Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.793
Filter
Add more filters

Publication year range
1.
Cell ; 187(20): 5540-5553.e10, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39236707

ABSTRACT

In 2022, mpox virus (MPXV) spread worldwide, causing 99,581 mpox cases in 121 countries. Modified vaccinia Ankara (MVA) vaccine use reduced disease in at-risk populations but failed to deliver complete protection. Lag in manufacturing and distribution of MVA resulted in additional MPXV spread, with 12,000 reported cases in 2023 and an additional outbreak in Central Africa of clade I virus. These outbreaks highlight the threat of zoonotic spillover by Orthopoxviruses. mRNA-1769, an mRNA-lipid nanoparticle (LNP) vaccine expressing MPXV surface proteins, was tested in a lethal MPXV primate model. Similar to MVA, mRNA-1769 conferred protection against challenge and further mitigated symptoms and disease duration. Antibody profiling revealed a collaborative role between neutralizing and Fc-functional extracellular virion (EV)-specific antibodies in viral restriction and ospinophagocytic and cytotoxic antibody functions in protection against lesions. mRNA-1769 enhanced viral control and disease attenuation compared with MVA, highlighting the potential for mRNA vaccines to mitigate future pandemic threats.


Subject(s)
Antibodies, Viral , Vaccination , Vaccinia virus , Animals , Vaccinia virus/immunology , Vaccinia virus/genetics , Antibodies, Viral/immunology , mRNA Vaccines , Mpox (monkeypox)/prevention & control , Mpox (monkeypox)/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Neutralizing/immunology , Nanoparticles/chemistry , Female , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA, Messenger/immunology , Macaca mulatta , Macaca fascicularis , Liposomes
2.
Nat Immunol ; 23(1): 50-61, 2022 01.
Article in English | MEDLINE | ID: mdl-34853448

ABSTRACT

NP105-113-B*07:02-specific CD8+ T cell responses are considered among the most dominant in SARS-CoV-2-infected individuals. We found strong association of this response with mild disease. Analysis of NP105-113-B*07:02-specific T cell clones and single-cell sequencing were performed concurrently, with functional avidity and antiviral efficacy assessed using an in vitro SARS-CoV-2 infection system, and were correlated with T cell receptor usage, transcriptome signature and disease severity (acute n = 77, convalescent n = 52). We demonstrated a beneficial association of NP105-113-B*07:02-specific T cells in COVID-19 disease progression, linked with expansion of T cell precursors, high functional avidity and antiviral effector function. Broad immune memory pools were narrowed postinfection but NP105-113-B*07:02-specific T cells were maintained 6 months after infection with preserved antiviral efficacy to the SARS-CoV-2 Victoria strain, as well as Alpha, Beta, Gamma and Delta variants. Our data show that NP105-113-B*07:02-specific T cell responses associate with mild disease and high antiviral efficacy, pointing to inclusion for future vaccine design.


Subject(s)
HLA-B7 Antigen/immunology , Immunodominant Epitopes/immunology , Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Aged , Amino Acid Sequence , Antibodies, Viral/immunology , Antibody Affinity/immunology , COVID-19/immunology , COVID-19/pathology , Cell Line, Transformed , Female , Gene Expression Profiling , Humans , Immunologic Memory/immunology , Male , Middle Aged , Receptors, Antigen, T-Cell/immunology , Severity of Illness Index , Vaccinia virus/genetics , Vaccinia virus/immunology , Vaccinia virus/metabolism
3.
Nat Immunol ; 20(5): 602-612, 2019 05.
Article in English | MEDLINE | ID: mdl-30886418

ABSTRACT

Despite intense interest in antiviral T cell priming, the routes by which virions move in lymph nodes (LNs) are imperfectly understood. Current models fail to explain how virus-infected cells rapidly appear within the LN interior after viral infection. To better understand virion trafficking in the LN, we determined the locations of virions and infected cells after administration to mice of vaccinia virus or Zika virus. Notably, many rapidly infected cells in the LN interior were adjacent to LN conduits. Through the use of confocal and electron microscopy, we clearly visualized virions within conduits. Functionally, CD8+ T cells rapidly and preferentially associated with vaccinia virus-infected cells in the LN paracortex, which led to T cell activation in the LN interior. These results reveal that it is possible for even large virions to flow through LN conduits and infect dendritic cells within the T cell zone to prime CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Virion/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Female , Lymph Nodes/ultrastructure , Lymph Nodes/virology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Vaccinia virus/immunology , Vaccinia virus/physiology , Virion/physiology , Virion/ultrastructure , Virus Diseases/immunology , Virus Diseases/virology , Zika Virus/immunology , Zika Virus/physiology
4.
Cell ; 167(3): 684-694.e9, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768891

ABSTRACT

Monkeypox (MPXV) and cowpox (CPXV) are emerging agents that cause severe human infections on an intermittent basis, and variola virus (VARV) has potential for use as an agent of bioterror. Vaccinia immune globulin (VIG) has been used therapeutically to treat severe orthopoxvirus infections but is in short supply. We generated a large panel of orthopoxvirus-specific human monoclonal antibodies (Abs) from immune subjects to investigate the molecular basis of broadly neutralizing antibody responses for diverse orthopoxviruses. Detailed analysis revealed the principal neutralizing antibody specificities that are cross-reactive for VACV, CPXV, MPXV, and VARV and that are determinants of protection in murine challenge models. Optimal protection following respiratory or systemic infection required a mixture of Abs that targeted several membrane proteins, including proteins on enveloped and mature virion forms of virus. This work reveals orthopoxvirus targets for human Abs that mediate cross-protective immunity and identifies new candidate Ab therapeutic mixtures to replace VIG.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Specificity , Poxviridae Infections/immunology , Cowpox/immunology , Cowpox virus/immunology , Cross Reactions , Humans , Leukocytes, Mononuclear/immunology , Mpox (monkeypox)/immunology , Monkeypox virus/immunology , Smallpox/immunology , Vaccinia/immunology , Vaccinia virus/immunology , Variola virus/immunology
5.
Nat Immunol ; 17(7): 851-860, 2016 07.
Article in English | MEDLINE | ID: mdl-27158840

ABSTRACT

T cell antigen receptor (TCR) signaling drives distinct responses depending on the differentiation state and context of CD8(+) T cells. We hypothesized that access of signal-dependent transcription factors (TFs) to enhancers is dynamically regulated to shape transcriptional responses to TCR signaling. We found that the TF BACH2 restrains terminal differentiation to enable generation of long-lived memory cells and protective immunity after viral infection. BACH2 was recruited to enhancers, where it limited expression of TCR-driven genes by attenuating the availability of activator protein-1 (AP-1) sites to Jun family signal-dependent TFs. In naive cells, this prevented TCR-driven induction of genes associated with terminal differentiation. Upon effector differentiation, reduced expression of BACH2 and its phosphorylation enabled unrestrained induction of TCR-driven effector programs.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/physiology , Transcription Factor AP-1/metabolism , Vaccinia virus/immunology , Vaccinia/immunology , Adaptive Immunity , Animals , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cells, Cultured , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Protein p65(gag-jun) , Signal Transduction/genetics , Transcription Factor AP-1/genetics
6.
Immunity ; 51(3): 548-560.e4, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471106

ABSTRACT

Immunotherapy can reinvigorate dormant responses to cancer, but response rates remain low. Oncolytic viruses, which replicate in cancer cells, induce tumor lysis and immune priming, but their immune consequences are unclear. We profiled the infiltrate of aggressive melanomas induced by oncolytic Vaccinia virus using RNA sequencing and found substantial remodeling of the tumor microenvironment, dominated by effector T cell influx. However, responses to oncolytic viruses were incomplete due to metabolic insufficiencies induced by the tumor microenvironment. We identified the adipokine leptin as a potent metabolic reprogramming agent that supported antitumor responses. Leptin metabolically reprogrammed T cells in vitro, and melanoma cells expressing leptin were immunologically controlled in mice. Engineering oncolytic viruses to express leptin in tumor cells induced complete responses in tumor-bearing mice and supported memory development in the tumor infiltrate. Thus, leptin can provide metabolic support to tumor immunity, and oncolytic viruses represent a platform to deliver metabolic therapy.


Subject(s)
Leptin/immunology , Melanoma/immunology , Oncolytic Viruses/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Immunotherapy/methods , Male , Mice , Mice, Inbred C57BL , Tumor Microenvironment/immunology , Vaccinia virus/immunology
7.
Immunity ; 50(5): 1249-1261.e5, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30952606

ABSTRACT

Regulated activation of the cytokine TGF-ß by integrins αvß6 and αvß8 expressed on keratinocytes is required for residence of epidermal-resident memory T cells, but whether skin-derived signals also affect recirculating memory cells in the skin remains unclear. Here, we show that after resolution of skin vaccinia virus (VV) infection, antigen-specific circulating memory CD8+ T cells migrated into skin. In mice lacking αvß6 and αvß8 integrins (Itgb6-/-Itgb8fl/fl-K14-cre), the absence of epidermal-activated TGF-ß resulted in a gradual loss of E- or P-selectin-binding central and peripheral memory populations, which were rescued when skin entry was inhibited. Skin recirculating memory cells were required for optimal host defense against skin VV infection. These data demonstrate that skin migration can persist after resolution of local skin infection and that the cytokine environment within this nonlymphoid tissue shapes the differentiation state and persistence of the central and peripheral memory-T-cell pool.


Subject(s)
Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Integrins/metabolism , Keratinocytes/metabolism , Transforming Growth Factor beta/metabolism , Vaccinia virus/immunology , Animals , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/enzymology , Cell Differentiation/immunology , Cytokines/immunology , Enzyme Activation , Female , Integrins/genetics , Male , Mice , Mice, Inbred C57BL , Skin/cytology , Skin/immunology
8.
Mol Cell ; 74(1): 19-31.e7, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30878284

ABSTRACT

Viral infection triggers host defenses through pattern-recognition receptor-mediated cytokine production, inflammasome activation, and apoptosis of the infected cells. Inflammasome-activated caspases are known to cleave cyclic GMP-AMP synthase (cGAS). Here, we found that apoptotic caspases are critically involved in regulating both DNA and RNA virus-triggered host defenses, in which activated caspase-3 cleaved cGAS, MAVS, and IRF3 to prevent cytokine overproduction. Caspase-3 was exclusively required in human cells, whereas caspase-7 was involved only in murine cells to inactivate cGAS, reflecting distinct regulatory mechanisms in different species. Caspase-mediated cGAS cleavage was enhanced in the presence of dsDNA. Alternative MAVS cleavage sites were used to ensure the inactivation of this critical protein. Elevated type I IFNs were detected in caspase-3-deficient cells without any infection. Casp3-/- mice consistently showed increased resistance to viral infection and experimental autoimmune encephalomyelitis. Our results demonstrate that apoptotic caspases control innate immunity and maintain immune homeostasis against viral infection.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Caspases/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Nucleotidyltransferases/metabolism , Virus Diseases/enzymology , Adaptor Proteins, Signal Transducing/genetics , Animals , Caspase 2/genetics , Caspase 2/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Caspases/genetics , Female , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Male , Mice, Inbred C57BL , Nucleotidyltransferases/genetics , Sendai virus/immunology , Sendai virus/pathogenicity , Signal Transduction , THP-1 Cells , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/virology
9.
Nat Immunol ; 15(6): 538-45, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24777530

ABSTRACT

Double-stranded DNA (dsDNA) in the cytoplasm triggers the production of interleukin 1ß (IL-1ß) as an antiviral host response, and deregulation of the pathways involved can promote inflammatory disease. Here we report a direct cytosolic interaction between the DNA-damage sensor Rad50 and the innate immune system adaptor CARD9. Transfection of dendritic cells with dsDNA or infection of dendritic cells with a DNA virus induced the formation of dsDNA-Rad50-CARD9 signaling complexes for activation of the transcription factor NF-κB and the generation of pro-IL-1ß. Primary cells conditionally deficient in Rad50 or lacking CARD9 consequently exhibited defective DNA-induced production of IL-1ß, and Card9(-/-) mice had impaired inflammatory responses after infection with a DNA virus in vivo. Our results define a cytosolic DNA-recognition pathway for inflammation and a physical and functional connection between a conserved DNA-damage sensor and the innate immune response to pathogens.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , DNA Repair Enzymes/immunology , DNA, Viral/immunology , DNA-Binding Proteins/immunology , Interleukin-1beta/biosynthesis , Vaccinia virus/immunology , Acid Anhydride Hydrolases , Adaptor Proteins, Signal Transducing/immunology , Animals , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/genetics , Cell Line , Cytosol/immunology , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Enzyme Activation , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , NF-kappa B/immunology , Signal Transduction , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 9/biosynthesis , Vaccinia virus/genetics
10.
Nat Immunol ; 15(5): 457-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24705297

ABSTRACT

SGK1 is an AGC kinase that regulates the expression of membrane sodium channels in renal tubular cells in a manner dependent on the metabolic checkpoint kinase complex mTORC2. We hypothesized that SGK1 might represent an additional mTORC2-dependent regulator of the differentiation and function of T cells. Here we found that after activation by mTORC2, SGK1 promoted T helper type 2 (TH2) differentiation by negatively regulating degradation of the transcription factor JunB mediated by the E3 ligase Nedd4-2. Simultaneously, SGK1 repressed the production of interferon-γ (IFN-γ) by controlling expression of the long isoform of the transcription factor TCF-1. Consistent with those findings, mice with selective deletion of SGK1 in T cells were resistant to experimentally induced asthma, generated substantial IFN-γ in response to viral infection and more readily rejected tumors.


Subject(s)
Asthma/immunology , Immediate-Early Proteins/metabolism , Melanoma, Experimental/immunology , Multiprotein Complexes/immunology , Poxviridae Infections/immunology , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Vaccinia virus/immunology , Adaptive Immunity/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 1-alpha , Immediate-Early Proteins/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases , Protein Serine-Threonine Kinases/genetics , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Burden/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Eur J Immunol ; 54(10): e2451135, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39086059

ABSTRACT

Inflammasomes are essential for host defense, recognizing foreign or stress signals to trigger immune responses, including maturation of IL-1 family cytokines and pyroptosis. Here, NLRP1 is emerging as an important sensor of viral infection in barrier tissues. NLRP1 is activated by various stimuli, including viral double-stranded (ds) RNA, ribotoxic stress, and inhibition of dipeptidyl peptidases 8 and 9 (DPP8/9). However, certain viruses, most notably the vaccinia virus, have evolved strategies to subvert inflammasome activation or effector functions. Using the modified vaccinia virus Ankara (MVA) as a model, we investigated how the vaccinia virus inhibits inflammasome activation. We confirmed that the early gene F1L plays a critical role in inhibiting NLRP1 inflammasome activation. Interestingly, it blocks dsRNA and ribotoxic stress-dependent NLRP1 activation without affecting its DPP9-inhibition-mediated activation. Complementation and loss-of-function experiments demonstrated the sufficiency and necessity of F1L in blocking NLRP1 activation. Furthermore, we found that F1L-deficient, but not wild-type MVA, induced ZAKα activation. Indeed, an F1L-deficient virus was found to disrupt protein translation more prominently than an unmodified virus, suggesting that F1L acts in part upstream of ZAKα. These findings underscore the inhibitory role of F1L on NLRP1 inflammasome activation and provide insight into viral evasion of host defenses and the intricate mechanisms of inflammasome activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Inflammasomes , NLR Proteins , Vaccinia virus , Vaccinia virus/immunology , Inflammasomes/immunology , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , NLR Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/immunology , HEK293 Cells , Viral Proteins/immunology , Viral Proteins/metabolism , Viral Proteins/genetics , Vaccinia/immunology , Animals , Mice , Immune Evasion
12.
J Virol ; 98(7): e0052124, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38874361

ABSTRACT

The reoccurrence of successive waves of SARS-CoV-2 variants suggests the exploration of more vaccine alternatives is imperative. Modified vaccinia virus Ankara (MVA) is a virus vector exhibiting excellent safety as well as efficacy for vaccine development. Here, a series of recombinant MVAs (rMVAs) expressing monomerized or trimerized S proteins from different SARS-CoV-2 variants are engineered. Trimerized S expressed from rMVAs is found predominantly as trimers on the surface of infected cells. Remarkably, immunization of mice with rMVAs demonstrates that S expressed in trimer elicits higher levels of binding IgG and IgA, as well as neutralizing antibodies for matched and mismatched S proteins than S in the monomer. In addition, trimerized S expressed by rMVA induces enhanced cytotoxic T-cell responses than S in the monomer. Importantly, the rMVA vaccines expressing trimerized S exhibit superior protection against a lethal SARS-CoV-2 challenge as the immunized animals all survive without displaying any pathological conditions. This study suggests that opting for trimerized S may represent a more effective approach and highlights that the MVA platform serves as an ideal foundation to continuously advance SARS-CoV-2 vaccine development. IMPORTANCE: MVA is a promising vaccine vector and has been approved as a vaccine for smallpox and mpox. Our analyses suggested that recombinant MVA expressing S in trimer (rMVA-ST) elicited robust cellular and humoral immunity and was more effective than MVA-S-monomer. Importantly, the rMVA-ST vaccine was able to stimulate decent cross-reactive neutralization against pseudoviruses packaged using S from different sublineages, including Wuhan, Delta, and Omicron. Remarkably, mice immunized with rMVA-ST were completely protected from a lethal challenge of SARS-CoV-2 without displaying any pathological conditions. Our results demonstrated that an MVA vectored vaccine expressing trimerized S is a promising vaccine candidate for SARS-CoV-2 and the strategy might be adapted for future vaccine development for coronaviruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccinia virus , Animals , Vaccinia virus/genetics , Vaccinia virus/immunology , Mice , Antibodies, Viral/immunology , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Female , Humans , Mice, Inbred BALB C , Protein Multimerization , Immunoglobulin G/immunology , T-Lymphocytes, Cytotoxic/immunology , Immunoglobulin A/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/genetics , Genetic Vectors
13.
Immunity ; 44(2): 207-8, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26885849

ABSTRACT

Cytotoxic CD8+ T lymphocytes (CTLs) have long been believed to be extremely efficient killers. Forster and colleagues (Halle et al., 2016) used in vivo imaging to tell a different story, in which each CTL killed only 2-16 targets a day, and several CTLs per target were needed to get the job done.


Subject(s)
Herpesviridae Infections/immunology , Muromegalovirus/immunology , Perforin/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , Humans
14.
Immunity ; 45(4): 847-860, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27692611

ABSTRACT

Despite the crucial role of tissue-resident memory T (Trm) cells in protective immunity, their priming remains poorly understood. Here, we have shown differential priming requirements for Trm versus circulating memory CD8+ T cells. In vaccinia cutaneous-infected mice, DNGR-1-mediated crosspresentation was required for optimal Trm cell priming but not for their skin differentiation or for circulating memory T cell generation. DNGR-1+ dendritic cells (DCs) promoted T-bet transcription-factor induction and retention of CD8+ T cells in the lymph nodes (LNs). Inhibition of LN egress enhanced Trm cell generation, whereas genetic or antibody blockade of DNGR-1 or specific signals provided during priming by DNGR-1+ DCs, such as interleukin-12 (IL-12), IL-15, or CD24, impaired Trm cell priming. DNGR-1 also regulated Trm cell generation during influenza infection. Moreover, protective immunity depended on optimal Trm cell induction by DNGR-1+ DCs. Our results reveal specific priming requirements for CD8+ Trm cells during viral infection and vaccination.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Virus Diseases/immunology , Animals , CD24 Antigen/immunology , Cross-Priming/immunology , Interleukin-12/immunology , Interleukin-15/immunology , Lymph Nodes/immunology , Lymph Nodes/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Skin/immunology , Skin/virology , Vaccinia/immunology , Vaccinia/virology , Vaccinia virus/immunology , Virus Diseases/virology
15.
Immunity ; 44(2): 233-45, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26872694

ABSTRACT

According to in vitro assays, T cells are thought to kill rapidly and efficiently, but the efficacy and dynamics of cytotoxic T lymphocyte (CTL)-mediated killing of virus-infected cells in vivo remains elusive. We used two-photon microscopy to quantify CTL-mediated killing in mice infected with herpesviruses or poxviruses. On average, one CTL killed 2-16 virus-infected cells per day as determined by real-time imaging and by mathematical modeling. In contrast, upon virus-induced MHC class I downmodulation, CTLs failed to destroy their targets. During killing, CTLs remained migratory and formed motile kinapses rather than static synapses with targets. Viruses encoding the calcium sensor GCaMP6s revealed strong heterogeneity in individual CTL functional capacity. Furthermore, the probability of death of infected cells increased for those contacted by more than two CTLs, indicative of CTL cooperation. Thus, direct visualization of CTLs during killing of virus-infected cells reveals crucial parameters of CD8(+) T cell immunity.


Subject(s)
Herpesviridae Infections/immunology , Muromegalovirus/immunology , Perforin/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , Calcium Signaling , Cell Communication , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Immune Evasion , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence, Multiphoton , Perforin/genetics , T-Lymphocyte Subsets/virology , T-Lymphocytes, Cytotoxic/virology
16.
Nature ; 566(7743): 259-263, 2019 02.
Article in English | MEDLINE | ID: mdl-30728498

ABSTRACT

Cytosolic DNA triggers innate immune responses through the activation of cyclic GMP-AMP synthase (cGAS) and production of the cyclic dinucleotide second messenger 2',3'-cyclic GMP-AMP (cGAMP)1-4. 2',3'-cGAMP is a potent inducer of immune signalling; however, no intracellular nucleases are known to cleave 2',3'-cGAMP and prevent the activation of the receptor stimulator of interferon genes (STING)5-7. Here we develop a biochemical screen to analyse 24 mammalian viruses, and identify poxvirus immune nucleases (poxins) as a family of 2',3'-cGAMP-degrading enzymes. Poxins cleave 2',3'-cGAMP to restrict STING-dependent signalling and deletion of the poxin gene (B2R) attenuates vaccinia virus replication in vivo. Crystal structures of vaccinia virus poxin in pre- and post-reactive states define the mechanism of selective 2',3'-cGAMP degradation through metal-independent cleavage of the 3'-5' bond, converting 2',3'-cGAMP into linear Gp[2'-5']Ap[3']. Poxins are conserved in mammalian poxviruses. In addition, we identify functional poxin homologues in the genomes of moths and butterflies and the baculoviruses that infect these insects. Baculovirus and insect host poxin homologues retain selective 2',3'-cGAMP degradation activity, suggesting an ancient role for poxins in cGAS-STING regulation. Our results define poxins as a family of 2',3'-cGAMP-specific nucleases and demonstrate a mechanism for how viruses evade innate immunity.


Subject(s)
Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Membrane Proteins/metabolism , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction/immunology , Vaccinia virus/enzymology , Animals , Baculoviridae/enzymology , Butterflies/enzymology , Cell Line , Conserved Sequence , Crystallography, X-Ray , DNA, Viral/immunology , Female , Genes, Viral/genetics , Humans , Immune Evasion , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Models, Molecular , Moths/enzymology , Second Messenger Systems , Vaccinia virus/genetics , Vaccinia virus/growth & development , Vaccinia virus/immunology , Virus Replication/genetics
17.
Mol Ther ; 32(7): 2406-2422, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38734899

ABSTRACT

Vaccinia viruses (VACVs) are versatile therapeutic agents and different features of various VACV strains allow for a broad range of therapeutic applications. Modified VACV Ankara (MVA) is a particularly altered VACV strain that is highly immunogenic, incapable of replicating in mammalian hosts, and broadly used as a safe vector for vaccination. Alternatively, Western Reserve (WR) or Copenhagen (Cop) are VACV strains that efficiently replicate in cancer cells and, therefore, are used to develop oncolytic viruses. However, the immune evasion capacity of WR or Cop hinders their ability to elicit antitumor immune responses, which is crucial for efficacy in the clinic. Here, we describe a new VACV strain named Immune-Oncolytic VACV Ankara (IOVA), which combines efficient replication in cancer cells with induction of immunogenic tumor cell death (ICD). IOVA was engineered from an MVA ancestor and shows superior cytotoxicity in tumor cells. In addition, the IOVA genome incorporates mutations that lead to massive fusogenesis of tumor cells, which contributes to improved antitumor effects. In syngeneic mouse tumor models, the induction of ICD results in robust antitumor immunity directed against tumor neo-epitopes and eradication of large established tumors. These data present IOVA as an improved immunotherapeutic oncolytic vector.


Subject(s)
Immunogenic Cell Death , Oncolytic Virotherapy , Oncolytic Viruses , Vaccinia virus , Vaccinia virus/genetics , Vaccinia virus/immunology , Animals , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Mice , Humans , Oncolytic Virotherapy/methods , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/immunology , Virus Replication , Genetic Vectors/genetics
18.
Mol Ther ; 32(6): 1779-1789, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38659224

ABSTRACT

Since the outbreak of monkeypox (mpox) in 2022, widespread concern has been placed on imposing an urgent demand for specific vaccines that offer safer and more effective protection. Using an efficient and scalable circular RNA (circRNA) platform, we constructed four circRNA vaccines that could induce robust neutralizing antibodies as well as T cell responses by expressing different surface proteins of mpox virus (MPXV), resulting in potent protection against vaccinia virus (VACV) in mice. Strikingly, the combination of the four circular RNA vaccines demonstrated the best protection against VACV challenge among all the tested vaccines. Our study provides a favorable approach for developing MPXV-specific vaccines by using a circular mRNA platform and opens up novel avenues for future vaccine research.


Subject(s)
Antibodies, Neutralizing , Monkeypox virus , RNA, Circular , Vaccinia virus , Animals , Mice , Vaccinia virus/genetics , Vaccinia virus/immunology , RNA, Circular/genetics , Antibodies, Neutralizing/immunology , Monkeypox virus/immunology , Monkeypox virus/genetics , Antibodies, Viral/immunology , Vaccinia/prevention & control , Vaccinia/immunology , Mpox (monkeypox)/prevention & control , Mpox (monkeypox)/immunology , Viral Vaccines/immunology , Viral Vaccines/genetics , Humans , Disease Models, Animal , Female , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Cell Mol Life Sci ; 81(1): 400, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39264480

ABSTRACT

Dendritic cells (DCs) play a crucial role in orchestrating immune responses, particularly in promoting IFNγ-producing-CD8 cytotoxic T lymphocytes (CTLs) and IFNγ-producing-CD4 T helper 1 (Th1) cells, which are essential for defending against viral infections. Additionally, the nuclear envelope protein lamin A/C has been implicated in T cell immunity. Nevertheless, the intricate interplay between innate and adaptive immunity in response to viral infections, particularly the role of lamin A/C in DC functions within this context, remains poorly understood. In this study, we demonstrate that mice lacking lamin A/C in myeloid LysM promoter-expressing cells exhibit a reduced capacity to induce Th1 and CD8 CTL responses, leading to impaired clearance of acute primary Vaccinia virus (VACV) infection. Remarkably, in vitro-generated granulocyte macrophage colony-stimulating factor bone marrow-derived DCs (GM-CSF BMDCs) show high levels of lamin A/C. Lamin A/C absence on GM-CSF BMDCs does not affect the expression of costimulatory molecules on the cell membrane but it reduces the cellular ability to form immunological synapses with naïve CD4 T cells. Lamin A/C deletion induces alterations in NFκB nuclear localization, thereby influencing NF-κB-dependent transcription. Furthermore, lamin A/C ablation modifies the gene accessibility of BMDCs, predisposing these cells to mount a less effective antiviral response upon TLR stimulation. This study highlights the critical role of DCs in interacting with CD4 T cells during antiviral responses and proposes some mechanisms through which lamin A/C may modulate DC function via gene accessibility and transcriptional regulation.


Subject(s)
Dendritic Cells , Lamin Type A , Mice, Inbred C57BL , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Lamin Type A/metabolism , Lamin Type A/genetics , Mice , NF-kappa B/metabolism , Vaccinia virus/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Mice, Knockout , Vaccinia/immunology , Th1 Cells/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunological Synapses/metabolism , Immunological Synapses/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
20.
Proc Natl Acad Sci U S A ; 119(24): e2202069119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35679343

ABSTRACT

Current vaccines have greatly diminished the severity of the COVID-19 pandemic, even though they do not entirely prevent infection and transmission, likely due to insufficient immunity in the upper respiratory tract. Here, we compare intramuscular and intranasal administration of a live, replication-deficient modified vaccinia virus Ankara (MVA)-based Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) spike (S) vaccine to raise protective immune responses in the K18-hACE2 mouse model. Using a recombinant MVA expressing firefly luciferase for tracking, live imaging revealed luminescence of the respiratory tract of mice within 6 h and persisting for 3 d following intranasal inoculation, whereas luminescence remained at the site of intramuscular vaccination. Intramuscular vaccination induced S-binding-Immunoglobulin G (IgG) and neutralizing antibodies in the lungs, whereas intranasal vaccination also induced Immunoglobulin A (IgA) and higher levels of antigen-specific CD3+CD8+IFN-γ+ T cells. Similarly, IgG and neutralizing antibodies were present in the blood of mice immunized intranasally and intramuscularly, but IgA was detected only after intranasal inoculation. Intranasal boosting increased IgA after intranasal or intramuscular priming. While intramuscular vaccination prevented morbidity and cleared SARS-CoV-2 from the respiratory tract within several days after challenge, intranasal vaccination was more effective as neither infectious virus nor viral messenger (m)RNAs were detected in the nasal turbinates or lungs as early as 2 d after challenge, indicating prevention or rapid elimination of SARS-CoV-2 infection. Additionally, we determined that neutralizing antibody persisted for more than 6 mo and that serum induced to the Wuhan S protein neutralized pseudoviruses expressing the S proteins of variants, although with less potency, particularly for Beta and Omicron.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunoglobulin A , Respiratory System , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccinia virus , Administration, Intranasal , Angiotensin-Converting Enzyme 2/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/prevention & control , COVID-19/transmission , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice , Mice, Transgenic , Respiratory System/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccination/methods , Vaccinia virus/genetics , Vaccinia virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL