401
|
Borriello F, Pietrasanta C, Lai JCY, Walsh LM, Sharma P, O'Driscoll DN, Ramirez J, Brightman S, Pugni L, Mosca F, Burkhart DJ, Dowling DJ, Levy O. Identification and Characterization of Stimulator of Interferon Genes As a Robust Adjuvant Target for Early Life Immunization. Front Immunol 2017; 8:1772. [PMID: 29312305 PMCID: PMC5732947 DOI: 10.3389/fimmu.2017.01772] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/27/2017] [Indexed: 11/16/2022] Open
Abstract
Immunization is key to preventing infectious diseases, a leading cause of death early in life. However, due to age-specific immunity, vaccines often demonstrate reduced efficacy in newborns and young infants as compared to adults. Here, we combined in vitro and in vivo approaches to identify adjuvant candidates for early life immunization. We employed newborn and adult bone marrow-derived dendritic cells (BMDCs) to perform a screening of pattern recognition receptor agonists and found that the stimulator of interferon genes ligand 2′3′-cGAMP (hereafter cGAMP) induces a comparable expression of surface maturation markers in newborn and adult BMDCs. Then, we utilized the trivalent recombinant hemagglutinin (rHA) influenza vaccine, Flublok, as a model antigen to investigate the role of cGAMP in adult and early life immunization. cGAMP adjuvantation alone could increase rHA-specific antibody titers in adult but not newborn mice. Remarkably, as compared to alum or cGAMP alone, immunization with cGAMP formulated with alum (Alhydrogel) enhanced newborn rHA-specific IgG2a/c titers ~400-fold, an antibody subclass associated with the development of IFNγ-driven type 1 immunity in vivo and endowed with higher effector functions, by 42 days of life. Highlighting the amenability for successful vaccine formulation and delivery, we next confirmed that cGAMP adsorbs onto alum in vitro. Accordingly, immunization early in life with (cGAMP+alum) promoted IFNγ production by CD4+ T cells and increased the proportions and absolute numbers of CD4+ CXCR5+ PD-1+ T follicular helper and germinal center (GC) GL-7+ CD138+ B cells, suggesting an enhancement of the GC reaction. Adjuvantation effects were apparently specific for IgG2a/c isotype switching without effect on antibody affinity maturation, as there was no effect on rHA-specific IgG avidity. Overall, our studies suggest that cGAMP when formulated with alum may represent an effective adjuvantation system to foster humoral and cellular aspects of type 1 immunity for early life immunization.
Collapse
Affiliation(s)
- Francesco Borriello
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Napoli, Italy.,WAO Center of Excellence, Naples, Italy
| | - Carlo Pietrasanta
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Jacqueline C Y Lai
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Lois M Walsh
- Biomedical & Pharmaceutical Science Skaggs School of Pharmacy, University of Montana, Missoula, MT, United States
| | - Pankaj Sharma
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - David N O'Driscoll
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Juan Ramirez
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Spencer Brightman
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Lorenza Pugni
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - David J Burkhart
- Biomedical & Pharmaceutical Science Skaggs School of Pharmacy, University of Montana, Missoula, MT, United States
| | - David J Dowling
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Ofer Levy
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Precision Vaccines Program, Divisions of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
402
|
Zhang WC, Du LJ, Zheng XJ, Chen XQ, Shi C, Chen BY, Sun XN, Li C, Zhang YY, Liu Y, Xiao H, Leng Q, Jiang X, Zhang Z, Sun S, Duan SZ. Elevated sodium chloride drives type I interferon signaling in macrophages and increases antiviral resistance. J Biol Chem 2017; 293:1030-1039. [PMID: 29203528 DOI: 10.1074/jbc.m117.805093] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 11/28/2017] [Indexed: 12/20/2022] Open
Abstract
Type I IFN production and signaling in macrophages play critical roles in innate immune responses. High salt (i.e. high concentrations of NaCl) has been proposed to be an important environmental factor that influences immune responses in multiple ways. However, it remains unknown whether high salt regulates type I IFN production and signaling in macrophages. Here, we demonstrated that high salt promoted IFNβ production and its signaling in both human and mouse macrophages, and consequentially primed macrophages for strengthened immune sensing and signaling when challenged with viruses or viral nucleic acid analogues. Using both pharmacological inhibitors and RNA interference we showed that these effects of high salt on IFNβ signaling were mediated by the p38 MAPK/ATF2/AP1 signaling pathway. Consistently, high salt increased resistance to vesicle stomatitis virus (VSV) infection in vitro. In vivo data indicated that a high-salt diet protected mice from lethal VSV infection. Taken together, these results identify high salt as a crucial regulator of type I IFN production and signaling, shedding important new light on the regulation of innate immune responses.
Collapse
Affiliation(s)
- Wu-Chang Zhang
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | - Lin-Juan Du
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011.,the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031
| | - Xiao-Jun Zheng
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011.,the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031
| | - Xiao-Qing Chen
- the Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, and
| | - Chaoji Shi
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | - Bo-Yan Chen
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | - Xue-Nan Sun
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011.,the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031
| | - Chao Li
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011.,the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031
| | - Yu-Yao Zhang
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011.,the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai 200031
| | - Yan Liu
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | - Hui Xiao
- the Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qibin Leng
- the Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinquan Jiang
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | - Zhiyuan Zhang
- From the Ninth People's Hospital, School of Stomatology and.,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | - Shuyang Sun
- From the Ninth People's Hospital, School of Stomatology and .,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| | - Sheng-Zhong Duan
- From the Ninth People's Hospital, School of Stomatology and .,the Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011
| |
Collapse
|
403
|
Abbassi-Daloii T, Yousefi S, Sekhavati MH, Tahmoorespur M. Impact of heat shock protein 60KD in combination with outer membrane proteins on immune response against Brucella melitensis. APMIS 2017; 126:65-75. [PMID: 29154438 DOI: 10.1111/apm.12778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/05/2017] [Indexed: 01/05/2023]
Abstract
Brucellosis caused by the bacterium Brucella affects various domestic and wild species. The outer membrane proteins 25 and 31 play key roles on stimulation of cell-mediated immune response against Brucella. GroEL as one of the major Brucella antigens stimulates the immune system and increases intracellular survival of bacteria. In the present study, we assumed injection of GroEL in combination with OMP25 and OMP31 would offer higher immunity levels. So, the impact of GroEL with different concentrations of recombinant outer membrane proteins emulsified in Chitosan Nanoparticles on immune responses was evaluated in mice model. Results showed both univalent (except rGroEL) and divalent immunized groups induced higher IFN-γ, TNF-α, and IL-4 titers in comparison to negative control groups. While GroEL showed negative effect on TNF-α titer, there were positive increase trends in IFN-γ in some treatments. Analysis of humoral antibody response revealed both univalent and divalent immunized groups induced higher IgG2a titer than IgG1 titer, indicating strong bent of Th1 immune response. Also, results showed GroEL can have positive impact on lymphocyte proliferation response. Overall, mice immunization using individual OMP25 or OMP31 demonstrated more effective cell-mediated immunity, although some combinations of rGroEL and rOMP31 vaccines were more efficient than other divalent ones.
Collapse
Affiliation(s)
- Tooba Abbassi-Daloii
- Department of Animal Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Human Genetics, Leiden University of Medical Center, Leiden, The Netherlands
| | - Soheil Yousefi
- Department of Animal Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | |
Collapse
|
404
|
cGAS/STING Pathway in Cancer: Jekyll and Hyde Story of Cancer Immune Response. Int J Mol Sci 2017; 18:ijms18112456. [PMID: 29156566 PMCID: PMC5713423 DOI: 10.3390/ijms18112456] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/11/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022] Open
Abstract
The last two decades have witnessed enormous growth in the field of cancer immunity. Mechanistic insights of cancer immunoediting have not only enhanced our understanding but also paved the way to target and/or harness the innate immune system to combat cancer, called cancer immunotherapy. Cyclic GMP-AMP synthase (cGAS)/Stimulator of interferon genes(STING) pathway has recently emerged as nodal player in cancer immunity and is currently being explored as potential therapeutic target. Although therapeutic activation of this pathway has shown promising anti-tumor effects in vivo, evidence also indicates the role of this pathway in inflammation mediated carcinogenesis. This review highlights our current understanding of cGAS/STING pathway in cancer, its therapeutic targeting and potential alternate approaches to target this pathway. Optimal therapeutic targeting and artificial tunability of this pathway still demand in depth understanding of cGAS/STING pathway regulation and homeostasis.
Collapse
|
405
|
Castro F, Pinto ML, Silva AM, Pereira CL, Teixeira GQ, Gomez-Lazaro M, Santos SG, Barbosa MA, Gonçalves RM, Oliveira MJ. Pro-inflammatory chitosan/poly(γ-glutamic acid) nanoparticles modulate human antigen-presenting cells phenotype and revert their pro-invasive capacity. Acta Biomater 2017; 63:96-109. [PMID: 28919508 DOI: 10.1016/j.actbio.2017.09.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
Anticancer immune responses depend on efficient presentation of tumor antigens and co-stimulatory signals provided by antigen-presenting cells (APCs). However, it is described that immature dendritic cells (DCs) and macrophages at the tumor site may have an immunosuppressive profile, which limits the activity of effector T cells and supports tumor progression. Therapeutic targeting of these innate immune cells, either aiming at their elimination or re-polarization towards an immunostimulatory profile, has been pointed as an attractive approach to control tumor progression. In the present work, we assessed the potential of Chitosan (Ch)/Poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) to modulate macrophages and DCs inflammatory profile and to impair their ability to promote cancer cell invasion. Interestingly, Ch/γ-PGA NPs, prepared by co-acervation method, induced an immunostimulatory DCs phenotype, enhancing the expression of the co-stimulatory molecules CD86, CD40 and HLA-DR, and the secretion of the pro-inflammatory cytokines TNF-α, IL-12p40 and IL-6. Furthermore, Ch/γ-PGA NPs re-educated IL-10-stimulated macrophages towards a pro-inflammatory profile, decreasing the expression of CD163 and promoting the secretion of IL-12p40 and TNF-α. These alterations in the immune cells phenotype promoted CD4+ and CD8+ T cell activation/proliferation and partially inhibited APCs' ability to induce colorectal cancer cell invasion. Overall, our findings open new perspectives on the use of Ch/γ-PGA NPs as an immunomodulatory therapy for antigen-presenting cells reprogramming, providing a new tool for anticancer therapies. STATEMENT OF SIGNIFICANCE The immune system is responsible to detect and destroy abnormal cells preventing the development of cancer. However, the immunosuppressive tumor microenvironment can compromise the immune response favoring tumor progression. Thus, immune system modulation towards an immunostimulatory profile can improve anticancer therapies. This research focus on the development of chitosan/poly(γ-glutamic acid) nanoparticles (NPs) to modulate human antigen-presenting cells (APCs) phenotype and to counteract their pro-invasive capacity. Interestingly, Ch/γ-PGA NPs had a prominent effect in inducing macrophages and dendritic cells immunostimulatory phenotype, thus favoring T cell proliferation and inhibiting colorectal cancer cell invasion. We propose that their combination with other immunomodulatory drugs or conventional anticancer therapies can improve patients' outcome.
Collapse
|
406
|
West AP. Mitochondrial dysfunction as a trigger of innate immune responses and inflammation. Toxicology 2017; 391:54-63. [DOI: 10.1016/j.tox.2017.07.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/22/2017] [Accepted: 07/24/2017] [Indexed: 12/19/2022]
|
407
|
Rongvaux A. Innate immunity and tolerance toward mitochondria. Mitochondrion 2017; 41:14-20. [PMID: 29054471 DOI: 10.1016/j.mito.2017.10.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 01/19/2023]
Abstract
Mitochondria are intracellular organelles that originate from a bacterial symbiont, and they retain multiple features of this bacterial ancestry. The immune system evolved to detect the presence of invading pathogens, including bacteria, to eliminate them by a diversity of antimicrobial mechanisms and to mount long-term protective immunity. Due to their bacterial ancestry, mitochondria are sensed by the innate immune system, and trigger inflammatory responses comparable to those induced by pathogenic bacteria. In both cases, innate sensing mechanisms involve Toll-Like Receptors, Formyl Peptide Receptors, inflammasomes or the cGAS/STING pathway. Stressed mitochondria release mitochondrial molecules, such as cardiolipin and mitochondrial DNA, which are sensed as cellular damage potentially caused by infections. Recent research has identified several conditions in which mitochondrial stress-induced immunity is essential to effective antimicrobial defenses. But, in pathological conditions, the abnormal activation of the innate immune system by damaged mitochondria results in auto-inflammatory or autoimmune diseases. To prevent undesirable mitochondria-targeted responses, immune tolerance toward mitochondria must be established, involving regulation of mitophagy and mitochondrial permeability, as well as activation of specific nucleases and pro-apoptotic caspases. Overall, recent findings identify mitochondria as central in the induction of innate immunity, and provide new insights as to how immune responses to these multi-functional organelles might be exploited therapeutically in various disease states.
Collapse
Affiliation(s)
- Anthony Rongvaux
- Fred Hutchinson Cancer Research Center, Program in Immunology, Clinical Research Division, Seattle, WA 98109, United States; University of Washington School of Medicine, Department of Immunology, Seattle, WA 98109, United States.
| |
Collapse
|
408
|
Cupino TL, Watson BA, Cupino AC, Oda K, Ghamsary MG, Soriano S, Kirsch WM. Stability and bioactivity of chitosan as a transfection agent in primary human cell cultures: A case for chitosan-only controls. Carbohydr Polym 2017; 180:376-384. [PMID: 29103517 DOI: 10.1016/j.carbpol.2017.10.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/21/2017] [Accepted: 10/04/2017] [Indexed: 01/04/2023]
Abstract
Chitosan polymers (Cs), from which microparticles (CsM) may be precipitated to deliver various intracellular payloads, are generally considered biologically inert. We examined the impact of cell culture conditions on CsM size and the effect of chitosan on CD59 expression in primary human smooth muscle cells. We found that particle concentration and incubation time in biological buffers augmented particle size. Between pH 7.0 and pH 7.5, CsM size increased abruptly. We utilized CsM containing a plasmid with a gene for CD59 (pCsM) to transfect cells. Both CD59 mRNA and the number of CD59-positive cells were increased after pCsM treatment. Unexpectedly, CsM also augmented the number of CD59-positive cells. Cs alone enhanced CD59 expression more potently than either pCSM or CsM. This observation strongly suggests that chitosan is in fact bioactive and that chitosan-only controls should be included to avoid misattributing the activity of the delivery agent with that of the payload.
Collapse
Affiliation(s)
- Tanya L Cupino
- Neurosurgery Center for Research, Training and Education, Loma Linda University School of Medicine, Loma Linda, CA, United States; Division of Microbiology and Molecular Genetics, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States.
| | - Billy A Watson
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States; Division of Human Anatomy, Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Alan C Cupino
- Department of Epidemiology and Biostatistics, Loma Linda University School of Public Health, Loma Linda, CA, United States
| | - Keiji Oda
- Department of Epidemiology and Biostatistics, Loma Linda University School of Public Health, Loma Linda, CA, United States
| | - Mark G Ghamsary
- Department of Epidemiology and Biostatistics, Loma Linda University School of Public Health, Loma Linda, CA, United States (Retired)
| | - Salvador Soriano
- Division of Human Anatomy, Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Wolff M Kirsch
- Neurosurgery Center for Research, Training and Education, Loma Linda University School of Medicine, Loma Linda, CA, United States.
| |
Collapse
|
409
|
Chitosan immunomodulatory properties: perspectives on the impact of structural properties and dosage. Future Sci OA 2017; 4:FSO225. [PMID: 29255618 PMCID: PMC5729595 DOI: 10.4155/fsoa-2017-0064] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 11/17/2022] Open
|
410
|
Lundahl MLE, Scanlan EM, Lavelle EC. Therapeutic potential of carbohydrates as regulators of macrophage activation. Biochem Pharmacol 2017; 146:23-41. [PMID: 28893617 DOI: 10.1016/j.bcp.2017.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/06/2017] [Indexed: 02/06/2023]
Abstract
It is well established for a broad range of disease states, including cancer and Mycobacterium tuberculosis infection, that pathogenesis is bolstered by polarisation of macrophages towards an anti-inflammatory phenotype, known as M2. As these innate immune cells are relatively long-lived, their re-polarisation to pro-inflammatory, phagocytic and bactericidal "classically activated" M1 macrophages is an attractive therapeutic approach. On the other hand, there are scenarios where the resolving inflammation, wound healing and tissue remodelling properties of M2 macrophages are beneficial - for example the successful introduction of biomedical implants. Although there are numerous endogenous and exogenous factors that have an impact on the macrophage polarisation spectrum, this review will focus specifically on prominent macrophage-modulating carbohydrate motifs with a view towards highlighting structure-function relationships and therapeutic potential.
Collapse
Affiliation(s)
- Mimmi L E Lundahl
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland; School of Chemistry and Trinity Biomedical Sciences Institute, Trinity College, Pearse St, Dublin 2, Ireland
| | - Eoin M Scanlan
- School of Chemistry and Trinity Biomedical Sciences Institute, Trinity College, Pearse St, Dublin 2, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin 2, Ireland.
| |
Collapse
|
411
|
Jin Z, Li D, Dai C, Cheng G, Wang X, Zhao K. Response of live Newcastle disease virus encapsulated in N -2-hydroxypropyl dimethylethyl ammonium chloride chitosan nanoparticles. Carbohydr Polym 2017; 171:267-280. [DOI: 10.1016/j.carbpol.2017.05.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/18/2017] [Accepted: 05/05/2017] [Indexed: 12/16/2022]
|
412
|
Chauhan N, Banerjee P, Khatri VK, Canciamille A, Gilles J, Kalyanasundaram R. Improving the efficacy of a prophylactic vaccine formulation against lymphatic filariasis. Parasitol Res 2017; 116:2821-2830. [PMID: 28828575 DOI: 10.1007/s00436-017-5593-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/10/2017] [Indexed: 12/15/2022]
Abstract
Mass drug administration (MDA) is the current strategy for interrupting the transmission of lymphatic filariasis (LF) infection and control of the disease in endemic areas. However, subject non-compliance has resulted in the presence of several "transmission hotspots" in the endemic regions threatening the reemergence of LF. This situation is further complicated by the fact that the drugs used in MDA are not effective against adult LF worms, a major concern for the control strategy. Thus, there is clearly a need for an effective and sustainable approach to control LF. Prophylactic vaccine combined with targeted treatment of infected patients and vector control is suggested as a more sustainable strategy to eliminate LF infection from endemic regions. A multivalent vaccine (rBmHAT) developed in our laboratory conferred about 90% protection in rodents. However, when we tested the rBmHAT vaccine along with alum in rhesus macaques, only about 40% protection was achieved and the immune response obtained was Th2 biased. In an attempt to improve the vaccine, in this study, we tested two vaccine antigens (rBmHAT and rBmHAX) along with two adjuvant formulations [alum + GLA (AL019) and mannosylated chitosan (MCA)] in a mouse model. Our results show that rBmHAT is a better vaccine antigen than rBmHAX. Combination of rBmHAT with AL019 or MCA adjuvants gave 94 and 88% protection, respectively, against challenge infections. Immunized animals developed antigen-specific memory T cells that secreted significant levels of IL-4, IFN-γ, and IL-17 suggesting the generation of a balanced Th1/Th2 responses following immunization. A major advantage of MCA adjuvant is that the vaccine booster doses can be administered orally. These studies thus showed that rBmHAT is a better vaccine antigen and can be given in combination with AL019 or MCA adjuvant to obtain excellent results.
Collapse
Affiliation(s)
- Nikhil Chauhan
- Department of Biomedical Sciences, University of Illinois College of Medicine, 1601 Parkview Avenue, Rockford, IL, 61107, USA
| | - Priyankana Banerjee
- Department of Biomedical Sciences, University of Illinois College of Medicine, 1601 Parkview Avenue, Rockford, IL, 61107, USA
| | - Vishal K Khatri
- Department of Biomedical Sciences, University of Illinois College of Medicine, 1601 Parkview Avenue, Rockford, IL, 61107, USA
| | - Andrew Canciamille
- Department of Biomedical Sciences, University of Illinois College of Medicine, 1601 Parkview Avenue, Rockford, IL, 61107, USA
| | - Jessica Gilles
- Department of Biomedical Sciences, University of Illinois College of Medicine, 1601 Parkview Avenue, Rockford, IL, 61107, USA
| | - Ramaswamy Kalyanasundaram
- Department of Biomedical Sciences, University of Illinois College of Medicine, 1601 Parkview Avenue, Rockford, IL, 61107, USA.
| |
Collapse
|
413
|
Ng KW, Marshall EA, Bell JC, Lam WL. cGAS-STING and Cancer: Dichotomous Roles in Tumor Immunity and Development. Trends Immunol 2017; 39:44-54. [PMID: 28830732 DOI: 10.1016/j.it.2017.07.013] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023]
Abstract
cGMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) sensing has emerged as a key regulator of innate immune responses to both exogenous and endogenous DNA. Recent studies reveal critical roles for this pathway in natural antitumor immunity across cancer types as well as in immune checkpoint blockade therapy. However, it is also clear that some tumors evade cGAS-STING-mediated immune responses, and immunomodulatory therapeutics are currently being explored to target this pathway. Finally, we also discuss recent observations that cGAS-STING-mediated inflammation may promote tumor initiation, growth, and metastasis in certain malignancies and how this may complicate the utility of this pathway in therapeutic development.
Collapse
Affiliation(s)
- Kevin W Ng
- Department of Integrative Oncology, BC Cancer Agency, Vancouver, Canada; These authors contributed equally to this work
| | - Erin A Marshall
- Department of Integrative Oncology, BC Cancer Agency, Vancouver, Canada; These authors contributed equally to this work
| | - John C Bell
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Wan L Lam
- Department of Integrative Oncology, BC Cancer Agency, Vancouver, Canada.
| |
Collapse
|
414
|
Erman A, Hergouth VK, Blango MG, Kos MK, Mulvey MA, Veranic P. Repeated Treatments with Chitosan in Combination with Antibiotics Completely Eradicate Uropathogenic Escherichia coli From Infected Mouse Urinary Bladders. J Infect Dis 2017; 216:375-381. [PMID: 28119486 PMCID: PMC5853829 DOI: 10.1093/infdis/jix023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/10/2017] [Indexed: 01/09/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC), the primary causative agents of urinary tract infections, colonize and invade the epithelial cells of the bladder urothelium. Infection of immature urothelial cells can result in the formation of persistent intracellular reservoirs that are refractory to antibiotic treatments. Previously, we defined a novel therapeutic strategy that used the bladder cell exfoliant chitosan to deplete UPEC reservoirs. However, although a single treatment of chitosan followed by ciprofloxacin administration had a marked effect on reducing UPEC titers within the bladder, this treatment failed to prevent relapsing bacteriuria. We show here that repeated use of chitosan in conjunction with the antibiotic ciprofloxacin completely eradicates UPEC from the urinary tract and prevents the development of relapsing bouts of bacteriuria. In addition, microscopy revealed rapid restoration of bladder integrity following chitosan treatment, indicating that chitosan can be used to effectively combat recalcitrant bladder infections without causing lasting harm to the urothelium.
Collapse
Affiliation(s)
- Andreja Erman
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Veronika Križan Hergouth
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Matthew G Blango
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City
| | | | - Matthew A Mulvey
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City
| | - Peter Veranic
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Slovenia
| |
Collapse
|
415
|
Abdel-Latif M, El-Shahawi G, Aboelhadid SM, Abdel-Tawab H. Immunoprotective Effect of Chitosan Particles onHymenolepis nana- Infected Mice. Scand J Immunol 2017; 86:83-90. [DOI: 10.1111/sji.12568] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/08/2017] [Indexed: 01/04/2023]
Affiliation(s)
- M. Abdel-Latif
- Department of Zoology; Faculty of Science; Beni-Suef University; Beni-Suef Egypt
| | - G. El-Shahawi
- Department of Zoology; Faculty of Science; Beni-Suef University; Beni-Suef Egypt
| | - S. M. Aboelhadid
- Department of Parasitology; Faculty of Veterinary Medicine; Beni-Suef University; Beni-Suef Egypt
| | - H. Abdel-Tawab
- Department of Zoology; Faculty of Science; Beni-Suef University; Beni-Suef Egypt
| |
Collapse
|
416
|
Anti-cancer efficacy of biotinylated chitosan nanoparticles in liver cancer. Oncotarget 2017; 8:59068-59085. [PMID: 28938619 PMCID: PMC5601715 DOI: 10.18632/oncotarget.19146] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 06/02/2017] [Indexed: 01/13/2023] Open
Abstract
The present study investigated the synthesis of biotinylated chitosan (Bio-CS) from chitosan using a nanomaterial skeleton with biotin and the successful targeting of the formulation in liver cancer cells. Bio-CS was validated by fourier transformed infrared spectroscopy and hydrogen-1 nuclear magnetic resonance spectroscopy. Bio-CS and plasmid DNA were used to construct Bio-CS/plasmid DNA nanoparticles according to the optimal molar ratio of 1:1 and the optimal pH-value of 5.5. Under these conditions, the parameters mean particle size, potential, encapsulation rate and drug loading, were 82.9 nm, +21.8 mV, 85.7% and 35.4%, respectively. Bio-CS exhibited an apparent liver cancer targeting effect in vitro and in vivo, as demonstrated by confocal laser scanning, green fluorescent protein transfection, and in vivo imaging assays. In addition, the Bio-CS/plasmid DNA nanoparticles significantly increased the survival period of the orthotropic liver cancer mouse model compared with the plasmid DNA, with no apparent side effects on the cells. Bio-CS nanomaterials stimulated an immune response in hepatoma cells via increased expression of GM-CSF, IL-21 and Rae-1 markers. The data suggest that Bio-CS increased the inhibition of liver cancer cell proliferation in vitro and the activation of the cellular immunity in vivo.
Collapse
|
417
|
Bernocchi B, Carpentier R, Betbeder D. Nasal nanovaccines. Int J Pharm 2017; 530:128-138. [PMID: 28698066 DOI: 10.1016/j.ijpharm.2017.07.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 01/08/2023]
Abstract
Nasal administration of vaccines is convenient for the potential stimulation of mucosal and systemic immune protection. Moreover the easy accessibility of the intranasal route renders it optimal for pandemic vaccination. Nanoparticles have been identified as ideal delivery systems and adjuvants for vaccine application. Heterogeneous protocols have been used for animal studies. This complicates the understanding of the formulation influence on the immune response and the comparison of the different nanoparticles approaches developed. Moreover anatomical and immunological differences between rodents and humans provide an additional hurdle in the rational development of nasal nanovaccines. This review will give a comprehensive expertise of the state of the art in nasal nanovaccines in animals and humans focusing on the nanomaterial used.
Collapse
Affiliation(s)
- B Bernocchi
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France
| | - R Carpentier
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France.
| | - D Betbeder
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France; University of Artois, 62000 Arras, France
| |
Collapse
|
418
|
Association of chitosan and aluminium as a new adjuvant strategy for improved vaccination. Int J Pharm 2017; 527:103-114. [DOI: 10.1016/j.ijpharm.2017.05.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/19/2022]
|
419
|
Luo M, Wang H, Wang Z, Cai H, Lu Z, Li Y, Du M, Huang G, Wang C, Chen X, Porembka MR, Lea J, Frankel AE, Fu YX, Chen ZJ, Gao J. A STING-activating nanovaccine for cancer immunotherapy. NATURE NANOTECHNOLOGY 2017; 12:648-654. [PMID: 28436963 PMCID: PMC5500418 DOI: 10.1038/nnano.2017.52] [Citation(s) in RCA: 637] [Impact Index Per Article: 79.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 03/03/2017] [Indexed: 05/17/2023]
Abstract
The generation of tumour-specific T cells is critically important for cancer immunotherapy. A major challenge in achieving a robust T-cell response is the spatiotemporal orchestration of antigen cross-presentation in antigen-presenting cells with innate stimulation. Here, we report a minimalist nanovaccine, comprising a simple physical mixture of an antigen and a synthetic polymeric nanoparticle, PC7A NP, which generates a strong cytotoxic T-cell response with low systemic cytokine expression. Mechanistically, the PC7A NP achieves efficient cytosolic delivery of tumour antigens to antigen-presenting cells in draining lymph nodes, leading to increased surface presentation while simultaneously activating type I interferon-stimulated genes. This effect is dependent on stimulator of interferon genes (STING), but not the Toll-like receptor or the mitochondrial antiviral-signalling protein (MAVS) pathway. The nanovaccine led to potent tumour growth inhibition in melanoma, colon cancer and human papilloma virus-E6/E7 tumour models. The combination of the PC7A nanovaccine and an anti-PD-1 antibody showed great synergy, with 100% survival over 60 days in a TC-1 tumour model. Rechallenging of these tumour-free animals with TC-1 cells led to complete inhibition of tumour growth, suggesting the generation of long-term antitumour memory. The STING-activating nanovaccine offers a simple, safe and robust strategy in boosting anti-tumour immunity for cancer immunotherapy.
Collapse
Affiliation(s)
- Min Luo
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Hua Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Zhaohui Wang
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Haocheng Cai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Zhigang Lu
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Yang Li
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Mingjian Du
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Gang Huang
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Chensu Wang
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Xiang Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Matthew R. Porembka
- Department of Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Jayanthi Lea
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Arthur E. Frankel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Zhijian J. Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
- Correspondence and requests for materials should be addressed to Z.J.C () or J.G. ()
| | - Jinming Gao
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
- Correspondence and requests for materials should be addressed to Z.J.C () or J.G. ()
| |
Collapse
|
420
|
Marinho FV, Benmerzoug S, Oliveira SC, Ryffel B, Quesniaux VFJ. The Emerging Roles of STING in Bacterial Infections. Trends Microbiol 2017. [PMID: 28625530 DOI: 10.1016/j.tim.2017.05.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The STING (Stimulator of Interferon Genes) protein connects microorganism cytosolic sensing with effector functions of the host cell by sensing directly cyclic dinucleotides (CDNs), originating from pathogens or from the host upon DNA recognition. Although STING activation favors effective immune responses against viral infections, its role during bacterial diseases is controversial, ranging from protective to detrimental effects for the host. In this review, we summarize important features of the STING activation pathway and recent highlights about the role of STING in bacterial infections by Chlamydia, Listeria, Francisella, Brucella, Shigella, Salmonella, Streptococcus, and Neisseria genera, with a special focus on mycobacteria.
Collapse
Affiliation(s)
- Fabio V Marinho
- CNRS, UMR7355, Orleans, France; Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sulayman Benmerzoug
- CNRS, UMR7355, Orleans, France; Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bernhard Ryffel
- CNRS, UMR7355, Orleans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orleans, France
| | - V F J Quesniaux
- CNRS, UMR7355, Orleans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orleans, France.
| |
Collapse
|
421
|
Lebre F, Sridharan R, Sawkins MJ, Kelly DJ, O'Brien FJ, Lavelle EC. The shape and size of hydroxyapatite particles dictate inflammatory responses following implantation. Sci Rep 2017; 7:2922. [PMID: 28592868 PMCID: PMC5462791 DOI: 10.1038/s41598-017-03086-0] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
The extent of regeneration following biomaterial implantation is dependent on the microenvironment surrounding the implant. Since implant composition can have a profound effect on inflammation, it is essential to understand this process as a non-resolving inflammatory response can lead to fibrous encapsulation and insufficient integration. Incorporation of particulates into implants confers structural and functional benefits, thus optimizing particulate characteristics to enhance immune mediated efficacy is important. We investigated the relationship between the nature of hydroxyapatite (HA) particles and the innate immune response, focusing on how particle size (0.1 µm, 5 µm, 20 µm, 100 µm) and morphology (needle-shaped/spherical; smooth/rough surface) modulates inflammatory responses. We observed a shape and size-dependent activation of the NLRP3 inflammasome and IL-1β secretion; while needle-shaped and smaller HA particles significantly enhanced cytokine secretion, larger particles did not. Moreover, HA particle characteristics profoundly influenced patterns of innate immune cell recruitment and cytokine production following injection. While small, needle-shaped particles induced a strong inflammatory response, this was not observed with smooth, spherical particles of comparable size or with larger particles. These findings indicate that hydroxyapatite particle characteristics dictate immune cell recruitment and the ensuing inflammatory response, providing an opportunity to tailor HA particle characteristics to regulate immune responses induced after biomaterial implantation.
Collapse
Affiliation(s)
- Filipa Lebre
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02 R590, Ireland.,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Dublin 2, D02 PN40, Ireland.,Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Rukmani Sridharan
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02 R590, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Michael J Sawkins
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02 R590, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02 R590, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Fergal J O'Brien
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02 R590, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02 R590, Ireland. .,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Dublin 2, D02 PN40, Ireland. .,Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland.
| |
Collapse
|
422
|
Potential of glucans as vaccine adjuvants: A review of the α-glucans case. Carbohydr Polym 2017; 165:103-114. [DOI: 10.1016/j.carbpol.2017.02.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 01/06/2023]
|
423
|
Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol 2017; 17:363-375. [PMID: 28393922 DOI: 10.1038/nri.2017.21] [Citation(s) in RCA: 720] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mitochondrial DNA (mtDNA) - which is well known for its role in oxidative phosphorylation and maternally inherited mitochondrial diseases - is increasingly recognized as an agonist of the innate immune system that influences antimicrobial responses and inflammatory pathology. On entering the cytoplasm, extracellular space or circulation, mtDNA can engage multiple pattern-recognition receptors in cell-type- and context-dependent manners to trigger pro-inflammatory and type I interferon responses. Here, we review the expanding research field of mtDNA in innate immune responses to highlight new mechanistic insights and discuss the physiological and pathological relevance of this exciting area of mitochondrial biology.
Collapse
|
424
|
|
425
|
Babu A, Ramesh R. Multifaceted Applications of Chitosan in Cancer Drug Delivery and Therapy. Mar Drugs 2017; 15:E96. [PMID: 28346381 PMCID: PMC5408242 DOI: 10.3390/md15040096] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 12/15/2022] Open
Abstract
Chitosan is a versatile polysaccharide of biological origin. Due to the biocompatible and biodegradable nature of chitosan, it is intensively utilized in biomedical applications in scaffold engineering as an absorption enhancer, and for bioactive and controlled drug release. In cancer therapy, chitosan has multifaceted applications, such as assisting in gene delivery and chemotherapeutic delivery, and as an immunoadjuvant for vaccines. The present review highlights the recent applications of chitosan and chitosan derivatives in cancer therapy.
Collapse
Affiliation(s)
- Anish Babu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
- Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
426
|
Martin TL, Jee J, Kim E, Steiner HE, Cormet-Boyaka E, Boyaka PN. Sublingual targeting of STING with 3'3'-cGAMP promotes systemic and mucosal immunity against anthrax toxins. Vaccine 2017; 35:2511-2519. [PMID: 28343781 DOI: 10.1016/j.vaccine.2017.02.064] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 02/06/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
Anthrax is caused by Bacillus anthracis, a zoonotic bacterial pathogen affecting humans and livestock worldwide. The current human anthrax vaccine, anthrax vaccine adsorbed (AVA), is an injected vaccine with a cumbersome administration schedule and fails to promote mucosal immunity. Bacterial enterotoxins, which stimulate production of the cyclic nucleotide cAMP are effective experimental mucosal vaccine adjuvants, but their inherent toxicity has precluded their use in humans. We investigated whether cyclic dinucleotides that target Stimulator of Interferon Gamma Genes (STING) in mammalian cells could represent an alternative to bacterial enterotoxins as adjuvant for sublingual immunization and promotion of mucosal immunity and secretory IgA responses in addition to systemic immunity. We found that sublingual immunization of mice with Bacillus anthracis protective antigen (PA) and the STING ligand 3'3'-cGAMP promotes PA-specific serum IgG Ab responses of the same magnitude as those induced after immunization with PA and the experimental adjuvant cholera toxin (CT). Interestingly, this STING ligand also promoted serum anti-PA IgA and IgA-producing cells in the bone marrow. Furthermore, the saliva of mice immunized with the STING ligand exhibited similar levels of PA-specific IgA Abs as groups immunized with CT as adjuvant. The adjuvant activity of 3'3'-cGAMP was associated with mixed Th1, Th2, and Th17 responses. This STING ligand also induced rapid IFN-β and IL-10 responses in sublingual tissues and cervical lymph nodes, and TGF-β responses in the cervical lymph nodes, which could contribute to promoting IgA responses after sublingual immunization.
Collapse
Affiliation(s)
- Tara L Martin
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Junbae Jee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Haley E Steiner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
427
|
Luo M, Samandi LZ, Wang Z, Chen ZJ, Gao J. Synthetic nanovaccines for immunotherapy. J Control Release 2017; 263:200-210. [PMID: 28336379 DOI: 10.1016/j.jconrel.2017.03.033] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/14/2017] [Accepted: 03/19/2017] [Indexed: 01/15/2023]
Abstract
Although vaccination is historically one of the most successful strategies for the prevention of infectious diseases, development of vaccines for cancer and many chronic infections, such as HIV, malaria, and tuberculosis, has remained a challenge. Strong and long-lasting antigen-specific T cell responses are critical for therapy of these diseases. A major challenge in achieving a robust CD8+ T cell response is the requirement of spatio-temporal orchestration of antigen cross-presentation in antigen-presenting cells with innate stimulation. Here, we discuss the development of nanoparticle vaccine (nanovaccine) that modulates the innate immune system and enhances adaptive immunity with reduced toxicity. We address how nanovaccines can integrate multiple functions, such as lymph node targeting, antigen presentation, and stimulation of innate immunity, to achieve a robust T cell response for immunotherapy.
Collapse
Affiliation(s)
- Min Luo
- Department of Pharmacology, Simmons Comprehensive Cancer Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Layla Z Samandi
- Department of Pharmacology, Simmons Comprehensive Cancer Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Zhaohui Wang
- Department of Pharmacology, Simmons Comprehensive Cancer Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Zhijian J Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA; Howard Hughes Medical Institute, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Jinming Gao
- Department of Pharmacology, Simmons Comprehensive Cancer Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
428
|
Fong D, Grégoire-Gélinas P, Cheng AP, Mezheritsky T, Lavertu M, Sato S, Hoemann CD. Lysosomal rupture induced by structurally distinct chitosans either promotes a type 1 IFN response or activates the inflammasome in macrophages. Biomaterials 2017; 129:127-138. [PMID: 28340358 DOI: 10.1016/j.biomaterials.2017.03.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/20/2017] [Accepted: 03/14/2017] [Indexed: 12/12/2022]
Abstract
Chitosan is a family of glucosamine and N-acetyl glucosamine polysaccharides with poorly understood immune modulating properties. Here, functional U937 macrophage responses were analyzed in response to a novel library of twenty chitosans with controlled degree of deacetylation (DDA, 60-98%), molecular weight (1 to >100 kDa), and acetylation pattern (block vs. random). Specific chitosan preparations (10 or 190 kDa 80% block DDA and 3, 5, or 10 kDa 98% DDA) either induced macrophages to release CXCL10 and IL-1ra at 5-50 μg/mL, or activated the inflammasome to release IL-1β and PGE2 at 50-150 μg/mL. Chitosan induction of these factors required lysosomal acidification. CXCL10 production was preceded by lysosomal rupture as shown by time-dependent co-localization of galectin-3 and chitosan and slowed autophagy flux, and specifically depended on IFN-β paracrine activity and STAT-2 activation that could be suppressed by PGE2. Chitosan induced a type I IFN paracrine response or inflammasome response depending on the extent of lysosomal rupture and cytosolic foreign body invasion. This study identifies the structural motifs that lead to chitosan-driven cytokine responses in macrophages and indicates that lysosomal rupture is a key mechanism that determines the endogenous release of either IL-1ra or IL-1β.
Collapse
Affiliation(s)
- David Fong
- Institute of Biomedical Engineering, École Polytechnique, Montreal, QC, H3T 1J4, Canada
| | | | - Alexandre P Cheng
- Department of Chemical Engineering, École Polytechnique, Montreal, QC, H3T 1J4, Canada
| | - Tal Mezheritsky
- Department of Chemical Engineering, École Polytechnique, Montreal, QC, H3T 1J4, Canada
| | - Marc Lavertu
- Department of Chemical Engineering, École Polytechnique, Montreal, QC, H3T 1J4, Canada
| | - Sachiko Sato
- Research Centre for Infectious Diseases, Faculty of Medicine, Laval University, Quebec, QC, G1V 4G2, Canada
| | - Caroline D Hoemann
- Institute of Biomedical Engineering, École Polytechnique, Montreal, QC, H3T 1J4, Canada; Department of Chemical Engineering, École Polytechnique, Montreal, QC, H3T 1J4, Canada; FRQ-S Biomedical Research Group/ Groupe de Recherche en Sciences et Technologies Biomédicales, École Polytechnique, Montreal, QC, H3T 1J4, Canada.
| |
Collapse
|
429
|
Impact of Type I Interferon on the Safety and Immunogenicity of an Experimental Live-Attenuated Herpes Simplex Virus 1 Vaccine in Mice. J Virol 2017; 91:JVI.02342-16. [PMID: 28122977 DOI: 10.1128/jvi.02342-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/17/2017] [Indexed: 12/12/2022] Open
Abstract
Viral fitness dictates virulence and capacity to evade host immune defenses. Understanding the biological underpinnings of such features is essential for rational vaccine development. We have previously shown that the live-attenuated herpes simplex virus 1 (HSV-1) mutant lacking the nuclear localization signal (NLS) on the ICP0 gene (0ΔNLS) is sensitive to inhibition by interferon beta (IFN-β) in vitro and functions as a highly efficacious experimental vaccine. Here, we characterize the host immune response and in vivo pathogenesis of HSV-1 0ΔNLS relative to its fully virulent parental strain in C57BL/6 mice. Additionally, we explore the role of type 1 interferon (IFN-α/β) signaling on virulence and immunogenicity of HSV-1 0ΔNLS and uncover a probable sex bias in the induction of IFN-α/β in the cornea during HSV-1 infection. Our data show that HSV-1 0ΔNLS lacks neurovirulence even in highly immunocompromised mice lacking the IFN-α/β receptor. These studies support the translational viability of the HSV-1 0ΔNLS vaccine strain by demonstrating that, while it is comparable to a virulent parental strain in terms of immunogenicity, HSV-1 0ΔNLS does not induce significant tissue pathology.IMPORTANCE HSV-1 is a common human pathogen associated with a variety of clinical presentations ranging in severity from periodic "cold sores" to lethal encephalitis. Despite the consistent failures of HSV subunit vaccines in clinical trials spanning the past 28 years, opposition to live-attenuated HSV vaccines predicated on unfounded safety concerns currently limits their widespread acceptance. Here, we demonstrate that a live-attenuated HSV-1 vaccine has great translational potential.
Collapse
|
430
|
Abstract
During viral and bacterial infections, pathogen-derived cytosolic nucleic acids are recognized by the intracellular RNA sensors retinoic acid-inducible gene I and melanoma-differentiated gene 5 and intracellular DNA sensors, including cyclic-di-GMP-AMP synthase, absent in melanoma 2, interferon (IFN)-gamma inducible protein 16, polymerase III, and so on. Binding of intracellular nucleic acids to these sensors activates downstream signaling cascades, resulting in the production of type I IFNs and pro-inflammatory cytokines to induce appropriate systematic immune responses. While these sensors also recognize endogenous nucleic acids and activate immune responses, they can discriminate between self- and non-self-nucleic acids. However, dysfunction of these sensors or failure of regulatory mechanisms causes aberrant activation of immune response and autoimmune disorders. In this review, we focus on how intracellular immune sensors recognize exogenous nucleic acids and activate the innate immune system, and furthermore, how autoimmune diseases result from dysfunction of these sensors.
Collapse
Affiliation(s)
- Daisuke Ori
- a Laboratory of Molecular Immunobiology , Graduate School of Biological Sciences, Nara Institute of Science and Technology (NAIST), Takayama-cho , Ikoma , Nara , Japan
| | - Motoya Murase
- a Laboratory of Molecular Immunobiology , Graduate School of Biological Sciences, Nara Institute of Science and Technology (NAIST), Takayama-cho , Ikoma , Nara , Japan
| | - Taro Kawai
- a Laboratory of Molecular Immunobiology , Graduate School of Biological Sciences, Nara Institute of Science and Technology (NAIST), Takayama-cho , Ikoma , Nara , Japan
| |
Collapse
|
431
|
Patel S, Blaauboer SM, Tucker HR, Mansouri S, Ruiz-Moreno JS, Hamann L, Schumann RR, Opitz B, Jin L. The Common R71H-G230A-R293Q Human TMEM173 Is a Null Allele. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:776-787. [PMID: 27927967 PMCID: PMC5225030 DOI: 10.4049/jimmunol.1601585] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/06/2016] [Indexed: 01/14/2023]
Abstract
TMEM173 encodes MPYS/STING and is an innate immune sensor for cyclic dinucleotides (CDNs) playing a critical role in infection, inflammation, and cancer. The R71H-G230A-R293Q (HAQ) of TMEM173 is the second most common human TMEM173 allele. In this study, using data from the 1000 Genomes Project we found that homozygous HAQ individuals account for ∼16.1% of East Asians and ∼2.8% of Europeans whereas Africans have no homozygous HAQ individuals. Using B cells from homozygous HAQ carriers, we found, surprisingly, that HAQ/HAQ carriers express extremely low MPYS protein and have a decreased TMEM173 transcript. Consequently, the HAQ/HAQ B cells do not respond to CDNs. We subsequently generated an HAQ knock-in mouse expressing a mouse equivalent of the HAQ allele (mHAQ). The mHAQ mouse has decreased MPYS protein in B cells, T cells, Ly6Chi monocytes, bone marrow-derived dendritic cells, and lung tissue. The mHAQ mouse also does not respond to CDNs in vitro and in vivo. Lastly, Pneumovax 23, with an efficacy that depends on TMEM173, is less effective in mHAQ mice than in wild type mice. We conclude that HAQ is a null TMEM173 allele. Our findings have a significant impact on research related to MPYS-mediated human diseases and medicine.
Collapse
Affiliation(s)
- Seema Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610
| | - Steven M Blaauboer
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Heidi R Tucker
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Samira Mansouri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Juan Sebastian Ruiz-Moreno
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité University Medicine Berlin, 13353 Berlin, Germany; and
| | - Lutz Hamann
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité University Medicine Berlin, 13353 Berlin, Germany; and
| | - Ralf R Schumann
- Institute of Microbiology and Hygiene, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité University Medicine Berlin, 13353 Berlin, Germany; and
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610;
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| |
Collapse
|
432
|
Shi GN, Zhang CN, Xu R, Niu JF, Song HJ, Zhang XY, Wang WW, Wang YM, Li C, Wei XQ, Kong DL. Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials 2017; 113:191-202. [DOI: 10.1016/j.biomaterials.2016.10.047] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 12/27/2022]
|
433
|
Tao W, Fu T, He Z, Hu R, Jia L, Hong Y. Evaluation of Immunostimulatory Effects of N-(2-Hydroxy) Propyl-3-Trimethylammonium Chitosan Chloride for Improving Live Attenuated Hepatitis A Virus Vaccine Efficacy. Viral Immunol 2016; 30:120-126. [PMID: 27918250 DOI: 10.1089/vim.2016.0099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study was to evaluate the immunostimulatory effects of N-(2-hydroxy) propyl-3-trimethylammonium chitosan chloride (HTCC) as an adjuvant for improving a commercial live attenuated hepatitis A virus (HAV) vaccine efficacy in mice. Mice in the experimental group were intraperitoneally immunized with a solution of HTCC and live attenuated HAV vaccine. And for those injected with sterile water, HTCC or live attenuated HAV vaccine were treated as mock group, negative group, and positive group in turn. The serum HAV-specific IgG titers and the ratios of the serum HAV-specific IgG2a/IgG1 in the experimental group were significantly increased (p = 0.00042 and p = 0.040, respectively). Splenocyte proliferation stimulation index in experimental group was higher than positive group (p = 0.021), and significantly higher than mock group and negative group (p = 0.0078 and p = 0.0050, respectively). The percentages of CD4+ T lymphocytes in the peripheral blood in experimental group were significantly higher than positive group, negative group, and mock group (p = 0.012, p = 0.012, and p = 0.045, respectively). Compared to the other three groups, experimental group showed a slightly higher ratio of CD4+/CD8+, but there were no significant differences (p > 0.05). In the percentages of CD8+ T lymphocytes, there were no significant differences among the four groups (p > 0.05). HTCC can enhance live attenuated HAV vaccine to generate stronger humoral responses and induce a Th1-biased immune response, as well as IgG2a class switching, compared with the live attenuated HAV vaccine alone. This study validated an important concept for further development of a safe and potent vaccine adjuvant.
Collapse
Affiliation(s)
- Wei Tao
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Ting Fu
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Zhuojing He
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Ruxi Hu
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Lan Jia
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Yan Hong
- Institute of Bioengineering , Zhejiang Academy of Medical Sciences, Hangzhou, China
| |
Collapse
|
434
|
Connor LM, Tang SC, Cognard E, Ochiai S, Hilligan KL, Old SI, Pellefigues C, White RF, Patel D, Smith AAT, Eccles DA, Lamiable O, McConnell MJ, Ronchese F. Th2 responses are primed by skin dendritic cells with distinct transcriptional profiles. J Exp Med 2016; 214:125-142. [PMID: 27913566 PMCID: PMC5206495 DOI: 10.1084/jem.20160470] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 08/20/2016] [Accepted: 11/01/2016] [Indexed: 01/25/2023] Open
Abstract
Connor et al. show that transcriptomic profiling of DCs exposed to two different Th2 stimuli in vivo reveals large numbers of differentially expressed genes but few similarities between conditions. The dendritic cell signals required for the in vivo priming of IL-4–producing T cells are unknown. We used RNA sequencing to characterize DCs from skin LN of mice exposed to two different Th2 stimuli: the helminth parasite Nippostrongylus brasiliensis (Nb) and the contact sensitizer dibutyl phthalate (DBP)-FITC. Both Nb and DBP-FITC induced extensive transcriptional changes that involved multiple DC subsets. Surprisingly, these transcriptional changes were highly distinct in the two models, with only a small number of genes being similarly regulated in both conditions. Pathway analysis of expressed genes identified no shared pathways between Nb and DBP-FITC, but revealed a type-I IFN (IFN-I) signature unique to DCs from Nb-primed mice. Blocking the IFN-I receptor at the time of Nb treatment had little effect on DC migration and antigen transport to the LN, but inhibited the up-regulation of IFN-I–induced markers on DCs and effectively blunted Th2 development. In contrast, the response to DBP-FITC was not affected by IFN-I receptor blockade, a finding consistent with the known dependence of this response on the innate cytokine TSLP. Thus, the priming of Th2 responses is associated with distinct transcriptional signatures in DCs in vivo, reflecting the diverse environments in which Th2 immune responses are initiated.
Collapse
Affiliation(s)
- Lisa M Connor
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Shiau-Choot Tang
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | | | - Sotaro Ochiai
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand.,Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington 6242, New Zealand
| | - Kerry L Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand.,Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington 6242, New Zealand
| | - Samuel I Old
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | | | - Ruby F White
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Deepa Patel
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | | | - David A Eccles
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Olivier Lamiable
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Melanie J McConnell
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand .,School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
435
|
Brotons-Canto A, Martín-Arbella N, Gamazo C, Irache JM. New pharmaceutical approaches for the treatment of food allergies. Expert Opin Drug Deliv 2016; 15:675-686. [PMID: 27732129 DOI: 10.1080/17425247.2016.1247805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Allergic diseases constitute one of the most common causes of chronic illness in developed countries. The main mechanism determining allergy is an imbalance between Th1 and Th2 response towards Th2. AREAS COVERED This review describes the mechanisms underlying the natural tolerance to food components and the development of an allergic response in sensitized individuals. Furthermore, therapeutic approaches proposed to manage these abnormal immunologic responses food are also presented and discussed. EXPERT OPINION In the past, management of food allergies has consisted of the education of patients to avoid the ingestion of the culprit food and to initiate the therapy (e.g. self-injectable epinephrine) in case of accidental ingestion. In recent years, sublingual/oral immunotherapies based on the continuous administration of small amounts of the allergen have been developed. However, the long periods of time needed to obtain significant desensitization and the generation of adverse effects, limit their use. In order to solve these drawbacks, strategies to induce tolerance are being studied, such as the use of either adjuvant immunotherapy in order to facilitate the reversion of the Th2 response towards Th1 or the use of monoclonal antibodies to block the main immunogenic elements.
Collapse
Affiliation(s)
- Ana Brotons-Canto
- a Department of Pharmacy and Pharmaceutical Technology , University of Navarra , Pamplona , Spain
| | - Nekane Martín-Arbella
- a Department of Pharmacy and Pharmaceutical Technology , University of Navarra , Pamplona , Spain
| | - Carlos Gamazo
- b Department of Microbiology , University of Navarra , Pamplona , Spain
| | - Juan M Irache
- a Department of Pharmacy and Pharmaceutical Technology , University of Navarra , Pamplona , Spain
| |
Collapse
|
436
|
Speth MT, Repnik U, Griffiths G. Layer-by-layer nanocoating of live Bacille-Calmette-Guérin mycobacteria with poly(I:C) and chitosan enhances pro-inflammatory activation and bactericidal capacity in murine macrophages. Biomaterials 2016; 111:1-12. [PMID: 27716523 DOI: 10.1016/j.biomaterials.2016.09.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/28/2016] [Accepted: 09/30/2016] [Indexed: 12/11/2022]
Abstract
Tuberculosis (TB) is a major disease burden globally causing more than 1.5 million deaths per year. The attenuated live vaccine strain Bacille Calmette-Guérin (BCG), although providing protection against childhood TB, is largely ineffective against adult pulmonary TB. A major aim therefore is to increase the potency of the BCG vaccine to generate stronger and more sustained immunity against TB. Here, we investigated the use of layer-by-layer (LbL) nanocoating of the surface of live BCG with several layers of polyinosinic-polycytidylic acid (poly(I:C)), a strong inducer of cell-mediated immunity, and the biodegradable polysaccharide chitosan to enhance BCG immunogenicity. Nanocoating of live BCG did not affect bacterial viability or growth in vitro but induced killing of the BCG in infected mouse bone marrow-derived macrophages and enhanced macrophage production of pro-inflammatory cytokines and expression of surface co-stimulatory molecules relative to uncoated BCG. In addition, poly(I:C) surface-coated BCG, but not BCG alone or together with soluble poly(I:C), induced high production of nitric oxide (NO) and IL-12. These results argue that BCG and surface absorbed poly(I:C) act in a synergistic manner to elicit pro-inflammatory macrophage activation. In conclusion, nanocoating of live BCG with the immunostimulatory agent poly(I:C) may be an appropriate strategy to enhance and modulate host responses to the BCG vaccine.
Collapse
Affiliation(s)
- Martin Tobias Speth
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| |
Collapse
|
437
|
Abstract
The host takes use of pattern recognition receptors (PRRs) to defend against pathogen invasion or cellular damage. Among microorganism-associated molecular patterns detected by host PRRs, nucleic acids derived from bacteria or viruses are tightly supervised, providing a fundamental mechanism of host defense. Pathogenic DNAs are supposed to be detected by DNA sensors that induce the activation of NFκB or TBK1-IRF3 pathway. DNA sensor cGAS is widely expressed in innate immune cells and is a key sensor of invading DNAs in several cell types. cGAS binds to DNA, followed by a conformational change that allows the synthesis of cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) from adenosine triphosphate and guanosine triphosphate. cGAMP is a strong activator of STING that can activate IRF3 and subsequent type I interferon production. Here we describe recent progresses in DNA sensors especially cGAS in the innate immune responses against pathogenic DNAs.
Collapse
|
438
|
Abstract
Vaccination is a biological process that administrates antigenic materials to stimulate an individual's immune system to develop immunity to a specific pathogen. It is the most effective tool to prevent illness and death from infectious diseases or diseases leading to cancers. Because many recombinant and synthetic antigens are poorly immunogenic, adjuvant is essentially added to vaccine formula that can potentiate the immune responses, offer better protection against pathogens and reduce the amount of antigens needed for protective immunity. To date, there are nearly 100 different types of adjuvants associated with about 400 vaccines that are either commercially available or under development. Among these adjuvants, many of them are particulates and nano-scale in nature. Nanoparticles represent a wide range of materials with novel physicochemical properties that exhibit immunostimulatory effects. However, the mechanistic understandings on how their physicochemical properties affect immunopotentiation remain elusive. In this article, we aim to review current development status of nanomaterial-based vaccine adjuvants, and further discuss their acting mechanisms, understanding of which will benefit the rational design of effective vaccine adjuvants with improved immunogenicity for prevention of infectious disease as well as therapeutic cancer treatment.
Collapse
Affiliation(s)
- Bingbing Sun
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, California, 90095, United States
- Center for Environmental Implications of Nanotechnology (CEIN), California NanoSystems Institute (CNSI), University of California, Los Angeles, California, 90095, United States
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, California, 90095, United States
- Center for Environmental Implications of Nanotechnology (CEIN), California NanoSystems Institute (CNSI), University of California, Los Angeles, California, 90095, United States
| |
Collapse
|
439
|
Abstract
Adjuvants promote adaptive immunity through the triggering of innate signals that are largely poorly understood. In this issue of Immunity, Lavelle and colleagues describe an unexpected role for the DNA sensing cGAS-STING pathway in the mechanism of action of the Th1 cell-promoting polysaccharide adjuvant chitosan.
Collapse
|
440
|
Islam N, Ferro V. Recent advances in chitosan-based nanoparticulate pulmonary drug delivery. NANOSCALE 2016; 8:14341-58. [PMID: 27439116 DOI: 10.1039/c6nr03256g] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The advent of biodegradable polymer-encapsulated drug nanoparticles has made the pulmonary route of administration an exciting area of drug delivery research. Chitosan, a natural biodegradable and biocompatible polysaccharide has received enormous attention as a carrier for drug delivery. Recently, nanoparticles of chitosan (CS) and its synthetic derivatives have been investigated for the encapsulation and delivery of many drugs with improved targeting and controlled release. Herein, recent advances in the preparation and use of micro-/nanoparticles of chitosan and its derivatives for pulmonary delivery of various therapeutic agents (drugs, genes, vaccines) are reviewed. Although chitosan has wide applications in terms of formulations and routes of drug delivery, this review is focused on pulmonary delivery of drug-encapsulated nanoparticles of chitosan and its derivatives. In addition, the controversial toxicological effects of chitosan nanoparticles for lung delivery will also be discussed.
Collapse
Affiliation(s)
- Nazrul Islam
- Pharmacy Discipline, School of Clinical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | | |
Collapse
|
441
|
Royer DJ, Conrady CD, Carr DJJ. Herpesvirus-Associated Lymphadenitis Distorts Fibroblastic Reticular Cell Microarchitecture and Attenuates CD8 T Cell Responses to Neurotropic Infection in Mice Lacking the STING-IFNα/β Defense Pathways. THE JOURNAL OF IMMUNOLOGY 2016; 197:2338-52. [PMID: 27511736 DOI: 10.4049/jimmunol.1600574] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/06/2016] [Indexed: 01/02/2023]
Abstract
Type I IFN (IFN-α/β)-driven immune responses to acute viral infection are critical to counter replication and prevent dissemination. However, the mechanisms underlying host resistance to HSV type 1 (HSV-1) are incompletely understood. In this study, we show that mice with deficiencies in IFN-α/β signaling or stimulator of IFN genes (STING) exhibit exacerbated neurovirulence and atypical lymphotropic dissemination of HSV-1 following ocular infection. Synergy between IFN-α/β signaling and efficacy of early adaptive immune responses to HSV-1 were dissected using bone marrow chimeras and adoptive cell transfer approaches to profile clonal expansion, effector function, and recruitment of HSV-specific CD8(+) T cells. Lymphotropic viral dissemination was commensurate with abrogated CD8(+) T cell responses and pathological alterations of fibroblastic reticular cell networks in the draining lymph nodes. Our results show that resistance to HSV-1 in the trigeminal ganglia during acute infection is conferred in part by STING and IFN-α/β signaling in both bone marrow-derived and -resident cells, which coalesce to support a robust HSV-1-specific CD8(+) T cell response.
Collapse
Affiliation(s)
- Derek J Royer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104; and Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Christopher D Conrady
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104; and
| | - Daniel J J Carr
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104; and Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
442
|
Wiens KE, Ernst JD. The Mechanism for Type I Interferon Induction by Mycobacterium tuberculosis is Bacterial Strain-Dependent. PLoS Pathog 2016; 12:e1005809. [PMID: 27500737 PMCID: PMC4976988 DOI: 10.1371/journal.ppat.1005809] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 07/13/2016] [Indexed: 12/30/2022] Open
Abstract
Type I interferons (including IFNαβ) are innate cytokines that may contribute to pathogenesis during Mycobacterium tuberculosis (Mtb) infection. To induce IFNβ, Mtb must gain access to the host cytosol and trigger stimulator of interferon genes (STING) signaling. A recently proposed model suggests that Mtb triggers STING signaling through bacterial DNA binding cyclic GMP-AMP synthase (cGAS) in the cytosol. The aim of this study was to test the generalizability of this model using phylogenetically distinct strains of the Mtb complex (MTBC). We infected bone marrow derived macrophages with strains from MTBC Lineages 2, 4 and 6. We found that the Lineage 6 strain induced less IFNβ, and that the Lineage 2 strain induced more IFNβ, than the Lineage 4 strain. The strains did not differ in their access to the host cytosol and IFNβ induction by each strain required both STING and cGAS. We also found that the three strains shed similar amounts of bacterial DNA. Interestingly, we found that the Lineage 6 strain was associated with less mitochondrial stress and less mitochondrial DNA (mtDNA) in the cytosol compared with the Lineage 4 strain. Treating macrophages with a mitochondria-specific antioxidant reduced cytosolic mtDNA and inhibited IFNβ induction by the Lineage 2 and 4 strains. We also found that the Lineage 2 strain did not induce more mitochondrial stress than the Lineage 4 strain, suggesting that additional pathways contribute to higher IFNβ induction. These results indicate that the mechanism for IFNβ by Mtb is more complex than the established model suggests. We show that mitochondrial dynamics and mtDNA contribute to IFNβ induction by Mtb. Moreover, we show that the contribution of mtDNA to the IFNβ response varies by MTBC strain and that additional mechanisms exist for Mtb to induce IFNβ.
Collapse
Affiliation(s)
- Kirsten E. Wiens
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
- Division of Infectious Disease, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Joel D. Ernst
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
- Division of Infectious Disease, Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
443
|
Gutjahr A, Tiraby G, Perouzel E, Verrier B, Paul S. Triggering Intracellular Receptors for Vaccine Adjuvantation. Trends Immunol 2016; 37:573-587. [PMID: 27474233 DOI: 10.1016/j.it.2016.07.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/16/2016] [Accepted: 07/06/2016] [Indexed: 12/15/2022]
Abstract
Immune adjuvants are components that stimulate, potentiate, or modulate the immune response to an antigen. They are key elements of vaccines in both the prophylactic and therapeutic domains. In the past decade substantial progress in our understanding of innate immunity has paved the way for the design of next-generation adjuvants that stimulate a wide range of receptors. Within the framework of vaccine adjuvant design, this review outlines the interest of targeting endosomal and intracellular receptors to enhance and guide the immune response. We present and compare the molecules as well as potential combinations which are currently in the spotlight. We emphasize how targeting the appropriate receptor can direct immunity towards the appropriate response, such as a cytotoxic or mucosal response.
Collapse
Affiliation(s)
- Alice Gutjahr
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Unité Mixte de Recherche 5305, Université Lyon 1, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie et Chimie des Protéines (IBCP)-Lyon, France; InvivoGen, Toulouse, France; Groupe Immunité des Muqueuses et Agents Pathogènes, Institut National de la Santé et de la Recherche Médicale (INSERM) Centre d'Investigation Clinique 1408 Vaccinologie, Faculté de Médecine de Saint-Etienne-Saint-Etienne, France
| | | | | | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Unité Mixte de Recherche 5305, Université Lyon 1, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie et Chimie des Protéines (IBCP)-Lyon, France
| | - Stéphane Paul
- Groupe Immunité des Muqueuses et Agents Pathogènes, Institut National de la Santé et de la Recherche Médicale (INSERM) Centre d'Investigation Clinique 1408 Vaccinologie, Faculté de Médecine de Saint-Etienne-Saint-Etienne, France.
| |
Collapse
|
444
|
Wang ZB, Shan P, Li SZ, Zhou Y, Deng X, Li JL, Zhang Y, Gao JS, Xu J. The mechanism of action of acid-soluble chitosan as an adjuvant in the formulation of nasally administered vaccine against HBV. RSC Adv 2016. [DOI: 10.1039/c6ra14419e] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recently, numerous attempts have been made to evaluate the potential of chitosan as an adjuvant; however, few have explored the mechanism underlying the adjuvant activity of chitosan.
Collapse
Affiliation(s)
- Zhi-Biao Wang
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Pu Shan
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Su-Zhen Li
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Ya Zhou
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Xia Deng
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Ji-Lai Li
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Yu Zhang
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Jin-Shuang Gao
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| | - Jing Xu
- China National Vaccine and Serum Institute
- Beijing 101111
- China
| |
Collapse
|