151
|
Kervevan J, Chakrabarti LA. Role of CD4+ T Cells in the Control of Viral Infections: Recent Advances and Open Questions. Int J Mol Sci 2021; 22:E523. [PMID: 33430234 PMCID: PMC7825705 DOI: 10.3390/ijms22020523] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/26/2022] Open
Abstract
CD4+ T cells orchestrate adaptive immune responses through their capacity to recruit and provide help to multiple immune effectors, in addition to exerting direct effector functions. CD4+ T cells are increasingly recognized as playing an essential role in the control of chronic viral infections. In this review, we present recent advances in understanding the nature of CD4+ T cell help provided to antiviral effectors. Drawing from our studies of natural human immunodeficiency virus (HIV) control, we then focus on the role of high-affinity T cell receptor (TCR) clonotypes in mediating antiviral CD4+ T cell responses. Last, we discuss the role of TCR affinity in determining CD4+ T cell differentiation, reviewing the at times divergent studies associating TCR signal strength to the choice of a T helper 1 (Th1) or a T follicular helper (Tfh) cell fate.
Collapse
Affiliation(s)
- Jérôme Kervevan
- Control of Chronic Viral Infections Group (CIVIC), Virus and Immunity Unit, Institut Pasteur, 75724 Paris, France;
- CNRS UMR, 3569 Paris, France
| | - Lisa A. Chakrabarti
- Control of Chronic Viral Infections Group (CIVIC), Virus and Immunity Unit, Institut Pasteur, 75724 Paris, France;
- CNRS UMR, 3569 Paris, France
| |
Collapse
|
152
|
Abstract
OBJECTIVES Immune activation, among others driven by interferon (IFN)-α and IFN-γ activation, is a main feature of progressive HIV infection. Suppressor of cytokine signaling (SOCS) 1 and 3 are negative feedback regulators of the IFN-α and IFN-γ axis. Here, we analyzed the role of 9 single-nucleotide polymorphisms (SNPs) within SOCS-1 and SOCS-3 genes for their association with an HIV progression rate in a cohort of 318 rapid vs 376 slow progressors from the Swiss HIV Cohort Study. DESIGN AND METHODS We analyzed 9 SNPs, which we have identified in Swiss blood donors, in a cohort of HIV-infected patients (n = 1144), which have been categorized according to the decline in CD4 T-cell counts. In all the conducted analyses, we focused on the comparison between rapid and slow progressors with regard to SNPs in SOCS-1 and SOCS-3 and with regard to haplotypes using multivariate logistic regression models. RESULTS Three SOCS-1 SNPs (rs193779, rs33989964, and rs4780355) are associated with a risk reduction for rapid progression. Two of these SNPs, rs33989964 and rs4780355, are in strong linkage disequilibrium, forming a frequent haplotype. Homozygous carriers of this haplotype are also associated with a risk reduction for rapid progression. By contrast, the minor TT genotype of rs33977706 is associated with twice the risk for rapid progression. No associations have been observed for the 4 SOCS-3 SNPs or the major SOCS-3 haplotypes. CONCLUSIONS Our data suggest that SNPs in SOCS-1 are associated with HIV disease progression and speak in favor that immune activation is causal for the progressive immunodeficiency.
Collapse
|
153
|
Merritt E, Londoño MC, Childs K, Whitehouse G, Kodela E, Sánchez-Fueyo A, Martínez-Llordella M. On the impact of hepatitis C virus and heterologous immunity on alloimmune responses following liver transplantation. Am J Transplant 2021; 21:247-257. [PMID: 32524678 DOI: 10.1111/ajt.16134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 01/25/2023]
Abstract
Virus-induced heterologous immunity is considered a barrier to transplantation tolerance. Yet, hepatitis C (HCV)-infected liver transplant (LT) patients occasionally achieve operational tolerance. We investigated the mechanisms through which HCV infection modulates donor-specific T cell responses following LT and the influence of HCV eradication. We generated T cell lines from HCV-infected LT and non-LT patients before and after HCV eradication and quantified alloreactive responses using cell lines expressing single-HLA class-I antigens in the presence/absence of PD-1/CTLA-4 blockade. HCV-specific CD8+ T cells cross-reacted with allogeneic class-I HLA molecules. HCV-positive LT recipients exhibited a higher proportion of CD8+ T cells coexpressing inhibitory receptors (PD-1/CTLA4) than HCV-negative LT, and their expression correlated with CXCL10 plasma levels. This resulted in decreased antidonor and third-party proliferative responses, which were significantly reversed by HCV eradication. PD-1/CTLA-4 blockade increased the proportion of HCV-specific CD8+ T cells reacting against donor only before viral clearance. In conclusion, HCV infection results in the generation of HCV-specific CD8+ T cells capable of reacting against allogeneic HLA molecules. Following LT, this results in a PD-1/CTLA4-dependent decrease in alloimmune responses. Our findings challenge the notion that heterologous immunity is necessarily detrimental in LT and provide an explanation for the association between HCV eradication and immune-mediated allograft damage.
Collapse
Affiliation(s)
- Elliot Merritt
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty e Sciences & Medicine, Institute of Liver Studies, King's College London, London, UK
| | - Maria-Carlota Londoño
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty e Sciences & Medicine, Institute of Liver Studies, King's College London, London, UK
- Liver Unit, Hospital Clínic Barcelona, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Kate Childs
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty e Sciences & Medicine, Institute of Liver Studies, King's College London, London, UK
| | - Gavin Whitehouse
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty e Sciences & Medicine, Institute of Liver Studies, King's College London, London, UK
| | - Elisavet Kodela
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty e Sciences & Medicine, Institute of Liver Studies, King's College London, London, UK
| | - Alberto Sánchez-Fueyo
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty e Sciences & Medicine, Institute of Liver Studies, King's College London, London, UK
| | - Marc Martínez-Llordella
- MRC Centre for Transplantation, Department of Inflammation Biology, Faculty e Sciences & Medicine, Institute of Liver Studies, King's College London, London, UK
| |
Collapse
|
154
|
Fu B, Wang D, Shen X, Guo C, Liu Y, Ye Y, Sun R, Li J, Tian Z, Wei H. Immunomodulation Induced During Interferon-α Therapy Impairs the Anti-HBV Immune Response Through CD24 +CD38 hi B Cells. Front Immunol 2020; 11:591269. [PMID: 33424840 PMCID: PMC7786281 DOI: 10.3389/fimmu.2020.591269] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Type I interferon is widely used for antiviral therapy, yet has yielded disappointing results toward chronic HBV infection. Here we identify that PEG-IFNα-2b therapy toward persistent infection in humans is a double-edged sword of both immunostimulation and immunomodulation. Our studies of this randomised trial showed persistent PEG-IFNα-2b therapy induced large number of CD24+CD38hi B cells and launched a CD24+CD38hi B cells centered immunosuppressive response, including downregulating functions of T cells and NK cells. Patients with low induced CD24+CD38hi B cells have achieved an improved therapeutic effect. Specifically, using the anti-CD24 antibody to deplete CD24+CD38hi B cells without harming other B cell subsets suggest a promising strategy to improve the therapeutic effects. Our findings show that PEG-IFNα-2b therapy toward persistent infection constitutes an immunomodulation effect, and strategies to identifying the molecular basis for the antiviral versus immunomodulatory effects of PEG-IFNα-2b to selectively manipulate these opposing activities provide an opportunity to ameliorate anti-virus immunity and control viral infection.
Collapse
Affiliation(s)
- Binqing Fu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Dongyao Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiaokun Shen
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Chuang Guo
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yanyan Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ying Ye
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
155
|
Fox LE, Locke MC, Lenschow DJ. Context Is Key: Delineating the Unique Functions of IFNα and IFNβ in Disease. Front Immunol 2020; 11:606874. [PMID: 33408718 PMCID: PMC7779635 DOI: 10.3389/fimmu.2020.606874] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Type I interferons (IFNs) are critical effector cytokines of the immune system and were originally known for their important role in protecting against viral infections; however, they have more recently been shown to play protective or detrimental roles in many disease states. Type I IFNs consist of IFNα, IFNβ, IFNϵ, IFNκ, IFNω, and a few others, and they all signal through a shared receptor to exert a wide range of biological activities, including antiviral, antiproliferative, proapoptotic, and immunomodulatory effects. Though the individual type I IFN subtypes possess overlapping functions, there is growing appreciation that they also have unique properties. In this review, we summarize some of the mechanisms underlying differential expression of and signaling by type I IFNs, and we discuss examples of differential functions of IFNα and IFNβ in models of infectious disease, cancer, and autoimmunity.
Collapse
Affiliation(s)
- Lindsey E. Fox
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Marissa C. Locke
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Deborah J. Lenschow
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
156
|
Kapoor T, Corrado M, Pearce EL, Pearce EJ, Grosschedl R. MZB1 enables efficient interferon α secretion in stimulated plasmacytoid dendritic cells. Sci Rep 2020; 10:21626. [PMID: 33318509 PMCID: PMC7736851 DOI: 10.1038/s41598-020-78293-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 11/23/2020] [Indexed: 12/19/2022] Open
Abstract
MZB1 is an endoplasmic reticulum (ER)-resident protein that plays an important role in the humoral immune response by enhancing the interaction of the μ immunoglobulin (Ig) heavy chain with the chaperone GRP94 and by augmenting the secretion of IgM. Here, we show that MZB1 is also expressed in plasmacytoid dendritic cells (pDCs). Mzb1−/− pDCs have a defect in the secretion of interferon (IFN) α upon Toll-like receptor (TLR) 9 stimulation and a reduced ability to enhance B cell differentiation towards plasma cells. Mzb1−/− pDCs do not properly expand the ER upon TLR9 stimulation, which may be accounted for by an impaired activation of ATF6, a regulator of the unfolded protein response (UPR). Pharmacological inhibition of ATF6 cleavage in stimulated wild type pDCs mimics the diminished IFNα secretion by Mzb1−/− pDCs. Thus, MZB1 enables pDCs to secrete high amounts of IFNα by mitigating ER stress via the ATF6-mediated UPR.
Collapse
Affiliation(s)
- Tanya Kapoor
- Department of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Mauro Corrado
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Rudolf Grosschedl
- Department of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108, Freiburg, Germany.
| |
Collapse
|
157
|
He X, Xia L, Tumas KC, Wu J, Su XZ. Type I Interferons and Malaria: A Double-Edge Sword Against a Complex Parasitic Disease. Front Cell Infect Microbiol 2020; 10:594621. [PMID: 33344264 PMCID: PMC7738626 DOI: 10.3389/fcimb.2020.594621] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Type I interferons (IFN-Is) are important cytokines playing critical roles in various infections, autoimmune diseases, and cancer. Studies have also shown that IFN-Is exhibit 'conflicting' roles in malaria parasite infections. Malaria parasites have a complex life cycle with multiple developing stages in two hosts. Both the liver and blood stages of malaria parasites in a vertebrate host stimulate IFN-I responses. IFN-Is have been shown to inhibit liver and blood stage development, to suppress T cell activation and adaptive immune response, and to promote production of proinflammatory cytokines and chemokines in animal models. Different parasite species or strains trigger distinct IFN-I responses. For example, a Plasmodium yoelii strain can stimulate a strong IFN-I response during early infection, whereas its isogenetic strain does not. Host genetic background also greatly influences IFN-I production during malaria infections. Consequently, the effects of IFN-Is on parasitemia and disease symptoms are highly variable depending on the combination of parasite and host species or strains. Toll-like receptor (TLR) 7, TLR9, melanoma differentiation-associated protein 5 (MDA5), and cyclic GMP-AMP synthase (cGAS) coupled with stimulator of interferon genes (STING) are the major receptors for recognizing parasite nucleic acids (RNA/DNA) to trigger IFN-I responses. IFN-I levels in vivo are tightly regulated, and various novel molecules have been identified to regulate IFN-I responses during malaria infections. Here we review the major findings and progress in ligand recognition, signaling pathways, functions, and regulation of IFN-I responses during malaria infections.
Collapse
Affiliation(s)
- Xiao He
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Lu Xia
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Keyla C. Tumas
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Jian Wu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Xin-Zhuan Su
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
158
|
Qi Y, Liu B, Sun Q, Xiong X, Chen Q. Immune Checkpoint Targeted Therapy in Glioma: Status and Hopes. Front Immunol 2020; 11:578877. [PMID: 33329549 PMCID: PMC7729019 DOI: 10.3389/fimmu.2020.578877] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
Glioma is the most malignant primary tumor of the central nervous system and is characterized by an extremely low overall survival. Recent breakthroughs in cancer therapy using immune checkpoint blockade have attracted significant attention. However, despite representing the most promising (immunotherapy) treatment for cancer, the clinical application of immune checkpoint blockade in glioma patients remains challenging due to the "cold phenotype" of glioma and multiple factors inducing resistance, both intrinsic and acquired. Therefore, comprehensive understanding of the tumor microenvironment and the unique immunological status of the brain will be critical for the application of glioma immunotherapy. More sensitive biomarkers to monitor the immune response, as well as combining multiple immunotherapy strategies, may accelerate clinical progress and enable development of effective and safe treatments for glioma patients.
Collapse
Affiliation(s)
- Yangzhi Qi
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
159
|
Llopiz D, Ruiz M, Silva L, Repáraz D, Aparicio B, Egea J, Lasarte JJ, Redin E, Calvo A, Angel M, Berzofsky JA, Stroncek D, Sarobe P. Inhibition of adjuvant-induced TAM receptors potentiates cancer vaccine immunogenicity and therapeutic efficacy. Cancer Lett 2020; 499:279-289. [PMID: 33232788 DOI: 10.1016/j.canlet.2020.11.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 01/19/2023]
Abstract
Analyzing immunomodulatory elements operating during antitumor vaccination in prostate cancer patients and murine models we identified IL-10-producing DC as a subset with poorer immunogenicity and clinical efficacy. Inhibitory TAM receptors MER and AXL were upregulated on murine IL-10+ DC. Thus, we analyzed conditions inducing these molecules and the potential benefit of their blockade during vaccination. MER and AXL upregulation was more efficiently induced by a vaccine containing Imiquimod than by a poly(I:C)-containing vaccine. Interestingly, MER expression was found on monocyte-derived DC, and was dependent on IL-10. TAM blockade improved Imiquimod-induced DC activation in vitro and in vivo, resulting in increased vaccine-induced T-cell responses, which were further reinforced by concomitant IL-10 inhibition. In different tumor models, a triple therapy (including vaccination, TAM inhibition and IL-10 blockade) provided the strongest therapeutic effect, associated with enhanced T-cell immunity and enhanced CD8+ T cell tumor infiltration. Finally, MER levels in DC used for vaccination in cancer patients correlated with IL-10 expression, showing an inverse association with vaccine-induced clinical response. These results suggest that TAM receptors upregulated during vaccination may constitute an additional target in combinatorial therapeutic vaccination strategies.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Imiquimod/administration & dosage
- Immunogenicity, Vaccine/drug effects
- Immunotherapy/methods
- Interleukin-10/metabolism
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Transgenic
- Poly I-C/administration & dosage
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/therapy
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Pyrimidines
- Quinolines
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptor Protein-Tyrosine Kinases/genetics
- Up-Regulation/drug effects
- Up-Regulation/immunology
- c-Mer Tyrosine Kinase/antagonists & inhibitors
- c-Mer Tyrosine Kinase/genetics
- Axl Receptor Tyrosine Kinase
Collapse
Affiliation(s)
- Diana Llopiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Marta Ruiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Leyre Silva
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - David Repáraz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Belén Aparicio
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Josune Egea
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Juan J Lasarte
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Esther Redin
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; CIBERONC, ISCIII, Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Alfonso Calvo
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; CIBERONC, ISCIII, Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Matthew Angel
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Center for Cancer Research Collaborative Bioinformatics Resource, Leidos Biomedical Research, Inc., FNLCR, Frederick, MD, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - David Stroncek
- Center for Cellular Engineering, Department of Transfusion Medicine, NIH Clinical Center, Bethesda, MD, USA
| | - Pablo Sarobe
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.
| |
Collapse
|
160
|
Liu YZ, Han DT, Shi DR, Hong B, Qian Y, Wu ZG, Yao SH, Tang TM, Wang MH, Xu XM, Yao HP. Pathological significance of abnormal recepteur d’origine nantais and programmed death ligand 1 expression in colorectal cancer. World J Gastrointest Oncol 2020; 12:1216-1236. [PMID: 33250957 PMCID: PMC7667461 DOI: 10.4251/wjgo.v12.i11.1216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Programmed death ligand 1 (PD-L1) immunotherapy remains poorly efficacious in colorectal cancer (CRC). The recepteur d’origine nantais (RON) receptor tyrosine kinase plays an important role in regulating tumor immunity.
AIM To identify the patterns of RON and PD-L1 expression and explore their clinical significance in CRC.
METHODS Gene expression data from the Gene Expression Omnibus database (GEO; n = 290) and patients at the First Affiliated Hospital, Zhejiang University School of Medicine (FAHZUSM; n = 381) were analyzed to determine the prognostic value of RON and PD-L1 expression within the tumor microenvironment of CRC. HT29 cell line was treated with BMS-777607 to explore the relationship between RON activity and PD-L1 expression. Signaling pathways and protein expression perturbed by RON inhibition were evaluated by cellular immunofluorescence and Western blot.
RESULTS In the GEO patient cohort, cut-off values for RON and PD-L1 expression were determined to be 7.70 and 4.3, respectively. Stratification of patients based on these cutoffs demonstrated that high expression of RON and PD-L1 was associated with a poor prognosis. In the FAHZUSM cohort, rates of high expression of RON in tumor cells, high PD-L1 expression in tumor cells and tumor infiltrating monocytes, and both high RON and high PD-L1 expression in the tumor microenvironment were 121 (32%), 43 (11%), 91 (24%), and 51 (13.4%), respectively. High expression of RON was significantly correlated with high expression of PD-L1 in the tumor cell compartment (P < 0.001). High expression of RON and that of PD-L1 were independent prognostic factors for poorer overall survival. Concurrent high expression of both RON and PD-L1 in the tumor microenvironment was significantly associated with a poor prognosis. In vitro, BMS-777607 inhibited the phosphorylation of RON, inhibited PD-L1 expression, and attenuated activation of the ERK1/2 and AKT signaling pathways in CRC cells.
CONCLUSION RON, PD-L1, and their crosstalk are significant in predicting the prognostic value of CRC. Moreover, phosphorylation of RON upregulates PD-L1 expression, which provides a novel approach to immunotherapy in CRC.
Collapse
Affiliation(s)
- Yi-Zhi Liu
- Department of Cancer Biology Research, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Da-Ting Han
- Department of Cancer Biology Research, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Dan-Rong Shi
- Department of Cancer Biology Research, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Bo Hong
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yun Qian
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Zhi-Gang Wu
- Department of Cancer Biology Research, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Shu-Hao Yao
- Department of Stomatology, Wenzhou Medical University Renji College, Wenzhou 325035, Zhejiang Province, China
| | - Tao-Ming Tang
- Department of Cancer Biology Research, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ming-Hai Wang
- Cancer Biology Research Center and Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States
| | - Xiang-Ming Xu
- Department of Cancer Biology Research, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Hang-Ping Yao
- Department of Cancer Biology Research, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
161
|
Sumida TS, Dulberg S, Schupp J, Stillwell HA, Axisa PP, Comi M, Lincoln M, Unterman A, Kaminski N, Madi A, Kuchroo VK, Hafler DA. Type I Interferon Transcriptional Network Regulates Expression of Coinhibitory Receptors in Human T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.10.30.362947. [PMID: 33140047 PMCID: PMC7605554 DOI: 10.1101/2020.10.30.362947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
While inhibition of T cell co-inhibitory receptors has revolutionized cancer therapy, the mechanisms governing their expression on human T cells have not been elucidated. Type 1 interferon (IFN-I) modulates T cell immunity in viral infection, autoimmunity, and cancer, and may facilitate induction of T cell exhaustion in chronic viral infection 1,2 . Here we show that IFN-I regulates co-inhibitory receptors expression on human T cells, inducing PD-1/TIM-3/LAG-3 while surprisingly inhibiting TIGIT expression. High-temporal-resolution mRNA profiling of IFN-I responses enabled the construction of dynamic transcriptional regulatory networks uncovering three temporal transcriptional waves. Perturbation of key transcription factors on human primary T cells revealed both canonical and non-canonical IFN-I transcriptional regulators, and identified unique regulators that control expression of co-inhibitory receptors. To provide direct in vivo evidence for the role of IFN-I on co-inhibitory receptors, we then performed single cell RNA-sequencing in subjects infected with SARS-CoV-2, where viral load was strongly associated with T cell IFN-I signatures. We found that the dynamic IFN-I response in vitro closely mirrored T cell features with acute IFN-I linked viral infection, with high LAG3 and decreased TIGIT expression. Finally, our gene regulatory network identified SP140 as a key regulator for differential LAG3 and TIGIT expression. The construction of co-inhibitory regulatory networks induced by IFN-I with identification of unique transcription factors controlling their expression may provide targets for enhancement of immunotherapy in cancer, infectious diseases, and autoimmunity.
Collapse
Affiliation(s)
- Tomokazu S. Sumida
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Shai Dulberg
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jonas Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Helen A. Stillwell
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Pierre-Paul Axisa
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Michela Comi
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Matthew Lincoln
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Avraham Unterman
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Asaf Madi
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David A. Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
162
|
Wang X, Caffrey-Carr AK, Liu KW, Espinosa V, Croteau W, Dhingra S, Rivera A, Cramer RA, Obar JJ. MDA5 Is an Essential Sensor of a Pathogen-Associated Molecular Pattern Associated with Vitality That Is Necessary for Host Resistance against Aspergillus fumigatus. THE JOURNAL OF IMMUNOLOGY 2020; 205:3058-3070. [PMID: 33087405 DOI: 10.4049/jimmunol.2000802] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022]
Abstract
RIG-I-like receptors (RLR) are cytosolic RNA sensors that signal through the MAVS adaptor to activate IFN responses against viruses. Whether the RLR family has broader effects on host immunity against other pathogen families remains to be fully explored. In this study, we demonstrate that MDA5/MAVS signaling was essential for host resistance against pulmonary Aspergillus fumigatus challenge through the regulation of antifungal leukocyte responses in mice. Activation of MDA5/MAVS signaling was driven by dsRNA from live A. fumigatus serving as a key vitality-sensing pattern recognition receptor. Interestingly, induction of type I IFNs after A. fumigatus challenge was only partially dependent on MDA5/MAVS signaling, whereas type III IFN expression was entirely dependent on MDA5/MAVS signaling. Ultimately, type I and III IFN signaling drove the expression of CXCL10. Furthermore, the MDA5/MAVS-dependent IFN response was critical for the induction of optimal antifungal neutrophil killing of A. fumigatus spores. In conclusion, our data broaden the role of the RLR family to include a role in regulating antifungal immunity against A. fumigatus.
Collapse
Affiliation(s)
- Xi Wang
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Alayna K Caffrey-Carr
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756.,Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59718; and
| | - Ko-Wei Liu
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Vanessa Espinosa
- Center for Immunity and Inflammation, Rutgers - New Jersey Medical School, Newark, NJ 07103
| | - Walburga Croteau
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Sourabh Dhingra
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Amariliz Rivera
- Center for Immunity and Inflammation, Rutgers - New Jersey Medical School, Newark, NJ 07103
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756
| | - Joshua J Obar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756;
| |
Collapse
|
163
|
Raju S, Verbaro DJ, Egawa T. PD-1 Signaling Promotes Control of Chronic Viral Infection by Restricting Type-I-Interferon-Mediated Tissue Damage. Cell Rep 2020; 29:2556-2564.e3. [PMID: 31775026 PMCID: PMC6894421 DOI: 10.1016/j.celrep.2019.10.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 10/03/2019] [Accepted: 10/22/2019] [Indexed: 01/15/2023] Open
Abstract
Immune responses are essential for pathogen elimination but also cause tissue damage, leading to disease or death. However, it is unclear how the host immune system balances control of infection and protection from the collateral tissue damage. Here, we show that PD-1-mediated restriction of immune responses is essential for durable control of chronic LCMV infection in mice. In contrast to responses in the chronic phase, PD-1 blockade in the subacute phase of infection paradoxically results in viral persistence. This effect is associated with damage to lymphoid architecture and subsequently decreases adaptive immune responses. Moreover, this tissue damage is type I interferon dependent, as sequential blockade of the interferon receptor and PD-1 pathways prevents immunopathology and enhances control of infection. We conclude that PD-1-mediated suppression is required as an immunoregulatory mechanism for sustained responses to chronic viral infection by antagonizing type-I interferon-dependent immunopathology. Using stage-specific PD-1 blockade in LCMV-infected mice, Raju et al. uncover the requirement for PD-1-mediated suppression of CD8 T cells for durable immune response to chronic viral infection, as well as the requirement for IFNAR signaling in programming of CD8 T cells toward effectors that cause immunopathology.
Collapse
Affiliation(s)
- Saravanan Raju
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Daniel J Verbaro
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Takeshi Egawa
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
164
|
Verdon DJ, Mulazzani M, Jenkins MR. Cellular and Molecular Mechanisms of CD8 + T Cell Differentiation, Dysfunction and Exhaustion. Int J Mol Sci 2020; 21:ijms21197357. [PMID: 33027962 PMCID: PMC7582856 DOI: 10.3390/ijms21197357] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
T cells follow a triphasic distinct pathway of activation, proliferation and differentiation before becoming functionally and phenotypically “exhausted” in settings of chronic infection, autoimmunity and in cancer. Exhausted T cells progressively lose canonical effector functions, exhibit altered transcriptional networks and epigenetic signatures and gain constitutive expression of a broad coinhibitory receptor suite. This review outlines recent advances in our understanding of exhausted T cell biology and examines cellular and molecular mechanisms by which a state of dysfunction or exhaustion is established, and mechanisms by which exhausted T cells may still contribute to pathogen or tumour control. Further, this review describes our understanding of exhausted T cell heterogeneity and outlines the mechanisms by which checkpoint blockade differentially engages exhausted T cell subsets to overcome exhaustion and recover T cell function.
Collapse
Affiliation(s)
- Daniel J. Verdon
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Matthias Mulazzani
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Misty R. Jenkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
- Institute of Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
- Correspondence:
| |
Collapse
|
165
|
Exposing Hidden Targets: Combining epigenetic and immunotherapy to overcome cancer resistance. Semin Cancer Biol 2020; 65:114-122. [DOI: 10.1016/j.semcancer.2020.01.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/27/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022]
|
166
|
Guo K, Shen G, Kibbie J, Gonzalez T, Dillon SM, Smith HA, Cooper EH, Lavender K, Hasenkrug KJ, Sutter K, Dittmer U, Kroehl M, Kechris K, Wilson CC, Santiago ML. Qualitative Differences Between the IFNα subtypes and IFNβ Influence Chronic Mucosal HIV-1 Pathogenesis. PLoS Pathog 2020; 16:e1008986. [PMID: 33064743 PMCID: PMC7592919 DOI: 10.1371/journal.ppat.1008986] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 10/28/2020] [Accepted: 09/16/2020] [Indexed: 12/27/2022] Open
Abstract
The Type I Interferons (IFN-Is) are innate antiviral cytokines that include 12 different IFNα subtypes and IFNβ that signal through the IFN-I receptor (IFNAR), inducing hundreds of IFN-stimulated genes (ISGs) that comprise the 'interferome'. Quantitative differences in IFNAR binding correlate with antiviral activity, but whether IFN-Is exhibit qualitative differences remains controversial. Moreover, the IFN-I response is protective during acute HIV-1 infection, but likely pathogenic during the chronic stages. To gain a deeper understanding of the IFN-I response, we compared the interferomes of IFNα subtypes dominantly-expressed in HIV-1-exposed plasmacytoid dendritic cells (1, 2, 5, 8 and 14) and IFNβ in the earliest cellular targets of HIV-1 infection. Primary gut CD4 T cells from 3 donors were treated for 18 hours ex vivo with individual IFN-Is normalized for IFNAR signaling strength. Of 1,969 IFN-regulated genes, 246 'core ISGs' were induced by all IFN-Is tested. However, many IFN-regulated genes were not shared between the IFNα subtypes despite similar induction of canonical antiviral ISGs such as ISG15, RSAD2 and MX1, formally demonstrating qualitative differences between the IFNα subtypes. Notably, IFNβ induced a broader interferome than the individual IFNα subtypes. Since IFNβ, and not IFNα, is upregulated during chronic HIV-1 infection in the gut, we compared core ISGs and IFNβ-specific ISGs from colon pinch biopsies of HIV-1-uninfected (n = 13) versus age- and gender-matched, antiretroviral-therapy naïve persons with HIV-1 (PWH; n = 19). Core ISGs linked to inflammation, T cell activation and immune exhaustion were elevated in PWH, positively correlated with plasma lipopolysaccharide (LPS) levels and gut IFNβ levels, and negatively correlated with gut CD4 T cell frequencies. In sharp contrast, IFNβ-specific ISGs linked to protein translation and anti-inflammatory responses were significantly downregulated in PWH, negatively correlated with gut IFNβ and LPS, and positively correlated with plasma IL6 and gut CD4 T cell frequencies. Our findings reveal qualitative differences in interferome induction by diverse IFN-Is and suggest potential mechanisms for how IFNβ may drive HIV-1 pathogenesis in the gut.
Collapse
Affiliation(s)
- Kejun Guo
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Guannan Shen
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Jon Kibbie
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Tania Gonzalez
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Stephanie M. Dillon
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Harry A. Smith
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Emily H. Cooper
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Kerry Lavender
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Canada
| | - Kim J. Hasenkrug
- Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, Hamilton, MT, United States of America
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisberg-Essen, Essen, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisberg-Essen, Essen, Germany
| | - Miranda Kroehl
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Katerina Kechris
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Cara C. Wilson
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, United States of America
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Mario L. Santiago
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, United States of America
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| |
Collapse
|
167
|
Palacio N, Dangi T, Chung YR, Wang Y, Loredo-Varela JL, Zhang Z, Penaloza-MacMaster P. Early type I IFN blockade improves the efficacy of viral vaccines. J Exp Med 2020; 217:152035. [PMID: 32820330 PMCID: PMC7953731 DOI: 10.1084/jem.20191220] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/09/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
Type I interferons (IFN-I) are a major antiviral defense and are critical for the activation of the adaptive immune system. However, early viral clearance by IFN-I could limit antigen availability, which could in turn impinge upon the priming of the adaptive immune system. In this study, we hypothesized that transient IFN-I blockade could increase antigen presentation after acute viral infection. To test this hypothesis, we infected mice with viruses coadministered with a single dose of IFN-I receptor–blocking antibody to induce a short-term blockade of the IFN-I pathway. This resulted in a transient “spike” in antigen levels, followed by rapid antigen clearance. Interestingly, short-term IFN-I blockade after coronavirus, flavivirus, rhabdovirus, or arenavirus infection induced a long-lasting enhancement of immunological memory that conferred improved protection upon subsequent reinfections. Short-term IFN-I blockade also improved the efficacy of viral vaccines. These findings demonstrate a novel mechanism by which IFN-I regulate immunological memory and provide insights for rational vaccine design.
Collapse
Affiliation(s)
- Nicole Palacio
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Young Rock Chung
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yidan Wang
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Juan Luis Loredo-Varela
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Zhongyao Zhang
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
168
|
Jondle CN, Tarakanova VL. Innate immunity and alpha/gammaherpesviruses: first impressions last a lifetime. Curr Opin Virol 2020; 44:81-89. [PMID: 32777757 DOI: 10.1016/j.coviro.2020.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 06/25/2020] [Accepted: 07/04/2020] [Indexed: 12/26/2022]
Abstract
Innate immune system is considered the first line of defense during viral invasion, with the wealth of the literature demonstrating innate immune control of diverse viruses during acute infection. What is far less clear is the role of innate immune system during chronic virus infections. This short review focuses on alphaherpesviruses and gammaherpesviruses, two highly prevalent herpesvirus subfamilies that, following a brief, once in a lifetime period of acute lytic infection, establish life-long latent infection that is characterized by sporadic reactivation in an immunocompetent host. In spite of many similarities, these two viral families are characterized by distinct cellular tropism and pathogenesis. Here we focus on the published in vivo studies to review known interactions of these two viral subfamilies with the innate immunity of the intact host, both during acute and, particularly, chronic virus infection.
Collapse
Affiliation(s)
- Christopher N Jondle
- Department of Microbiology and Immunology, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI, 53226, United States
| | - Vera L Tarakanova
- Department of Microbiology and Immunology, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI, 53226, United States; Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, United States.
| |
Collapse
|
169
|
Sato T, Ishikawa S, Asano J, Yamamoto H, Fujii M, Sato T, Yamamoto K, Kitagaki K, Akashi T, Okamoto R, Ohteki T. Regulated IFN signalling preserves the stemness of intestinal stem cells by restricting differentiation into secretory-cell lineages. Nat Cell Biol 2020; 22:919-926. [PMID: 32690888 DOI: 10.1038/s41556-020-0545-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/17/2020] [Indexed: 11/09/2022]
Abstract
Intestinal stem cells (ISCs) are located at the crypt base and fine-tune the balance of their self-renewal and differentiation1,2, but the physiological mechanism involved in regulating that balance remains unknown. Here we describe a transcriptional regulator that preserves the stemness of ISCs by restricting their differentiation into secretory-cell lineages. Interferon regulatory factor 2 (IRF2) negatively regulates interferon signalling3, and mice completely lacking Irf24 or with a selective Irf2 deletion in their intestinal epithelial cells have significantly fewer crypt Lgr5hi ISCs than control mice. Although the integrity of intestinal epithelial cells was unimpaired at steady state in Irf2-deficient mice, regeneration of their intestinal epithelia after 5-fluorouracil-induced damage was severely impaired. Similarly, extended treatment with low-dose poly(I:C) or chronic infection of lymphocytic choriomeningitis virus clone 13 (LCMV C13)5 caused a functional decline of ISCs in wild-type mice. In contrast, massive accumulations of immature Paneth cells were found at the crypt base of Irf2-/- as well as LCMV C13-infected wild-type mice, indicating that excess interferon signalling directs ISCs towards a secretory-cell fate. Collectively, our findings indicate that regulated interferon signalling preserves ISC stemness by restricting secretory-cell differentiation.
Collapse
Affiliation(s)
- Taku Sato
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,PRESTO, Japan Science and Technology Agency, Saitama, Japan
| | - Shun Ishikawa
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Jumpei Asano
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirona Yamamoto
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masayuki Fujii
- Department of Surgical Oncology, The University of Tokyo, Tokyo, Japan.,Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Keisuke Kitagaki
- Division of Surgical Pathology, Tokyo Medical and Dental University Hospital, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takumi Akashi
- Division of Surgical Pathology, Tokyo Medical and Dental University Hospital, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| |
Collapse
|
170
|
Kusnadi A, Ramírez-Suástegui C, Fajardo V, Chee SJ, Meckiff BJ, Simon H, Pelosi E, Seumois G, Ay F, Vijayanand P, Ottensmeier CH. Severely ill COVID-19 patients display augmented functional properties in SARS-CoV-2-reactive CD8 + T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.09.194027. [PMID: 32676602 PMCID: PMC7359524 DOI: 10.1101/2020.07.09.194027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The molecular properties of CD8 + T cells that respond to SARS-CoV-2 infection are not fully known. Here, we report on the single-cell transcriptomes of >80,000 virus-reactive CD8 + T cells from 39 COVID-19 patients and 10 healthy subjects. COVID-19 patients segregated into two groups based on whether the dominant CD8 + T cell response to SARS-CoV-2 was 'exhausted' or not. SARS-CoV-2-reactive cells in the exhausted subset were increased in frequency and displayed lesser cytotoxicity and inflammatory features in COVID-19 patients with mild compared to severe illness. In contrast, SARS-CoV-2-reactive cells in the non-exhausted subsets from patients with severe disease showed enrichment of transcripts linked to co-stimulation, pro-survival NF-κB signaling, and anti-apoptotic pathways, suggesting the generation of robust CD8 + T cell memory responses in patients with severe COVID-19 illness. CD8 + T cells reactive to influenza and respiratory syncytial virus from healthy subjects displayed polyfunctional features. Cells with such features were mostly absent in SARS-CoV-2 responsive cells from both COVID-19 patients and healthy controls non-exposed to SARS-CoV-2. Overall, our single-cell analysis revealed substantial diversity in the nature of CD8 + T cells responding to SARS-CoV-2.
Collapse
Affiliation(s)
- Anthony Kusnadi
- La Jolla Institute for Immunology, La Jolla, CA, USA
- These authors jointly contributed to the work
| | - Ciro Ramírez-Suástegui
- La Jolla Institute for Immunology, La Jolla, CA, USA
- These authors jointly contributed to the work
| | - Vicente Fajardo
- La Jolla Institute for Immunology, La Jolla, CA, USA
- These authors jointly contributed to the work
| | - Serena J Chee
- NIHR and CRUK Southampton Experimental Cancer Medicine Center, Faculty of Medicine, University of Southampton, Southampton, UK
- These authors jointly contributed to the work
| | | | - Hayley Simon
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Emanuela Pelosi
- Southampton Specialist Virology Centre, Department of Infection, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Ferhat Ay
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Liverpool Head and Neck Center, Institute of Translational Medicine, University of Liverpool & Clatterbridge Cancer Center NHS Foundation Trust, Liverpool, United Kingdom
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- These authors jointly directed the work
| | - Christian H Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Liverpool Head and Neck Center, Institute of Translational Medicine, University of Liverpool & Clatterbridge Cancer Center NHS Foundation Trust, Liverpool, United Kingdom
- These authors jointly directed the work
| |
Collapse
|
171
|
Wu W, Metcalf JP. The Role of Type I IFNs in Influenza: Antiviral Superheroes or Immunopathogenic Villains? J Innate Immun 2020; 12:437-447. [PMID: 32564033 PMCID: PMC7747089 DOI: 10.1159/000508379] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/03/2020] [Indexed: 12/29/2022] Open
Abstract
The important role of interferons (IFNs) in antiviral innate immune defense is well established. Although recombinant IFN-α was approved for cancer and chronic viral infection treatment by regulatory agencies in many countries starting in 1986, no IFNs are approved for treatment of influenza A virus (IAV) infection. This is partially due to the complex effects of IFNs in acute influenza infection. IAV attacks the human respiratory system and causes significant morbidity and mortality globally. During influenza infection, depending on the strain of IAV and the individual host, type I IFNs can have protective antiviral effects or can contribute to immunopathology. In the context of virus infection, the immune system has complicated mechanisms regulating the expression and effects of type I IFN to maximize the antiviral response by both activating and enhancing beneficial innate cell function, while limiting immunopathological responses that lead to exaggerated tissue damage. In this review, we summarize the complicated, but important, role of type I IFNs in influenza infections. This includes both protective and harmful effects of these important cytokines during infection.
Collapse
Affiliation(s)
- Wenxin Wu
- Department of Medicine, Pulmonary, Critical Care and Sleep Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA,
| | - Jordan P Metcalf
- Department of Medicine, Pulmonary, Critical Care and Sleep Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Pulmonary Section, Medicine Service, Veterans Affairs Medical Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
172
|
Azar P, Mejía JE, Cenac C, Shaiykova A, Youness A, Laffont S, Essat A, Izopet J, Passaes C, Müller-Trutwin M, Delobel P, Meyer L, Guéry JC. TLR7 dosage polymorphism shapes interferogenesis and HIV-1 acute viremia in women. JCI Insight 2020; 5:136047. [PMID: 32554924 DOI: 10.1172/jci.insight.136047] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/06/2020] [Indexed: 12/25/2022] Open
Abstract
Type I IFN (IFN-I) production by plasmacytoid DCs (pDCs) occurs during acute HIV-1 infection in response to TLR7 stimulation, but the role of pDC-derived IFN-I in controlling or promoting HIV-1 infection is ambiguous. We report here a sex-biased interferogenic phenotype for a frequent single-nucleotide polymorphism of human TLR7, rs179008, displaying an impact on key parameters of acute HIV-1 infection. We show allele rs179008 T to determine lower TLR7 protein abundance in cells from women, specifically - likely by diminishing TLR7 mRNA translation efficiency through codon usage. The hypomorphic TLR7 phenotype is mirrored by decreased TLR7-driven IFN-I production by female pDCs. Among women from the French ANRS PRIMO cohort of acute HIV-1 patients, carriage of allele rs179008 T associated with lower viremia, cell-associated HIV-1 DNA, and CXCL10 (IP-10) plasma concentrations. RNA viral load was decreased by 0.85 log10 (95% CI, -1.51 to -0.18) among T/T homozygotes, who also exhibited a lower frequency of acute symptoms. TLR7 emerges as an important control locus for acute HIV-1 viremia, and the clinical phenotype for allele rs179008 T, carried by 30%-50% of European women, supports a beneficial effect of toning down TLR7-driven IFN-I production by pDCs during acute HIV-1 infection.
Collapse
Affiliation(s)
- Pascal Azar
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France
| | - José Enrique Mejía
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France
| | - Claire Cenac
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France
| | - Arnoo Shaiykova
- Centre de Recherche en Epidémiologie et Santé des Populations (CESP), Université Paris-Sud, Université Paris-Saclay, INSERM, Le Kremlin-Bicêtre, France
| | - Ali Youness
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France
| | - Sophie Laffont
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France
| | - Asma Essat
- Centre de Recherche en Epidémiologie et Santé des Populations (CESP), Université Paris-Sud, Université Paris-Saclay, INSERM, Le Kremlin-Bicêtre, France
| | - Jacques Izopet
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France.,Laboratoire de Virologie, CHU Purpan, Toulouse, France
| | - Caroline Passaes
- Institut Pasteur, Unité HIV Inflammation et Persistance, Paris, France
| | | | - Pierre Delobel
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France.,Service des Maladies Infectieuses et Tropicales, CHU Purpan, Toulouse, France
| | - Laurence Meyer
- Centre de Recherche en Epidémiologie et Santé des Populations (CESP), Université Paris-Sud, Université Paris-Saclay, INSERM, Le Kremlin-Bicêtre, France
| | - Jean-Charles Guéry
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UMR 1043 INSERM, CNRS, Toulouse, France
| |
Collapse
|
173
|
Major J, Crotta S, Llorian M, McCabe TM, Gad HH, Priestnall SL, Hartmann R, Wack A. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection. Science 2020; 369:712-717. [PMID: 32527928 PMCID: PMC7292500 DOI: 10.1126/science.abc2061] [Citation(s) in RCA: 357] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/08/2020] [Indexed: 12/27/2022]
Abstract
Interferons (IFNs) are central to antiviral immunity. Viral recognition elicits IFN production, which in turn triggers the transcription of IFN-stimulated genes (ISGs), which engage in various antiviral functions. Type I IFNs (IFN-α and IFN-β) are widely expressed and can result in immunopathology during viral infections. By contrast, type III IFN (IFN-λ) responses are primarily restricted to mucosal surfaces and are thought to confer antiviral protection without driving damaging proinflammatory responses. Accordingly, IFN-λ has been proposed as a therapeutic in coronavirus disease 2019 (COVID-19) and other such viral respiratory diseases (see the Perspective by Grajales-Reyes and Colonna). Broggi et al. report that COVID-19 patient morbidity correlates with the high expression of type I and III IFNs in the lung. Furthermore, IFN-λ secreted by dendritic cells in the lungs of mice exposed to synthetic viral RNA causes damage to the lung epithelium, which increases susceptibility to lethal bacterial superinfections. Similarly, using a mouse model of influenza infection, Major et al. found that IFN signaling (especially IFN-λ) hampers lung repair by inducing p53 and inhibiting epithelial proliferation and differentiation. Complicating this picture, Hadjadj et al. observed that peripheral blood immune cells from severe and critical COVID-19 patients have diminished type I IFN and enhanced proinflammatory interleukin-6– and tumor necrosis factor-α–fueled responses. This suggests that in contrast to local production, systemic production of IFNs may be beneficial. The results of this trio of studies suggest that the location, timing, and duration of IFN exposure are critical parameters underlying the success or failure of therapeutics for viral respiratory infections. Science, this issue p. 706, p. 712, p. 718; see also p. 626 Excessive cytokine signaling frequently exacerbates lung tissue damage during respiratory viral infection. Type I (IFN-α and IFN-β) and III (IFN-λ) interferons are host-produced antiviral cytokines. Prolonged IFN-α and IFN-β responses can lead to harmful proinflammatory effects, whereas IFN-λ mainly signals in epithelia, thereby inducing localized antiviral immunity. In this work, we show that IFN signaling interferes with lung repair during influenza recovery in mice, with IFN-λ driving these effects most potently. IFN-induced protein p53 directly reduces epithelial proliferation and differentiation, which increases disease severity and susceptibility to bacterial superinfections. Thus, excessive or prolonged IFN production aggravates viral infection by impairing lung epithelial regeneration. Timing and duration are therefore critical parameters of endogenous IFN action and should be considered carefully for IFN therapeutic strategies against viral infections such as influenza and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Jack Major
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Stefania Crotta
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Miriam Llorian
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Teresa M McCabe
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Hans Henrik Gad
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Simon L Priestnall
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK.,Experimental Histopathology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
174
|
Yan B, Freiwald T, Chauss D, Wang L, West E, Bibby J, Olson M, Kordasti S, Portilla D, Laurence A, Lionakis MS, Kemper C, Afzali B, Kazemian M. SARS-CoV2 drives JAK1/2-dependent local and systemic complement hyper-activation. RESEARCH SQUARE 2020:rs.3.rs-33390. [PMID: 32702726 PMCID: PMC7336704 DOI: 10.21203/rs.3.rs-33390/v1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with coronavirus disease 2019 (COVID-19) present with a range of devastating acute clinical manifestations affecting the lungs, liver, kidneys and gut. The best-characterized entry receptor for the disease-causing virus SARS-CoV2, angiotensin converting enzyme (ACE) 2, is highly expressed in these tissues. However, the pathways that underlie the disease are still poorly understood. Here we show that the complement system is unexpectedly one of the intracellular pathways most highly induced by SARS-CoV2 infection in lung epithelial and liver cells. Within cells of the bronchoalveolar lavage of patients, distinct signatures of complement activation in myeloid, lymphoid and epithelial cells tracked with disease severity. Modelling the regulome of host genes induced by COVID-19 and the drugs that could normalize these genes both implicated the JAK1/2-STAT1 signaling system downstream of type I interferon receptors, and NF-kB. Ruxolitinib, a JAK1/2 inhibitor and the top predicted pharmaceutical candidate, normalized interferon signature genes, IL-6 (the best characterized severity marker in COVID-19) and all complement genes induced by SARS-CoV2, but did not affect NF-kB-regulated genes. We predict that combination therapy with JAK inhibitors and other agents with the potential to normalize NF-kB-signaling, such as anti-viral agents, may serve as an effective clinical strategy.
Collapse
Affiliation(s)
- Bingyu Yan
- Departments of Biochemistry and Computer Science, Purdue
University, West Lafayette, IN, USA
| | - Tilo Freiwald
- Immunoregulation Section, Kidney Diseases Branch, National
Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD,
USA
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National
Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD,
USA
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue
University, West Lafayette, IN, USA
| | - Erin West
- Laboratory of Molecular Immunology and the Immunology Center,
National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH),
Bethesda, MD, USA
| | - Jack Bibby
- Laboratory of Molecular Immunology and the Immunology Center,
National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH),
Bethesda, MD, USA
| | - Matthew Olson
- Department of Biological Sciences, Purdue University, West
Lafayette, IN, USA
| | - Shahram Kordasti
- School of Immunology and Microbial Sciences, Faculty of Life
Sciences and Medicine, King’s College London, London, UK
| | - Didier Portilla
- Immunoregulation Section, Kidney Diseases Branch, National
Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD,
USA
- Division of Nephrology and the Center for Immunity, Inflammation
and Regenerative Medicine, University of Virginia, VA, USA
| | - Arian Laurence
- Nuffield Department of Medicine, University of Oxford, UK
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology
and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH,
Bethesda, MD, USA
| | - Claudia Kemper
- Laboratory of Molecular Immunology and the Immunology Center,
National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH),
Bethesda, MD, USA
- Institute for Systemic Inflammation Research, University of
Lübeck, Lübeck, Germany
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National
Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD,
USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue
University, West Lafayette, IN, USA
| |
Collapse
|
175
|
Reis AL, Goatley LC, Jabbar T, Lopez E, Rathakrishnan A, Dixon LK. Deletion of the Gene for the Type I Interferon Inhibitor I329L from the Attenuated African Swine Fever Virus OURT88/3 Strain Reduces Protection Induced in Pigs. Vaccines (Basel) 2020; 8:vaccines8020262. [PMID: 32486154 PMCID: PMC7349983 DOI: 10.3390/vaccines8020262] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 01/01/2023] Open
Abstract
Live attenuated vaccines are considered to be the fastest route to the development of a safe and efficacious African swine fever (ASF) vaccine. Infection with the naturally attenuated OURT88/3 strain induces protection against challenge with virulent isolates from the same or closely related genotypes. However, adverse clinical signs following immunisation have been observed. Here, we attempted to increase the OURT88/3 safety profile by deleting I329L, a gene previously shown to inhibit the host innate immune response. The resulting virus, OURT88/3ΔI329L, was tested in vitro to evaluate the replication and expression of type I interferon (IFN) and in vivo by immunisation and lethal challenge experiments in pigs. No differences were observed regarding replication; however, increased amounts of both IFN-β and IFN-α were observed in macrophages infected with the deletion mutant virus. Unexpectedly, the deletion of I329L markedly reduced protection against challenge with the virulent OURT88/1 isolate. This was associated with a decrease in both antibody levels against VP72 and the number of IFN-γ-producing cells in the blood of non-protected animals. Furthermore, a significant increase in IL-10 levels in serum was observed in pigs immunised with OURT88/3ΔI329L following challenge. Interestingly, the deletion of the I329L gene failed to attenuate the virulent Georgia/2007 isolate.
Collapse
Affiliation(s)
- Ana Luisa Reis
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK; (L.C.G.); (T.J.); (A.R.); (L.K.D.)
- Correspondence:
| | - Lynnette C. Goatley
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK; (L.C.G.); (T.J.); (A.R.); (L.K.D.)
| | - Tamara Jabbar
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK; (L.C.G.); (T.J.); (A.R.); (L.K.D.)
| | - Elisabeth Lopez
- IRTA, Centre de Recerca en Sanitat Animal (IRTA-CReSA), Campus de la Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain;
| | - Anusyah Rathakrishnan
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK; (L.C.G.); (T.J.); (A.R.); (L.K.D.)
| | - Linda K. Dixon
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK; (L.C.G.); (T.J.); (A.R.); (L.K.D.)
| |
Collapse
|
176
|
Hickerson BT, Sefing EJ, Bailey KW, Van Wettere AJ, Penichet ML, Gowen BB. Type I interferon underlies severe disease associated with Junín virus infection in mice. eLife 2020; 9:55352. [PMID: 32452770 PMCID: PMC7297529 DOI: 10.7554/elife.55352] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
Junín virus (JUNV) is one of five New World mammarenaviruses (NWMs) that causes fatal hemorrhagic disease in humans and is the etiological agent of Argentine hemorrhagic fever (AHF). The pathogenesis underlying AHF is poorly understood; however, a prolonged, elevated interferon-α (IFN-α) response is associated with a negative disease outcome. A feature of all NWMs that cause viral hemorrhagic fever is the use of human transferrin receptor 1 (hTfR1) for cellular entry. Here, we show that mice expressing hTfR1 develop a lethal disease course marked by an increase in serum IFN-α concentration when challenged with JUNV. Further, we provide evidence that the type I IFN response is central to the development of severe JUNV disease in hTfR1 mice. Our findings identify hTfR1-mediated entry and the type I IFN response as key factors in the pathogenesis of JUNV infection in mice.
Collapse
Affiliation(s)
- Brady T Hickerson
- Department of Animal, Dairy and Veterinary Sciences, Utah State UniversityLoganUnited States
| | - Eric J Sefing
- Department of Animal, Dairy and Veterinary Sciences, Utah State UniversityLoganUnited States
| | - Kevin W Bailey
- Department of Animal, Dairy and Veterinary Sciences, Utah State UniversityLoganUnited States
| | - Arnaud J Van Wettere
- Department of Animal, Dairy and Veterinary Sciences, Utah State UniversityLoganUnited States
- Utah Veterinary Diagnostic Laboratory, Utah State UniversityLoganUnited States
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles (UCLA)Los AngelesUnited States
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLALos AngelesUnited States
- UCLA Molecular Biology InstituteLos AngelesUnited States
- UCLA Jonsson Comprehensive Cancer CenterLos AngelesUnited States
- UCLA AIDS InstituteLos AngelesUnited States
| | - Brian B Gowen
- Department of Animal, Dairy and Veterinary Sciences, Utah State UniversityLoganUnited States
| |
Collapse
|
177
|
Riedelberger M, Penninger P, Tscherner M, Hadriga B, Brunnhofer C, Jenull S, Stoiber A, Bourgeois C, Petryshyn A, Glaser W, Limbeck A, Lynes MA, Schabbauer G, Weiss G, Kuchler K. Type I Interferons Ameliorate Zinc Intoxication of Candida glabrata by Macrophages and Promote Fungal Immune Evasion. iScience 2020; 23:101121. [PMID: 32428860 PMCID: PMC7232100 DOI: 10.1016/j.isci.2020.101121] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/09/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022] Open
Abstract
Host and fungal pathogens compete for metal ion acquisition during infectious processes, but molecular mechanisms remain largely unknown. Here, we show that type I interferons (IFNs-I) dysregulate zinc homeostasis in macrophages, which employ metallothionein-mediated zinc intoxication of pathogens as fungicidal response. However, Candida glabrata can escape immune surveillance by sequestering zinc into vacuoles. Interestingly, zinc-loading is inhibited by IFNs-I, because a Janus kinase 1 (JAK1)-dependent suppression of zinc homeostasis affects zinc distribution in macrophages as well as generation of reactive oxygen species (ROS). In addition, systemic fungal infections elicit IFN-I responses that suppress splenic zinc homeostasis, thereby altering macrophage zinc pools that otherwise exert fungicidal actions. Thus, IFN-I signaling inadvertently increases fungal fitness both in vitro and in vivo during fungal infections. Our data reveal an as yet unrecognized role for zinc intoxication in antifungal immunity and suggest that interfering with host zinc homeostasis may offer therapeutic options to treat invasive fungal infections.
Collapse
Affiliation(s)
- Michael Riedelberger
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Philipp Penninger
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Michael Tscherner
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Bernhard Hadriga
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Carina Brunnhofer
- Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Sabrina Jenull
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anton Stoiber
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Christelle Bourgeois
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Andriy Petryshyn
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Walter Glaser
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Andreas Limbeck
- Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, CT, USA
| | - Gernot Schabbauer
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, and Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Karl Kuchler
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria.
| |
Collapse
|
178
|
Jiang R, Han B, Song M, Xue B, Zhang Y, Ding Y, Chen J, Zhu J, Liu J, Nie Q, Han X, Jin X, Shan X, Guo W, Zhang E, Zhang Z, Zhang C, Zhang J, Wang B, Dong S, Li J, Li X, Li X. Efficacy and safety of aerosol inhalation of recombinant human interferon α1b (IFNα1b) injection for noninfluenza viral pneumonia, a multicenter, randomized, double-blind, placebo-controlled trial. JOURNAL OF INFLAMMATION-LONDON 2020; 17:19. [PMID: 32431566 PMCID: PMC7221328 DOI: 10.1186/s12950-020-00249-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Background To investigate the efficacy and safety of aerosol inhalation of recombinant human interferon α1b (IFNα1b) injection for noninfluenza viral pneumonia. Methods One hundred sixty-four patients with noninfluenza viral pneumonia were divided into IFNα1b and control groups. The IFNα1b group received routine treatment + aerosol inhalation of recombinant human IFNα1b injection (50 μg × 2 injections, bid). The control group received routine treatment + IFN analog (two injections, bid). Overall response rate (ORR) of five kinds clinical symptoms. Further outcomes were daily average score and the response rate of each of the symptoms above. Results A total of 163 patients were included in the full analysis set (FAS) and 151 patients were included in the per-protocol set (PPS). After 7 days of treatment, ORR of clinical symptoms was higher in IFNα1b group than that in control group for both the FAS and PPS. Moreover, after 7 days of treatment, the daily score of three efficacy indexes including expectoration, respiratory rate, and pulmonary rales were improved. The ORRs for expectoration and pulmonary rales were higher in the IFNα1b group than in the control group (P < 0.05). There were no significant differences of the ORRs for coughing, chest pain and respiratory rate between the two groups (P > 0.05). The incidence of adverse events was 6.5% (n = 5) in IFNα1b group and 3.5% (n = 3) in control group (P > 0.05). Conclusion Aerosol inhalation of recombinant human IFNα1b is safe and it can improve the clinical symptoms of noninfluenza viral pneumonia.
Collapse
Affiliation(s)
- Rongmeng Jiang
- 1Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, No. 8 East Jingshun Street, Chaoyang District, Beijing, 100015 China
| | - Bing Han
- 1Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, No. 8 East Jingshun Street, Chaoyang District, Beijing, 100015 China
| | - Meihua Song
- 1Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, No. 8 East Jingshun Street, Chaoyang District, Beijing, 100015 China
| | - Bing Xue
- 2Department of Respiratory Medicine, Beijing Chuiyangliu Hospital, Beijing, 100022 China
| | - Yongxiang Zhang
- 3Department of Respiratory Medicine, People's Hospital of Beijing Daxing District, Beijing, 102600 China
| | - Yanyan Ding
- 3Department of Respiratory Medicine, People's Hospital of Beijing Daxing District, Beijing, 102600 China
| | - Jin Chen
- 4Department of Respiratory Medicine, Fuxing Hospital, Capital Medical University, Beijing, 100038 China
| | - Jing Zhu
- 4Department of Respiratory Medicine, Fuxing Hospital, Capital Medical University, Beijing, 100038 China
| | - Jianhua Liu
- Department of Respiratory Medicine, Beijing Huairou District Hospital, Beijing, 101400 China
| | - Qingrong Nie
- Department of Respiratory Medicine, Liangxiang Hospital of Fangshan District, Beijing, 102401 China
| | - Xuefeng Han
- Department of Respiratory Medicine, Liangxiang Hospital of Fangshan District, Beijing, 102401 China
| | - Xiuhong Jin
- Department of Respiratory Medicine, Beijing Pinggu Hospital, Beijing, 101200 China
| | - Xiaoyin Shan
- Department of Respiratory Medicine, Beijing Pinggu Hospital, Beijing, 101200 China
| | - Weian Guo
- 8Department of Respiratory Medicine, Peking University Shougang Hospital, Beijing, 100144 China
| | - Erming Zhang
- 8Department of Respiratory Medicine, Peking University Shougang Hospital, Beijing, 100144 China
| | - Zuoqing Zhang
- Department of Respiratory Medicine, Beijing Shijingshan Hospital, Beijing, 100043 China
| | - Changhong Zhang
- Department of Respiratory Medicine, Beijing Shijingshan Hospital, Beijing, 100043 China
| | - Jie Zhang
- 10Department of Respiratory Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070 China
| | - Baozeng Wang
- 11Department of Infectious Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070 China
| | - Shuwen Dong
- 10Department of Respiratory Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070 China
| | - Jiandong Li
- 12Department of Respiratory Medicine, General Hospital of Beijing Military Region, Beijing, 100700 China
| | - Xiaoguang Li
- 13Department of Infectious Disease, Peking University Third Hospital, Beijing, 100191 China
| | - Xingwang Li
- 1Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, No. 8 East Jingshun Street, Chaoyang District, Beijing, 100015 China
| |
Collapse
|
179
|
Heyman O, Horev Y, Koren N, Barel O, Aizenbud I, Aizenbud Y, Brandwein M, Shapira L, Hovav A, Wilensky A. Niche Specific Microbiota-Dependent and Independent Bone Loss around Dental Implants and Teeth. J Dent Res 2020; 99:1092-1101. [DOI: 10.1177/0022034520920577] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Oral mucosal homeostasis is achieved by complex immunologic mechanisms, orchestrating host immunity to adapt to the physiologic functions of the various specialized niches in the oral cavity. Dental implants introduce a novel mucosal niche to the immune system to deal with. Nevertheless, the immune mechanisms engaged toward implants and whether they have broader effects are not well defined. Using a murine model, we found an accumulation of neutrophils and RANKL-expressing T and B lymphocytes in the implant-surrounding mucosa, accompanied by local bone loss. Surprisingly, the presence of implants had an impact on remote periodontal sites, as elevated inflammation and accelerated bone loss were detected in intact distant teeth. This was due to microbial dysbiosis induced by the implants, since antibiotic treatment prevented bone loss around teeth. However, antibiotic treatment failed to prevent the loss of implant-supporting bone, highlighting the distinct mechanisms mediating bone loss at each site. Further analysis revealed that implants induced chronic lymphocyte activation and increased mRNA expression of IFN-α and accumulation of IFN-α–producing plasmacytoid dendritic cells, which we previously reported as bone-destructive immune responses. Collectively, this study demonstrates that implants have a strong and broad impact on oral mucosal homeostasis, inducing periodontal bone loss in a niche-specific manner that is both microbiota dependent and independent.
Collapse
Affiliation(s)
- O. Heyman
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| | - Y. Horev
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| | - N. Koren
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - O. Barel
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - I. Aizenbud
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Y. Aizenbud
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - M. Brandwein
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - L. Shapira
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| | - A.H. Hovav
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - A. Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University–Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
180
|
Benci JL, Johnson LR, Choa R, Xu Y, Qiu J, Zhou Z, Xu B, Ye D, Nathanson KL, June CH, Wherry EJ, Zhang NR, Ishwaran H, Hellmann MD, Wolchok JD, Kambayashi T, Minn AJ. Opposing Functions of Interferon Coordinate Adaptive and Innate Immune Responses to Cancer Immune Checkpoint Blockade. Cell 2020; 178:933-948.e14. [PMID: 31398344 DOI: 10.1016/j.cell.2019.07.019] [Citation(s) in RCA: 335] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 04/24/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
Abstract
Interferon-gamma (IFNG) augments immune function yet promotes T cell exhaustion through PDL1. How these opposing effects are integrated to impact immune checkpoint blockade (ICB) is unclear. We show that while inhibiting tumor IFNG signaling decreases interferon-stimulated genes (ISGs) in cancer cells, it increases ISGs in immune cells by enhancing IFNG produced by exhausted T cells (TEX). In tumors with favorable antigenicity, these TEX mediate rejection. In tumors with neoantigen or MHC-I loss, TEX instead utilize IFNG to drive maturation of innate immune cells, including a PD1+TRAIL+ ILC1 population. By disabling an inhibitory circuit impacting PD1 and TRAIL, blocking tumor IFNG signaling promotes innate immune killing. Thus, interferon signaling in cancer cells and immune cells oppose each other to establish a regulatory relationship that limits both adaptive and innate immune killing. In melanoma and lung cancer patients, perturbation of this relationship is associated with ICB response independent of tumor mutational burden.
Collapse
Affiliation(s)
- Joseph L Benci
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lexus R Johnson
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ruth Choa
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuanming Xu
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingya Qiu
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zilu Zhou
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bihui Xu
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Darwin Ye
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine L Nathanson
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E John Wherry
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy R Zhang
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hemant Ishwaran
- Division of Biostatistics, Department of Epidemiology and Public Health, University of Miami, Miami, FL 33136, USA
| | - Matthew D Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andy J Minn
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
181
|
Cooper L, Good-Jacobson KL. Dysregulation of humoral immunity in chronic infection. Immunol Cell Biol 2020; 98:456-466. [PMID: 32275789 DOI: 10.1111/imcb.12338] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Chronic viral infections disrupt the ability of the humoral immune response to produce neutralizing antibody or form effective immune memory, preventing viral clearance and making vaccine design difficult. Multiple components of the B-cell response are affected by pathogens that are not cleared from the host. Changes in the microenvironment shift production of B cells to short-lived plasma cells early in the response. Polyclonal B cells are recruited into both the plasma cell and germinal center compartments, inhibiting the formation of a targeted, high-affinity response. Finally, memory B cells shift toward an "atypical" phenotype, which may in turn result in changes to the functional properties of this population. While similar properties of B-cell dysregulation have been described across different types of persistent infections, key questions about the underlying mechanisms remain. This review will discuss the recent advances in this field, as well as highlight the critical questions about the interplay between viral load, microenvironment, the polyclonal response and atypical memory B cells that are yet to be answered. Design of new preventative treatments will rely on identifying the extrinsic and intrinsic modulators that push B cells toward an ineffective response, and thus identify new ways to guide them back onto the best path for clearance of virus and formation of effective immune memory.
Collapse
Affiliation(s)
- Lucy Cooper
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Kim L Good-Jacobson
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
182
|
Namineni S, O'Connor T, Faure-Dupuy S, Johansen P, Riedl T, Liu K, Xu H, Singh I, Shinde P, Li F, Pandyra A, Sharma P, Ringelhan M, Muschaweckh A, Borst K, Blank P, Lampl S, Neuhaus K, Durantel D, Farhat R, Weber A, Lenggenhager D, Kündig TM, Staeheli P, Protzer U, Wohlleber D, Holzmann B, Binder M, Breuhahn K, Assmus LM, Nattermann J, Abdullah Z, Rolland M, Dejardin E, Lang PA, Lang KS, Karin M, Lucifora J, Kalinke U, Knolle PA, Heikenwalder M. A dual role for hepatocyte-intrinsic canonical NF-κB signaling in virus control. J Hepatol 2020; 72:960-975. [PMID: 31954207 DOI: 10.1016/j.jhep.2019.12.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 12/02/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Hepatic innate immune control of viral infections has largely been attributed to Kupffer cells, the liver-resident macrophages. However, hepatocytes, the parenchymal cells of the liver, also possess potent immunological functions in addition to their known metabolic functions. Owing to their abundance in the liver and known immunological functions, we aimed to investigate the direct antiviral mechanisms employed by hepatocytes. METHODS Using lymphocytic choriomeningitis virus (LCMV) as a model of liver infection, we first assessed the role of myeloid cells by depletion prior to infection. We investigated the role of hepatocyte-intrinsic innate immune signaling by infecting mice lacking canonical NF-κB signaling (IkkβΔHep) specifically in hepatocytes. In addition, mice lacking hepatocyte-specific interferon-α/β signaling-(IfnarΔHep), or interferon-α/β signaling in myeloid cells-(IfnarΔMyel) were infected. RESULTS Here, we demonstrate that LCMV activates NF-κB signaling in hepatocytes. LCMV-triggered NF-κB activation in hepatocytes did not depend on Kupffer cells or TNFR1 signaling but rather on Toll-like receptor signaling. LCMV-infected IkkβΔHep livers displayed strongly elevated viral titers due to LCMV accumulation within hepatocytes, reduced interferon-stimulated gene (ISG) expression, delayed intrahepatic immune cell influx and delayed intrahepatic LCMV-specific CD8+ T cell responses. Notably, viral clearance and ISG expression were also reduced in LCMV-infected primary hepatocytes lacking IKKβ, demonstrating a hepatocyte-intrinsic effect. Similar to livers of IkkβΔHep mice, enhanced hepatocytic LCMV accumulation was observed in livers of IfnarΔHep mice, whereas IfnarΔMyel mice were able to control LCMV infection. Hepatocytic NF-κB signaling was also required for efficient ISG induction in HDV-infected dHepaRG cells and interferon-α/β-mediated inhibition of HBV replication in vitro. CONCLUSIONS Together, these data show that hepatocyte-intrinsic NF-κB is a vital amplifier of interferon-α/β signaling, which is pivotal for strong early ISG responses, immune cell infiltration and hepatic viral clearance. LAY SUMMARY Innate immune cells have been ascribed a primary role in controlling viral clearance upon hepatic infections. We identified a novel dual role for NF-κB signaling in infected hepatocytes which was crucial for maximizing interferon responses and initiating adaptive immunity, thereby efficiently controlling hepatic virus replication.
Collapse
Affiliation(s)
- Sukumar Namineni
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany; Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Tracy O'Connor
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Suzanne Faure-Dupuy
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pål Johansen
- Department of Dermatology, University Hospital Zurich and University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Tobias Riedl
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kaijing Liu
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Haifeng Xu
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Indrabahadur Singh
- Emmy Noether Research Group Epigenetic Machineries and Cancer, Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Prashant Shinde
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätstr.1, 40225 Düsseldorf, Germany
| | - Fanghui Li
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Aleksandra Pandyra
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Piyush Sharma
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany; Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA, 38105
| | - Marc Ringelhan
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany; Department of Internal Medicine II, University Hospital rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Andreas Muschaweckh
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Katharina Borst
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Patrick Blank
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Sandra Lampl
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Katharina Neuhaus
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Durantel
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Rayan Farhat
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Daniela Lenggenhager
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich and University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Peter Staeheli
- Institute of Virology, University of Freiburg, Freiburg, Germany
| | - Ulrike Protzer
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany
| | - Dirk Wohlleber
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Bernhard Holzmann
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Marco Binder
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Jacob Nattermann
- Department of Internal Medicine, University of Bonn, Bonn, Germany
| | | | - Maude Rolland
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Institute, University of Liège, 4000 Liège, Belgium
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Institute, University of Liège, 4000 Liège, Belgium
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätstr.1, 40225 Düsseldorf, Germany
| | - Karl S Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Julie Lucifora
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany; Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany.
| |
Collapse
|
183
|
Kumar D, Romero Y, Schuck KN, Smalley H, Subedi B, Fleming SD. Drivers and regulators of humoral innate immune responses to infection and cancer. Mol Immunol 2020; 121:99-110. [PMID: 32199212 PMCID: PMC7207242 DOI: 10.1016/j.molimm.2020.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/21/2022]
Abstract
The complement cascade consists of cell bound and serum proteins acting together to protect the host from pathogens, remove cancerous cells and effectively links innate and adaptive immune responses. Despite its usefulness in microbial neutralization and clearance of cancerous cells, excessive complement activation causes an immune imbalance and tissue damage in the host. Hence, a series of complement regulatory proteins present at a higher concentration in blood plasma and on cell surfaces tightly regulate the cascade. The complement cascade can be initiated by B-1 B cell production of natural antibodies. Natural antibodies arise spontaneously without any known exogenous antigenic or microbial stimulus and protect against invading pathogens, clear apoptotic cells, provide tissue homeostasis, and modulate adaptive immune functions. Natural IgM antibodies recognize microbial and cancer antigens and serve as an activator of complement mediated lysis. This review will discuss advances in complement activation and regulation in bacterial and viral infections, and cancer. We will also explore the crosstalk of natural antibodies with bacterial populations and cancer.
Collapse
MESH Headings
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Apoptosis/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Bacterial Infections/immunology
- Complement Activation
- Complement System Proteins/immunology
- Complement System Proteins/metabolism
- Humans
- Immunity, Humoral
- Immunity, Innate
- Immunoglobulin M/immunology
- Immunoglobulin M/metabolism
- Neoplasms/immunology
- Receptors, Complement/immunology
- Receptors, Complement/metabolism
- Tumor Escape
- Virus Diseases/immunology
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Yeni Romero
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA
| | - Kaitlynn N Schuck
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Haley Smalley
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Bibek Subedi
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
184
|
TLR4 abrogates the Th1 immune response through IRF1 and IFN-β to prevent immunopathology during L. infantum infection. PLoS Pathog 2020; 16:e1008435. [PMID: 32210480 PMCID: PMC7135367 DOI: 10.1371/journal.ppat.1008435] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/06/2020] [Accepted: 02/25/2020] [Indexed: 12/26/2022] Open
Abstract
A striking feature of human visceral leishmaniasis (VL) is chronic inflammation in the spleen and liver, and VL patients present increased production levels of multiple inflammatory mediators, which contribute to tissue damage and disease severity. Here, we combined an experimental model with the transcriptional profile of human VL to demonstrate that the TLR4-IFN-β pathway regulates the chronic inflammatory process and is associated with the asymptomatic form of the disease. Tlr4-deficient mice harbored fewer parasites in their spleen and liver than wild-type mice. TLR4 deficiency enhanced the Th1 immune response against the parasite, which was correlated with an increased activation of dendritic cells (DCs). Gene expression analyses demonstrated that IRF1 and IFN-β were expressed downstream of TLR4 after infection. Accordingly, IRF1- and IFNAR-deficient mice harbored fewer parasites in the target organs than wild-type mice due to having an increased Th1 immune response. However, the absence of TLR4 or IFNAR increased the serum transaminase levels in infected mice, indicating the presence of liver damage in these animals. In addition, IFN-β limits IFN-γ production by acting directly on Th1 cells. Using RNA sequencing analysis of human samples, we demonstrated that the transcriptional signature for the TLR4 and type I IFN (IFN-I) pathways was positively modulated in asymptomatic subjects compared with VL patients and thus provide direct evidence demonstrating that the TLR4-IFN-I pathway is related to the nondevelopment of the disease. In conclusion, our results demonstrate that the TLR4-IRF1 pathway culminates in IFN-β production as a mechanism for dampening the chronic inflammatory process and preventing immunopathology development. Visceral leishmaniasis (VL) is one of the most lethal neglected tropical diseases and is caused by Leishmania parasites. Most subjects infected with Leishmania present subclinical VL symptoms, and their immune response is mediated by Th1 cells and immunoregulatory mechanisms. However, when infection progresses to disease, VL patients present increased levels of inflammatory mediators in the serum which are related to the severity of disease. During infection, Toll-like receptors (TLRs) interact with Leishmania parasites and contribute to the outcome of the disease. Herein, we report that TLR4 signaling hampers the chronic immune response during VL to prevent immunopathology. TLR4 triggers the activation of IRF1 and thus induces the transcription of IFN-β, which in turn acts directly on Th1 cells to limit the production of IFN-γ. In addition, a transcription analysis of human VL samples provides direct evidence demonstrating that the TLR4-IFN-I pathway is related to the asymptomatic form of the disease. Collectively, our findings reveal that TLR4 hampers the Th1 immune response through IRF1 and IFN-β to prevent immunopathology during VL.
Collapse
|
185
|
Pathogenic Role of Type I Interferons in HIV-Induced Immune Impairments in Humanized Mice. Curr HIV/AIDS Rep 2020; 16:224-229. [PMID: 31055732 DOI: 10.1007/s11904-019-00444-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE OF REVIEW Recent findings on the critical pathogenic role of type 1 interferons (IFN-I) in HIV-1 persistence in humanized mice suggest that inhibiting IFN-I signaling transiently will reverse HIV-induced inflammatory diseases and rescue anti-HIV immunity to control HIV-1 reservoirs. RECENT FINDINGS In both humanized mice and in monkeys, IFN-I signaling is functionally defined to play an important role in suppressing early HIV-1 and SIV infection. During persistent infection in humanized mice, however, IFN-I signaling is revealed to induce T cell depletion and impairment. Interestingly, in HIV-infected mice with effective combination antiretroviral therapy (cART), blocking IFN-I signaling reverses HIV-induced inflammation, rescues anti-HIV T cells, and reduces HIV-1 reservoirs. These findings functionally define the role of IFN-I in HIV-1 reservoir persistence and suggest that blocking IFN-I signaling will provide a novel therapeutic strategy to (i) reverse inflammation-associated diseases in HIV patients under cART, (ii) rescue host anti-HIV immunity, and (iii) reduce or control HIV-1 reservoirs.
Collapse
|
186
|
Morgado FN, da Silva AVA, Porrozzi R. Infectious Diseases and the Lymphoid Extracellular Matrix Remodeling: A Focus on Conduit System. Cells 2020; 9:cells9030725. [PMID: 32187985 PMCID: PMC7140664 DOI: 10.3390/cells9030725] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022] Open
Abstract
The conduit system was described in lymphoid organs as a tubular and reticular set of structures compounded by collagen, laminin, perlecan, and heparin sulfate proteoglycan wrapped by reticular fibroblasts. This tubular system is capable of rapidly transport small molecules such as viruses, antigens, chemokines, cytokines, and immunoglobulins through lymphoid organs. This structure plays an important role in guiding the cells to their particular niches, therefore participating in cell cooperation, antigen presentation, and cellular activation. The remodeling of conduits has been described in chronic inflammation and infectious diseases to improve the transport of antigens to specific T and B cells in lymphoid tissue. However, malnutrition and infectious agents may induce extracellular matrix remodeling directly or indirectly, leading to the microarchitecture disorganization of secondary lymphoid organs and their conduit system. In this process, the fibers and cells that compound the conduit system may also be altered, which affects the development of a specific immune response. This review aims to discuss the extracellular matrix remodeling during infectious diseases with an emphasis on the alterations of molecules from the conduit system, which damages the cellular and molecular transit in secondary lymphoid organs compromising the immune response.
Collapse
Affiliation(s)
- Fernanda N. Morgado
- Correspondence: (F.N.M.); (R.P.); Tel.: +55-2138658226 (F.N.M.); +55-2138658203 (R.P.)
| | | | - Renato Porrozzi
- Correspondence: (F.N.M.); (R.P.); Tel.: +55-2138658226 (F.N.M.); +55-2138658203 (R.P.)
| |
Collapse
|
187
|
Zhou L, Zhang Y, Wang Y, Zhang M, Sun W, Dai T, Wang A, Wu X, Zhang S, Wang S, Zhou F. A Dual Role of Type I Interferons in Antitumor Immunity. ACTA ACUST UNITED AC 2020; 4:e1900237. [PMID: 33245214 DOI: 10.1002/adbi.201900237] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFN-Is) are a family of cytokines that exert direct antiviral effects and regulate innate and adaptive immune responses through direct and indirect mechanisms. It is generally believed that IFN-Is repress tumor development via restricting tumor proliferation and inducing antitumor immune responses. However, recent emerging evidence suggests that IFN-Is play a dual role in antitumor immunity. That is, in the early stage of tumorigenesis, IFN-Is promote the antitumor immune response by enhancing antigen presentation in antigen-presenting cells and activating CD8+ T cells. However, in the late stage of tumor progression, persistent expression of IFN-Is induces the expression of immunosuppressive factors (PD-L1, IDO, and IL-10) on the surface of dendritic cells and other bone marrow cells and inhibits their antitumor immunity. This review outlines these dual functions of IFN-Is in antitumor immunity and elucidates the involved mechanisms, as well as their applications in tumor therapy.
Collapse
Affiliation(s)
- Lili Zhou
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yuqi Zhang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yongqiang Wang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Meirong Zhang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Wenhuan Sun
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Tong Dai
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Aijun Wang
- Department of Surgery, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Xiaojin Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Suping Zhang
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for international Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518055, China
| | - Shuai Wang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Fangfang Zhou
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
188
|
Elsaesser HJ, Mohtashami M, Osokine I, Snell LM, Cunningham CR, Boukhaled GM, McGavern DB, Zúñiga-Pflücker JC, Brooks DG. Chronic virus infection drives CD8 T cell-mediated thymic destruction and impaired negative selection. Proc Natl Acad Sci U S A 2020; 117:5420-5429. [PMID: 32094187 PMCID: PMC7071912 DOI: 10.1073/pnas.1913776117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chronic infection provokes alterations in inflammatory and suppressive pathways that potentially affect the function and integrity of multiple tissues, impacting both ongoing immune control and restorative immune therapies. Here we demonstrate that chronic lymphocytic choriomeningitis virus infection rapidly triggers severe thymic depletion, mediated by CD8 T cell-intrinsic type I interferon (IFN) and signal transducer and activator of transcription 2 (Stat2) signaling. Occurring temporal to T cell exhaustion, thymic cellularity reconstituted despite ongoing viral replication, with a rapid secondary thymic depletion following immune restoration by anti-programmed death-ligand 1 (PDL1) blockade. Therapeutic hematopoietic stem cell transplant (HSCT) during chronic infection generated new antiviral CD8 T cells, despite sustained virus replication in the thymus, indicating an impairment in negative selection. Consequently, low amounts of high-affinity self-reactive T cells also escaped the thymus following HSCT during chronic infection. Thus, by altering the stringency and partially impairing negative selection, the host generates new virus-specific T cells to replenish the fight against the chronic infection, but also has the potentially dangerous effect of enabling the escape of self-reactive T cells.
Collapse
Affiliation(s)
- Heidi J Elsaesser
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Mahmood Mohtashami
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8 Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Ivan Osokine
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Laura M Snell
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Cameron R Cunningham
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Giselle M Boukhaled
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20824
| | - Juan Carlos Zúñiga-Pflücker
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8 Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - David G Brooks
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2M9, Canada;
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8 Canada
| |
Collapse
|
189
|
Li SY, Zhang ZN, Jiang YJ, Fu YJ, Shang H. Transcriptional insights into the CD8 + T cell response in mono-HIV and HCV infection. J Transl Med 2020; 18:96. [PMID: 32093694 PMCID: PMC7038596 DOI: 10.1186/s12967-020-02252-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/31/2020] [Indexed: 12/04/2022] Open
Abstract
Background Disease progression in the absence of therapy varies significantly in mono-HIV and HCV infected individuals. Virus-specific CD8+ T cells play an important role in restricting lentiviral replication and determining the rate of disease progression during HIV and HCV mono- and co-infection. Thus, understanding the similarities in the characteristics of CD8+ T cells in mono-HIV and HCV infection at the transcriptomic level contributes to the development of antiviral therapy. In this study, a meta-analysis of CD8+ T cell gene expression profiles derived from mono-HIV and HCV infected individuals at different stages of disease progression, was conducted to understand the common changes experienced by CD8+ T cells. Methods Five microarray datasets, reporting CD8+ T cell mRNA expression of the mono-HIV and HCV infected patients, were retrieved from Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) were identified via integrative meta-analysis of expression data (INMEX) program. Network analysis methods were used to assess protein–protein interaction (PPI) networks, Gene Ontology (GO) terms and pathway enrichment for DEGs. MirDIP and miRDB online prediction tools were used to predict potential microRNAs (miRNAs) targeting hub genes. Results First, we identified 625 and 154 DEGs in the CD8+ T cells originating from mono-HIV and HCV chronic progressor patients, respectively, compared to healthy individuals. Among them, interferon-stimulated genes (ISGs) including ISG15, IFIT3, ILI44L, CXCL8, FPR1 and TLR2, were upregulated after mono-HIV and HCV infection. Pathway enrichment analysis of DEGs showed that the “cytokine–cytokine receptor interaction” and “NF-kappa B” signaling pathways were upregulated after mono-HIV and HCV infection. In addition, we identified 92 and 50 DEGs in the CD8+ T cells of HIV non-progressor and HCV resolver patients, respectively, compared with corresponding chronic progressors. We observed attenuated mitosis and reduced ISG expression in HIV non-progressors and HCV resolvers compared with the corresponding chronic progressors. Finally, we identified miRNA-143-3p, predicted to target both IFIT3 in HIV and STAT5A in HCV infection. Conclusions We identified DEGs and transcriptional patterns in mono-HIV and HCV infected individuals at different stages of disease progression and identified miRNA-143-3p with potential to intervene disease progression, which provides a new strategy for developing targeted therapies.
Collapse
Affiliation(s)
- Si-Yao Li
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Zi-Ning Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yong-Jun Jiang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Ya-Jing Fu
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Hong Shang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China. .,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
190
|
Drouin M, Saenz J, Chiffoleau E. C-Type Lectin-Like Receptors: Head or Tail in Cell Death Immunity. Front Immunol 2020; 11:251. [PMID: 32133013 PMCID: PMC7040094 DOI: 10.3389/fimmu.2020.00251] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
C-type lectin-like receptors (CLRs) represent a family of transmembrane pattern recognition receptors, expressed primarily by myeloid cells. They recognize not only pathogen moieties for host defense, but also modified self-antigens such as damage-associated molecular patterns released from dead cells. Upon ligation, CLR signaling leads to the production of inflammatory mediators to shape amplitude, duration and outcome of the immune response. Thus, following excessive injury, dysregulation of these receptors leads to the development of inflammatory diseases. Herein, we will focus on four CLRs of the "Dectin family," shown to decode the immunogenicity of cell death. CLEC9A on dendritic cells links F-actin exposed by dying cells to favor cross-presentation of dead-cell associated antigens to CD8+ T cells. Nevertheless, CLEC9A exerts also feedback mechanisms to temper neutrophil recruitment and prevent additional tissue damage. MINCLE expressed by macrophages binds nuclear SAP130 released by necrotic cells to potentiate pro-inflammatory responses. However, the consequent inflammation can exacerbate pathogenesis of inflammatory diseases. Moreover, in a tumor microenvironment, MINCLE induces macrophage-induced immune suppression and cancer progression. Similarly, triggering of LOX-1 by oxidized LDL, amplifies pro-inflammatory response but promotes tumor immune escape and metastasis. Finally, CLEC12A that recognizes monosodium urate crystals formed during cell death, inhibits activating signals to prevent detrimental inflammation. Interestingly, CLEC12A also sustains type-I IFN response to finely tune immune responses in case of viral-induced collateral damage. Therefore, CLRs acting in concert as sensors of injury, could be used in a targeted way to treat numerous diseases such as allergies, obesity, tumors, and autoimmunity.
Collapse
Affiliation(s)
- Marion Drouin
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Javier Saenz
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Elise Chiffoleau
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
191
|
Chronic Lymphocytic Choriomeningitis Infection Causes Susceptibility to Mousepox and Impairs Natural Killer Cell Maturation and Function. J Virol 2020; 94:JVI.01831-19. [PMID: 31776282 DOI: 10.1128/jvi.01831-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 11/25/2019] [Indexed: 11/20/2022] Open
Abstract
Chronic viral infections. like those of humans with cytomegalovirus, human immunodeficiency virus (even when under antiretroviral therapy), and hepatitis C virus or those of mice with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13), result in immune dysfunction that predisposes the host to severe infections with unrelated pathogens. It is known that C57BL/6 (B6) mice are resistant to mousepox, a lethal disease caused by the orthopoxvirus ectromelia virus (ECTV), and that this resistance requires natural killer (NK) cells and other immune cells. We show that most B6 mice chronically infected with CL13 succumb to mousepox but that most of those that recovered from acute infection with the LCMV Armstrong (Arm) strain survive. We also show that B6 mice chronically infected with CL13 and those that recovered from Arm infection have a reduced frequency and a reduced number of NK cells. However, at steady state, NK cells in mice that have recovered from Arm infection mature normally and, in response to ECTV, get activated, become more mature, proliferate, and increase their cytotoxicity in vivo Conversely, in mice chronically infected with CL13, NK cells are immature and residually activated, and following ECTV infection, they do not mature, proliferate, or increase their cytotoxicity. Given the well-established importance of NK cells in resistance to mousepox, these data suggest that the NK cell dysfunction caused by CL13 persistence may contribute to the susceptibility of CL13-infected mice to mousepox. Whether chronic infections similarly affect NK cells in humans should be explored.IMPORTANCE Infection of adult mice with the clone 13 (CL13) strain of lymphocytic choriomeningitis virus (LCMV) is extensively used as a model of chronic infection. In this paper, we show that mice chronically infected with CL13 succumb to challenge with ectromelia virus (ECTV; the agent of mousepox) and that natural killer (NK) cells in CL13-infected mice are reduced in numbers and have an immature and partially activated phenotype but do respond to ECTV. These data may provide additional clues why humans chronically infected with certain pathogens are less resistant to viral diseases.
Collapse
|
192
|
Loss of Resistance to Mousepox during Chronic Lymphocytic Choriomeningitis Virus Infection Is Associated with Impaired T-Cell Responses and Can Be Rescued by Immunization. J Virol 2020; 94:JVI.01832-19. [PMID: 31826990 DOI: 10.1128/jvi.01832-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 11/29/2019] [Indexed: 01/21/2023] Open
Abstract
It is well established that chronic viral infections can cause immune suppression, resulting in increased susceptibility to other infectious diseases. However, the effects of chronic viral infection on T-cell responses and vaccination against highly pathogenic viruses are not well understood. We have recently shown that C57BL/6 (B6) mice lose their natural resistance to wild-type (WT) ectromelia virus (ECTV) when chronically infected with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13). Here we compared the T-cell response to ECTV in previously immunologically naive mice that were chronically infected with CL13 or that were convalescent from acute infection with the Armstrong (Arm) strain of LCMV. Our results show that mice that were chronically infected with CL13 but not those that had recovered from Arm infection have highly defective ECTV-specific CD8+ and CD4+ T-cell responses to WT ECTV. These defects are at least partly due to the chronic infection environment. In contrast to mice infected with WT ECTV, mice chronically infected with CL13 survived without signs of disease when infected with ECTV-Δ036, a mutant ECTV strain that is highly attenuated. Strikingly, mice chronically infected with CL13 mounted a strong CD8+ T-cell response to ECTV-Δ036 and survived without signs of disease after a subsequent challenge with WT ECTV. Our work suggests that enhanced susceptibility to acute viral infections in chronically infected individuals can be partly due to poor T-cell responses but that sufficient T-cell function can be recovered and resistance to acute infection can be restored by immunization with highly attenuated vaccines.IMPORTANCE Chronic viral infections may result in immunosuppression and enhanced susceptibility to infections with other pathogens. For example, we have recently shown that mice chronically infected with lymphocytic choriomeningitis virus (LCMV) clone 13 (CL13) are highly susceptible to mousepox, a disease that is caused by ectromelia virus and that is the mouse homolog of human smallpox. Here we show chronic CL13 infection severely disrupts the expansion, proliferation, activation, and cytotoxicity of T cells in response due at least in part to the suppressive effects of the chronic infection milieu. Notably, despite this profound immunodeficiency, mice chronically infected with CL13 could be protected by vaccination with a highly attenuated variant of ECTV. These results demonstrate that protective vaccination of immunosuppressed individuals is possible, provided that proper immunization tools are used.
Collapse
|
193
|
Integrating context of tumor biology and vaccine design to shape multidimensional immunotherapies. FUTURE DRUG DISCOVERY 2020. [DOI: 10.4155/fdd-2019-0031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Advances in cancer therapy have offered great promise but only modest clinical benefits as monotherapies to date. Patients usually respond well to therapies targeted at specific mutations, but only for a short time. Conversely, immunotherapies help fewer patients, but increase survival. Combination therapies, which could offer the best of both worlds, are currently limited by substantial toxicity. While recent advances in genomics and proteomics have yielded an unprecedented depth of enabling datasets, it has also shifted the focus toward in silico predictions. Designing the next wave of multidimensional immunotherapies will require leveraging this knowledge while providing a renewed emphasis on tumor biology and vaccine design. This includes careful selection of tumor clinical stage in the context of pre-existing tumor microenvironments, target antigen and technology platform selections to maximize their effect, and treatment staging. Here, we review strategies on how to approach an increasingly complex landscape of immunotherapeutic agents for use in combination therapies.
Collapse
|
194
|
Distinct Roles of Interferon Alpha and Beta in Controlling Chikungunya Virus Replication and Modulating Neutrophil-Mediated Inflammation. J Virol 2019; 94:JVI.00841-19. [PMID: 31619554 DOI: 10.1128/jvi.00841-19] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/04/2019] [Indexed: 12/19/2022] Open
Abstract
Type I interferons (IFNs) are key mediators of the innate immune response. Although members of this family of cytokines signal through a single shared receptor, biochemical and functional variation exists in response to different IFN subtypes. While previous work has demonstrated that type I IFNs are essential to control infection by chikungunya virus (CHIKV), a globally emerging alphavirus, the contributions of individual IFN subtypes remain undefined. To address this question, we evaluated CHIKV pathogenesis in mice lacking IFN-β (IFN-β knockout [IFN-β-KO] mice or mice treated with an IFN-β-blocking antibody) or IFN-α (IFN regulatory factor 7 knockout [IRF7-KO] mice or mice treated with a pan-IFN-α-blocking antibody). Mice lacking either IFN-α or IFN-β developed severe clinical disease following infection with CHIKV, with a marked increase in foot swelling compared to wild-type mice. Virological analysis revealed that mice lacking IFN-α sustained elevated infection in the infected ankle and in distant tissues. In contrast, IFN-β-KO mice displayed minimal differences in viral burdens within the ankle or at distal sites and instead had an altered cellular immune response. Mice lacking IFN-β had increased neutrophil infiltration into musculoskeletal tissues, and depletion of neutrophils in IFN-β-KO but not IRF7-KO mice mitigated musculoskeletal disease caused by CHIKV. Our findings suggest disparate roles for the IFN subtypes during CHIKV infection, with IFN-α limiting early viral replication and dissemination and IFN-β modulating neutrophil-mediated inflammation.IMPORTANCE Type I interferons (IFNs) possess a range of biological activity and protect against a number of viruses, including alphaviruses. Despite signaling through a shared receptor, there are established biochemical and functional differences among the IFN subtypes. The significance of our research is in demonstrating that IFN-α and IFN-β both have protective roles during acute chikungunya virus (CHIKV) infection but do so by distinct mechanisms. IFN-α limits CHIKV replication and dissemination, whereas IFN-β protects from CHIKV pathogenesis by limiting inflammation mediated by neutrophils. Our findings support the premise that the IFN subtypes have distinct biological activities in the antiviral response.
Collapse
|
195
|
Abstract
Interleukin (IL)-10 is an essential anti-inflammatory cytokine and functions as a negative regulator of immune responses to microbial antigens. IL-10 is particularly important in maintaining the intestinal microbe-immune homeostasis. Loss of IL-10 promotes the development of inflammatory bowel disease (IBD) as a consequence of an excessive immune response to the gut microbiota. IL-10 also functions more generally to prevent excessive inflammation during the course of infection. Although IL-10 can be produced by virtually all cells of the innate and adaptive immune system, T cells constitute a non-redundant source for IL-10 in many cases. The various roles of T cell-derived IL-10 will be discussed in this review. Given that IL-10 is at the center of maintaining the delicate balance between effective immunity and tissue protection, it is not surprising that IL-10 expression is highly dynamic and tightly regulated. We summarize the environmental signals and molecular pathways that regulate IL-10 expression. While numerous studies have provided us with a deep understanding of IL-10 biology, the majority of findings have been made in murine models, prompting us to highlight gaps in our knowledge about T cell-derived IL-10 in the human system.
Collapse
|
196
|
Kuka M, De Giovanni M, Iannacone M. The role of type I interferons in CD4 + T cell differentiation. Immunol Lett 2019; 215:19-23. [PMID: 30771379 PMCID: PMC7234836 DOI: 10.1016/j.imlet.2019.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFNs) released upon viral infections play different and opposing roles in disease outcome. This pleiotropic effect is mainly influenced by the cellular sources, timing and target cells for these molecules. The effect of type I IFN signaling on the activation and differentiation of antiviral CD4+ T cells remains ill defined, with studies reporting either a beneficial or a detrimental role, depending on the context of infection. This review will highlight several recent studies that have investigated the role of type I IFNs in the priming and polarization of CD4+ T cells and discuss areas of uncertainty that require further investigation.
Collapse
Affiliation(s)
- Mirela Kuka
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - Marco De Giovanni
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, 20132, Italy.
| |
Collapse
|
197
|
Dagenais-Lussier X, Loucif H, Cadorel H, Blumberger J, Isnard S, Bego MG, Cohen ÉA, Routy JP, van Grevenynghe J, for the Montreal Primary Infection Study Group. USP18 is a significant driver of memory CD4 T-cell reduced viability caused by type I IFN signaling during primary HIV-1 infection. PLoS Pathog 2019; 15:e1008060. [PMID: 31658294 PMCID: PMC6837632 DOI: 10.1371/journal.ppat.1008060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 11/07/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023] Open
Abstract
The loss of Memory CD4 T-cells (Mem) is a major hallmark of HIV-1 immuno-pathogenesis and occurs early during the first months of primary infection. A lot of effort has been put into understanding the molecular mechanisms behind this loss, yet they still have not been fully identified. In this study, we unveil the unreported role of USP18 in the deleterious effects of sustained type I IFN signaling on Mem, including HIV-1-specific CD4 T-cells. We find that interfering with IFN-I signaling pathway in infected patients, notably by targeting the interferon-stimulated gene USP18, resulted in reduced PTEN expression similar to those observed in uninfected control donors. We show that AKT activation in response to cytokine treatment, T-cell receptor (TcR) triggering, as well as HIV-1 Gag stimulation was significantly improved in infected patients when PTEN or USP18 were inhibited. Finally, our data demonstrate that higher USP18 in Mem from infected patients prevent proper cell survival and long-lasting maintenance in an AKT-dependent manner. Altogether, we establish a direct role for type I IFN/USP18 signaling in the maintenance of total and virus-specific Mem and provide a new mechanism for the reduced survival of these populations during primary HIV-1 infection. In this study, we expend our knowledge of how type I interferons (IFN-I) leads to memory CD4 T-cell defective survival by unveiling the molecular mechanism behind such impairments, placing USP18 at its center. Our data further deciphers the specific USP18-related mechanism that is responsible for such impairments by implicating AKT inhibition in a PTEN-dependent manner. Our findings also point to a potential use of neutralizing anti-interferon α/β receptor antibodies to rescue the defective memory CD4 T-cell survival during HIV-1 infection, even in HIV-1 specific CD4 T-cell. To conclude, our findings provide the characterization of the molecular pathway leading to disturbances caused by sustained IFN-I signaling which occurs early during primary HIV-1 infection, complementing current knowledge which placed sustained IFN-I signaling as detrimental to the host during this infection.
Collapse
Affiliation(s)
- Xavier Dagenais-Lussier
- Institut national de la recherche scientifique (INRS)-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, Canada
| | - Hamza Loucif
- Institut national de la recherche scientifique (INRS)-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, Canada
| | - Hugo Cadorel
- Institut national de la recherche scientifique (INRS)-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, Canada
| | - Juliette Blumberger
- Institut national de la recherche scientifique (INRS)-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, Canada
| | - Stéphane Isnard
- Chronic Viral Illness Service and Division of Hematology, McGill University Health Centre, Glen site, Montréal, Québec, Canada
| | - Mariana Gé Bego
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Éric A. Cohen
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Division of Hematology, McGill University Health Centre, Glen site, Montréal, Québec, Canada
| | - Julien van Grevenynghe
- Institut national de la recherche scientifique (INRS)-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, Canada
- * E-mail:
| | | |
Collapse
|
198
|
Liu D, Gibb DR, Escamilla-Rivera V, Liu J, Santhanakrishnan M, Shi Z, Xu L, Eisenbarth SC, Hendrickson JE. Type 1 IFN signaling critically regulates influenza-induced alloimmunization to transfused KEL RBCs in a murine model. Transfusion 2019; 59:3243-3252. [PMID: 31403208 PMCID: PMC6785373 DOI: 10.1111/trf.15482] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Only a fraction of red blood cell (RBC) transfusion recipients form alloantibodies, and variables determining responsiveness or nonresponsiveness are poorly understood. We and others have previously shown in animal models that pretreatment with toll-like receptor agonists that mimic different types of infections impacts the magnitude or frequency of RBC alloantibody responses. We hypothesized that influenza infection, coexistent with transfusion, would impact responses to transfused RBCs in a manner dependent on Type 1(α/β) interferon (IFN) signaling and tested this in a murine model. STUDY DESIGN AND METHODS Wild-type mice or mice lacking the ability to respond to Type 1 IFN were infected with influenza prior to the transfusion of transgenic murine RBCs (K1) expressing the human KEL glycoprotein or the triple fusion HOD protein. Alloantibody responses were measured longitudinally after transfusion by flow cytometric crossmatch, and posttransfusion RBC recovery and survival was evaluated. RESULTS Influenza-infected mice transfused with K1 RBCs developed robust anti-KEL alloantibodies, whereas animals transfused in the absence of infection remained nonresponders; influenza-associated RBC alloimmunization was also observed after transfusion of HOD RBCs. Recipient Type 1 IFN production was critical to the mechanism of action of influenza-induced RBC alloimmunization, with alloimmunization being significantly decreased in mice unable to sense Type 1 IFN (through antibody blockade or genetic approaches). CONCLUSION These and other data suggest that Type 1 IFN responses to toll-like receptor agonists or infections regulate RBC alloantibody responses. Studies investigating whether such a correlation exists in humans may be informative.
Collapse
Affiliation(s)
- Dong Liu
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT
| | - David R. Gibb
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT
- Cedars-Sinai Medical Center, Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, Los Angeles, CA
| | | | - Jingchun Liu
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT
| | | | - Zhimin Shi
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT
- Nanfang Hospital of Southern Medical University, Department of Primary Care, Guangzhou, Guangdong, China
| | - Lan Xu
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT
| | - Stephanie C. Eisenbarth
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT
- Yale University School of Medicine, Department of Immunobiology, New Haven, CT
| | - Jeanne E. Hendrickson
- Yale University School of Medicine, Department of Laboratory Medicine, New Haven, CT
- Yale University School of Medicine, Department of Pediatrics, New Haven, CA
| |
Collapse
|
199
|
Abstract
'Immune checkpoint blockade' for cancer describes the use of therapeutic antibodies that disrupt negative immune regulatory checkpoints and unleash pre-existing antitumour immune responses. Antibodies targeting the checkpoint molecules cytotoxic T lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD1) and PD1 ligand 1 (PD-L1) have had early success in the clinic, which has led to approval by the US Food and Drug Administration of multiple agents in several cancer types. Yet, clinicians still have very limited tools to discriminate a priori patients who will and will not respond to treatment. This has fuelled a wave of research into the molecular mechanisms of tumour-intrinsic resistance to immune checkpoint blockade, leading to the rediscovery of biological processes critical to antitumour immunity, namely interferon signalling and antigen presentation. Other efforts have shed light on the immunological implications of canonical cancer signalling pathways, such as WNT-β-catenin signalling, cell cycle regulatory signalling, mitogen-activated protein kinase signalling and pathways activated by loss of the tumour suppressor phosphoinositide phosphatase PTEN. Here we review each of these molecular mechanisms of resistance and explore ongoing approaches to overcome resistance to immune checkpoint blockade and expand the spectrum of patients who can benefit from immune checkpoint blockade.
Collapse
|
200
|
Dickow J, Francois S, Kaiserling RL, Malyshkina A, Drexler I, Westendorf AM, Lang KS, Santiago ML, Dittmer U, Sutter K. Diverse Immunomodulatory Effects of Individual IFNα Subtypes on Virus-Specific CD8 + T Cell Responses. Front Immunol 2019; 10:2255. [PMID: 31608062 PMCID: PMC6771563 DOI: 10.3389/fimmu.2019.02255] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/06/2019] [Indexed: 12/21/2022] Open
Abstract
Clinical administration of Interferon α (IFNα) resulted in limited therapeutic success against some viral infections. Immune modulation of CD8+ T cell responses during IFNα therapy is believed to play a pivotal role in promoting viral clearance. However, these clinical studies primarily focused on IFNα subtype 2. To date, the immunomodulatory roles of the remaining 10-13 IFNα subtypes remains poorly understood, thereby precluding assessments of their potential for more effective treatments. Here, we report that virus-specific CD8+ T cell responses were influenced to various extents by individual IFNα subtypes. IFNα4, 6, and 9 had the strongest effects on CD8+ T cells, including antiproliferative effects, improved cytokine production and cytotoxicity. Interestingly, augmented cytokine responses were dependent on IFNα subtype stimulation of dendritic cells (DCs), while antiproliferative effects and cytotoxicity were mediated by IFNAR signaling in either CD8+ T cells or DCs. Thus, precise modulation of virus-specific CD8+ T cell responses may be feasible for specific antiviral immunotherapies through careful selection and administration of individual IFNα subtypes.
Collapse
Affiliation(s)
- Julia Dickow
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sandra Francois
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Rouven-Luca Kaiserling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anna Malyshkina
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ingo Drexler
- Institute of Virology, University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Düsseldorf, Germany
| | - Astrid Maria Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karl Sebastian Lang
- Institute for Immunology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mario L. Santiago
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|