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Pôle de Biologie-Pathologie, Créteil, France
| | - François Renault-Mihara
- Inserm, U1130, Neuroscience Paris Seine, IBPS, Paris, France
- Université Pierre et Marie Curie, UM119, Neuroscience Paris Seine, IBPS, Paris, France
- CNRS, UMR8246, Neuroscience Paris Seine, IBPS, Paris, France
| | - Silvina Dos Reis Tavares
- Inserm, U1130, Neuroscience Paris Seine, IBPS, Paris, France
- Université Pierre et Marie Curie, UM119, Neuroscience Paris Seine, IBPS, Paris, France
- CNRS, UMR8246, Neuroscience Paris Seine, IBPS, Paris, France
| | | | - France Noizat-Pirenne
- Université Paris-Est, Créteil, France
- Inserm U955, Créteil, France
- Etablissement Français du Sang, Créteil, France
| | - Jorge Boczkowski
- Université Paris-Est, Créteil, France
- Inserm U955, Créteil, France
| | - Georges Guellaën
- Université Paris-Est, Créteil, France
- Inserm U955, Créteil, France
| | - Hervé Chneiweiss
- Inserm, U1130, Neuroscience Paris Seine, IBPS, Paris, France
- Université Pierre et Marie Curie, UM119, Neuroscience Paris Seine, IBPS, Paris, France
- CNRS, UMR8246, Neuroscience Paris Seine, IBPS, Paris, France
- * E-mail: (SLG); (HC)
| | - Sabine Le Gouvello
- Université Paris-Est, Créteil, France
- Inserm U955, Créteil, France
- AP-HP, Hôpital H. Mondor- A. Chenevier, Pôle de Biologie-Pathologie, Créteil, France
- * E-mail: (SLG); (HC)
| |
Collapse
|
277
|
Dhillon SK, Gunn AJ, Jung Y, Mathai S, Bennet L, Fraser M. Lipopolysaccharide-Induced Preconditioning Attenuates Apoptosis and Differentially Regulates TLR4 and TLR7 Gene Expression after Ischemia in the Preterm Ovine Fetal Brain. Dev Neurosci 2015; 37:497-514. [PMID: 26184807 DOI: 10.1159/000433422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/13/2015] [Indexed: 11/19/2022] Open
Abstract
Acute exposure to subclinical infection modulates subsequent hypoxia-ischemia (HI) injury in a time-dependent manner, likely by cross-talk through Toll-like receptors (TLRs), but the specific pathways are unclear in the preterm-equivalent brain. In the present study, we tested the hypothesis that repeated low-dose exposure to lipopolysaccharide (LPS) before acute ischemia would be associated with induction of specific TLRs that are potentially neuroprotective. Fetal sheep at 0.65 gestation (term is ∼145 days) received intravenous boluses of low-dose LPS for 5 days (day 1, 50 ng/kg; days 2-5, 100 ng/kg) or the same volume of saline. Either 4 or 24 h after the last bolus of LPS, complete carotid occlusion was induced for 22 min. Five days after LPS, brains were collected. Pretreatment with LPS for 5 days decreased cellular apoptosis, microglial activation and reactive astrogliosis in response to HI injury induced 24 but not 4 h after the last dose of LPS. This was associated with upregulation of TLR4, TLR7 and IFN-β mRNA, and increased fetal plasma IFN-β concentrations. The association of reduced white matter apoptosis and astrogliosis after repeated low-dose LPS finishing 24 h but not 4 h before cerebral ischemia, with central and peripheral induction of IFN-β, suggests the possibility that IFN-β may be an important mediator of endogenous neuroprotection in the developing brain.
Collapse
|
278
|
A Plasmacytoid Dendritic Cells-Type I Interferon Axis Is Critically Implicated in the Pathogenesis of Systemic Lupus Erythematosus. Int J Mol Sci 2015; 16:14158-70. [PMID: 26110387 PMCID: PMC4490545 DOI: 10.3390/ijms160614158] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/01/2015] [Accepted: 06/16/2015] [Indexed: 01/12/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease that is characterized by the generation of immune responses to various nuclear components. Impaired clearance of apoptotic cells and loss of tolerance to self-antigens are involved both in the initiation and in the propagation of the disease. Dendritic cells (DCs) are key factors in the balance between autoimmunity and tolerance and play a role linking innate and adaptive immunity. DCs, particularly plasmacytoid DCs (pDCs), are the main source of type I interferon (IFN) cytokines, which contribute to the immunopathogenesis of SLE. There is accumulating evidence that pDCs and type I IFN cytokines take the leading part in the development of SLE. In this review, we discuss recent data regarding the role of pDCs and type I IFN cytokines in the pathogenesis of SLE and the potential for employing therapies targeting against aberrant regulation of the pDC-type I IFN axis for treating SLE.
Collapse
|
279
|
Széles L, Meissner F, Dunand-Sauthier I, Thelemann C, Hersch M, Singovski S, Haller S, Gobet F, Fuertes Marraco SA, Mann M, Garcin D, Acha-Orbea H, Reith W. TLR3-Mediated CD8+ Dendritic Cell Activation Is Coupled with Establishment of a Cell-Intrinsic Antiviral State. THE JOURNAL OF IMMUNOLOGY 2015; 195:1025-33. [PMID: 26101320 DOI: 10.4049/jimmunol.1402033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 05/20/2015] [Indexed: 01/13/2023]
Abstract
Because of their unique capacity to cross-present Ags to CD8(+) T cells, mouse lymphoid tissue-resident CD8(+) dendritic cells (DCs) and their migratory counterparts are critical for priming antiviral T cell responses. High expression of the dsRNA sensor TLR3 is a distinctive feature of these cross-presenting DC subsets. TLR3 engagement in CD8(+) DCs promotes cross-presentation and the acquisition of effector functions required for driving antiviral T cell responses. In this study, we performed a comprehensive analysis of the TLR3-induced antiviral program and cell-autonomous immunity in CD8(+) DC lines and primary CD8(+) DCs. We found that TLR3-ligand polyinosinic-polycytidylic acid and human rhinovirus infection induced a potent antiviral protection against Sendai and vesicular stomatitis virus in a TLR3 and type I IFN receptor-dependent manner. Polyinosinic-polycytidylic acid-induced antiviral genes were identified by mass spectrometry-based proteomics and transcriptomics in the CD8(+) DC line. Nanostring nCounter experiments confirmed that these antiviral genes were induced by TLR3 engagement in primary CD8(+) DCs, and indicated that many are secondary TLR3-response genes requiring autocrine IFN-β stimulation. TLR3-activation thus establishes a type I IFN-dependent antiviral program in a DC subtype playing crucial roles in priming adaptive antiviral immune responses. This mechanism is likely to shield the priming of antiviral responses against inhibition or abrogation by the viral infection. It could be particularly relevant for viruses detected mainly by TLR3, which may not trigger type I IFN production by DCs that lack TLR3, such as plasmacytoid DCs or CD8(-) DCs.
Collapse
Affiliation(s)
- Lajos Széles
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Felix Meissner
- Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| | - Isabelle Dunand-Sauthier
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christoph Thelemann
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Micha Hersch
- Computational Biology Group, Department of Medical Genetics, University of Lausanne, CH-1005 Lausanne, Switzerland; and
| | - Simon Singovski
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Sergio Haller
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Florian Gobet
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | | | - Matthias Mann
- Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, D-82152 Martinsried, Germany
| | - Dominique Garcin
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Hans Acha-Orbea
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland;
| |
Collapse
|
280
|
Martin-Gayo E, Buzon MJ, Ouyang Z, Hickman T, Cronin J, Pimenova D, Walker BD, Lichterfeld M, Yu XG. Potent Cell-Intrinsic Immune Responses in Dendritic Cells Facilitate HIV-1-Specific T Cell Immunity in HIV-1 Elite Controllers. PLoS Pathog 2015; 11:e1004930. [PMID: 26067651 PMCID: PMC4466270 DOI: 10.1371/journal.ppat.1004930] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/01/2015] [Indexed: 12/29/2022] Open
Abstract
The majority of HIV-1 elite controllers (EC) restrict HIV-1 replication through highly functional HIV-1-specific T cell responses, but mechanisms supporting the evolution of effective HIV-1-specific T cell immunity in these patients remain undefined. Cytosolic immune recognition of HIV-1 in conventional dendritic cells (cDC) can facilitate priming and expansion of HIV-1-specific T cells; however, HIV-1 seems to be able to avoid intracellular immune recognition in cDCs in most infected individuals. Here, we show that exposure of cDCs from EC to HIV-1 leads to a rapid and sustained production of type I interferons and upregulation of several interferon-stimulated effector genes. Emergence of these cell-intrinsic immune responses was associated with a reduced induction of SAMHD1 and LEDGF/p75, and an accumulation of viral reverse transcripts, but inhibited by pharmacological blockade of viral reverse transcription or siRNA-mediated silencing of the cytosolic DNA sensor cGAS. Importantly, improved cell-intrinsic immune recognition of HIV-1 in cDCs from elite controllers translated into stronger abilities to stimulate and expand HIV-1-specific CD8 T cell responses. These data suggest an important role of cell-intrinsic type I interferon secretion in dendritic cells for the induction of effective HIV-1-specific CD8 T cells, and may be helpful for eliciting functional T cell immunity against HIV-1 for preventative or therapeutic clinical purposes.
Collapse
Affiliation(s)
- Enrique Martin-Gayo
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Maria Jose Buzon
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Zhengyu Ouyang
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Taylor Hickman
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Jacqueline Cronin
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Dina Pimenova
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
281
|
Errett JS, Gale M. Emerging complexity and new roles for the RIG-I-like receptors in innate antiviral immunity. Virol Sin 2015; 30:163-73. [PMID: 25997992 PMCID: PMC7090589 DOI: 10.1007/s12250-015-3604-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/13/2015] [Indexed: 12/24/2022] Open
Abstract
Innate immunity is critical for the control of virus infection and operates to restrict viral susceptibility and direct antiviral immunity for protection from acute or chronic viral-associated diseases including cancer. RIG-I like receptors (RLRs) are cytosolic RNA helicases that function as pathogen recognition receptors to detect RNA pathogen associated molecular patterns (PAMPs) of virus infection. The RLRs include RIG-I, MDA5, and LGP2. They function to recognize and bind to PAMP motifs within viral RNA in a process that directs the RLR to trigger downstream signaling cascades that induce innate immunity that controls viral replication and spread. Products of RLR signaling also serve to modulate the adaptive immune response to infection. Recent studies have additionally connected RLRs to signaling cascades that impart inflammatory and apoptotic responses to virus infection. Viral evasion of RLR signaling supports viral outgrowth and pathogenesis, including the onset of viral-associated cancer.
Collapse
Affiliation(s)
- John S Errett
- Center for Innate Immunity and Immune Disease, Department of Immunology, School of Medicine, University of Washington, Seattle, 98109, USA
| | | |
Collapse
|
282
|
Violacein Treatment Modulates Acute and Chronic Inflammation through the Suppression of Cytokine Production and Induction of Regulatory T Cells. PLoS One 2015; 10:e0125409. [PMID: 25938431 PMCID: PMC4418714 DOI: 10.1371/journal.pone.0125409] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/23/2015] [Indexed: 12/20/2022] Open
Abstract
Inflammation is a necessary process to control infection. However, exacerbated inflammation, acute or chronic, promotes deleterious effects in the organism. Violacein (viola), a quorum sensing metabolite from the Gram-negative bacterium Chromobacterium violaceum, has been shown to protect mice from malaria and to have beneficial effects on tumors. However, it is not known whether this drug possesses anti-inflammatory activity. In this study, we investigated whether viola administration is able to reduce acute and chronic autoimmune inflammation. For that purpose, C57BL/6 mice were intraperitoneally injected with 1 μg of LPS and were treated with viola (3.5mg/kg) via i.p. at the same time-point. Three hours later, the levels of inflammatory cytokines in the sera and phenotypical characterization of leukocytes were determined. Mice treated with viola presented a significant reduction in the production of inflammatory cytokines compared with untreated mice. Interestingly, although viola is a compound derived from bacteria, it did not induce inflammation upon administration to naïve mice. To test whether viola would protect mice from an autoimmune inflammation, Experimental Autoimmune Encephalomyelitis (EAE)-inflicted mice were given viola i.p. at disease onset, at the 10th day from immunization. Viola-treated mice developed mild EAE disease in contrast with placebo-treated mice. The frequencies of dendritic cells and macrophages were unaltered in EAE mice treated with viola. However, the sole administration of viola augmented the levels of splenic regulatory T cells (CD4+Foxp3+). We also found that adoptive transfer of viola-elicited regulatory T cells significantly reduced EAE. Our study shows, for the first time, that violacein is able to modulate acute and chronic inflammation. Amelioration relied in suppression of cytokine production (in acute inflammation) and stimulation of regulatory T cells (in chronic inflammation). New studies must be conducted in order to assess the possible use of viola in therapeutic approaches in human autoimmune diseases.
Collapse
|
283
|
Dahlgren MW, Gustafsson-Hedberg T, Livingston M, Cucak H, Alsén S, Yrlid U, Johansson-Lindbom B. T follicular helper, but not Th1, cell differentiation in the absence of conventional dendritic cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:5187-99. [PMID: 25917099 DOI: 10.4049/jimmunol.1401938] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/18/2015] [Indexed: 11/19/2022]
Abstract
Development of long-lived humoral immunity is dependent on CXCR5-expressing T follicular helper (Tfh) cells, which develop concomitantly to effector Th cells that support cellular immunity. Conventional dendritic cells (cDCs) are critical APCs for initial priming of naive CD4(+) T cells but, importantly, also provide accessory signals that govern effector Th cell commitment. To define the accessory role of cDCs during the concurrent development of Tfh and effector Th1 cells, we performed high-dose Ag immunization in conjunction with the Th1-biased adjuvant polyinosinic:polycytidylic acid (pI:C). In the absence of cDCs, pI:C failed to induce Th1 cell commitment and IgG2c production. However, cDC depletion did not impair Tfh cell differentiation or germinal center formation, and long-lived IgG1 responses of unaltered affinity developed in mice lacking cDCs at the time point for immunization. Thus, cDCs are required for the pI:C-driven Th1 cell fate commitment but have no crucial accessory function in relation to Tfh cell differentiation.
Collapse
Affiliation(s)
| | - Tobias Gustafsson-Hedberg
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Megan Livingston
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Helena Cucak
- Immunology Section, Lund University, 221 84 Lund, Sweden; and
| | - Samuel Alsén
- Immunology Section, Lund University, 221 84 Lund, Sweden; and Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | | |
Collapse
|
284
|
Wang Y, Lavender P, Watson J, Arno M, Lehner T. Stress-activated Dendritic Cells (DC) Induce Dual Interleukin (IL)-15- and IL1β-mediated Pathways, Which May Elicit CD4+ Memory T Cells and Interferon (IFN)-stimulated Genes. J Biol Chem 2015; 290:15595-15609. [PMID: 25907558 DOI: 10.1074/jbc.m115.645754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Indexed: 01/03/2023] Open
Abstract
The prevailing evidence suggests that immunological memory does not require antigenic re-stimulation but is maintained by low level tonic stimulation. We examined the hypothesis that stress agents contribute to tonic cellular activation and maintain immunological memory. Stimulation of monocyte-derived dendritic cells (DC) with stress agents elicits reactive oxygen species and HSP70. NFκB is activated, which up-regulates membrane-associated (ma) IL-15, caspase-1 and IL-1β. Co-culture of stress-treated DC with mononuclear cells activates IL-15 and IL-1β receptors on CD4(+) T cells, eliciting CD40L, proliferation, and up-regulation of CD45RO(+) memory T cells. The transcription factors Tbet(high) and RORγt are up-regulated, whereas FoxP3 is down-regulated, resulting in enhanced Th1 and Th17 expression and the corresponding cytokines. The interaction between maIL-15 expressed by DC and IL-15R on CD4(+) T cells results in one pathway and the corresponding cells expressing IL-1β and IL1βR as a second pathway. Importantly, inhibition studies with IL-15 antibodies and IL-1βR inhibitor suggest that both pathways may be required for optimum CD4(+) CD45RO(+) memory T cell expression. Type 1 IFN expression in splenic CD11c DC of stress-treated mice demonstrated a significant increase of IFN-α in CD11c CD317(+) and CD8α(+) DC. Analysis of RNA in human CD4(+) memory T cells showed up-regulation of type 1 IFN-stimulated genes and inhibition with histone methyltransferase inhibitor. We suggest the paradigm that stress-induced tonic stimulation might be responsible for the robust persistence of the immune response in vaccination and that epigenetic changes are involved in maintaining CD4(+) T cell memory.
Collapse
Affiliation(s)
- Yufei Wang
- Mucosal Immunology Unit, Kings College London, London SE1 1UL, United Kingdom
| | - Paul Lavender
- MRC, and Asthma UK Centre in Allergic Mechanisms of Asthma, Kings College London, London SE1 1UL, United Kingdom
| | - Julie Watson
- MRC, and Asthma UK Centre in Allergic Mechanisms of Asthma, Kings College London, London SE1 1UL, United Kingdom
| | - Matthew Arno
- Genomics Centre, Kings College London, London SE1 1UL, United Kingdom
| | - Thomas Lehner
- Mucosal Immunology Unit, Kings College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
285
|
Chang CJ, Sun B, Robertsen B. Adjuvant activity of fish type I interferon shown in a virus DNA vaccination model. Vaccine 2015; 33:2442-8. [PMID: 25865470 DOI: 10.1016/j.vaccine.2015.03.093] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/24/2015] [Accepted: 03/27/2015] [Indexed: 12/17/2022]
Abstract
There is a need for more efficient vaccines to combat viral diseases of Atlantic salmon and other farmed fish. DNA vaccines are highly effective against salmonid rhabdoviruses, but have shown less effect against other viruses. In the present work we have studied if type I IFNs might be used as adjuvants in fish DNA vaccines. For this purpose we chose a DNA vaccine model based on the hemagglutinin-esterase (HE) gene of infectious salmon anemia virus (ISAV) as antigen. Salmon presmolts were injected with a plasmid encoding HE alone or together with a plasmid encoding Atlantic salmon type I IFN (IFNa1, IFNb or IFNc). Sera were harvested after 7-10 weeks for measurements of antibody against ISAV and the fish were challenged with ISAV to measure protective effects of the vaccines. The results showed that all three IFN plasmids delivered together with HE plasmid potently enhanced protection of salmon against ISAV mediated mortality and stimulated an increase in IgM antibodies against the virus. In contrast, HE plasmid alone gave low antibody titers and a minor protection against ISAV. This demonstrates that type I IFNs stimulate adaptive immune responses in fish, which may be a benefit also in other fish DNA vaccines. Quantitative RT-PCR studies showed that the salmon IFNs caused an increased influx of B-cells and cytotoxic T-cells at the muscle injection site, which may in part explain the adjuvant effect of the IFNs.
Collapse
Affiliation(s)
- Chia-Jung Chang
- Norwegian College of Fishery Science, University of Tromsø, 9037 Tromsø, Norway
| | - Baojian Sun
- Norwegian College of Fishery Science, University of Tromsø, 9037 Tromsø, Norway
| | - Børre Robertsen
- Norwegian College of Fishery Science, University of Tromsø, 9037 Tromsø, Norway.
| |
Collapse
|
286
|
|
287
|
Wilson KL, Xiang SD, Plebanski M. Montanide, Poly I:C and nanoparticle based vaccines promote differential suppressor and effector cell expansion: a study of induction of CD8 T cells to a minimal Plasmodium berghei epitope. Front Microbiol 2015; 6:29. [PMID: 25705207 PMCID: PMC4319470 DOI: 10.3389/fmicb.2015.00029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/09/2015] [Indexed: 01/02/2023] Open
Abstract
The development of practical and flexible vaccines to target liver stage malaria parasites would benefit from an ability to induce high levels of CD8 T cells to minimal peptide epitopes. Herein we compare different adjuvant and carrier systems in a murine model for induction of interferon gamma (IFN-γ) producing CD8 T cells to the minimal immuno-dominant peptide epitope from the circumsporozoite protein (CSP) of Plasmodium berghei, pb9 (SYIPSAEKI, referred to as KI). Two pro-inflammatory adjuvants, Montanide and Poly I:C, and a non-classical, non-inflammatory nanoparticle based carrier (polystyrene nanoparticles, PSNPs), were compared side-by-side for their ability to induce potentially protective CD8 T cell responses after two immunizations. KI in Montanide (Montanide + KI) or covalently conjugated to PSNPs (PSNPs-KI) induced such high responses, whereas adjuvanting with Poly I:C or PSNPs without conjugation was ineffective. This result was consistent with an observed induction of an immunosuppressed environment by Poly I:C in the draining lymph node (dLN) 48 h post injection, which was reflected by increased frequencies of myeloid derived suppressor cells (MDSCs) and a proportion of inflammation reactive regulatory T cells (Treg) expressing the tumor necrosis factor receptor 2 (TNFR2), as well as decreased dendritic cell (DC) maturation. The other inflammatory adjuvant, Montanide, also promoted proportional increases in the TNFR2+ Treg subpopulation, but not MDSCs, in the dLN. By contrast, injection with non-inflammatory PSNPs did not cause these changes. Induction of high CD8 T cell responses, using minimal peptide epitopes, can be achieved by non-inflammatory carrier nanoparticles, which in contrast to some conventional inflammatory adjuvants, do not expand either MDSCs or inflammation reactive Tregs at the site of priming.
Collapse
Affiliation(s)
- Kirsty L Wilson
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC Australia
| | - Sue D Xiang
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC Australia
| | - Magdalena Plebanski
- Department of Immunology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC Australia
| |
Collapse
|
288
|
He S, Mao X, Sun H, Shirakawa T, Zhang H, Wang X. Potential therapeutic targets in the process of nucleic acid recognition: opportunities and challenges. Trends Pharmacol Sci 2015; 36:51-64. [DOI: 10.1016/j.tips.2014.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 10/18/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023]
|
289
|
Chen S, Zhao Q, Qi Y, Liu F, Wang M, Jia R, Zhu D, Liu M, Chen X, Cheng A. Immunobiological activity and antiviral regulation efforts of Chinese goose (Anser cygnoides) CD8α during NGVEV and GPV infection. Poult Sci 2015; 94:17-24. [DOI: 10.3382/ps/peu024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
290
|
Saravanan P, Sreenivasa B, Selvan RT, Basagoudanavar SH, Hosamani M, Reddy ND, Nathanielsz J, Derozier C, Venkataramanan R. Protective immune response to liposome adjuvanted high potency foot-and-mouth disease vaccine in Indian cattle. Vaccine 2015; 33:670-7. [DOI: 10.1016/j.vaccine.2014.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 01/26/2023]
|
291
|
Dhodapkar MV, Sznol M, Zhao B, Wang D, Carvajal RD, Keohan ML, Chuang E, Sanborn RE, Lutzky J, Powderly J, Kluger H, Tejwani S, Green J, Ramakrishna V, Crocker A, Vitale L, Yellin M, Davis T, Keler T. Induction of antigen-specific immunity with a vaccine targeting NY-ESO-1 to the dendritic cell receptor DEC-205. Sci Transl Med 2014; 6:232ra51. [PMID: 24739759 DOI: 10.1126/scitranslmed.3008068] [Citation(s) in RCA: 298] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immune-based therapies for cancer are generating substantial interest because of the success of immune checkpoint inhibitors. This study aimed to enhance anticancer immunity by exploiting the capacity of dendritic cells (DCs) to initiate T cell immunity by efficient uptake and presentation of endocytosed material. Delivery of tumor-associated antigens to DCs using receptor-specific monoclonal antibodies (mAbs) in the presence of DC-activating agents elicits robust antigen-specific immune responses in preclinical models. DEC-205 (CD205), a molecule expressed on DCs, has been extensively studied for its role in antigen processing and presentation. CDX-1401 is a vaccine composed of a human mAb specific for DEC-205 fused to the full-length tumor antigen NY-ESO-1. This phase 1 trial assessed the safety, immunogenicity, and clinical activity of escalating doses of CDX-1401 with the Toll-like receptor (TLR) agonists resiquimod (TLR7/8) and Hiltonol (poly-ICLC, TLR3) in 45 patients with advanced malignancies refractory to available therapies. Treatment induced humoral and cellular immunity to NY-ESO-1 in patients with confirmed NY-ESO-1-expressing tumors across various dose levels and adjuvant combinations. No dose-limiting or grade 3 toxicities were reported. Thirteen patients experienced stabilization of disease, with a median duration of 6.7 months (range, 2.4+ to 13.4 months). Two patients had tumor regression (~20% shrinkage in target lesions). Six of eight patients who received immune-checkpoint inhibitors within 3 months after CDX-1401 administration had objective tumor regression. This first-in-human study of a protein vaccine targeting DCs demonstrates its feasibility, safety, and biological activity and provides rationale for combination immunotherapy strategies including immune checkpoint blockade.
Collapse
|
292
|
Desbien AL, Reed SJ, Bailor HR, Dubois Cauwelaert N, Laurance JD, Orr MT, Fox CB, Carter D, Reed SG, Duthie MS. Squalene emulsion potentiates the adjuvant activity of the TLR4 agonist, GLA, via inflammatory caspases, IL-18, and IFN-γ. Eur J Immunol 2014; 45:407-17. [PMID: 25367751 DOI: 10.1002/eji.201444543] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 10/03/2014] [Accepted: 10/30/2014] [Indexed: 12/26/2022]
Abstract
The synthetic TLR4 agonist glucopyranosyl lipid adjuvant (GLA) is a potent Th1-response-inducing adjuvant when formulated in a squalene oil-in-water emulsion (SE). While the innate signals triggered by TLR4 engagement are well studied, the contribution of SE remains unclear. To better understand the effect of SE on the adjuvant properties of GLA-SE, we compared the innate and adaptive immune responses elicited by immunization with different formulations: GLA without oil, SE alone or the combination, GLA-SE, in mice. Within the innate response to adjuvants, only GLA-SE displayed features of inflammasome activation, evidenced by early IL-18 secretion and IFN-γ production in memory CD8(+) T cells and neutrophils. Such early IFN-γ production was ablated in caspase-1/11(-/-) mice and in IL-18R1(-/-) mice. Furthermore, caspase-1/11 and IL-18 were also required for full Th1 CD4(+) T-cell induction via GLA-SE. Thus, we demonstrate that IL-18 and caspase-1/11 are components of the response to immunization with the TLR4 agonist/squalene oil-in-water based adjuvant, GLA-SE, providing implications for other adjuvants that combine oils with TLR agonists.
Collapse
|
293
|
Tomasello E, Pollet E, Vu Manh TP, Uzé G, Dalod M. Harnessing Mechanistic Knowledge on Beneficial Versus Deleterious IFN-I Effects to Design Innovative Immunotherapies Targeting Cytokine Activity to Specific Cell Types. Front Immunol 2014; 5:526. [PMID: 25400632 PMCID: PMC4214202 DOI: 10.3389/fimmu.2014.00526] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/07/2014] [Indexed: 12/15/2022] Open
Abstract
Type I interferons (IFN-I) were identified over 50 years ago as cytokines critical for host defense against viral infections. IFN-I promote anti-viral defense through two main mechanisms. First, IFN-I directly reinforce or induce de novo in potentially all cells the expression of effector molecules of intrinsic anti-viral immunity. Second, IFN-I orchestrate innate and adaptive anti-viral immunity. However, IFN-I responses can be deleterious for the host in a number of circumstances, including secondary bacterial or fungal infections, several autoimmune diseases, and, paradoxically, certain chronic viral infections. We will review the proposed nature of protective versus deleterious IFN-I responses in selected diseases. Emphasis will be put on the potentially deleterious functions of IFN-I in human immunodeficiency virus type 1 (HIV-1) infection, and on the respective roles of IFN-I and IFN-III in promoting resolution of hepatitis C virus (HCV) infection. We will then discuss how the balance between beneficial versus deleterious IFN-I responses is modulated by several key parameters including (i) the subtypes and dose of IFN-I produced, (ii) the cell types affected by IFN-I, and (iii) the source and timing of IFN-I production. Finally, we will speculate how integration of this knowledge combined with advanced biochemical manipulation of the activity of the cytokines should allow designing innovative immunotherapeutic treatments in patients. Specifically, we will discuss how induction or blockade of specific IFN-I responses in targeted cell types could promote the beneficial functions of IFN-I and/or dampen their deleterious effects, in a manner adapted to each disease.
Collapse
Affiliation(s)
- Elena Tomasello
- UM2, Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University , Marseille , France ; U1104, Institut National de la Santé et de la Recherche Médicale (INSERM) , Marseille , France ; UMR7280, Centre National de la Recherche Scientifique (CNRS) , Marseille , France
| | - Emeline Pollet
- UM2, Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University , Marseille , France ; U1104, Institut National de la Santé et de la Recherche Médicale (INSERM) , Marseille , France ; UMR7280, Centre National de la Recherche Scientifique (CNRS) , Marseille , France
| | - Thien-Phong Vu Manh
- UM2, Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University , Marseille , France ; U1104, Institut National de la Santé et de la Recherche Médicale (INSERM) , Marseille , France ; UMR7280, Centre National de la Recherche Scientifique (CNRS) , Marseille , France
| | - Gilles Uzé
- UMR 5235, Centre National de la Recherche Scientifique (CNRS), University Montpellier II , Montpellier , France
| | - Marc Dalod
- UM2, Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University , Marseille , France ; U1104, Institut National de la Santé et de la Recherche Médicale (INSERM) , Marseille , France ; UMR7280, Centre National de la Recherche Scientifique (CNRS) , Marseille , France
| |
Collapse
|
294
|
He LZ, Weidlick J, Sisson C, Marsh HC, Keler T. Toll-like receptor agonists shape the immune responses to a mannose receptor-targeted cancer vaccine. Cell Mol Immunol 2014; 12:719-28. [PMID: 25345808 PMCID: PMC4716615 DOI: 10.1038/cmi.2014.100] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 08/26/2014] [Accepted: 09/15/2014] [Indexed: 12/17/2022] Open
Abstract
Previous studies have documented that selective delivery of protein antigens to cells expressing mannose receptor (MR) can lead to enhanced immune responses. We postulated that agents that influenced the MR expression level, and the activation and migration status of MR-expressing antigen presenting cells, would modulate immune responses to MR-targeted vaccines. To address this question, we investigated the effect of clinically used adjuvants in human MR transgenic (hMR-Tg) mice immunized with an MR-targeting cancer vaccine composed of the human anti-MR monoclonal antibody B11 fused with the oncofetal protein, human chorionic gonadotropin beta chain (hCGβ), and referred to as B11-hCGβ. We found that humoral responses to low doses of B11-hCGβ could be enhanced by prior administration of GM-CSF, which upregulated MR expression in vivo. However, co-administration of the Toll-like receptor (TLR) agonists, poly-ICLC and/or CpG with B11-hCGβ was required to elicit Th1 immunity, as measured by antigen-specific T-cell production of IFN-γ. The TLR agonists were shown to increase the number of vaccine-containing cells in the draining lymph nodes of immunized hMR-Tg mice. In particular, with B11-hCGβ and poly-ICLC, a dramatic increase in vaccine-positive cells was observed in the T-cell areas of the lymph nodes, compared to the vaccine alone or combined with GM-CSF. Importantly, the absence of the TLR agonists during the priming immunization led to antigen-specific tolerance. Therefore, this study provides insight into the mechanisms by which adjuvants can augment immune responses to B11-hCGβ and have implications for the rationale design of clinical studies combining MR-targeted vaccination with TLR agonists.
Collapse
Affiliation(s)
- Li-Zhen He
- Celldex Therapeutics, Inc., Hampton, NJ, USA
| | | | | | | | - Tibor Keler
- Celldex Therapeutics, Inc., Hampton, NJ, USA
| |
Collapse
|
295
|
Furlan SN, Mandraju R, Brewer T, Roybal K, Troutman TD, Hu W, Palm NW, Unni A, Pasare C. Enhancement of anti-tumor CD8 immunity by IgG1-mediated targeting of Fc receptors. MAbs 2014; 6:108-18. [PMID: 24284965 DOI: 10.4161/mabs.27052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dendritic cells (DCs) function as professional antigen presenting cells and are critical for linking innate immune responses to the induction of adaptive immunity. Many current cancer DC vaccine strategies rely on differentiating DCs, feeding them tumor antigens ex vivo, and infusing them into patients. Importantly, this strategy relies on prior knowledge of suitable “tumor-specific” antigens to prime an effective anti-tumor response. DCs express a variety of receptors specific for the Fc region of immunoglobulins, and antigen uptake via Fc receptors is highly efficient and facilitates antigen presentation to T cells. Therefore, we hypothesized that expression of the mouse IgG1 Fc region on the surface of tumors would enhance tumor cell uptake by DCs and other myeloid cells and promote the induction of anti-tumor T cell responses. To test this, we engineered a murine lymphoma cell line expressing surface IgG1 Fc and discovered that such tumor cells were taken up rapidly by DCs, leading to enhanced cross-presentation of tumor-derived antigen to CD8+ T cells. IgG1-Fc tumors failed to grow in vivo and prophylactic vaccination of mice with IgG1-Fc tumors resulted in rejection of unmanipulated tumor cells. Furthermore, IgG1-Fc tumor cells were able to slow the growth of an unmanipulated primary tumor when used as a therapeutic tumor vaccine. Our data demonstrate that engagement of Fc receptors by tumors expressing the Fc region of IgG1 is a viable strategy to induce efficient and protective anti-tumor CD8+ T cell responses without prior knowledge of tumor-specific antigens.
Collapse
|
296
|
Kasamatsu J, Azuma M, Oshiumi H, Morioka Y, Okabe M, Ebihara T, Matsumoto M, Seya T. INAM plays a critical role in IFN-γ production by NK cells interacting with polyinosinic-polycytidylic acid-stimulated accessory cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:5199-207. [PMID: 25320282 DOI: 10.4049/jimmunol.1400924] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Polyinosinic-polycytidylic acid strongly promotes the antitumor activity of NK cells via TLR3/Toll/IL-1R domain-containing adaptor molecule 1 and melanoma differentiation-associated protein-5/mitochondrial antiviral signaling protein pathways. Polyinosinic-polycytidylic acid acts on accessory cells such as dendritic cells (DCs) and macrophages (Mφs) to secondarily activate NK cells. In a previous study in this context, we identified a novel NK-activating molecule, named IFN regulatory factor 3-dependent NK-activating molecule (INAM), a tetraspanin-like membrane glycoprotein (also called Fam26F). In the current study, we generated INAM-deficient mice and investigated the in vivo function of INAM. We found that cytotoxicity against NK cell-sensitive tumor cell lines was barely decreased in Inam(-/-) mice, whereas the number of IFN-γ-producing cells was markedly decreased in the early phase. Notably, deficiency of INAM in NK and accessory cells, such as CD8α(+) conventional DCs and Mφs, led to a robust decrease in IFN-γ production. In conformity with this phenotype, INAM effectively suppressed lung metastasis of B16F10 melanoma cells, which is controlled by NK1.1(+) cells and IFN-γ. These results suggest that INAM plays a critical role in NK-CD8α(+) conventional DC (and Mφ) interaction leading to IFN-γ production from NK cells in vivo. INAM could therefore be a novel target molecule for cancer immunotherapy against IFN-γ-suppressible metastasis.
Collapse
Affiliation(s)
- Jun Kasamatsu
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masahiro Azuma
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Hiroyuki Oshiumi
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Yuka Morioka
- Division of Disease Model Innovation, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masaru Okabe
- Research Institute for Microbial Disease, Osaka University, Osaka 565-0871, Japan
| | - Takashi Ebihara
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Misako Matsumoto
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Tsukasa Seya
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan;
| |
Collapse
|
297
|
Martins KAO, Bavari S, Salazar AM. Vaccine adjuvant uses of poly-IC and derivatives. Expert Rev Vaccines 2014; 14:447-59. [PMID: 25308798 DOI: 10.1586/14760584.2015.966085] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pathogen-associated molecular patterns (PAMPs) are stand-alone immunomodulators or 'danger signals,' that are increasingly recognized as critical components of many modern vaccines. Polyinosinic-polycytidylic acid (poly-IC) is a synthetic dsRNA that can activate multiple elements of the host defense in a pattern that parallels that of a viral infection. When properly combined with an antigen, it can be utilized as a PAMP-adjuvant, resulting in modulation and optimization of the antigen-specific immune response. We briefly review the preclinical and clinical uses of poly-IC and two poly-IC derivatives, poly-IC12U (Ampligen) and poly-ICLC (Hiltonol), as vaccine adjuvants.
Collapse
|
298
|
Ammi R, De Waele J, Willemen Y, Van Brussel I, Schrijvers DM, Lion E, Smits ELJ. Poly(I:C) as cancer vaccine adjuvant: knocking on the door of medical breakthroughs. Pharmacol Ther 2014; 146:120-31. [PMID: 25281915 DOI: 10.1016/j.pharmthera.2014.09.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 09/22/2014] [Indexed: 01/03/2023]
Abstract
Although cancer vaccination has yielded promising results in patients, the objective response rates are low. The right choice of adjuvant might improve the efficacy. Here, we review the biological rationale, as well as the preclinical and clinical results of polyinosinic:polycytidylic acid and its derivative poly-ICLC as cancer vaccine adjuvants. These synthetic immunological danger signals enhanced vaccine-induced anti-tumor immune responses and contributed to tumor elimination in animal tumor models and patients. Supported by these results, poly-ICLC-containing cancer vaccines are currently extensively studied in the ongoing trials, making it highly plausible that poly-ICLC will be part of the future approved cancer immunotherapies.
Collapse
Affiliation(s)
- Rachid Ammi
- Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium
| | - Jorrit De Waele
- Center for Oncological Research, University of Antwerp, B-2610 Antwerp, Belgium
| | - Yannick Willemen
- Tumor Immunology Group, Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2650 Edegem, Belgium
| | - Ilse Van Brussel
- Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium
| | - Dorien M Schrijvers
- Laboratory of Physiopharmacology, University of Antwerp, B-2610 Antwerp, Belgium
| | - Eva Lion
- Tumor Immunology Group, Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2650 Edegem, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, B-2650 Edegem, Belgium
| | - Evelien L J Smits
- Center for Oncological Research, University of Antwerp, B-2610 Antwerp, Belgium; Tumor Immunology Group, Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, B-2650 Edegem, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, B-2650 Edegem, Belgium.
| |
Collapse
|
299
|
Neeland MR, Elhay MJ, Meeusen ENT, de Veer MJ. Vaccination with liposomal poly(I:C) induces discordant maturation of migratory dendritic cell subsets and anti-viral gene signatures in afferent lymph cells. Vaccine 2014; 32:6183-92. [PMID: 25280435 DOI: 10.1016/j.vaccine.2014.09.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 09/18/2014] [Accepted: 09/19/2014] [Indexed: 01/14/2023]
Abstract
Vaccine formulations administered in the periphery must activate naive immune cells within the lymph node. In this study, we have directly cannulated the ovine lymphatic vessels to investigate the cellular and molecular mechanisms that transfer information from the periphery into the local draining lymph node via the afferent lymph. Inclusion of poly(I:C) into a liposomal vaccine formulation enhances the neutrophil-associated inflammatory immune response in afferent lymph and increases antigen uptake by migratory dendritic cells (DCs). Interestingly, antigen positive migratory DCs undergo discordant maturation, with peak expression of CD86 at 4 h and CD80 at 48-72 h post vaccination. Afferent lymph monocytes up-regulate expression of genes related to inflammatory and anti-viral immune phenotypes following vaccination however show no differentiation into APCs prior to their migration to the local lymph node as measured by surface MHC II expression. Finally, this study reveals the addition of poly(I:C) increases systemic antigen-specific humoral immunity. These findings provide a detailed understanding of the real time in vivo immune response induced by liposomes incorporating the innate immune agonist poly(I:C) utilising a vaccination setting comparable to that administered in humans.
Collapse
Affiliation(s)
- Melanie R Neeland
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Wellington Road Clayton VIC 3800, Australia
| | - Martin J Elhay
- Zoetis Research and Manufacturing Australia P/L, 45 Poplar Road, Parkville VIC 3052, Australia
| | - Els N T Meeusen
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Wellington Road Clayton VIC 3800, Australia
| | - Michael J de Veer
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Wellington Road Clayton VIC 3800, Australia.
| |
Collapse
|
300
|
|