1
|
Chaudhary V, Mishra B, Ah Kioon MD, Du Y, Ivashkiv LB, Crow MK, Barrat FJ. Mechanosensing regulates pDC activation in the skin through NRF2 activation. J Exp Med 2025; 222:e20240852. [PMID: 39670996 PMCID: PMC11639951 DOI: 10.1084/jem.20240852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/25/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024] Open
Abstract
Plasmacytoid DCs (pDCs) infiltrate the skin, chronically produce type I interferon (IFN-I), and promote skin lesions and fibrosis in autoimmune patients. However, what controls their activation in the skin is unknown. Here, we report that increased stiffness inhibits the production of IFN-I by pDCs. Mechanistically, mechanosensing activates stress pathways including NRF2, which induces the pentose phosphate pathway and reduces pyruvate levels, a product necessary for pDC responses. Modulating NRF2 activity in vivo controlled the pDC response, leading to resolution or chronic induction of IFN-I in the skin. In systemic sclerosis (SSc) patients, although NRF2 was induced in skin-infiltrating pDCs, as compared with blood pDCs, the IFN response was maintained. We observed that CXCL4, a profibrotic chemokine elevated in fibrotic skin, was able to overcome stiffness-mediated IFN-I inhibition, allowing chronic IFN-I responses by pDCs in the skin. Hence, these data identify a novel regulatory mechanism exerted by the skin microenvironment and identify points of dysregulation of this mechanism in patients with skin inflammation and fibrosis.
Collapse
Affiliation(s)
- Vidyanath Chaudhary
- HSS Research Institute, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Bikash Mishra
- HSS Research Institute, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Marie Dominique Ah Kioon
- HSS Research Institute, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Yong Du
- HSS Research Institute, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Lionel B. Ivashkiv
- HSS Research Institute, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mary K. Crow
- HSS Research Institute, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, NY, USA
| | - Franck J. Barrat
- HSS Research Institute, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
2
|
Jeon D, Hill E, McNeel DG. Toll-like receptor agonists as cancer vaccine adjuvants. Hum Vaccin Immunother 2024; 20:2297453. [PMID: 38155525 PMCID: PMC10760790 DOI: 10.1080/21645515.2023.2297453] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy to treat cancer patients. Among the wide range of immunological approaches, cancer vaccines have been investigated to activate and expand tumor-reactive T cells. However, most cancer vaccines have not shown significant clinical benefit as monotherapies. This is likely due to the antigen targets of vaccines, "self" proteins to which there is tolerance, as well as to the immunosuppressive tumor microenvironment. To help circumvent immune tolerance and generate effective immune responses, adjuvants for cancer vaccines are necessary. One representative adjuvant family is Toll-Like receptor (TLR) agonists, synthetic molecules that stimulate TLRs. TLRs are the largest family of pattern recognition receptors (PRRs) that serve as the sensors of pathogens or cellular damage. They recognize conserved foreign molecules from pathogens or internal molecules from cellular damage and propel innate immune responses. When used with vaccines, activation of TLRs signals an innate damage response that can facilitate the development of a strong adaptive immune response against the target antigen. The ability of TLR agonists to modulate innate immune responses has positioned them to serve as adjuvants for vaccines targeting infectious diseases and cancers. This review provides a summary of various TLRs, including their expression patterns, their functions in the immune system, as well as their ligands and synthetic molecules developed as TLR agonists. In addition, it presents a comprehensive overview of recent strategies employing different TLR agonists as adjuvants in cancer vaccine development, both in pre-clinical models and ongoing clinical trials.
Collapse
Affiliation(s)
- Donghwan Jeon
- Department of Oncology, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ethan Hill
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
3
|
Agrez M, Chandler C, Thurecht KJ, Fletcher NL, Liu F, Subramaniam G, Howard CB, Parker S, Turner D, Rzepecka J, Knox G, Nika A, Hall AM, Gooding H, Gallagher L. A novel immunomodulating peptide with potential to complement oligodeoxynucleotide-mediated adjuvanticity in vaccination strategies. Sci Rep 2024; 14:26737. [PMID: 39501043 PMCID: PMC11538426 DOI: 10.1038/s41598-024-78150-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
The identification of adjuvants to improve vaccination efficacy is a major unmet need. One approach is to augment the functionality of dendritic cells (DCs) by using Toll-like receptor-9 (TLR9) agonists such as cytosine-phosphate-guanine oligodeoxynucleotides (CpG ODNs) as adjuvants. Another approach is adjuvant selection based on production of bioactive interleukin-12 (IL-12). We report a D-peptide isomer, designated D-15800, that induces monocyte differentiation to the DC phenotype in vitro and more effectively stimulates IL-12p70 production upon T cell receptor (TCR) activation than the L-isomer. In the absence of TCR activation and either IL-12p70 or interleukin-2 production, only D-15800 activates CD4+ T and natural killer cells. In the presence of CpG ODN, D-15800 synergistically enhances production of interferon-alpha (IFN-α). Taken together with its biostability in human serum and depot retention upon injection, co-delivery of D-15800 with TLR9 agonists could serve to improve vaccine efficacy.
Collapse
Affiliation(s)
- Michael Agrez
- InterK Peptide Therapeutics Limited, Lane Cove West, NSW, Australia.
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia.
| | | | - Kristofer J Thurecht
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Feifei Liu
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Gayathri Subramaniam
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology and the ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, Brisbane, Australia
| | - Stephen Parker
- InterK Peptide Therapeutics Limited, Lane Cove West, NSW, Australia
| | | | | | - Gavin Knox
- Concept Life Sciences, Edinburgh, Scotland
| | | | | | | | | |
Collapse
|
4
|
Gong H, Griffin JD, Groer CE, Wu S, Downes GM, Markum G, Abdelaziz MM, Alhakamy NA, Forrest ML, Berkland CJ. Glatiramer Acetate Complexes CpG Oligodeoxynucleotides into Nanoparticles and Boosts Their TLR9-Driven Immunity. Mol Pharm 2024. [PMID: 39484963 DOI: 10.1021/acs.molpharmaceut.4c00841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Unmethylated cytosine-guanine oligodeoxynucleotides (CpG ODNs) have a storied history as agonists for Toll-like receptor 9 (TLR9). CpG ODNs have shown promising antitumor effects in preclinical studies by inducing potent proinflammatory immune responses. However, clinical success has been hindered by inconsistent efficacy and immune-related toxicities caused by systemic exposure to CpG ODNs. We previously identified that glatiramer acetate (GA), an FDA-approved, lysine-rich polypeptide, could complex class B CpG into cationic nanoparticles which persist at the intratumoral injection site while mitigating the induction of systemic proinflammatory cytokines in mouse tumor models. To extend GA applications across subtypes of CpG ODN (class A, B, and C), we evaluated physiochemical properties and identified the immunological signaling of GA and its complexes with different classes of CpG ODNs. We compared the physiochemical characteristics of three types of GA-CpG nanoparticles, followed by assessments of cell uptake efficiency and endolysosomal trafficking. We then performed successive in vitro and in vivo assays to evaluate immunological discrepancies. Complexation with GA preserved the immunological activity of CpG ODN subtypes while encapsulating them into cationic spherical nanoparticles. GA improved the cellular uptake of CpG ODNs, generally increased retention in early endosomes, and amplified immunological responses. A subsequent in vivo experiment confirmed the achievement of potent tumor suppression while mitigating systemic immune-related toxicities. Together, these data help elucidate the noncanonical role of GA to serve as a nucleic acid delivery scaffold that can improve the efficacy and safety of CpG adjuvant for clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Huan Gong
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | | | - Chad E Groer
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Sa Wu
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Grant M Downes
- Bioengineering Graduate Program, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Grace Markum
- Department of Chemical and Petroleum Engineering, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Moustafa M Abdelaziz
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Nabil A Alhakamy
- Kinimmune Inc., Saint Louis, Missouri 63141, United States
- Department of Pharmaceutics, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - M Laird Forrest
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Cory J Berkland
- Kinimmune Inc., Saint Louis, Missouri 63141, United States
- Department of Biomedical Engineering, Washington University, Saint Louis, Missouri 63105, United States
- Department of Chemistry, Washington University, Saint Louis, Missouri 63130, United States
| |
Collapse
|
5
|
Bandopadhyay P, Sarif J, D'Rozario R, Liu CSC, Sinha BP, Hoque MA, Chatterjee K, Choudhury S, Kumar H, Raychaudhuri D, Ganguly D. Cutting Edge: ATP13A2 Is an Endolysosomal Regulator of TLR9/7 Activation in Human Plasmacytoid Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:109-114. [PMID: 38950331 DOI: 10.4049/jimmunol.2300733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 05/05/2024] [Indexed: 07/03/2024]
Abstract
ATPase cation transporting 13A2 (ATP13A2) is an endolysosomal P-type ATPase known to be a polyamine transporter, explored mostly in neurons. As endolysosomal functions are also crucial in innate immune cells, we aimed to explore the potential role of ATP13A2 in the human immunocellular compartment. We found that human plasmacytoid dendritic cells (pDCs), the professional type I IFN-producing immune cells, especially have a prominent enrichment of ATP13A2 expression in endolysosomal compartments. ATP13A2 knockdown in human pDCs interferes with cytokine induction in response to TLR9/7 activation in response to bona fide ligands. ATP13A2 plays this crucial role in TLR9/7 activation in human pDCs by regulating endolysosomal pH and mitochondrial reactive oxygen generation. This (to our knowledge) hitherto unknown regulatory mechanism in pDCs involving ATP13A2 opens up a new avenue of research, given the crucial role of pDC-derived type I IFNs in protective immunity against infections as well as in the immunopathogenesis of myriad contexts of autoreactive inflammation.
Collapse
Affiliation(s)
- Purbita Bandopadhyay
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Jafar Sarif
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Ranit D'Rozario
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Chinky Shiu Chen Liu
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
| | - Bishnu P Sinha
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Md Asmaul Hoque
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Koustav Chatterjee
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata 700017, India
| | - Supriyo Choudhury
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata 700017, India
| | - Hrishikesh Kumar
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata 700017, India
| | - Deblina Raychaudhuri
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata 700091, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
6
|
Adams NM, Das A, Yun TJ, Reizis B. Ontogeny and Function of Plasmacytoid Dendritic Cells. Annu Rev Immunol 2024; 42:347-373. [PMID: 38941603 DOI: 10.1146/annurev-immunol-090122-041105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Plasmacytoid dendritic cells (pDCs) represent a unique cell type within the innate immune system. Their defining property is the recognition of pathogen-derived nucleic acids through endosomal Toll-like receptors and the ensuing production of type I interferon and other soluble mediators, which orchestrate innate and adaptive responses. We review several aspects of pDC biology that have recently come to the fore. We discuss emerging questions regarding the lineage affiliation and origin of pDCs and argue that these cells constitute an integral part of the dendritic cell lineage. We emphasize the specific function of pDCs as innate sentinels of virus infection, particularly their recognition of and distinct response to virus-infected cells. This essential evolutionary role of pDCs has been particularly important for the control of coronaviruses, as demonstrated by the recent COVID-19 pandemic. Finally, we highlight the key contribution of pDCs to systemic lupus erythematosus, in which therapeutic targeting of pDCs is currently underway.
Collapse
Affiliation(s)
- Nicholas M Adams
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| | - Annesa Das
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| | - Tae Jin Yun
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| |
Collapse
|
7
|
Ah Kioon MD, Laurent P, Chaudhary V, Du Y, Crow MK, Barrat FJ. Modulation of plasmacytoid dendritic cells response in inflammation and autoimmunity. Immunol Rev 2024; 323:241-256. [PMID: 38553621 DOI: 10.1111/imr.13331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The discovery of toll-like receptors (TLRs) and the subsequent recognition that endogenous nucleic acids (NAs) could serve as TLR ligands have led to essential insights into mechanisms of healthy immune responses as well as pathogenic mechanisms relevant to systemic autoimmune and inflammatory diseases. In systemic lupus erythematosus, systemic sclerosis, and rheumatoid arthritis, NA-containing immune complexes serve as TLR ligands, with distinct implications depending on the additional immune stimuli available. Plasmacytoid dendritic cells (pDCs), the robust producers of type I interferon (IFN-I), are providing critical insights relevant to TLR-mediated healthy immune responses and tissue repair, as well as generation of inflammation, autoimmunity and fibrosis, processes central to the pathogenesis of many autoimmune diseases. In this review, we describe recent data characterizing the role of platelets and NA-binding chemokines in modulation of TLR signaling in pDCs, as well as implications for how the IFN-I products of pDCs contribute to the generation of inflammation and wound healing responses by monocyte/macrophages. Chemokine modulators of TLR-mediated B cell tolerance mechanisms and interactions between TLR signaling and metabolic pathways are also considered. The modulators of TLR signaling and their contribution to the pathogenesis of systemic autoimmune diseases suggest new opportunities for identification of novel therapeutic targets.
Collapse
Affiliation(s)
| | - Paôline Laurent
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Vidyanath Chaudhary
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Yong Du
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Mary K Crow
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Franck J Barrat
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
8
|
Tiberio L, Laffranchi M, Zucchi G, Salvi V, Schioppa T, Sozzani S, Del Prete A, Bosisio D. Inhibitory receptors of plasmacytoid dendritic cells as possible targets for checkpoint blockade in cancer. Front Immunol 2024; 15:1360291. [PMID: 38504978 PMCID: PMC10948453 DOI: 10.3389/fimmu.2024.1360291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are the major producers of type I interferons (IFNs), which are essential to mount antiviral and antitumoral immune responses. To avoid exaggerated levels of type I IFNs, which pave the way to immune dysregulation and autoimmunity, pDC activation is strictly regulated by a variety of inhibitory receptors (IRs). In tumors, pDCs display an exhausted phenotype and correlate with an unfavorable prognosis, which largely depends on the accumulation of immunosuppressive cytokines and oncometabolites. This review explores the hypothesis that tumor microenvironment may reduce the release of type I IFNs also by a more pDC-specific mechanism, namely the engagement of IRs. Literature shows that many cancer types express de novo, or overexpress, IR ligands (such as BST2, PCNA, CAECAM-1 and modified surface carbohydrates) which often represent a strong predictor of poor outcome and metastasis. In line with this, tumor cells expressing ligands engaging IRs such as BDCA-2, ILT7, TIM3 and CD44 block pDC activation, while this blocking is prevented when IR engagement or signaling is inhibited. Based on this evidence, we propose that the regulation of IFN secretion by IRs may be regarded as an "innate checkpoint", reminiscent of the function of "classical" adaptive immune checkpoints, like PD1 expressed in CD8+ T cells, which restrain autoimmunity and immunopathology but favor chronic infections and tumors. However, we also point out that further work is needed to fully unravel the biology of tumor-associated pDCs, the neat contribution of pDC exhaustion in tumor growth following the engagement of IRs, especially those expressed also by other leukocytes, and their therapeutic potential as targets of combined immune checkpoint blockade in cancer immunotherapy.
Collapse
Affiliation(s)
- Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Laffranchi
- Department of Molecular Medicine, Laboratory Affiliated to Institute Pasteur-Italia, Sapienza University of Rome, Rome, Italy
| | - Giovanni Zucchi
- Department of Molecular Medicine, Laboratory Affiliated to Institute Pasteur-Italia, Sapienza University of Rome, Rome, Italy
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Tiziana Schioppa
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Laboratory Affiliated to Institute Pasteur-Italia, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
9
|
Monti M, Ferrari G, Grosso V, Missale F, Bugatti M, Cancila V, Zini S, Segala A, La Via L, Consoli F, Orlandi M, Valerio A, Tripodo C, Rossato M, Vermi W. Impaired activation of plasmacytoid dendritic cells via toll-like receptor 7/9 and STING is mediated by melanoma-derived immunosuppressive cytokines and metabolic drift. Front Immunol 2024; 14:1227648. [PMID: 38239354 PMCID: PMC10795195 DOI: 10.3389/fimmu.2023.1227648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Plasmacytoid dendritic cells (pDCs) infiltrate a large set of human cancers. Interferon alpha (IFN-α) produced by pDCs induces growth arrest and apoptosis in tumor cells and modulates innate and adaptive immune cells involved in anti-cancer immunity. Moreover, effector molecules exert tumor cell killing. However, the activation state and clinical relevance of pDCs infiltration in cancer is still largely controversial. In Primary Cutaneous Melanoma (PCM), pDCs density decreases over disease progression and collapses in metastatic melanoma (MM). Moreover, the residual circulating pDC compartment is defective in IFN-α production. Methods The activation of tumor-associated pDCs was evaluated by in silico and microscopic analysis. The expression of human myxovirus resistant protein 1 (MxA), as surrogate of IFN-α production, and proximity ligation assay (PLA) to test dsDNA-cGAS activation were performed on human melanoma biopsies. Moreover, IFN-α and CXCL10 production by in vitro stimulated (i.e. with R848, CpG-A, ADU-S100) pDCs exposed to melanoma cell lines supernatants (SN-mel) was tested by intracellular flow cytometry and ELISA. We also performed a bulk RNA-sequencing on SN-mel-exposed pDCs, resting or stimulated with R848. Glycolytic rate assay was performed on SN-mel-exposed pDCs using the Seahorse XFe24 Extracellular Flux Analyzer. Results Based on a set of microscopic, functional and in silico analyses, we demonstrated that the melanoma milieu directly impairs IFN-α and CXCL10 production by pDCs via TLR-7/9 and cGAS-STING signaling pathways. Melanoma-derived immunosuppressive cytokines and a metabolic drift represent relevant mechanisms enforcing pDC-mediated melanoma escape. Discussion These findings propose a new window of intervention for novel immunotherapy approaches to amplify the antitumor innate immune response in cutaneous melanoma (CM).
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giorgia Ferrari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valentina Grosso
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Nederlands Kanker Instituut, Amsterdam, Netherlands
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Stefania Zini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Agnese Segala
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Consoli
- Oncology Unit, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili di Brescia, Brescia, Italy
| | - Matteo Orlandi
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
10
|
Çakan E, Ah Kioon MD, Garcia-Carmona Y, Glauzy S, Oliver D, Yamakawa N, Vega Loza A, Du Y, Schickel JN, Boeckers JM, Yang C, Baldo A, Ivashkiv LB, Young RM, Staudt LM, Moody KL, Nündel K, Marshak-Rothstein A, van der Made CI, Hoischen A, Hayward A, Rossato M, Radstake TR, Cunningham-Rundles C, Ryu C, Herzog EL, Barrat FJ, Meffre E. TLR9 ligand sequestration by chemokine CXCL4 negatively affects central B cell tolerance. J Exp Med 2023; 220:e20230944. [PMID: 37773045 PMCID: PMC10541333 DOI: 10.1084/jem.20230944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
Central B cell tolerance is believed to be regulated by B cell receptor signaling induced by the recognition of self-antigens in immature B cells. Using humanized mice with defective MyD88, TLR7, or TLR9 expression, we demonstrate that TLR9/MYD88 are required for central B cell tolerance and the removal of developing autoreactive clones. We also show that CXCL4, a chemokine involved in systemic sclerosis (SSc), abrogates TLR9 function in B cells by sequestering TLR9 ligands away from the endosomal compartments where this receptor resides. The in vivo production of CXCL4 thereby impedes both TLR9 responses in B cells and the establishment of central B cell tolerance. We conclude that TLR9 plays an essential early tolerogenic function required for the establishment of central B cell tolerance and that correcting defective TLR9 function in B cells from SSc patients may represent a novel therapeutic strategy to restore B cell tolerance.
Collapse
Affiliation(s)
- Elif Çakan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Yolanda Garcia-Carmona
- Department of Clinical Immunology, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Salomé Glauzy
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David Oliver
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Natsuko Yamakawa
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Andrea Vega Loza
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yong Du
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | | | - Joshua M. Boeckers
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Chao Yang
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Alessia Baldo
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lionel B. Ivashkiv
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Ryan M. Young
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Krishna L. Moody
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, MA, USA
| | - Kerstin Nündel
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, MA, USA
| | - Ann Marshak-Rothstein
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, MA, USA
| | - Caspar I. van der Made
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anthony Hayward
- Warren Alper School of Medicine, Brown University, Providence, RI, USA
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Timothy R.D.J. Radstake
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Charlotte Cunningham-Rundles
- Department of Clinical Immunology, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine and Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Changwan Ryu
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Erica L. Herzog
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Section of Rheumatology, Allergy, and Clinical Immunology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
11
|
Wiest MJ, Baert L, Gu C, Gayler KM, Ham H, Gorvel L, Keddis MT, Griffing LW, Joo H, Gorvel JP, Billadeau DD, Kane RR, Oh S. Endosomal trafficking inhibitor EGA can control TLR7-mediated IFNα expression by human plasmacytoid dendritic cells. Front Immunol 2023; 14:1202197. [PMID: 38077311 PMCID: PMC10704457 DOI: 10.3389/fimmu.2023.1202197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Plasmacytoid dendritic cells (pDC) are the major producer of type 1 IFN in response to TLR7 agonists. Aberrant TLR7 activation and type 1 IFN expression by pDCs are linked to the pathogenesis of certain types of autoimmune diseases, including systemic lupus erythematosus (SLE). This study investigated the underlying mechanisms for TLR7-mediated cytokine expression by pDCs using a late endosome trafficking inhibitor, EGA (4-bromobenzaldehyde N-(2,6-dimethylphenyl) semicarbazone). We found that EGA treatment decreased IFNα expression by pDCs stimulated with imiquimod (R837), single-stranded RNA40, and influenza virus. EGA also decreased TNFα expression and secretion by R837-stimulated pDCs. Mechanistically, EGA treatment decreased phosphorylation of IKKα/β, STAT1, and p38, and prolonged degradation of IκBα. Furthermore, EGA treatment decreased the colocalization of 3F, a substituted adenine TLR7 agonist, with LAMP1+ compartments in pDCs. EGA was also capable of diminishing IFNα expression by SLE pDCs treated with R837 or live PR8/A/34 influenza viruses. Therefore, we concluded that trafficking of TLR7 agonists to LAMP1+ compartments is important for IFNα expression by pDCs. Data from this study support additional examinations of the potential benefits of EGA in treating type 1 IFN-associated inflammatory diseases in the future.
Collapse
Affiliation(s)
- Matthew J. Wiest
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Laurie Baert
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Chao Gu
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Kevin M. Gayler
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| | - Hyoungjun Ham
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Laurent Gorvel
- CRCM, Aix Marseille Universite, INSERM, Marseille, France
| | - Mira T. Keddis
- Department of Nephrology, Mayo Clinic, Scottsdale, AZ, United States
| | - Leroy W. Griffing
- Department of Rheumatology, Mayo Clinic, Scottsdale, AZ, United States
| | - HyeMee Joo
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| | | | | | - Robert R. Kane
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| | - SangKon Oh
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| |
Collapse
|
12
|
Cosgrove HA, Gingras S, Kim M, Bastacky S, Tilstra JS, Shlomchik MJ. B cell-intrinsic TLR7 expression drives severe lupus in TLR9-deficient mice. JCI Insight 2023; 8:e172219. [PMID: 37606042 PMCID: PMC10543715 DOI: 10.1172/jci.insight.172219] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/11/2023] [Indexed: 08/23/2023] Open
Abstract
The endosomal Toll-like receptor 7 (TLR7) is a major driver of murine and human systemic lupus erythematosus (SLE). The role of TLR7 in lupus pathogenesis is enhanced when the regulatory role of TLR9 is absent. TLR7 signaling in plasmacytoid DCs (pDC) is generally thought to be a major driver of the IFN response and disease pathology; however, the cell types in which TLR7 acts to mediate disease have not been distinguished. To address this, we selectively deleted TLR7 in either CD11c+ cells or CD19+ cells; using a TLR7-floxed allele, we created on the lupus-prone MRL/lpr background, along with a BM chimera strategy. Unexpectedly, TLR7 deficiency in CD11c+ cells had no impact on disease, while TLR7 deficiency in CD19+ B cells yielded mild suppression of proteinuria and a trend toward reduced glomerular disease. However, in TLR9-deficient MRL/lpr mice with accelerated SLE, B cell-specific TLR7 deficiency greatly improved disease. These results support revision of the mechanism by which TLR7 drives lupus and highlight a cis regulatory interaction between the protective TLR9 and the pathogenic TLR7 within the B cell compartment. They suggest B cell-directed, dual TLR7 antagonism/TLR9 agonism or dual TLR7/9 antagonism as a potential future therapeutic strategy to treat SLE.
Collapse
Affiliation(s)
| | | | | | | | - Jeremy S. Tilstra
- Department of Immunology
- Department of Medicine, and
- Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
13
|
Keppler M, Straß S, Geiger S, Fischer T, Späth N, Weinstein T, Schwamborn A, Guezguez J, Guse JH, Laufer S, Burnet M. Imidazoquinolines with improved pharmacokinetic properties induce a high IFNα to TNFα ratio in vitro and in vivo. Front Immunol 2023; 14:1168252. [PMID: 37409123 PMCID: PMC10319141 DOI: 10.3389/fimmu.2023.1168252] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/31/2023] [Indexed: 07/07/2023] Open
Abstract
TLR Agonists have promising activity in preclinical models of viral infection and cancer. However, clinical use is only in topical application. Systemic uses of TLR-ligands such as Resiquimod, have failed due to adverse effects that limited dose and thus, efficacy. This issue could be related to pharmacokinetic properties that include fast elimination leading to low AUC with simultaneously high cmax at relevant doses. The high cmax is associated with a sharp, poorly tolerated cytokine pulse, suggesting that a compound with a higher AUC/cmax-ratio could provide a more sustained and tolerable immune activation. Our approach was to design TLR7/8-agonist Imidazoquinolines intended to partition to endosomes via acid trapping using a macrolide-carrier. This can potentially extend pharmacokinetics and simultaneously direct the compounds to the target compartment. The compounds have hTLR7/8-agonist activity (EC50 of the most active compound in cellular assays: 75-120 nM hTLR7, 2.8-3.1 µM hTLR8) and maximal hTLR7 activation between 40 and 80% of Resiquimod. The lead candidates induce secretion of IFNα from human Leukocytes in the same range as Resiquimod but induce at least 10-fold less TNFα in this system, consistent with a higher specificity for human TLR7. This pattern was reproduced in vivo in a murine system, where small molecules are thought not to activate TLR8. We found that Imidazoquinolines conjugated to a macrolide or, substances carrying an unlinked terminal secondary amine, had longer exposure compared with Resiquimod. The kinetics of pro-inflammatory cytokine release for these substances in vivo were slower and more extended (for comparable AUCs, approximately half-maximal plasma concentrations). Maximal IFNα plasma levels were reached 4 h post application. Resiquimod-treated groups had by then returned to baseline from a peak at 1 h. We propose that the characteristic cytokine profile is likely a consequence of altered pharmacokinetics and, potentially, enhanced endosomal tropism of the novel substances. In particular, our substances are designed to partition to cellular compartments where the target receptor and a distinct combination of signaling molecules relevant to IFNα-release are located. These properties could address the tolerability issues of TLR7/8 ligands and provide insight into approaches to fine-tune the outcomes of TLR7/8 activation by small molecules.
Collapse
Affiliation(s)
| | - Simon Straß
- Synovo GmbH, Tübingen, Germany
- Pharmaceutical Chemistry, Institute for Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | - Stefan Laufer
- Pharmaceutical Chemistry, Institute for Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
| | | |
Collapse
|
14
|
Zhang W, Zhong R, Qu X, Xiang Y, Ji M. Effect of 8-Hydroxyguanine DNA Glycosylase 1 on the Function of Immune Cells. Antioxidants (Basel) 2023; 12:1300. [PMID: 37372030 DOI: 10.3390/antiox12061300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Excess reactive oxygen species (ROS) can cause an imbalance between oxidation and anti-oxidation, leading to the occurrence of oxidative stress in the body. The most common product of ROS-induced base damage is 8-hydroxyguanine (8-oxoG). Failure to promptly remove 8-oxoG often causes mutations during DNA replication. 8-oxoG is cleared from cells by the 8-oxoG DNA glycosylase 1 (OGG1)-mediated oxidative damage base excision repair pathway so as to prevent cells from suffering dysfunction due to oxidative stress. Physiological immune homeostasis and, in particular, immune cell function are vulnerable to oxidative stress. Evidence suggests that inflammation, aging, cancer, and other diseases are related to an imbalance in immune homeostasis caused by oxidative stress. However, the role of the OGG1-mediated oxidative damage repair pathway in the activation and maintenance of immune cell function is unknown. This review summarizes the current understanding of the effect of OGG1 on immune cell function.
Collapse
Affiliation(s)
- Weiran Zhang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Ranwei Zhong
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Ming Ji
- Department of Physiology, School of Basic Medicine, Central South University, Changsha 410078, China
| |
Collapse
|
15
|
Reyes C, Patarroyo MA. Adjuvants approved for human use: What do we know and what do we need to know for designing good adjuvants? Eur J Pharmacol 2023; 945:175632. [PMID: 36863555 DOI: 10.1016/j.ejphar.2023.175632] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/04/2023]
Abstract
Adjuvants represent one of the most significant biotechnological solutions regarding vaccine development, thereby broadening the amount of candidates which can now be used and tested in vaccine formulations targeting various pathogens, as antigens which were previously discarded due to their low or null immunogenicity can now be included. Adjuvant development research has grown side-by-side with an increasing body of knowledge regarding immune systems and their recognition of foreign microorganisms. Alum-derived adjuvants were used in human vaccines for many years, even though complete understanding of their vaccination-related mechanism of action was lacking. The amount of adjuvants approved for human use has increased recently in line with attempts to interact with and stimulate the immune system. This review is aimed at summarising what is known about adjuvants, focusing on those approved for use in humans, their mechanism of action and why they are so necessary for vaccine candidate formulations; it also discusses what the future may hold in this growing research field.
Collapse
Affiliation(s)
- César Reyes
- PhD Programme in Biotechnology, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá, DC 111321, Colombia; Three-dimensional Structures Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá, DC 111321, Colombia; Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222#55-37, Bogotá, DC 111166, Colombia.
| | - Manuel A Patarroyo
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá, DC 111321, Colombia; Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá, DC 111321, Colombia.
| |
Collapse
|
16
|
Yang L, Lang Y, Wu H, Xiang K, Wang Y, Yu M, Liu Y, Yang B, He L, Lu G, Ni Q, Chen X, Zhang L. Engineered Toll-like Receptor Nanoagonist Binding to Extracellular Matrix Elicits Safe and Robust Antitumor Immunity. ACS NANO 2023; 17:5340-5353. [PMID: 36913671 DOI: 10.1021/acsnano.2c08429] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cancer immunotherapy, such as the Toll-like receptor (TLR) agonist including CpG oligodeoxynucleotide, has shown potency in clinical settings. However, it is still confronted with multiple challenges, which include the limited efficacy and severe adverse events caused by the rapid clearance and systemic diffusion of CpG. Here we report an improved CpG-based immunotherapy approach composed of a synthetic extracellular matrix (ECM)-anchored DNA/peptide hybrid nanoagonist (EaCpG) via (1) a tailor designed DNA template that encodes tetramer CpG and additional short DNA moieties, (2) generation of elongated multimeric CpG through rolling circle amplification (RCA), (3) self-assembly of densely packaged CpG particles composed of tandem CpG building blocks and magnesium pyrophosphate, and (4) incorporation of multiple copies of ECM binding peptide through hybridization to short DNA moieties. The structurally well-defined EaCpG shows dramatically increased intratumoral retention and marginal systemic dissemination through peritumoral administration, leading to potent antitumor immune response and subsequent tumor elimination, with minimal treatment-related toxicity. Combined with conventional standard-of-care therapies, peritumor administration of EaCpG generates systemic immune responses that lead to a curative abscopal effect on distant untreated tumors in multiple cancer models, which is superior to the unmodified CpG. Taken together, EaCpG provides a facile and generalizable strategy to simultaneously potentiate the potency and safety of CpG for combinational cancer immunotherapies.
Collapse
Affiliation(s)
- Liu Yang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| | - Yue Lang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| | - Haoguang Wu
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| | - Kaiyan Xiang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| | - Yuanzheng Wang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| | - Mengqi Yu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yu Liu
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| | - Bowei Yang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Liangcan He
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China
| | - Guangming Lu
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| | - Qianqian Ni
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002 Jiangsu, China
| |
Collapse
|
17
|
Butkovich N, Tucker JA, Ramirez A, Li E, Meli VS, Nelson EL, Wang SW. Nanoparticle vaccines can be designed to induce pDC support of mDCs for increased antigen display. Biomater Sci 2023; 11:596-610. [PMID: 36476811 PMCID: PMC10775882 DOI: 10.1039/d2bm01132h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cancer vaccine immunotherapy facilitates the immune system's recognition of tumor-associated antigens, and the biomolecular design of these vaccines using nanoparticles is one important approach towards obtaining strong anti-tumor responses. Following activation of dendritic cells (DCs), a robust CD8+ T cell-mediated adaptive immune response is critical for tumor elimination. While the role of efficient antigen-presenting myeloid DCs (mDCs) is conventionally attributed towards vaccine efficacy, participation by highly cytokine-producing plasmacytoid DCs (pDCs) is less understood and is often overlooked. We examined vaccines based on the E2 protein nanoparticle platform that delivered encapsulated TLR9 agonist bacterial-like DNA (CpG1826 or CpG1018) or TLR7 agonist viral ssRNA to determine their efficacy over free agonists in activating both mDCs and pDCs for antigen presentation. Although mDCs were only activated by nanoparticle-encapsulated TLR9 agonists, pDCs were activated by all the individually tested constructs, and CpG1826 was shown to induce pDC cytokine production. Transfer of secreted factors from pDCs that were stimulated with a vaccine formulation comprising peptide antigen and CpG1826 enhanced mDC display of the antigen, particularly when delivered in nanoparticles. Only when treated with nanoparticle-conjugated vaccine could pDCs secrete factors to induce antigen display on naïve mDCs. These results reveal that pDCs can aid mDCs, highlighting the importance of activating both pDCs and mDCs in designing effective cancer vaccines, and demonstrate the advantage of using nanoparticle-based vaccine delivery.
Collapse
Affiliation(s)
- Nina Butkovich
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Jo Anne Tucker
- Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Aaron Ramirez
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Enya Li
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Edward L Nelson
- Department of Medicine, University of California, Irvine, CA 92697, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
- Institute for Immunology, University of California, Irvine, CA 92697, USA
| | - Szu-Wen Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
- Institute for Immunology, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| |
Collapse
|
18
|
Wiest MJ, Gu C, Ham H, Gorvel L, Keddis MT, Griffing LW, Joo H, Gorvel JP, Billadeau DD, Oh S. Disruption of endosomal trafficking with EGA alters TLR9 cytokine response in human plasmacytoid dendritic cells. Front Immunol 2023; 14:1144127. [PMID: 37020542 PMCID: PMC10067882 DOI: 10.3389/fimmu.2023.1144127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Plasmacytoid dendritic cells (pDCs) exhibit bifurcated cytokine responses to TLR9 agonists, an IRF7-mediated type 1 IFN response or a pro-inflammatory cytokine response via the activation of NF-κB. This bifurcated response has been hypothesized to result from either distinct signaling endosomes or endo-lysosomal trafficking delay of TLR9 agonists allowing for autocrine signaling to affect outcomes. Utilizing the late endosome trafficking inhibitor, EGA, we assessed the bifurcated cytokine responses of pDCs to TLR9 stimulation. EGA treatment of pDCs diminished both IFNα and pro-inflammatory cytokine expression induced by CpG DNAs (D- and K-type), CpG-DNAs complexed with DOTAP, and genomic DNAs complexed with LL37. Mechanistically, EGA suppressed phosphorylation of IKKα/β, STAT1, Akt, and p38, and decreased colocalization of CpG oligodeoxynucleotides with LAMP+ endo-lysosomes. EGA also diminished type 1 IFN expression by pDCs from systemic lupus erythematosus patients. Therefore, our findings help understand mechanisms for the bifurcated cytokine responses by pDCs and support future examination of the potential benefit of EGA in treating type 1 IFN-associated inflammatory diseases in the future.
Collapse
Affiliation(s)
- Matthew J. Wiest
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Baylor Institute of Biomedical Studies, Baylor University, Waco, TX, United States
| | - Chao Gu
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Hyoungjun Ham
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Laurent Gorvel
- CRCM, Aix Marseille Universite, INSERM, Marseille, France
| | - Mira T. Keddis
- Department of Nephrology, Mayo Clinic, Scottsdale, AZ, United States
| | - Leroy W. Griffing
- Department of Rheumatology, Mayo Clinic, Scottsdale, AZ, United States
| | - HyeMee Joo
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Baylor Institute of Biomedical Studies, Baylor University, Waco, TX, United States
| | | | | | - SangKon Oh
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
- Baylor Institute of Biomedical Studies, Baylor University, Waco, TX, United States
- *Correspondence: SangKon Oh,
| |
Collapse
|
19
|
Kumpunya S, Thim-uam A, Thumarat C, Leelahavanichkul A, Kalpongnukul N, Chantaravisoot N, Pisitkun T, Pisitkun P. cGAS deficiency enhances inflammasome activation in macrophages and inflammatory pathology in pristane-induced lupus. Front Immunol 2022; 13:1010764. [PMID: 36591278 PMCID: PMC9800982 DOI: 10.3389/fimmu.2022.1010764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Type I interferon (IFN) plays a vital role in the pathogenesis of systemic lupus erythematosus. Cyclic GMP AMP synthase (cGAS) is a cytosolic DNA sensor that recognizes dsDNA and creates cGAMP to activate STING-mediated type I IFN production. The activation of STING induces lupus disease in Fcgr2b deficient mice through the differentiation of dendritic cells. In contrast, Cgas-deficient mice could be generated more autoantibody production and proteinuria in pristane-induced lupus (PIL). These data suggested that the other dsDNA sensors could be involved in lupus development mechanisms. Methods This study aimed to identify the cGAS-mediated mechanisms contributing to lupus pathogenesis in PIL. The Cgas-deficient and WT mice were induced lupus disease with pristane and subsequently analyzed autoantibody, histopathology, and immunophenotypes. The lung tissues were analyzed with the expression profiles by RT-PCR and western blot. The bone marrow-derived macrophages were stimulated with inflammasome activators and observed pyroptosis. Results The Cgas-/- mice developed more severe pulmonary hemorrhage and autoantibody production than WT mice. The activated dendritic cells, IFN-g-, and IL-17a-producing T helper cells, and infiltrated macrophages in the lung were detected in Cgas-/- mice higher than in WT mice. We observed an increase in expression of Aim2, Casp11, and Ifi16 in the lung and serum IL-1a but IL-1b in pristane-injected Cgas-/- mice. The rise of Caspase-11 in the lung of pristane-injected Cgas-/- mice suggested noncanonical inflammasome activation. The activation of AIM2 and NLRP3 inflammasomes in bone marrow-derived macrophages (BMDMs) enhanced the number of dead cells in Cgas-/- mice compared with WT mice. Activation of the inflammasome significantly induced pyroptosis in Cgas-/- BMDMs. The dsDNA level, but not mitochondrial DNA, increased dramatically in pristane-injected Cgas-/- mice suggesting the dsDNA could be a ligand activating inflammasomes. The cGAS agonist-induced BMDM activation in the Cgas-/- mice indicated that the activation of DNA sensors other than cGAS enhanced activated macrophages. Conclusion These findings suggested that cGAS hampers the unusual noncanonical inflammasome activation through other DNA sensors.
Collapse
Affiliation(s)
- Sarinya Kumpunya
- Interdisciplinary Program of Biomedical Sciences, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Arthid Thim-uam
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Chisanu Thumarat
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nuttiya Kalpongnukul
- Interdisciplinary Program of Biomedical Sciences, Graduate School, Chulalongkorn University, Bangkok, Thailand,Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Naphat Chantaravisoot
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,Epithelial Systems Biology Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States,*Correspondence: Prapaporn Pisitkun, ; Trairak Pisitkun,
| | - Prapaporn Pisitkun
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand,Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand,*Correspondence: Prapaporn Pisitkun, ; Trairak Pisitkun,
| |
Collapse
|
20
|
Crow MK. Advances in lupus therapeutics: Achieving sustained control of the type I interferon pathway. Curr Opin Pharmacol 2022; 67:102291. [PMID: 36183477 DOI: 10.1016/j.coph.2022.102291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 01/25/2023]
Abstract
Achieving sustained control of disease activity in patients with systemic lupus erythematosus has been impeded by the complexity of its immunopathogenesis as well its clinical heterogeneity. In spite of these challenges, gains in understanding disease mechanisms have identified immune targets that are currently under study in trials of candidate therapeutics. Defining the type I interferon (IFN-I) pathway and autoantibodies specific for nucleic acid binding proteins as core pathogenic mediators allows an analysis of approaches that could control production of those mediators and improve patient outcomes. This review describes therapeutic targets and agents that could achieve control of the IFN-I pathway. Toll-like receptor 7, involved in IFN-I production and differentiation of B cells, and long-lived plasma cells, the producers of autoantibodies specific for RNA-binding proteins, components of the immune complex drivers of IFN-I, are particularly attractive therapeutic targets.
Collapse
Affiliation(s)
- Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery and Weill Cornell Medicine, 535 East 70th Street, New York, NY 10021, USA.
| |
Collapse
|
21
|
Chaudhary V, Ah Kioon MD, Hwang SM, Mishra B, Lakin K, Kirou KA, Zhang-Sun J, Wiseman RL, Spiera RF, Crow MK, Gordon JK, Cubillos-Ruiz JR, Barrat FJ. Chronic activation of pDCs in autoimmunity is linked to dysregulated ER stress and metabolic responses. J Exp Med 2022; 219:e20221085. [PMID: 36053251 PMCID: PMC9441715 DOI: 10.1084/jem.20221085] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/04/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) chronically produce type I interferon (IFN-I) in autoimmune diseases, including systemic sclerosis (SSc) and systemic lupus erythematosus (SLE). We report that the IRE1α-XBP1 branch of the unfolded protein response (UPR) inhibits IFN-α production by TLR7- or TLR9-activated pDCs. In SSc patients, UPR gene expression was reduced in pDCs, which inversely correlated with IFN-I-stimulated gene expression. CXCL4, a chemokine highly secreted in SSc patients, downregulated IRE1α-XBP1-controlled genes and promoted IFN-α production by pDCs. Mechanistically, IRE1α-XBP1 activation rewired glycolysis to serine biosynthesis by inducing phosphoglycerate dehydrogenase (PHGDH) expression. This process reduced pyruvate access to the tricarboxylic acid (TCA) cycle and blunted mitochondrial ATP generation, which are essential for pDC IFN-I responses. Notably, PHGDH expression was reduced in pDCs from patients with SSc and SLE, and pharmacological blockade of TCA cycle reactions inhibited IFN-I responses in pDCs from these patients. Hence, modulating the IRE1α-XBP1-PHGDH axis may represent a hitherto unexplored strategy for alleviating chronic pDC activation in autoimmune disorders.
Collapse
Affiliation(s)
- Vidyanath Chaudhary
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
| | - Sung-Min Hwang
- Sandra and Edward Meyer Cancer Center and Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY
| | - Bikash Mishra
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY
| | - Kimberly Lakin
- Department of Medicine, Division of Rheumatology and Scleroderma and Vasculitis Center, Hospital for Special Surgery, New York, NY
| | - Kyriakos A. Kirou
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, NY
| | - Jeffrey Zhang-Sun
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, NY
| | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Robert F. Spiera
- Department of Medicine, Division of Rheumatology and Scleroderma and Vasculitis Center, Hospital for Special Surgery, New York, NY
| | - Mary K. Crow
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Jessica K. Gordon
- Department of Medicine, Division of Rheumatology and Scleroderma and Vasculitis Center, Hospital for Special Surgery, New York, NY
| | - Juan R. Cubillos-Ruiz
- Sandra and Edward Meyer Cancer Center and Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
- Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY
| |
Collapse
|
22
|
Laurent P, Yang C, Rendeiro AF, Nilsson-Payant BE, Carrau L, Chandar V, Bram Y, tenOever BR, Elemento O, Ivashkiv LB, Schwartz RE, Barrat FJ. Sensing of SARS-CoV-2 by pDCs and their subsequent production of IFN-I contribute to macrophage-induced cytokine storm during COVID-19. Sci Immunol 2022; 7:eadd4906. [PMID: 36083891 PMCID: PMC9853436 DOI: 10.1126/sciimmunol.add4906] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Lung-infiltrating macrophages create a marked inflammatory milieu in a subset of patients with COVID-19 by producing a cytokine storm, which correlates with increased lethality. However, these macrophages are largely not infected by SARS-CoV-2, so the mechanism underlying their activation in the lung is unclear. Type I interferons (IFN-I) contribute to protecting the host against SARS-CoV-2 but may also have some deleterious effect, and the source of IFN-I in the lungs of infected patients is not well defined. Plasmacytoid dendritic cells (pDCs), a key cell type involved in antiviral responses, can produce IFN-I in response to SARS-CoV-2. We observed the infiltration of pDCs in the lungs of SARS-CoV-2-infected patients, which correlated with strong IFN-I signaling in lung macrophages. In patients with severe COVID-19, lung macrophages expressed a robust inflammatory signature, which correlated with persistent IFN-I signaling at the single-cell level. Hence, we observed the uncoupling in the kinetics of the infiltration of pDCs in the lungs and the associated IFN-I signature, with the cytokine storm in macrophages. We observed that pDCs were the dominant IFN-α-producing cells in response to the virus in the blood, whereas macrophages produced IFN-α only when in physical contact with infected epithelial cells. We also showed that IFN-α produced by pDCs, after the sensing of SARS-CoV-2 by TLR7, mediated changes in macrophages at both transcriptional and epigenetic levels, which favored their hyperactivation by environmental stimuli. Together, these data indicate that the priming of macrophages can result from the response by pDCs to SARS-CoV-2, leading to macrophage activation in patients with severe COVID-19.
Collapse
Affiliation(s)
- Paôline Laurent
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Chao Yang
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - André F. Rendeiro
- Institute for Computational Biomedicine and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Benjamin E. Nilsson-Payant
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave., New York, NY 10029, USA
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Lucia Carrau
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave., New York, NY 10029, USA
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Vasuretha Chandar
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Benjamin R. tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave., New York, NY 10029, USA
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Olivier Elemento
- Institute for Computational Biomedicine and Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- WorldQuant Initiative for Quantitative Prediction and Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY 10029, USA
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
| | - Lionel B. Ivashkiv
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
| | - Robert E. Schwartz
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| |
Collapse
|
23
|
Wang L, Noyer L, Wang YH, Tao AY, Li W, Zhu J, Saavedra P, Hoda ST, Yang J, Feske S. ORAI3 is dispensable for store-operated Ca2+ entry and immune responses by lymphocytes and macrophages. J Gen Physiol 2022; 154:213360. [PMID: 35861698 PMCID: PMC9532584 DOI: 10.1085/jgp.202213104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/24/2022] [Indexed: 01/23/2023] Open
Abstract
Ca2+ signals regulate the function of many immune cells and promote immune responses to infection, cancer, and autoantigens. Ca2+ influx in immune cells is mediated by store-operated Ca2+ entry (SOCE) that results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels. The CRAC channel is formed by three plasma membrane proteins, ORAI1, ORAI2, and ORAI3. Of these, ORAI1 is the best studied and plays important roles in immune function. By contrast, the physiological role of ORAI3 in immune cells remains elusive. We show here that ORAI3 is expressed in many immune cells including macrophages, B cells, and T cells. To investigate ORAI3 function in immune cells, we generated Orai3-/- mice. The development of lymphoid and myeloid cells in the thymus and bone marrow was normal in Orai3-/- mice, as was the composition of immune cells in secondary lymphoid organs. Deletion of Orai3 did not affect SOCE in B cells and T cells but moderately enhanced SOCE in macrophages. Orai3-deficient macrophages, B cells, and T cells had normal effector functions in vitro. Immune responses in vivo, including humoral immunity (T cell dependent or independent) and antitumor immunity, were normal in Orai3-/- mice. Moreover, Orai3-/- mice showed no differences in susceptibility to septic shock, experimental autoimmune encephalomyelitis, or collagen-induced arthritis. We conclude that despite its expression in myeloid and lymphoid cells, ORAI3 appears to be dispensable or redundant for physiological and pathological immune responses mediated by these cells.
Collapse
Affiliation(s)
- Liwei Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Lucile Noyer
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Yin-Hu Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Anthony Y. Tao
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Wenyi Li
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Jingjie Zhu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Pedro Saavedra
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Syed T. Hoda
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Jun Yang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, NY,Correspondence to Stefan Feske:
| |
Collapse
|
24
|
Du Y, Ah Kioon MD, Laurent P, Chaudhary V, Pierides M, Yang C, Oliver D, Ivashkiv LB, Barrat FJ. Chemokines form nanoparticles with DNA and can superinduce TLR-driven immune inflammation. J Exp Med 2022; 219:e20212142. [PMID: 35640018 PMCID: PMC9161158 DOI: 10.1084/jem.20212142] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/24/2022] [Accepted: 05/12/2022] [Indexed: 12/13/2022] Open
Abstract
Chemokines control the migratory patterns and positioning of immune cells to organize immune responses to pathogens. However, many chemokines have been associated with systemic autoimmune diseases that have chronic IFN signatures. We report that a series of chemokines, including CXCL4, CXCL10, CXCL12, and CCL5, can superinduce type I IFN (IFN-I) by TLR9-activated plasmacytoid DCs (pDCs), independently of their respective known chemokine receptors. Mechanistically, we show that chemokines such as CXCL4 mediate transcriptional and epigenetic changes in pDCs, mostly targeted to the IFN-I pathways. We describe that chemokines physically interact with DNA to form nanoparticles that promote clathrin-mediated cellular uptake and delivery of DNA in the early endosomes of pDCs. Using two separate mouse models of skin inflammation, we observed the presence of CXCL4 associated with DNA in vivo. These data reveal a noncanonical role for chemokines to serve as nucleic acid delivery vectors to modulate TLR signaling, with implications for the chronic presence of IFN-I by pDCs in autoimmune diseases.
Collapse
Affiliation(s)
- Yong Du
- HSS Research Institute, Hospital for Special Surgery, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY
| | | | - Paoline Laurent
- HSS Research Institute, Hospital for Special Surgery, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY
| | - Vidyanath Chaudhary
- HSS Research Institute, Hospital for Special Surgery, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY
| | - Michael Pierides
- HSS Research Institute, Hospital for Special Surgery, New York, NY
| | - Chao Yang
- HSS Research Institute, Hospital for Special Surgery, New York, NY
| | - David Oliver
- HSS Research Institute, Hospital for Special Surgery, New York, NY
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
| | - Lionel B. Ivashkiv
- HSS Research Institute, Hospital for Special Surgery, New York, NY
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
- Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY
| | - Franck J. Barrat
- HSS Research Institute, Hospital for Special Surgery, New York, NY
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY
| |
Collapse
|
25
|
Turley JL, Lavelle EC. Resolving adjuvant mode of action to enhance vaccine efficacy. Curr Opin Immunol 2022; 77:102229. [PMID: 35779364 DOI: 10.1016/j.coi.2022.102229] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022]
Abstract
Adjuvants are a miscellaneous range of molecules and materials that can enhance the magnitude, functionality, breadth and durability of immune responses. Despite the multiplicity of compounds with adjuvant properties, less than a dozen are in clinical use in vaccines against infectious diseases. While many factors have contributed to their slow development, among the major challenges are the high safety and efficacy standards set by current adjuvants in human vaccines and our limited understanding of how adjuvants mediate their effects. This review outlines why it is so difficult to elucidate their mechanism of action, highlights areas that require in-depth research and discusses recent advancements that are revitalising adjuvant development. It is hoped that a fuller understanding of adjuvant sensing, signalling and function will facilitate the design of vaccines that promote sustained protective immunity against challenging bacterial and viral pathogens.
Collapse
Affiliation(s)
- Joanna L Turley
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02R590, Ireland.
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02R590, Ireland.
| |
Collapse
|
26
|
Yang C, Bachu M, Du Y, Brauner C, Yuan R, Ah Kioon MD, Chesi G, Barrat FJ, Ivashkiv LB. CXCL4 synergizes with TLR8 for TBK1-IRF5 activation, epigenomic remodeling and inflammatory response in human monocytes. Nat Commun 2022; 13:3426. [PMID: 35701499 PMCID: PMC9195402 DOI: 10.1038/s41467-022-31132-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 06/06/2022] [Indexed: 01/11/2023] Open
Abstract
Regulation of endosomal Toll-like receptor (TLR) responses by the chemokine CXCL4 is implicated in inflammatory and fibrotic diseases, with CXCL4 proposed to potentiate TLR responses by binding to nucleic acid TLR ligands and facilitating their endosomal delivery. Here we report that in human monocytes/macrophages, CXCL4 initiates signaling cascades and downstream epigenomic reprogramming that change the profile of the TLR8 response by selectively amplifying inflammatory gene transcription and interleukin (IL)-1β production, while partially attenuating the interferon response. Mechanistically, costimulation by CXCL4 and TLR8 synergistically activates TBK1 and IKKε, repurposes these kinases towards an inflammatory response via coupling with IRF5, and activates the NLRP3 inflammasome. CXCL4 signaling, in a cooperative and synergistic manner with TLR8, induces chromatin remodeling and activates de novo enhancers associated with inflammatory genes. Our findings thus identify new regulatory mechanisms of TLR responses relevant for cytokine storm, and suggest targeting the TBK1-IKKε-IRF5 axis may be beneficial in inflammatory diseases.
Collapse
Affiliation(s)
- Chao Yang
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Mahesh Bachu
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Yong Du
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Caroline Brauner
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Ruoxi Yuan
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Giancarlo Chesi
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Franck J Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Lionel B Ivashkiv
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA.
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Sabatini A, Guerrera G, Corsetti M, Ruocco G, De Bardi M, Renzi S, Cavalieri D, Battistini L, Angelini DF, Volpe E. Human Conventional and Plasmacytoid Dendritic Cells Differ in Their Ability to Respond to Saccharomyces cerevisiae. Front Immunol 2022; 13:850404. [PMID: 35634316 PMCID: PMC9131191 DOI: 10.3389/fimmu.2022.850404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
Saccharomyces cerevisiae is a commensal yeast colonizer of mucosal surfaces and an emerging opportunistic pathogen in the mucosa and bloodstream. The role of S. cerevisiae has been largely characterized in peripheral blood mononuclear cells and monocyte-derived dendritic cells, where yeast cells induce the production of inflammatory cytokines through the interaction with mannose receptors, chitin receptors, DC SIGN, and dectin1. However, the response of blood-circulating dendritic cells (DCs) to S. cerevisiae has never been investigated. Among blood DCs, conventional DCs (cDCs) are producers of inflammatory cytokines, while plasmacytoid DCs (pDCs) are a specialized population producing a large amount of interferon (IFN)-α, which is involved in the antiviral immune response. Here we report that both human DC subsets are able to sense S. cerevisiae. In particular, cDCs produce interleukin (IL)-6, express activation markers, and promotes T helper 17 cell polarization in response to yeasts, behaving similarly to monocyte-derived DCs as previously described. Interestingly, pDCs, not cDCs, sense fungal nucleic acids, leading to the generation of P1-pDCs (PD-L1+CD80–), a pDC subset characterized by the production of IFN-α and the induction of a Th profile producing IL-10. These results highlight a novel role of pDCs in response to S. cerevisiae that could be important for the regulation of the host microbiota–immune system balance and of anti-fungal immune response.
Collapse
Affiliation(s)
- Andrea Sabatini
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy.,Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Gisella Guerrera
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Marta Corsetti
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Gabriella Ruocco
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Sonia Renzi
- Department of Biology, University of Florence, Florence, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Daniela Francesca Angelini
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
28
|
Facciolà A, Visalli G, Laganà A, Di Pietro A. An Overview of Vaccine Adjuvants: Current Evidence and Future Perspectives. Vaccines (Basel) 2022; 10:vaccines10050819. [PMID: 35632575 PMCID: PMC9147349 DOI: 10.3390/vaccines10050819] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Vaccinations are one of the most important preventive tools against infectious diseases. Over time, many different types of vaccines have been developed concerning the antigen component. Adjuvants are essential elements that increase the efficacy of vaccination practises through many different actions, especially acting as carriers, depots, and stimulators of immune responses. For many years, few adjuvants have been included in vaccines, with aluminium salts being the most commonly used adjuvant. However, recent research has focused its attention on many different new compounds with effective adjuvant properties and improved safety. Modern technologies such as nanotechnologies and molecular biology have forcefully entered the production processes of both antigen and adjuvant components, thereby improving vaccine efficacy. Microparticles, emulsions, and immune stimulators are currently in the spotlight for their huge potential in vaccine production. Although studies have reported some potential side effects of vaccine adjuvants such as the recently recognised ASIA syndrome, the huge worth of vaccines remains unquestionable. Indeed, the recent COVID-19 pandemic has highlighted the importance of vaccines, especially in regard to managing future potential pandemics. In this field, research into adjuvants could play a leading role in the production of increasingly effective vaccines.
Collapse
Affiliation(s)
- Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Correspondence:
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| | - Antonio Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Multi-Specialist Clinical Institute for Orthopaedic Trauma Care (COT), 98124 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| |
Collapse
|
29
|
Tu ATT, Hoshi K, Shobo M, Yamazaki T. G-quadruplex-based CpG oligodeoxynucleotide/DOTAP complex strongly stimulates immunity in CpG motif-specific and loop-length-dependent manners. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102508. [PMID: 34906721 DOI: 10.1016/j.nano.2021.102508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/11/2021] [Accepted: 11/19/2021] [Indexed: 06/14/2023]
Abstract
Guanine-quadruplex (G4) oligodeoxynucleotides (ODNs) that contain unmethylated cytosine-phosphate-guanine motifs (G4 CpG ODN) with phosphodiester backbones are safer than the phosphorothioate (PT)-modified CpG ODNs recently used as vaccine adjuvants. However, cellular uptake and the nuclease stability of G4 CpG ODNs are still insufficient, resulting in lower immunostimulatory activity than PT-modified CpG ODNs. We aimed to enhance the immunostimulatory properties of G4 CpG ODNs by complexing with the cationic liposome 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP). The complex acquired nuclease resistance and improved cellular uptake. The immunostimulatory activity of the G4 CpG ODN-DOTAP lipoplexes was enhanced to a level comparable to that of PT-modified ODNs. In addition, the lipoplexes based on unmodified G4 CpG ODNs demonstrated CpG motif-specific immunostimulant activity, although PT-modified ODNs lacking the CpG motif could activate human immune cells. Interestingly, G4 CpG ODN-DOTAP lipoplexes induced interferon-α production in a loop-length dependent manner.
Collapse
Affiliation(s)
- Anh Thi Tram Tu
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan; Division of Life Science, Hokkaido university, Sapporo, Japan
| | - Kazuaki Hoshi
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan
| | - Miwako Shobo
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan
| | - Tomohiko Yamazaki
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan; Division of Life Science, Hokkaido university, Sapporo, Japan.
| |
Collapse
|
30
|
Acharya M, Jackson SW. Regulatory strategies limiting endosomal Toll-like receptor activation in B cells. Immunol Rev 2022; 307:66-78. [PMID: 35040152 PMCID: PMC8986562 DOI: 10.1111/imr.13065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/26/2022]
Abstract
The recognition of pathogen-associated nucleic acid (NA) promotes effective immunity against invading pathogens. However, endosomal Toll-like receptor (TLR) activation by self-NA also underlies the pathogenesis of systemic autoimmune diseases, such as systemic lupus erythematosus (SLE). For this reason, the activation thresholds of NA-sensing TLRs must be tightly regulated to balance protective and pathogenic immune responses. In this study, we will provide an overview of the evolutionary mechanisms designed to limit the aberrant activation of endosomal TLRs by self-ligands, focusing on four broad strategies. These include the following: 1) the production of nucleases able to degrade self-DNA and RNA; 2) the cell-specific regulation of endosomal TLR expression; 3) the spatial and temporal control of TLR positioning at a sub-cellular level; and 4) the modulation of downstream TLR signaling cascades. Given the critical role of B cells in lupus pathogenesis, where possible, we will describe evidence for B cell-specific induction of these regulatory mechanisms. We will also highlight our own work showing how modulation of B cell endolysosomal flux tunes NA-sensing TLR activation signals. In the face of inevitable generation of self-NA during normal cellular turnover, these parallel mechanisms are vital to protect against pathogenic inflammation.
Collapse
Affiliation(s)
- Mridu Acharya
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.,Seattle Children's Research Institute, Seattle, Washington, USA
| | - Shaun W Jackson
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.,Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
31
|
Baculovirus Vectors Induce the Production of Interferons in Swine: Their Potential in the Development of Antiviral Strategies. Vet Sci 2021; 8:vetsci8110278. [PMID: 34822651 PMCID: PMC8617851 DOI: 10.3390/vetsci8110278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
The huge variety of viruses affecting swine represents a global threat. Since vaccines against highly contagious viruses last several days to induce protective immune responses, antiviral strategies for rapid control of outbreak situations are needed. The baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV), an insect virus, has been demonstrated to be an effective vaccine vector for mammals. Besides the ability to display or transduce heterologous antigens, it also induces strong innate immune responses and provides IFN-mediated protection against lethal challenges with viruses like foot-and-mouth disease virus (FMDV) in mice. Thus, the aim of this study was to evaluate the ability of AcMNPV to induce IFN production and elicit antiviral activity in porcine peripheral blood mononuclear cells (PBMCs). Our results demonstrated that AcMNPV induced an IFN-α-mediated antiviral activity in PBMCs in vitro. Moreover, the inoculation of AcMNPV in piglets led to the production of type I and II IFNs in sera from inoculated animals and antiviral activities against vesicular stomatitis virus (VSV) and FMDV measured by in vitro assays. Finally, it was demonstrated that the pseudotyping of AcMNPV with VSV-G protein, but not the enrichment of the AcMNPV genome with specific immunostimulatory CpG motifs for the porcine TLR9, improved the ability to induce IFN-α production in PBMCs in vitro. Together, these results suggest that AcMNPV is a promising tool for the induction of IFNs in antiviral strategies, with the potential to be biotechnologically improved.
Collapse
|
32
|
Giza HM, Bozzacco L. Unboxing dendritic cells: Tales of multi-faceted biology and function. Immunology 2021; 164:433-449. [PMID: 34309853 PMCID: PMC8517577 DOI: 10.1111/imm.13394] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Often referred to as the bridge between innate and adaptive immunity, dendritic cells (DCs) are professional antigen-presenting cells (APCs) that constitute a unique, yet complex cell system. Among other APCs, DCs display the unique property of inducing protective immune responses against invading microbes, or cancer cells, while safeguarding the proper homeostatic equilibrium of the immune system and maintaining self-tolerance. Unsurprisingly, DCs play a role in many diseases such as autoimmunity, allergy, infectious disease and cancer. This makes them attractive but challenging targets for therapeutics. Since their initial discovery, research and understanding of DC biology have flourished. We now recognize the presence of multiple subsets of DCs distributed across tissues. Recent studies of phenotype and gene expression at the single cell level have identified heterogeneity even within the same DC type, supporting the idea that DCs have evolved to greatly expand the flexibility of the immune system to react appropriately to a wide range of threats. This review is meant to serve as a quick and robust guide to understand the basic divisions of DC subsets and their role in the immune system. Between mice and humans, there are some differences in how these subsets are identified and function, and we will point out specific distinctions as necessary. Throughout the text, we are using both fundamental and therapeutic lens to describe overlaps and distinctions and what this could mean for future research and therapies.
Collapse
|
33
|
Huppert LA, Mariotti V, Chien AJ, Soliman HH. Emerging immunotherapeutic strategies for the treatment of breast cancer. Breast Cancer Res Treat 2021; 191:243-255. [PMID: 34716870 DOI: 10.1007/s10549-021-06406-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022]
Abstract
Immunotherapy has resulted in unprecedented gains in long-term outcomes for many cancer types and has revolutionized the treatment landscape of solid tumor oncology. Checkpoint inhibition in combination with chemotherapy has proven to be effective for the treatment of a subset of advanced triple-negative breast cancer in the first-line setting. This initial success is likely just the tip of the iceberg as there is much that remains unknown about how to best harness the immune system as a therapeutic strategy in all breast cancer subtypes. Therefore, numerous ongoing studies are currently underway to evaluate the safety and efficacy of immunotherapy in breast cancer. In this review, we will discuss emerging immunotherapeutic strategies for breast cancer treatment including the following: (1) Intratumoral therapies, (2) Anti-tumor vaccines, (3) B-specific T-cell engagers, and (4) Chimeric antigen receptor T-cell therapy, and (5) Emerging systemic immunotherapy strategies. For each topic, we will review the existing preclinical and clinical literature, discuss ongoing clinical trials, and highlight future directions in the field.
Collapse
Affiliation(s)
- Laura A Huppert
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | - A Jo Chien
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Hatem H Soliman
- Department of Breast Oncology, H Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
34
|
Ong GH, Lian BSX, Kawasaki T, Kawai T. Exploration of Pattern Recognition Receptor Agonists as Candidate Adjuvants. Front Cell Infect Microbiol 2021; 11:745016. [PMID: 34692565 PMCID: PMC8526852 DOI: 10.3389/fcimb.2021.745016] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Adjuvants are used to maximize the potency of vaccines by enhancing immune reactions. Components of adjuvants include pathogen-associated molecular patterns (PAMPs) and damage-associate molecular patterns (DAMPs) that are agonists for innate immune receptors. Innate immune responses are usually activated when pathogen recognition receptors (PRRs) recognize PAMPs derived from invading pathogens or DAMPs released by host cells upon tissue damage. Activation of innate immunity by PRR agonists in adjuvants activates acquired immune responses, which is crucial to enhance immune reactions against the targeted pathogen. For example, agonists for Toll-like receptors have yielded promising results as adjuvants, which target PRR as adjuvant candidates. However, a comprehensive understanding of the type of immunological reaction against agonists for PRRs is essential to ensure the safety and reliability of vaccine adjuvants. This review provides an overview of the current progress in development of PRR agonists as vaccine adjuvants, the molecular mechanisms that underlie activation of immune responses, and the enhancement of vaccine efficacy by these potential adjuvant candidates.
Collapse
Affiliation(s)
- Guang Han Ong
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Benedict Shi Xiang Lian
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Takumi Kawasaki
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| |
Collapse
|
35
|
Tseng JC, Yang JX, Liu YL, Su YW, Lee AYL, Chen YW, Liu KJ, Luo Y, Hong YR, Chuang TH. Sharpening up tumor microenvironment to enhance the efficacy of immune checkpoint blockade on head and neck cancer using a CpG-oligodeoxynucleotide. Cancer Immunol Immunother 2021; 71:1115-1128. [PMID: 34581869 PMCID: PMC9016021 DOI: 10.1007/s00262-021-03062-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/17/2021] [Indexed: 12/09/2022]
Abstract
Head and neck cancers are a type of life-threatening cancers characterized by an immunosuppressive tumor microenvironment. Only less than 20% of the patients respond to immune checkpoint blockade therapy, indicating the need for a strategy to increase the efficacy of immunotherapy for this type of cancers. Previously, we identified a type B CpG-oligodeoxynucleotide (CpG-ODN) called CpG-2722, which has the universal activity of eliciting an immune response in grouper, mouse, and human cells. In this study, we further characterized and compared its cytokine-inducing profiles with different types of CpG-ODNs. The antitumor effect of CpG-2722 was further investigated alone and in combination with an immune checkpoint inhibitor in a newly developed syngeneic orthotopic head and neck cancer animal model. Along with other inflammatory cytokines, CpG-2722 induces the gene expressions of interleukin-12 and different types of interferons, which are critical for the antitumor response. Both CpG-2722 and anti-programmed death (PD)-1 alone suppressed tumor growth. Their tumor suppression efficacies were further enhanced when CpG-2722 and anti-PD-1 were used in combination. Mechanistically, CpG-2722 shaped a tumor microenvironment that is favorable for the action of anti-PD-1, which included promoting the expression of different cytokines such as IL-12, IFN-β, and IFN-γ, and increasing the presence of plasmacytoid dendritic cells, M1 macrophages, and CD8 positive T cells. Overall, CpG-2722 provided a priming effect for CD8 positive T cells by sharpening the tumor microenvironment, whereas anti-PD-1 released the brake for their tumor-killing effect, resulting in an enhanced efficacy of the combined CpG-2722 and anti-PD-1.
Collapse
Affiliation(s)
- Jen-Chih Tseng
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Jing-Xing Yang
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Yi-Ling Liu
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Ya-Wen Chen
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Yi-Ren Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
- Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
36
|
Greene TT, Zuniga EI. Type I Interferon Induction and Exhaustion during Viral Infection: Plasmacytoid Dendritic Cells and Emerging COVID-19 Findings. Viruses 2021; 13:1839. [PMID: 34578420 PMCID: PMC8472174 DOI: 10.3390/v13091839] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 01/12/2023] Open
Abstract
Type I Interferons (IFN-I) are a family of potent antiviral cytokines that act through the direct restriction of viral replication and by enhancing antiviral immunity. However, these powerful cytokines are a caged lion, as excessive and sustained IFN-I production can drive immunopathology during infection, and aberrant IFN-I production is a feature of several types of autoimmunity. As specialized producers of IFN-I plasmacytoid (p), dendritic cells (DCs) can secrete superb quantities and a wide breadth of IFN-I isoforms immediately after infection or stimulation, and are the focus of this review. Notably, a few days after viral infection pDCs tune down their capacity for IFN-I production, producing less cytokines in response to both the ongoing infection and unrelated secondary stimulations. This process, hereby referred to as "pDC exhaustion", favors viral persistence and associates with reduced innate responses and increased susceptibility to secondary opportunistic infections. On the other hand, pDC exhaustion may be a compromise to avoid IFN-I driven immunopathology. In this review we reflect on the mechanisms that initially induce IFN-I and subsequently silence their production by pDCs during a viral infection. While these processes have been long studied across numerous viral infection models, the 2019 coronavirus disease (COVID-19) pandemic has brought their discussion back to the fore, and so we also discuss emerging results related to pDC-IFN-I production in the context of COVID-19.
Collapse
Affiliation(s)
| | - Elina I. Zuniga
- Division of Biological Sciences, University of California, San Diego, CA 92093, USA;
| |
Collapse
|
37
|
Tran TTP, Tran TH, Kremer EJ. IgG-Complexed Adenoviruses Induce Human Plasmacytoid Dendritic Cell Activation and Apoptosis. Viruses 2021; 13:1699. [PMID: 34578281 PMCID: PMC8472521 DOI: 10.3390/v13091699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
Following repeat exposure to many human adenoviruses (HAdVs), most adults harbour long-lived B- and T-cell responses. Combined, this response typically protects us for years from re-infection by the same HAdV type. In spite of these immune responses, some HAdV types are associated with persistent infections that constitute a life-threatening risk when an individual's T-cell response is compromised. By contrast, patients with B-cell deficiencies do not appear to be at a greater risk of HAdV disease. This dichotomy begs the question of the secondary role of anti-HAdV antibodies during host defence. In this study, we explored IgG-complexed (IC)-HAdV5 and primary human plasmacytoid dendritic cell (pDC) interactions. We found that IC-HAdV5 are efficiently internalized in pDCs, stimulate their activation through TLR9 signalling, and cause apoptosis. These data may help reconcile the enigma of robust immune response to HAdVs, while concurrently allowing persistence.
Collapse
Affiliation(s)
- Thi Thu Phuong Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, 34090 Montpellier, France; (T.T.P.T.); (T.H.T.)
- Department of Life Sciences, University of Science and Technology of Hanoi Vietnam Academy of Science and Technology, Hanoi 100000, Vietnam
| | - Tuan Hiep Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, 34090 Montpellier, France; (T.T.P.T.); (T.H.T.)
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
- PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, Hanoi 11313, Vietnam
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, 34090 Montpellier, France; (T.T.P.T.); (T.H.T.)
| |
Collapse
|
38
|
Liu B, Huang J, Ashraf A, Rahaman O, Lou J, Wang L, Cai P, Wen J, Anwaar S, Liu X, Ni H, Ganguly D, Zhao J, Yang CY. The RNase MCPIP3 promotes skin inflammation by orchestrating myeloid cytokine response. Nat Commun 2021; 12:4105. [PMID: 34215755 PMCID: PMC8253787 DOI: 10.1038/s41467-021-24352-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
CCCH zinc finger proteins resolve immune responses by degrading the mRNAs of inflammatory cytokines such as tumor necrosis factor (TNF) and interleukin (IL)-6. Here we report that one such family member, monocyte chemotactic protein-induced protein 3 (MCPIP3, also named ZC3H12C or Regnase-3), promotes skin inflammation by simultaneously enhancing TNF in macrophages and repressing IL-6 in plasmacytoid dendritic cells (pDCs). MCPIP3 is positively associated with psoriasis pathogenesis, and highly expressed by macrophages and pDCs. MCPIP3-deficient macrophages produce less TNF and IL-12p40. However, MCPIP3-deficient pDCs secrete significantly more IL-6. This enhanced intradermal IL-6 may alleviate imiquimod-induced skin inflammation. As a result, MCPIP3-deficient mice are protected from imiquimod-induced psoriasiform lesions. Furthermore, early exposure to pDC-derived IL-6 suppresses macrophage-derived TNF and IL-12p40. Mechanistically, MCPIP3 could directly degrade mRNAs of IL-6, Regnase-1, and IκBζ. In turn, Regnase-1 could degrade MCPIP3 mRNAs. Our study identifies a critical post-transcriptional mechanism that synchronizes myeloid cytokine secretion to initiate autoimmune skin inflammation. Zinc finger proteins are involved in the resolution of immune responses and function by degrading mRNA of inflammatory cytokines. Here the authors show MCPIP3 promotes skin inflammation via modification of cytokine profiles in pDCs and macrophages.
Collapse
Affiliation(s)
- Bo Liu
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Jiancheng Huang
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Amina Ashraf
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Oindrila Rahaman
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Jing Lou
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Ling Wang
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Peiliang Cai
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Jinping Wen
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Shoaib Anwaar
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Xiaoli Liu
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Hai Ni
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China
| | - Dipyaman Ganguly
- IICB-Translational Research Unit of Excellence, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Jijun Zhao
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Cliff Y Yang
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, Guangdong, China. .,Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, China.
| |
Collapse
|
39
|
Dieu RS, Wais V, Sørensen MZ, Marczynska J, Dubik M, Kavan S, Thomassen M, Burton M, Kruse T, Khorooshi R, Owens T. Central Nervous System-Endogenous TLR7 and TLR9 Induce Different Immune Responses and Effects on Experimental Autoimmune Encephalomyelitis. Front Neurosci 2021; 15:685645. [PMID: 34211367 PMCID: PMC8241214 DOI: 10.3389/fnins.2021.685645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/19/2021] [Indexed: 11/13/2022] Open
Abstract
Innate receptors, including Toll like receptors (TLRs), are implicated in pathogenesis of CNS inflammatory diseases such as multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). TLR response to pathogens or endogenous signals includes production of immunoregulatory mediators. One of these, interferon (IFN)β, a Type I IFN, plays a protective role in MS and EAE. We have previously shown that intrathecal administration of selected TLR ligands induced IFNβ and infiltration of blood-derived myeloid cells into the central nervous system (CNS), and suppressed EAE in mice. We have now extended these studies to evaluate a potential therapeutic role for CNS-endogenous TLR7 and TLR9. Intrathecal application of Imiquimod (TLR7 ligand) or CpG oligonucleotide (TLR9 ligand) into CNS of otherwise unmanipulated mice induced IFNβ expression, with greater magnitude in response to CpG. CD45+ cells in the meninges were identified as source of IFNβ. Intrathecal CpG induced infiltration of monocytes, neutrophils, CD4+ T cells and NK cells whereas Imiquimod did not recruit blood-derived CD45+ cells. CpG, but not Imiquimod, had a beneficial effect on EAE, when given at time of disease onset. This therapeutic effect of CpG on EAE was not seen in mice lacking the Type I IFN receptor. In mice with EAE treated with CpG, the proportion of monocytes was significantly increased in the CNS. Infiltrating cells were predominantly localized to spinal cord meninges and demyelination was significantly reduced compared to non-treated mice with EAE. Our findings show that TLR7 and TLR9 signaling induce distinct inflammatory responses in the CNS with different outcome in EAE and point to recruitment of blood-derived cells and IFNβ induction as possible mechanistic links between TLR9 stimulation and amelioration of EAE. The protective role of TLR9 signaling in the CNS may have application in treatment of diseases such as MS.
Collapse
Affiliation(s)
- Ruthe Storgaard Dieu
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Vian Wais
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Michael Zaucha Sørensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Joanna Marczynska
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Magdalena Dubik
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Stephanie Kavan
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mark Burton
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Torben Kruse
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
40
|
Pulendran B, S Arunachalam P, O'Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov 2021; 20:454-475. [PMID: 33824489 PMCID: PMC8023785 DOI: 10.1038/s41573-021-00163-y] [Citation(s) in RCA: 799] [Impact Index Per Article: 199.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Adjuvants are vaccine components that enhance the magnitude, breadth and durability of the immune response. Following its introduction in the 1920s, alum remained the only adjuvant licensed for human use for the next 70 years. Since the 1990s, a further five adjuvants have been included in licensed vaccines, but the molecular mechanisms by which these adjuvants work remain only partially understood. However, a revolution in our understanding of the activation of the innate immune system through pattern recognition receptors (PRRs) is improving the mechanistic understanding of adjuvants, and recent conceptual advances highlight the notion that tissue damage, different forms of cell death, and metabolic and nutrient sensors can all modulate the innate immune system to activate adaptive immunity. Furthermore, recent advances in the use of systems biology to probe the molecular networks driving immune response to vaccines ('systems vaccinology') are revealing mechanistic insights and providing a new paradigm for the vaccine discovery and development process. Here, we review the 'known knowns' and 'known unknowns' of adjuvants, discuss these emerging concepts and highlight how our expanding knowledge about innate immunity and systems vaccinology are revitalizing the science and development of novel adjuvants for use in vaccines against COVID-19 and future pandemics.
Collapse
Affiliation(s)
- Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Chemistry, Engineering & Medicine for Human Health, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
41
|
Amadio R, Piperno GM, Benvenuti F. Self-DNA Sensing by cGAS-STING and TLR9 in Autoimmunity: Is the Cytoskeleton in Control? Front Immunol 2021; 12:657344. [PMID: 34084165 PMCID: PMC8167430 DOI: 10.3389/fimmu.2021.657344] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Modified or misplaced DNA can be recognized as a danger signal by mammalian cells. Activation of cellular responses to DNA has evolved as a defense mechanism to microbial infections, cellular stress, and tissue damage, yet failure to control this mechanism can lead to autoimmune diseases. Several monogenic and multifactorial autoimmune diseases have been associated with type-I interferons and interferon-stimulated genes (ISGs) induced by deregulated recognition of self-DNA. Hence, understanding how cellular mechanism controls the pathogenic responses to self-nucleic acid has important clinical implications. Fine-tuned membrane trafficking and cellular compartmentalization are two major factors that balance activation of DNA sensors and availability of self-DNA ligands. Intracellular transport and organelle architecture are in turn regulated by cytoskeletal dynamics, yet the precise impact of actin remodeling on DNA sensing remains elusive. This review proposes a critical analysis of the established and hypothetical connections between self-DNA recognition and actin dynamics. As a paradigm of this concept, we discuss recent evidence of deregulated self-DNA sensing in the prototypical actin-related primary immune deficiency (Wiskott-Aldrich syndrome). We anticipate a broader impact of actin-dependent processes on tolerance to self-DNA in autoimmune disorders.
Collapse
Affiliation(s)
- Roberto Amadio
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Biomedical Sciences, Venetian Institute of Molecular Medicine, University of Padova, Padova, Italy
| | - Giulia Maria Piperno
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Federica Benvenuti
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| |
Collapse
|
42
|
Synthetic Hexanucleotides as a Tool to Overcome Excessive Neutrophil Activation Caused by CpG-Containing Oligonucleotides. Pathogens 2021; 10:pathogens10050530. [PMID: 33924760 PMCID: PMC8146577 DOI: 10.3390/pathogens10050530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/16/2022] Open
Abstract
Mimicking bacterial DNA, synthetic CpG-containing oligodeoxyribonucleotides (CpG-ODNs) have a powerful immunomodulatory potential. Their practical application is mainly associated with the production of vaccines, where they are used as adjuvants, as well as in local antimicrobial therapy. CpG-ODNs act on a wide variety of immune cells, including neutrophilic granulocytes. On the one hand, the stimulatory effect provides both the direct implementation of their antimicrobial and fungicidal mechanisms, and an avalanche-like strengthening of the immune signal due to interaction with other participants in the immune process. On the other hand, hyperactivation of neutrophilic granulocytes can have negative consequences. In particular, the formation of unreasonably high amounts of reactive oxygen species leads to tissue damages and, as a consequence, a spontaneous aggravation and prolongation of the inflammatory process. Under physiological conditions, a large number of DNA fragments are present in inflammation foci: both of microbial and self-tissue origin. We investigated effects of several short modified hexanucleotides on the main indicators of neutrophil activation, as well as their influence on the immunomodulatory activity of known synthetic CpG-ODNs. The results obtained show that short oligonucleotides partially inhibit the prooxidant effect of synthetic CpG-ODNs without significantly affecting the ability of the latter to overcome bacteria-induced pro-survival effects on neutrophilic granulocytes.
Collapse
|
43
|
Abstract
CpG Oligonucleotides (ODN) are immunomodulatory synthetic oligonucleotides specifically designed to stimulate Toll-like receptor 9. TLR9 is expressed on human plasmacytoid dendritic cells and B cells and triggers an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. This chapter reviews recent progress in understanding the mechanism of action of CpG ODN and provides an overview of human clinical trial results using CpG ODN to improve vaccines for the prevention/treatment of cancer, allergy, and infectious disease.
Collapse
Affiliation(s)
| | | | - Dennis M Klinman
- National Cancer Institute, NIH, Frederick, MD, USA.
- Leitman Klinman Consulting, Potomac, MD, USA.
| |
Collapse
|
44
|
Montamat G, Leonard C, Poli A, Klimek L, Ollert M. CpG Adjuvant in Allergen-Specific Immunotherapy: Finding the Sweet Spot for the Induction of Immune Tolerance. Front Immunol 2021; 12:590054. [PMID: 33708195 PMCID: PMC7940844 DOI: 10.3389/fimmu.2021.590054] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Prevalence and incidence of IgE-mediated allergic diseases have increased over the past years in developed and developing countries. Allergen-specific immunotherapy (AIT) is currently the only curative treatment available for allergic diseases that has long-term efficacy. Although AIT has been proven successful as an immunomodulatory therapy since its beginnings, it still faces several unmet needs and challenges today. For instance, some patients can experience severe side effects, others are non-responders, and prolonged treatment schedules can lead to lack of patient adherence and therapy discontinuation. A common strategy to improve AIT relies on the use of adjuvants and immune modulators to boost its effects and improve its safety. Among the adjuvants tested for their clinical efficacy, CpG oligodeoxynucleotide (CpG-ODN) was investigated with limited success and without reaching phase III trials for clinical allergy treatment. However, recently discovered immune tolerance-promoting properties of CpG-ODN place this adjuvant again in a prominent position as an immune modulator for the treatment of allergic diseases. Indeed, it has been shown that the CpG-ODN dose and concentration are crucial in promoting immune regulation through the recruitment of pDCs. While low doses induce an inflammatory response, high doses of CpG-ODN trigger a tolerogenic response that can reverse a pre-established allergic milieu. Consistently, CpG-ODN has also been found to stimulate IL-10 producing B cells, so-called B regulatory cells (Bregs). Accordingly, CpG-ODN has shown its capacity to prevent and revert allergic reactions in several animal models showing its potential as both preventive and active treatment for IgE-mediated allergy. In this review, we describe how CpG-ODN-based therapies for allergic diseases, despite having shown limited success in the past, can still be exploited further as an adjuvant or immune modulator in the context of AIT and deserves additional attention. Here, we discuss the past and current knowledge, which highlights CpG-ODN as a potential adjuvant to be reevaluated for the enhancement of AIT when used in appropriate conditions and formulations.
Collapse
Affiliation(s)
- Guillem Montamat
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Cathy Leonard
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Ludger Klimek
- Centre for Rhinology and Allergology, Wiesbaden, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
| |
Collapse
|
45
|
Montes-Cobos E, Huscher B, Engler JB, Woo MS, Binkle L, Bauer S, Kursawe N, Moles M, Friese MA, Ufer F. Voltage-Gated Proton Channel Hv1 Controls TLR9 Activation in Plasmacytoid Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:3001-3010. [PMID: 33127821 DOI: 10.4049/jimmunol.2000404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022]
Abstract
The voltage-gated proton channel Hv1 regulates proton fluxes across membranes, thereby influencing pH-dependent processes. Plasmacytoid dendritic cells (pDCs) require a particularly tight regulation of endosomal pH to ensure strong type I IFN secretion exclusively during infection, avoiding autoimmunity. However, whether Hv1 is important for pH control in pDCs is presently unknown. In this study, we show that mouse pDCs require Hv1 to achieve potent type I IFN responses after the recognition of foreign DNA by endosomal TLR9. Genetic disruption of Hvcn1, which encodes Hv1, impaired mouse pDC activation by CpG oligonucleotides in vitro and in vivo, reducing IFN-α secretion and the induction of IFN-stimulated genes. Mechanistically, Hvcn1 deficiency delayed endosomal acidification and enhanced intracellular reactive oxygen species production, consequently limiting protease activity and TLR9 signaling. Our study reveals a critical role of Hv1 during innate immune responses and places this channel as a key modulator of type I IFN production, the hallmark function of pDCs, commending Hv1 as an attractive target for modulating type I IFN-driven autoimmunity.
Collapse
Affiliation(s)
- Elena Montes-Cobos
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Britta Huscher
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marcel S Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lars Binkle
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Simone Bauer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Nina Kursawe
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michael Moles
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Friederike Ufer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
46
|
Chang CJ. Immune sensing of DNA and strategies for fish DNA vaccine development. FISH & SHELLFISH IMMUNOLOGY 2020; 101:252-260. [PMID: 32247047 DOI: 10.1016/j.fsi.2020.03.064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 03/24/2020] [Accepted: 03/30/2020] [Indexed: 05/21/2023]
Abstract
Studies of DNA vaccines have shown that understanding the mechanism of DNA vaccine-mediated action is the key for vaccine development. Current knowledge has shown the presence of antigen presenting cells (APCs) involving in B and T cells at the muscle injection site and the upregulation of type I interferon (IFN-I) that initiates antiviral response and benefits adaptive immunity in fish DNA vaccines. IFN-I may be triggered by expressed antigen such as the rhabdovirus G protein encoded DNA vaccine or by plasmid DNA itself through cytosolic DNA sensing. The investigating of Toll-like receptor 9, and 21 are the CpG-motif sensors in many fish species, and the cytosolic DNA receptors DDX41 and downstream STING signaling revealed the mechanisms for IFN-I production. This review article describes the recent finding of receptors for cytosolic DNA, the STING-TBK1-IRF signaling, and the possibility of turning these findings into strategies for the future development of DNA vaccines.
Collapse
Affiliation(s)
- Chia-Jung Chang
- Laboratory of Fish Immunology, Institute of Infectology, Friedrich-Loeffler-Institut, 17493, Greifswald-Insel Riems, Germany.
| |
Collapse
|
47
|
Chuang YC, Tseng JC, Huang LR, Huang CM, Huang CYF, Chuang TH. Adjuvant Effect of Toll-Like Receptor 9 Activation on Cancer Immunotherapy Using Checkpoint Blockade. Front Immunol 2020; 11:1075. [PMID: 32547560 PMCID: PMC7274158 DOI: 10.3389/fimmu.2020.01075] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy using checkpoint blockade has revolutionized cancer treatment, improving patient survival and quality of life. Nevertheless, the clinical outcomes of such immunotherapy are highly heterogeneous between patients. Depending on the cancer type, the patient response rates to this immunotherapy are limited to 20–30%. Based on the mechanism underlying the antitumor immune response, new therapeutic strategies have been designed with the aim of increasing the effectiveness and specificity of the antitumor immune response elicited by checkpoint blockade agents. The activation of toll-like receptor 9 (TLR9) by its synthetic agonists induces the antitumor response within the innate immunity arm, generating adjuvant effects and priming the adaptive immune response elicited by checkpoint blockade during the effector phase of tumor-cell killing. This review first describes the underlying mechanisms of action and current status of monotherapy using TLR9 agonists and immune checkpoint inhibitors for cancer immunotherapy. The rationale for combining these two agents is discussed, and evidence indicating the current status of such combination therapy as a novel cancer treatment strategy is presented.
Collapse
Affiliation(s)
- Yu-Chen Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Jen-Chih Tseng
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Li-Rung Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Chun-Ming Huang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
48
|
Wilson KL, Howard GP, Coatsworth H, Dinglasan RR, Mao HQ, Plebanski M. Biodegradable PLGA- b-PEG Nanoparticles Induce T Helper 2 (Th2) Immune Responses and Sustained Antibody Titers via TLR9 Stimulation. Vaccines (Basel) 2020; 8:E261. [PMID: 32485944 PMCID: PMC7349924 DOI: 10.3390/vaccines8020261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/20/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Sustained immune responses, particularly antibody responses, are key for protection against many endemic infectious diseases. Antibody responses are often accompanied by T helper (Th) cell immunity. Herein we study small biodegradable poly (ethylene glycol)-b-poly (lactic-co-glycolic acid) nanoparticles (PEG-b-PLGA NPs, 25-50 nm) as antigen- or adjuvant-carriers. The antigen carrier function of PEG-b-PLGA NPs was compared against an experimental benchmark polystyrene nanoparticles (PS NPs, 40-50 nm), both conjugated with the model antigen ovalbumin (OVA-PS NPs, and OVA-PEG-b-PLGA NPs). The OVA-PEG-b-PLGA NPs induced sustained antibody responses to Day 120 after two immunizations. The OVA-PEG-b-PLGA NPs as a self-adjuvanting vaccine further induced IL-4 producing T-helper cells (Th2), but not IFN-γ producing T-cells (Th1). The PEG-b-PLGA NPs as a carrier for CpG adjuvant (CpG-PEG-b-PLGA NPs) were also tested as mix-in vaccine adjuvants comparatively for protein antigens, or for protein-conjugated to PS NPs or to PEG-b-PLGA NPs. While the addition of this adjuvant NP did not further increase T-cell responses, it improved the consistency of antibody responses across all immunization groups. Together these data support further development of PEG-b-PLGA NPs as a vaccine carrier, particularly where it is desired to induce Th2 immunity and achieve sustained antibody titers in the absence of affecting Th1 immunity.
Collapse
Affiliation(s)
- Kirsty L. Wilson
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, Victoria 3084, Australia;
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3181, Australia
| | - Gregory P. Howard
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Heather Coatsworth
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Rhoel R. Dinglasan
- Emerging Pathogens Institute, Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, Victoria 3084, Australia;
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3181, Australia
| |
Collapse
|
49
|
Ye Y, Gaugler B, Mohty M, Malard F. Plasmacytoid dendritic cell biology and its role in immune-mediated diseases. Clin Transl Immunology 2020; 9:e1139. [PMID: 32489664 PMCID: PMC7248678 DOI: 10.1002/cti2.1139] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/26/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are a unique subset of dendritic cells specialised in secreting high levels of type I interferons. pDCs play a crucial role in antiviral immunity and have been implicated in the initiation and development of many autoimmune and inflammatory diseases. This review summarises the latest advances in recent years in several aspects of pDC biology, with special focus on pDC heterogeneity, pDC development via the lymphoid pathway, and newly identified proteins/pathways involved in pDC trafficking, nucleic acid sensing and interferon production. Finally, we also highlight the current understanding of pDC involvement in autoimmunity and alloreactivity, and opportunities for pDC‐targeting therapies in these diseases. These new insights have contributed to answers to several fundamental questions remaining in pDC biology and may pave the way to successful pDC‐targeting therapy in the future.
Collapse
Affiliation(s)
- Yishan Ye
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Bone Marrow Transplantation Center The First Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
| | - Béatrice Gaugler
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France
| | - Mohamad Mohty
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Service d'Hématologie Clinique et Thérapie Cellulaire AP-HP, Hôpital Saint-Antoine Sorbonne Université Paris France
| | - Florent Malard
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Service d'Hématologie Clinique et Thérapie Cellulaire AP-HP, Hôpital Saint-Antoine Sorbonne Université Paris France
| |
Collapse
|
50
|
Prevention of liver metastases through perioperative acute CpG-C immune stimulation. Cancer Immunol Immunother 2020; 69:2021-2031. [PMID: 32405793 DOI: 10.1007/s00262-020-02596-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 04/27/2020] [Indexed: 12/21/2022]
Abstract
Following excision of colorectal tumors, metastatic disease is prevalent, primarily occurs in the liver, and is highly predictive of poor prognosis. The perioperative period is now recognized as critical in determining the incidence of postoperative metastases and long-term cancer outcomes. Thus, various perioperative prophylactic interventions are currently studied during this time frame. However, immune stimulation during the perioperative period has rarely been attempted due to specific contraindications to surgery and various adverse effects. Here, to prevent liver metastases, we perioperatively employed a TLR-9 agonist, CpG-C, which exhibits minimal pyrogenic and other adverse effects in patients. We found that marginating-hepatic (MH) cells in BALB/c mice contained high percentage of NK cells, but exhibited negligible NK cytotoxicity, as previously reported in humans. However, a single CpG-C administration (25-100 µg/mouse) doubled MH-NK cell numbers, increased NK cell activation and maturation markers (NKp46, CD11b), decreased the inhibitory NKG2A ligand, and dramatically increased MH-NK-cell cytotoxicity against the syngeneic CT26 colon cancer line. Moreover, in operated mice, this innocuous intervention also markedly improved resistance to CT26 and MC38 hepatic metastases in BALB/c and C57BL/6 mice, respectively. Beneficial effects of CpG-C were mediated through activation of MH-NK cells, as indicated by an in vivo NK depletion study. Last, CpG-C protected against surgery-induced suppression of MH-NK cytotoxicity and improved their activation indices. Thus, we suggest that systemic perioperative CpG-C treatment should be considered and studied as a novel therapeutic approach to improve long-term cancer outcomes in colorectal cancer patients.
Collapse
|