Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.049
Filter
Add more filters

Publication year range
1.
Cell ; 187(6): 1363-1373.e12, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38366591

ABSTRACT

In response to the 2022 outbreak of mpox driven by unprecedented human-to-human monkeypox virus (MPXV) transmission, we designed BNT166, aiming to create a highly immunogenic, safe, accessible, and scalable next-generation vaccine against MPXV and related orthopoxviruses. To address the multiple viral forms and increase the breadth of immune response, two candidate multivalent mRNA vaccines were evaluated pre-clinically: a quadrivalent vaccine (BNT166a; encoding the MPXV antigens A35, B6, M1, H3) and a trivalent vaccine (BNT166c; without H3). Both candidates induced robust T cell responses and IgG antibodies in mice, including neutralizing antibodies to both MPXV and vaccinia virus. In challenge studies, BNT166a and BNT166c provided complete protection from vaccinia, clade I, and clade IIb MPXV. Furthermore, immunization with BNT166a was 100% effective at preventing death and at suppressing lesions in a lethal clade I MPXV challenge in cynomolgus macaques. These findings support the clinical evaluation of BNT166, now underway (NCT05988203).


Subject(s)
Monkeypox virus , Mpox (monkeypox) , Smallpox Vaccine , Animals , Humans , Mice , Macaca fascicularis , Monkeypox virus/genetics , Mpox (monkeypox)/immunology , Mpox (monkeypox)/prevention & control , Vaccines, Combined , Vaccinia virus/genetics
2.
Nat Immunol ; 25(2): 307-315, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38182667

ABSTRACT

The global outbreak of the mpox virus (MPXV) in 2022 highlights the urgent need for safer and more accessible new-generation vaccines. Here, we used a structure-guided multi-antigen fusion strategy to design a 'two-in-one' immunogen based on the single-chain dimeric MPXV extracellular enveloped virus antigen A35 bivalently fused with the intracellular mature virus antigen M1, called DAM. DAM preserved the natural epitope configuration of both components and showed stronger A35-specific and M1-specific antibody responses and in vivo protective efficacy against vaccinia virus (VACV) compared to co-immunization strategies. The MPXV-specific neutralizing antibodies elicited by DAM were 28 times higher than those induced by live VACV vaccine. Aluminum-adjuvanted DAM vaccines protected mice from a lethal VACV challenge with a safety profile, and pilot-scale production confirmed the high yield and purity of DAM. Thus, our study provides innovative insights and an immunogen candidate for the development of alternative vaccines against MPXV and other orthopoxviruses.


Subject(s)
Monkeypox virus , Vaccines , Animals , Mice , Viral Envelope Proteins , Antibodies, Viral , Vaccinia virus , Antigens, Viral , Immunity
3.
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
4.
Cell ; 179(7): 1537-1550.e19, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31835032

ABSTRACT

Poxviruses encode a multisubunit DNA-dependent RNA polymerase (vRNAP) that carries out viral gene expression in the host cytoplasm. We report cryo-EM structures of core and complete vRNAP enzymes from Vaccinia virus at 2.8 Å resolution. The vRNAP core enzyme resembles eukaryotic RNA polymerase II (Pol II) but also reveals many virus-specific features, including the transcription factor Rap94. The complete enzyme additionally contains the transcription factor VETF, the mRNA processing factors VTF/CE and NPH-I, the viral core protein E11, and host tRNAGln. This complex can carry out the entire early transcription cycle. The structures show that Rap94 partially resembles the Pol II initiation factor TFIIB, that the vRNAP subunit Rpo30 resembles the Pol II elongation factor TFIIS, and that NPH-I resembles chromatin remodeling enzymes. Together with the accompanying paper (Hillen et al., 2019), these results provide the basis for unraveling the mechanisms of poxvirus transcription and RNA processing.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Transcription Factors/chemistry , Vaccinia virus/ultrastructure , Viral Proteins/chemistry , Cryoelectron Microscopy , Multienzyme Complexes/chemistry , Multienzyme Complexes/ultrastructure , Single Molecule Imaging , Vaccinia virus/genetics , Vaccinia virus/metabolism
5.
Cell ; 179(7): 1525-1536.e12, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31835031

ABSTRACT

Poxviruses use virus-encoded multisubunit RNA polymerases (vRNAPs) and RNA-processing factors to generate m7G-capped mRNAs in the host cytoplasm. In the accompanying paper, we report structures of core and complete vRNAP complexes of the prototypic Vaccinia poxvirus (Grimm et al., 2019; in this issue of Cell). Here, we present the cryo-electron microscopy (cryo-EM) structures of Vaccinia vRNAP in the form of a transcribing elongation complex and in the form of a co-transcriptional capping complex that contains the viral capping enzyme (CE). The trifunctional CE forms two mobile modules that bind the polymerase surface around the RNA exit tunnel. RNA extends from the vRNAP active site through this tunnel and into the active site of the CE triphosphatase. Structural comparisons suggest that growing RNA triggers large-scale rearrangements on the surface of the transcription machinery during the transition from transcription initiation to RNA capping and elongation. Our structures unravel the basis for synthesis and co-transcriptional modification of poxvirus RNA.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Methyltransferases/chemistry , Multienzyme Complexes/chemistry , Nucleotidyltransferases/chemistry , Phosphoric Monoester Hydrolases/chemistry , Vaccinia virus/ultrastructure , Viral Proteins/chemistry , Cryoelectron Microscopy , Multienzyme Complexes/ultrastructure , RNA, Messenger/chemistry , Single Molecule Imaging , Transcription, Genetic , Vaccinia virus/genetics , Vaccinia virus/metabolism
6.
Cell ; 171(2): 398-413.e21, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28942919

ABSTRACT

A fundamental challenge in immunology is to decipher the principles governing immune responses at the whole-organism scale. Here, using a comparative infection model, we observe immune signal propagation within and between organs to obtain a dynamic map of immune processes at the organism level. We uncover two inter-organ mechanisms of protective immunity mediated by soluble and cellular factors. First, analyzing ligand-receptor connectivity across tissues reveals that type I IFNs trigger a whole-body antiviral state, protecting the host within hours after skin vaccination. Second, combining parabiosis, single-cell analyses, and gene knockouts, we uncover a multi-organ web of tissue-resident memory T cells that functionally adapt to their environment to stop viral spread across the organism. These results have implications for manipulating tissue-resident memory T cells through vaccination and open up new lines of inquiry for the analysis of immune responses at the organism level.


Subject(s)
Immunologic Memory , Interferon Type I/immunology , Vaccinia virus/physiology , Vaccinia/immunology , Vaccinia/prevention & control , Viral Vaccines/immunology , Administration, Cutaneous , Animals , Female , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Organ Specificity , Specific Pathogen-Free Organisms , T-Lymphocytes/immunology , Viral Vaccines/administration & dosage
8.
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
9.
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
10.
Immunity ; 54(2): 276-290.e5, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33434494

ABSTRACT

The oropharyngeal mucosa serves as a perpetual pathogen entry point and a critical site for viral replication and spread. Here, we demonstrate that type 1 innate lymphoid cells (ILC1s) were the major immune force providing early protection during acute oral mucosal viral infection. Using intravital microscopy, we show that ILC1s populated and patrolled the uninfected labial mucosa. ILC1s produced interferon-γ (IFN-γ) in the absence of infection, leading to the upregulation of key antiviral genes, which were downregulated in uninfected animals upon genetic ablation of ILC1s or antibody-based neutralization of IFN-γ. Thus, tonic IFN-γ production generates increased oral mucosal viral resistance even before infection. Our results demonstrate barrier-tissue protection through tissue surveillance in the absence of rearranged-antigen receptors and the induction of an antiviral state during homeostasis. This aspect of ILC1 biology raises the possibility that these cells do not share true functional redundancy with other tissue-resident lymphocytes.


Subject(s)
Interferon-gamma/metabolism , Lymphocytes/immunology , Oropharynx/immunology , Respiratory Mucosa/immunology , Vaccinia virus/physiology , Vaccinia/immunology , Animals , Cells, Cultured , Disease Resistance , Humans , Immunity, Innate , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/genetics , Th1 Cells/immunology
11.
Immunity ; 54(2): 247-258.e7, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33444549

ABSTRACT

The vaccine strain against smallpox, vaccinia virus (VACV), is highly immunogenic yet causes relatively benign disease. These attributes are believed to be caused by gene loss in VACV. Using a targeted small interfering RNA (siRNA) screen, we identified a viral inhibitor found in cowpox virus (CPXV) and other orthopoxviruses that bound to the host SKP1-Cullin1-F-box (SCF) machinery and the essential necroptosis kinase receptor interacting protein kinase 3 (RIPK3). This "viral inducer of RIPK3 degradation" (vIRD) triggered ubiquitination and proteasome-mediated degradation of RIPK3 and inhibited necroptosis. In contrast to orthopoxviruses, the distantly related leporipoxvirus myxoma virus (MYXV), which infects RIPK3-deficient hosts, lacks a functional vIRD. Introduction of vIRD into VACV, which encodes a truncated and defective vIRD, enhanced viral replication in mice. Deletion of vIRD reduced CPXV-induced inflammation, viral replication, and mortality, which were reversed in RIPK3- and MLKL-deficient mice. Hence, vIRD-RIPK3 drives pathogen-host evolution and regulates virus-induced inflammation and pathogenesis.


Subject(s)
Cowpox virus/physiology , Cowpox/immunology , RNA, Small Interfering/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Vaccinia virus/metabolism , Viral Proteins/metabolism , Animals , Evolution, Molecular , HEK293 Cells , Host-Pathogen Interactions , Humans , Inflammation , Mice , Mice, Knockout , Necroptosis/genetics , Orthopoxvirus , Phylogeny , Protein Kinases/genetics , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequence Analysis, RNA , Viral Proteins/genetics , Virus Replication
12.
Immunity ; 54(5): 962-975.e8, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33857420

ABSTRACT

Activation of the cyclic guanosine monophosphate (GMP)-AMP (cGAMP) sensor STING requires its translocation from the endoplasmic reticulum to the Golgi apparatus and subsequent polymerization. Using a genome-wide CRISPR-Cas9 screen to define factors critical for STING activation in cells, we identified proteins critical for biosynthesis of sulfated glycosaminoglycans (sGAGs) in the Golgi apparatus. Binding of sGAGs promoted STING polymerization through luminal, positively charged, polar residues. These residues are evolutionarily conserved, and selective mutation of specific residues inhibited STING activation. Purified or chemically synthesized sGAGs induced STING polymerization and activation of the kinase TBK1. The chain length and O-linked sulfation of sGAGs directly affected the level of STING polymerization and, therefore, its activation. Reducing the expression of Slc35b2 to inhibit GAG sulfation in mice impaired responses to vaccinia virus infection. Thus, sGAGs in the Golgi apparatus are necessary and sufficient to drive STING polymerization, providing a mechanistic understanding of the requirement for endoplasmic reticulum (ER)-to-Golgi apparatus translocation for STING activation.


Subject(s)
Glycosaminoglycans/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Nucleotides, Cyclic/metabolism , Animals , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Mice , Polymerization , Signal Transduction/physiology , Sulfate Transporters/metabolism , Vaccinia/metabolism , Vaccinia virus/pathogenicity
13.
Immunity ; 54(3): 542-556.e9, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33631118

ABSTRACT

A combination of vaccination approaches will likely be necessary to fully control the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Here, we show that modified vaccinia Ankara (MVA) vectors expressing membrane-anchored pre-fusion stabilized spike (MVA/S) but not secreted S1 induced strong neutralizing antibody responses against SARS-CoV-2 in mice. In macaques, the MVA/S vaccination induced strong neutralizing antibodies and CD8+ T cell responses, and conferred protection from SARS-CoV-2 infection and virus replication in the lungs as early as day 2 following intranasal and intratracheal challenge. Single-cell RNA sequencing analysis of lung cells on day 4 after infection revealed that MVA/S vaccination also protected macaques from infection-induced inflammation and B cell abnormalities and lowered induction of interferon-stimulated genes. These results demonstrate that MVA/S vaccination induces neutralizing antibodies and CD8+ T cells in the blood and lungs and is a potential vaccine candidate for SARS-CoV-2.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/prevention & control , Genetic Vectors/genetics , SARS-CoV-2/immunology , Vaccines, DNA/immunology , Vaccinia virus/genetics , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , COVID-19 Vaccines/genetics , Disease Models, Animal , Gene Expression , Gene Order , Immunophenotyping , Lung/immunology , Lung/pathology , Lung/virology , Macaca , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Vaccination/methods , Vaccines, DNA/genetics
14.
Cell ; 162(6): 1210-2, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26359981

ABSTRACT

Eickhoff et al. and Hor et al. use time-lapse intravital microscopy to show an unexpected choreography of CD4+ and CD8+ T cells "dancing" between different dendritic cell sub-populations during priming of cytotoxic immune responses to viruses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Dendritic Cells/immunology , Vaccinia virus/physiology , Vaccinia/immunology , Animals
15.
Cell ; 162(6): 1322-37, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26296422

ABSTRACT

Host defense against viruses and intracellular parasites depends on effector CD8(+) T cells, whose optimal clonal expansion, differentiation, and memory properties require signals from CD4(+) T cells. Here, we addressed the role of dendritic cell (DC) subsets in initial activation of the two T cell types and their co-operation. Surprisingly, initial priming of CD4(+) and CD8(+) T cells was spatially segregated within the lymph node and occurred on different DCs with temporally distinct patterns of antigen presentation via MHCI versus MHCII molecules. DCs that co-present antigen via both MHC molecules were detected at a later stage; these XCR1(+) DCs are the critical platform involved in CD4(+) T cell augmentation of CD8(+) T cell responses. These findings delineate the complex choreography of cellular interactions underlying effective cell-mediated anti-viral responses, with implications for basic DC subset biology, as well as for translational application to the development of vaccines that evoke optimal T cell immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Dendritic Cells/immunology , Vaccinia virus/physiology , Vaccinia/immunology , Animals , Antigen Presentation , Antigens, Viral/immunology , Dendritic Cells/cytology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Receptors, Chemokine/genetics , Spleen/cytology , Spleen/immunology
16.
Nat Immunol ; 17(4): 369-78, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26829768

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) senses cytosolic DNA during viral infection and catalyzes synthesis of the dinucleotide cGAMP, which activates the adaptor STING to initiate antiviral responses. Here we found that deficiency in the carboxypeptidase CCP5 or CCP6 led to susceptibility to DNA viruses. CCP5 and CCP6 were required for activation of the transcription factor IRF3 and interferons. Polyglutamylation of cGAS by the enzyme TTLL6 impeded its DNA-binding ability, whereas TTLL4-mediated monoglutamylation of cGAS blocked its synthase activity. Conversely, CCP6 removed the polyglutamylation of cGAS, whereas CCP5 hydrolyzed the monoglutamylation of cGAS, which together led to the activation of cGAS. Therefore, glutamylation and deglutamylation of cGAS tightly modulate immune responses to infection with DNA viruses.


Subject(s)
Carboxypeptidases/genetics , DNA Virus Infections/metabolism , DNA, Viral/immunology , Nucleotidyltransferases/metabolism , Peptide Synthases/metabolism , Animals , Cytosol , DNA Viruses/genetics , Fluorescent Antibody Technique , Herpes Simplex/metabolism , Immunoprecipitation , Interferon Regulatory Factor-3/immunology , Interferons/immunology , Mice , Mice, Knockout , Nucleotides, Cyclic/biosynthesis , Nucleotidyltransferases/immunology , Reverse Transcriptase Polymerase Chain Reaction , Simplexvirus/genetics , Vaccinia/metabolism , Vaccinia virus/genetics
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL