Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 742
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 31: 163-94, 2013.
Article in English | MEDLINE | ID: mdl-23298212

ABSTRACT

Natural killer (NK) cells are effector cells of the innate immune system and are important in the control of viral infections. Their relevance is reflected by the multiple mechanisms evolved by viruses to evade NK cell-mediated immune responses. Over recent years, our understanding of the interplay between NK cell immunity and viral pathogenesis has improved significantly. Here, we review the role of NK cells in the control of four important viral infections in humans: cytomegalovirus, influenza virus, HIV-1, and hepatitis C virus.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Virus Diseases/immunology , Virus Diseases/virology , Animals , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/prevention & control , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/prevention & control , Hepatitis C/immunology , Hepatitis C/pathology , Hepatitis C/prevention & control , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/prevention & control , Humans , Influenza, Human/immunology , Influenza, Human/pathology , Influenza, Human/prevention & control , Killer Cells, Natural/pathology , Virus Diseases/pathology
2.
Immunity ; 53(4): 864-877.e5, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32791036

ABSTRACT

The SARS-CoV-2 pandemic has resulted in millions of infections, yet the role of host immune responses in early COVID-19 pathogenesis remains unclear. By investigating 17 acute and 24 convalescent patients, we found that acute SARS-CoV-2 infection resulted in broad immune cell reduction including T, natural killer, monocyte, and dendritic cells (DCs). DCs were significantly reduced with functional impairment, and ratios of conventional DCs to plasmacytoid DCs were increased among acute severe patients. Besides lymphocytopenia, although neutralizing antibodies were rapidly and abundantly generated in patients, there were delayed receptor binding domain (RBD)- and nucleocapsid protein (NP)-specific T cell responses during the first 3 weeks after symptoms onset. Moreover, acute RBD- and NP-specific T cell responses included relatively more CD4 T cells than CD8 T cells. Our findings provided evidence that impaired DCs, together with timely inverted strong antibody but weak CD8 T cell responses, could contribute to acute COVID-19 pathogenesis and have implications for vaccine development.


Subject(s)
Betacoronavirus/pathogenicity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Dendritic Cells/immunology , Diabetes Mellitus/immunology , Hypertension/immunology , Pneumonia, Viral/immunology , Adult , Aged , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Betacoronavirus/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , COVID-19 , Convalescence , Coronavirus Infections/complications , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Dendritic Cells/pathology , Dendritic Cells/virology , Diabetes Complications , Diabetes Mellitus/diagnosis , Diabetes Mellitus/virology , Disease Progression , Female , Humans , Hypertension/complications , Hypertension/diagnosis , Hypertension/virology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Lymphocyte Activation , Lymphocyte Count , Male , Middle Aged , Monocytes/immunology , Monocytes/pathology , Monocytes/virology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , SARS-CoV-2 , Severity of Illness Index
3.
Immunity ; 45(1): 74-82, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27438766

ABSTRACT

Natural killer (NK) cells are important in host defense against pathogens, and they can subsequently differentiate into memory NK cells. The Ly49 and KIR gene families in rodents and humans encode both inhibitory and activating receptors for MHC class I. The physiological role of activating KIR or Ly49 receptors that recognize self-MHC class I during immune response to viral infections is unknown. Here, we address how the activating Ly49D receptor impacts the NK cell response to mouse cytomegalovirus (MCMV) infection by comparing the activation and differentiation of Ly49D-bearing NK cells in mice lacking or expressing H-2D(d), the cognate MHC class I ligand of Ly49D. After MCMV infection, Ly49D augmented IFN-γ production by MCMV-specific Ly49H(+) NK cells and preferentially promoted the generation of memory Ly49H(+) NK cells. Thus, activating receptors for self-MHC class I modulate the differentiation of MCMV-specific NK cells and are beneficial for host defense against MCMV infection.


Subject(s)
Herpesviridae Infections/immunology , Immunologic Memory , Killer Cells, Natural/physiology , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Autoantigens/genetics , Autoantigens/metabolism , Cell Differentiation , Cell Survival , Cells, Cultured , Histocompatibility Antigen H-2D/genetics , Histocompatibility Antigen H-2D/metabolism , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/virology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Rats
4.
J Virol ; 97(1): e0151922, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36511699

ABSTRACT

Natural killer (NK) cells are potent effector cells of the innate immune system possessing both cytotoxic and immunoregulatory capabilities, which contribute to their crucial role in controlling human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infections. However, despite significant evidence for NK cell modulation of HIV disease, their specific contribution to transmission and control of acute infection remains less clear. To elucidate the contribution of NK cells during acute SIV infection, we performed an acute necropsy study, where rhesus macaques (RM) were subjected to preinfection depletion of systemic NK cells using established methods of IL-15 neutralization, followed by subsequent challenge with barcoded SIVmac239X. Our study showed that depletion was highly effective, resulting in near total ablation of all NK cell subsets in blood, liver, oral, and rectal mucosae, and lymph nodes (LN) that persisted through the duration of the study. Meanwhile, frequencies and phenotypes of T cells remained virtually unchanged, indicating that our method of NK cell depletion had minimal off-target effects. Importantly, NK cell-depleted RM demonstrated an early and sustained 1 to 2 log increase in viremia over controls, but sequence analysis suggested no difference in the number of independent transmission events. Acute bulk, central memory (CM), and CCR5+ CD4+ T cell depletion was similar between experimental and control groups, while CD8+ T cell activation was higher in NK cell-depleted RM as measured by Ki67 and PD-1 expression. Using 27-plex Luminex analyses, we also found modestly increased inflammatory cytokines in NK cell-depleted RM compared to control animals. In the effort to determine the impact of NK cells on HIV/SIV transmission and acute viremia, future studies will be necessary to better harness these cells for future viral therapies. Collectively, these data suggest NK cells are important modulators of lentivirus dissemination and disease but may not have the capacity to independently eliminate individual transmission events. IMPORTANCE Natural killer (NK) cells as major effector cells of the innate immune system can contribute significantly to human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) control. However, a specific role for NK cells in blocking lentivirus transmission remains incompletely clear. In this study, we depleted NK cells prior to challenge with a barcoded SIV. Importantly, our studied showed systemic NK cell depletion was associated with a significant increase in acute viremia, but did not impact the number of independent transmission events. Collectively, these data suggest NK cells are critical modulators of early lentivirus replication but may not regulate individual transmission events at mucosal portals of entry.


Subject(s)
Killer Cells, Natural , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Humans , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , HIV Infections , Killer Cells, Natural/virology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/transmission , Viral Load , Viremia , Virus Replication
5.
Immunity ; 43(2): 331-42, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26253785

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that possess traits of adaptive immunity, such as clonal expansion, contraction, and generation of long-lived "memory" cells, processes poorly understood at the molecular level. Here, we found that as proliferating NK cells accumulated dysfunctional mitochondria during viral infection, a protective mitophagy pathway was induced during the contraction phase to promote their survival in a reactive oxygen species (ROS)-dependent manner. Inhibition of mechanistic target of rapamycin (mTOR) or activation of AMP-activated protein kinase (AMPK) during the contraction-to-memory phase transition of the antiviral response increased autophagic activity and enhanced memory NK cell numbers through an Atg3-dependent mechanism. Furthermore, we demonstrated a temporally regulated role for mitophagy-inducing proteins BCL2/adenovirus E1B 19-kDa interacting protein 3 (BNIP3) and BNIP3-like (BNIP3L) in the generation of robust NK cell memory. Thus, our study reveals the functional importance of mitophagy during the dynamic response of these cytolytic innate lymphocytes.


Subject(s)
Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitophagy/genetics , Muromegalovirus/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy-Related Proteins , Cells, Cultured , Immunologic Memory/genetics , Killer Cells, Natural/virology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/virology , Mitochondrial Proteins/genetics , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
6.
Immunity ; 42(3): 431-42, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25786175

ABSTRACT

Long-lived "memory-like" NK cells have been identified in individuals infected by human cytomegalovirus (HCMV), but little is known about how the memory-like NK cell pool is formed. Here, we have shown that HCMV-infected individuals have several distinct subsets of memory-like NK cells that are often deficient for multiple transcription factors and signaling proteins, including tyrosine kinase SYK, for which the reduced expression was stable over time and correlated with epigenetic modification of the gene promoter. Deficient expression of these proteins was largely confined to the recently discovered FcRγ-deficient NK cells that display enhanced antibody-dependent functional activity. Importantly, FcRγ-deficient NK cells exhibited robust preferential expansion in response to virus-infected cells (both HCMV and influenza) in an antibody-dependent manner. These findings suggest that the memory-like NK cell pool is shaped and maintained by a mechanism that involves both epigenetic modification of gene expression and antibody-dependent expansion.


Subject(s)
Antibodies/immunology , Cytomegalovirus Infections/genetics , Epigenesis, Genetic/immunology , Immunologic Memory , Killer Cells, Natural/immunology , Cell Proliferation , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , DNA Methylation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Profiling , Humans , Immunophenotyping , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/classification , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Microarray Analysis , NK Cell Lectin-Like Receptor Subfamily C/deficiency , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/immunology , Promoter Regions, Genetic , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/immunology , Signal Transduction , Syk Kinase
7.
Immunity ; 42(3): 443-56, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25786176

ABSTRACT

The mechanisms underlying human natural killer (NK) cell phenotypic and functional heterogeneity are unknown. Here, we describe the emergence of diverse subsets of human NK cells selectively lacking expression of signaling proteins after human cytomegalovirus (HCMV) infection. The absence of B and myeloid cell-related signaling protein expression in these NK cell subsets correlated with promoter DNA hypermethylation. Genome-wide DNA methylation patterns were strikingly similar between HCMV-associated adaptive NK cells and cytotoxic effector T cells but differed from those of canonical NK cells. Functional interrogation demonstrated altered cytokine responsiveness in adaptive NK cells that was linked to reduced expression of the transcription factor PLZF. Furthermore, subsets of adaptive NK cells demonstrated significantly reduced functional responses to activated autologous T cells. The present results uncover a spectrum of epigenetically unique adaptive NK cell subsets that diversify in response to viral infection and have distinct functional capabilities compared to canonical NK cell subsets.


Subject(s)
Antibodies/immunology , Cytomegalovirus Infections/genetics , Epigenesis, Genetic/immunology , Killer Cells, Natural/immunology , Kruppel-Like Transcription Factors/immunology , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity , Cell Proliferation , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , DNA Methylation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Profiling , Humans , Immunophenotyping , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/classification , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Microarray Analysis , NK Cell Lectin-Like Receptor Subfamily C/deficiency , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/immunology , Promoter Regions, Genetic , Promyelocytic Leukemia Zinc Finger Protein , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/immunology , Signal Transduction , Syk Kinase , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/immunology
8.
J Virol ; 96(12): e0039422, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35612313

ABSTRACT

The main target cells for Epstein-Barr virus (EBV) infection and persistence are B lymphocytes, although T and NK cells can also become infected. In this paper, we characterize the EBV present in 21 pediatric and adult patients who were treated in France for a range of diseases that involve infection of T or NK cells. Of these 21 cases, 5 pediatric patients (21%) and 11 adult patients (52%) were of Caucasian origin. In about 30% of the cases, some of the EBV genomes contain a large deletion. The deletions are different in every patient but tend to cluster near the BART region of the viral genome. Detailed investigation of a family in which several members have persistent T or NK cell infection by EBV indicates that the virus genome deletions arise or are selected independently in each individual patient. Genome sequence polymorphisms in the EBV in these T or NK cell diseases reflect the geographic origin of the patient and not a distinct type of EBV (the 21 cases studied included examples of both type 1 and type 2 EBV infection). Using virus produced from type 1 or type 2 EBV genomes cloned in bacterial artificial chromosome (BAC) vectors, we demonstrate infection of T cells in cord blood from healthy donors. Our results are consistent with transient infection of some T cells being part of normal asymptomatic infection by EBV in young children. IMPORTANCE EBV contributes to several types of human cancer. Some cancers and nonmalignant lymphoproliferative diseases involving T or NK cells contain EBV. These diseases are relatively frequent in Japan and China and have been shown sometimes to have deletions in the EBV genome in the disease cells. We identify further examples of deletions within the EBV genome associated with T or NK cell diseases, and we provide evidence that the virus genomes with these deletions are most likely selected in the individual cases, rather than being transmitted between people during infection. We demonstrate EBV infection of cord blood T cells by highly characterized, cloned EBV genomes and suggest that transient infection of T cells may be part of normal asymptomatic infection by EBV in young children.


Subject(s)
Epstein-Barr Virus Infections , Gene Deletion , Genome, Viral , Herpesvirus 4, Human , Lymphoproliferative Disorders , Adult , Asymptomatic Infections , Child , Herpesvirus 4, Human/genetics , Humans , Killer Cells, Natural/virology , Lymphoproliferative Disorders/virology , T-Lymphocytes/virology
9.
PLoS Pathog ; 17(8): e1009868, 2021 08.
Article in English | MEDLINE | ID: mdl-34415956

ABSTRACT

Epstein Barr virus (EBV) causes a highly prevalent and lifelong infection contributing to the development of some malignancies. In addition to the key role played by T cells in controlling this pathogen, NK cells mediate cytotoxicity and IFNγ production in response to EBV-infected B cells in lytic cycle, both directly and through antibody (Ab)-dependent activation. We recently described that EBV-specific Ab-dependent NK cell interaction with viral particles (VP) bound to B cells triggered degranulation and TNFα secretion but not B cell lysis nor IFNγ production. In this report we show that NK cell activation under these conditions reduced B cell transformation by EBV. NK cells eliminated VP from the surface of B cells through a specific and active process which required tyrosine kinase activation, actin polymerization and Ca2+, being independent of proteolysis and perforin. VP were displayed at the NK cell surface before being internalized and partially shuttled to early endosomes and lysosomes. VP transfer was encompassed by a trogocytosis process including the EBV receptor CD21, together with CD19 and CD20. Our study reveals a novel facet of the antibody-dependent NK cell mediated response to this viral infection.


Subject(s)
Antibodies, Viral/immunology , B-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , B-Lymphocytes/virology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Humans , Killer Cells, Natural/virology
10.
PLoS Pathog ; 17(1): e1009255, 2021 01.
Article in English | MEDLINE | ID: mdl-33508041

ABSTRACT

Cytomegalovirus (CMV) causes clinically important diseases in immune compromised and immune immature individuals. Based largely on work in the mouse model of murine (M)CMV, there is a consensus that myeloid cells are important for disseminating CMV from the site of infection. In theory, such dissemination should expose CMV to cell-mediated immunity and thus necessitate evasion of T cells and NK cells. However, this hypothesis remains untested. We constructed a recombinant MCMV encoding target sites for the hematopoietic specific miRNA miR-142-3p in the essential viral gene IE3. This virus disseminated poorly to the salivary gland following intranasal or footpad infections but not following intraperitoneal infection in C57BL/6 mice, demonstrating that dissemination by hematopoietic cells is essential for specific routes of infection. Remarkably, depletion of NK cells or T cells restored dissemination of this virus in C57BL/6 mice after intranasal infection, while dissemination occurred normally in BALB/c mice, which lack strong NK cell control of MCMV. These data show that cell-mediated immunity is responsible for restricting MCMV to hematopoietic cell-mediated dissemination. Infected hematopoietic cells avoided cell-mediated immunity via three immune evasion genes that modulate class I MHC and NKG2D ligands (m04, m06 and m152). MCMV lacking these 3 genes spread poorly to the salivary gland unless NK cells were depleted, but also failed to replicate persistently in either the nasal mucosa or salivary gland unless CD8+ T cells were depleted. Surprisingly, CD8+ T cells primed after intranasal infection required CD4+ T cell help to expand and become functional. Together, our data suggest that MCMV can use both hematopoietic cell-dependent and -independent means of dissemination after intranasal infection and that cell mediated immune responses restrict dissemination to infected hematopoietic cells, which are protected from NK cells during dissemination by viral immune evasion. In contrast, viral replication within mucosal tissues depends on evasion of T cells.


Subject(s)
Herpesviridae Infections/immunology , Immune Evasion , Immunity, Cellular , Muromegalovirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/virology , Herpesviridae Infections/virology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/genetics , Muromegalovirus/physiology , Virus Replication
11.
PLoS Pathog ; 17(5): e1009593, 2021 05.
Article in English | MEDLINE | ID: mdl-34015056

ABSTRACT

Type I interferons (IFN-I) are antiviral cytokines that signal through the ubiquitous IFN-I receptor (IFNAR). Following footpad infection with ectromelia virus (ECTV), a mouse-specific pathogen, C57BL/6 (B6) mice survive without disease, while B6 mice broadly deficient in IFNAR succumb rapidly. We now show that for survival to ECTV, only hematopoietic cells require IFNAR expression. Survival to ECTV specifically requires IFNAR in both natural killer (NK) cells and monocytes. However, intrinsic IFNAR signaling is not essential for adaptive immune cell responses or to directly protect non-hematopoietic cells such as hepatocytes, which are principal ECTV targets. Mechanistically, IFNAR-deficient NK cells have reduced cytolytic function, while lack of IFNAR in monocytes dampens IFN-I production and hastens virus dissemination. Thus, during a pathogenic viral infection, IFN-I coordinates innate immunity by stimulating monocytes in a positive feedback loop and by inducing NK cell cytolytic function.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Animals , Cytokines/immunology , Disease Resistance , Ectromelia, Infectious/virology , Female , Hepatocytes/immunology , Hepatocytes/virology , Immunity, Innate , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/virology , Receptor, Interferon alpha-beta/genetics
12.
PLoS Pathog ; 17(8): e1009724, 2021 08.
Article in English | MEDLINE | ID: mdl-34352041

ABSTRACT

Hemagglutinin (HA) is the immunodominant protein of the influenza virus. We previously showed that mice injected with a monoglycosylated influenza A HA (HAmg) produced cross-strain-reactive antibodies and were better protected than mice injected with a fully glycosylated HA (HAfg) during lethal dose challenge. We employed a single B-cell screening platform to isolate the cross-protective monoclonal antibody (mAb) 651 from mice immunized with the HAmg of A/Brisbane/59/2007 (H1N1) influenza virus (Bris/07). The mAb 651 recognized the head domain of a broad spectrum of HAs from groups 1 and 2 influenza A viruses and offered prophylactic and therapeutic efficacy against A/California/07/2009 (H1N1) (Cal/09) and Bris/07 infections in mice. The antibody did not possess neutralizing activity; however, antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis mediated by natural killer cells and alveolar macrophages were important in the protective efficacy of mAb 651. Together, this study highlighted the significance of effector functions for non-neutralizing antibodies to exhibit protection against influenza virus infection.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antibody-Dependent Cell Cytotoxicity , Influenza A virus/immunology , Killer Cells, Natural/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/virology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
13.
Immunity ; 41(6): 960-72, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25526309

ABSTRACT

Granzyme B (GzmB) is a protease with a well-characterized intracellular role in targeted destruction of compromised cells by cytotoxic lymphocytes. However, GzmB also cleaves extracellular matrix components, suggesting that it influences the interplay between cytotoxic lymphocytes and their environment. Here, we show that GzmB-null effector T cells and natural killer (NK) cells exhibited a cell-autonomous homing deficit in mouse models of inflammation and Ectromelia virus infection. Intravital imaging of effector T cells in inflamed cremaster muscle venules revealed that GzmB-null cells adhered normally to the vessel wall and could extend lamellipodia through it but did not cross it efficiently. In vitro migration assays showed that active GzmB was released from migrating cytotoxic lymphocytes and enabled chemokine-driven movement through basement membranes. Finally, proteomic analysis demonstrated that GzmB cleaved basement membrane constituents. Our results highlight an important role for GzmB in expediting cytotoxic lymphocyte diapedesis via basement membrane remodeling.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Granzymes/metabolism , Killer Cells, Natural/physiology , T-Lymphocytes, Cytotoxic/physiology , Animals , Basement Membrane/metabolism , Cell Movement/genetics , Cells, Cultured , Chemokines/metabolism , Extracellular Matrix Proteins/metabolism , Granzymes/genetics , Killer Cells, Natural/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , T-Lymphocytes, Cytotoxic/virology , Transendothelial and Transepithelial Migration/genetics
14.
Gastroenterology ; 161(5): 1567-1583.e9, 2021 11.
Article in English | MEDLINE | ID: mdl-34302839

ABSTRACT

BACKGROUND & AIMS: The hepatitis D virus (HDV) causes the most severe form of chronic hepatitis, often progressing to cirrhosis within 5 to 10 years. There is no curative treatment, and the mechanisms underlying the accelerated liver disease progression are unknown. METHODS: Innate and adaptive immune responses were studied in blood and liver of 24 patients infected with HDV and 30 uninfected controls by multiparameter flow cytometry in correlation with disease severity and stage. RESULTS: The 2 main intrahepatic innate immune-cell populations, mucosal-associated invariant T cells and natural killer (NK) cells, were reduced in the livers of patients infected with HDV compared with those of uninfected controls but were more frequently activated in the liver compared with the blood. Most intrahepatic cluster of differentiation (CD) 8-positive (CD8+) T cells were memory cells or terminal effector memory cells, and most of the activated and degranulating (CD107a+) HDV-specific and total CD8+ T cells were liver-resident (CD69+C-X-C motif chemokine receptor 6+). Unsupervised analysis of flow cytometry data identified an activated, memory-like, tissue-resident HDV-specific CD8+ T-cell cluster with expression of innate-like NK protein 30 (NKp30) and NK group 2D (NKG2D) receptors. The size of this population correlated with liver enzyme activity (r = 1.0). NKp30 and NKG2D expression extended beyond the HDV-specific to the total intrahepatic CD8+ T-cell population, suggesting global bystander activation. This was supported by the correlations between (i) NKG2D expression with degranulation of intrahepatic CD8+ T cells, (ii) frequency of degranulating CD8+ T cells with liver enzyme activity and the aspartate aminotransferase-to-platelet ratio index score, and by the in vitro demonstration of cytokine-induced NKG2D-dependent cytotoxicity. CONCLUSION: Antigen-nonspecific activation of liver-resident CD8+ T cells may contribute to inflammation and disease stage in HDV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hepatitis D, Chronic/immunology , Hepatitis Delta Virus/immunology , Killer Cells, Natural/immunology , Liver/immunology , Lymphocyte Activation , Mucosal-Associated Invariant T Cells/immunology , Adaptive Immunity , Adult , Aged , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Case-Control Studies , Cell Degranulation , Cell Line, Tumor , Cytokines/blood , Disease Progression , Female , Hepatitis D, Chronic/blood , Hepatitis D, Chronic/diagnosis , Hepatitis D, Chronic/virology , Hepatitis Delta Virus/pathogenicity , Host-Pathogen Interactions , Humans , Immunity, Innate , Immunologic Memory , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Liver/metabolism , Liver/virology , Male , Middle Aged , Mucosal-Associated Invariant T Cells/metabolism , Mucosal-Associated Invariant T Cells/virology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Natural Cytotoxicity Triggering Receptor 3/metabolism , Phenotype , Young Adult
15.
J Virol ; 95(17): e0081621, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34133900

ABSTRACT

The complete eradication of human immunodeficiency virus type 1 (HIV-1) is blocked by latent reservoirs in CD4+ T cells and myeloid lineage cells. Toll-like receptors (TLRs) can induce the reversal of HIV-1 latency and trigger the innate immune response. To the best of our knowledge, there is little evidence showing the "killing" effect of TLR1/2 agonists but only a small "shock" potential. To identify a new approach for eradicating the HIV latent reservoir, we evaluated the effectiveness of SMU-Z1, a novel small-molecule TLR1/2 agonist, in the "shock-and-kill" strategy. The results showed that SMU-Z1 could enhance latent HIV-1 transcription not only ex vivo in peripheral blood mononuclear cells from aviremic HIV-1-infected donors receiving combined antiretroviral therapy but also in vitro in cells of myeloid-monocytic origin targeting the NF-κB and mitogen-activated protein kinase pathways. Interestingly, the activation marker CD69 was significantly upregulated in natural killer (NK) cells, B cells, and monocytes 48 h after SMU-Z1 treatment. Furthermore, SMU-Z1 was able to activate T cells without global T cell activation, as well as increasing NK cell degranulation and gamma interferon (IFN-γ) production, which further block HIV-1-infected CD4+ lymphocytes. In summary, the present study found that SMU-Z1 can both enhance HIV-1 transcription and promote NK cell-mediated inhibition of HIV-1-infected autologous CD4+ T cells. These findings indicate that the novel TLR1/2 agonist SMU-Z1 is a promising latency-reversing agent (LRA) for eradication of HIV-1 reservoirs. IMPORTANCE Multiple in vivo studies showed that many LRAs used in the shock-and-kill approach could activate viral transcription but could not induce killing effectively. Therefore, a dual-function LRA is needed for elimination of HIV-1 reservoirs. We previously developed a small-molecule TLR1/2 agonist, SMU-Z1, and demonstrated that it could upregulate NK cells and CD8+ T cells with immune adjuvant and antitumor properties in vivo. In the present study, SMU-Z1 could activate innate immune cells without global T cell activation, induce production of proinflammatory and antiviral cytokines, and enhance the cytotoxic function of NK cells. We showed that SMU-Z1 displayed dual potential ex vivo in the shock of exposure of latently HIV-1-infected cells and in the kill of clearance of infected cells, which is critical for effective use in combination with therapeutic vaccines or broadly neutralizing antibody treatments aimed at curing AIDS.


Subject(s)
Anti-Retroviral Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Imidazoles/pharmacology , Killer Cells, Natural/immunology , Phenols/pharmacology , Toll-Like Receptor 1/agonists , Toll-Like Receptor 2/agonists , Virus Latency , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-1/immunology , Humans , Imidazoles/therapeutic use , Killer Cells, Natural/drug effects , Killer Cells, Natural/virology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Phenols/therapeutic use , Small Molecule Libraries/pharmacology , Viral Load , Virus Activation
16.
Nat Immunol ; 11(5): 395-402, 2010 May.
Article in English | MEDLINE | ID: mdl-20351692

ABSTRACT

Inflammasomes regulate the activity of caspase-1 and the maturation of interleukin 1beta (IL-1beta) and IL-18. AIM2 has been shown to bind DNA and engage the caspase-1-activating adaptor protein ASC to form a caspase-1-activating inflammasome. Using Aim2-deficient mice, we identify a central role for AIM2 in regulating caspase-1-dependent maturation of IL-1beta and IL-18, as well as pyroptosis, in response to synthetic double-stranded DNA. AIM2 was essential for inflammasome activation in response to Francisella tularensis, vaccinia virus and mouse cytomegalovirus and had a partial role in the sensing of Listeria monocytogenes. Moreover, production of IL-18 and natural killer cell-dependent production of interferon-gamma, events critical in the early control of virus replication, were dependent on AIM2 during mouse cytomegalovirus infection in vivo. Collectively, our observations demonstrate the importance of AIM2 in the sensing of both bacterial and viral pathogens and in triggering innate immunity.


Subject(s)
DNA Virus Infections/immunology , DNA Viruses/immunology , Francisella tularensis/immunology , Killer Cells, Natural/metabolism , Listeriosis/immunology , Macrophages/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Tularemia/immunology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Caspase 1/genetics , Caspase 1/immunology , Caspase 1/metabolism , Cell Line , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Cytoskeletal Proteins/genetics , DNA/immunology , DNA Virus Infections/genetics , DNA Virus Infections/metabolism , DNA Viruses/growth & development , DNA Viruses/pathogenicity , DNA-Binding Proteins , Francisella tularensis/pathogenicity , Humans , Immunity, Innate , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Listeriosis/genetics , Listeriosis/metabolism , Lymphocyte Activation/genetics , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , Tularemia/genetics , Tularemia/metabolism , Viral Load/genetics , Viral Load/immunology
17.
Rev Med Virol ; 31(6): e2236, 2021 11.
Article in English | MEDLINE | ID: mdl-33793006

ABSTRACT

Modifications in HLA-I expression are found in many viral diseases. They represent one of the immune evasion strategies most widely used by viruses to block antigen presentation and NK cell response, and SARS-CoV-2 is no exception. These alterations result from a combination of virus-specific factors, genetically encoded mechanisms, and the status of host defences and range from loss or upregulation of HLA-I molecules to selective increases of HLA-I alleles. In this review, I will first analyse characteristic features of altered HLA-I expression found in SARS-CoV-2. I will then discuss the potential factors underlying these defects, focussing on HLA-E and class-I-related (like) molecules and their receptors, the most documented HLA-I alterations. I will also draw attention to potential differences between cells transfected to express viral proteins and those presented as part of authentic infection. Consideration of these factors and others affecting HLA-I expression may provide us with improved possibilities for research into cellular immunity against viral variants.


Subject(s)
Antigenic Variation , COVID-19/immunology , Clonal Anergy , Histocompatibility Antigens Class I/immunology , Immune Evasion , SARS-CoV-2/genetics , Alleles , COVID-19/pathology , COVID-19/virology , Cytokines/genetics , Cytokines/immunology , Cytotoxicity, Immunologic , Gene Expression , Histocompatibility Antigens Class I/genetics , Humans , Immunity, Cellular , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/immunology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology
18.
J Infect Dis ; 223(7): 1183-1195, 2021 04 08.
Article in English | MEDLINE | ID: mdl-32777077

ABSTRACT

Direct acting antiviral therapies rapidly clear chronic hepatitis C virus (HCV) infection and restore natural killer (NK) cell function. We investigated NK-cell memory formation following HCV clearance by examining NK-cell phenotype and responses from control and chronic HCV patients before and after therapy following sustained virologic response at 12 weeks post therapy (SVR12). NK-cell phenotype at SVR12 differed significantly from paired pretreatment samples, with an increase in maturation markers CD16, CD57, and KLRG1. HCV patients possessed stronger cytotoxic responses against HCV-infected cells as compared to healthy controls; a response that further increased following SVR12. The antigen-specific response was mediated by KLRG1+ NK cells, as demonstrated by increased degranulation and proliferation in response to HCV antigen only. Our data suggest that KLRG1+ HCV-specific memory NK cells develop following viral infection, providing insight into their role in HCV clearance and relevance with regard to vaccine design.


Subject(s)
Antiviral Agents , Hepatitis C, Chronic , Immunological Memory Cells/drug effects , Killer Cells, Natural/drug effects , Antiviral Agents/therapeutic use , Hepacivirus , Hepatitis C, Chronic/drug therapy , Humans , Immunological Memory Cells/virology , Interferons , Killer Cells, Natural/virology , Lectins, C-Type , Receptors, Immunologic
19.
Eur J Immunol ; 50(5): 685-694, 2020 05.
Article in English | MEDLINE | ID: mdl-32012247

ABSTRACT

Syncytin-1 is the envelope protein of the human endogenous retrovirus W (HERV-W). It has been related to multiple sclerosis (MS) but its role in cellular immunity and its pathogenic mechanism in the autoimmune context are not fully understood. We analyzed syncytin-1 levels in peripheral blood mononuclear cells (PBMC) subsets from healthy donors, MS patients in relapse or remission, and patients with acute infections by flow cytometry. PBMC cultures were also prepared to analyze protein expression kinetics. MS patients had higher levels of syncytin-1 levels than controls. We found that syncytin-1 is elevated in monocytes during MS relapses and infections. Cells expressing syncytin-1, including monocytes, T and B lymphocytes, and NKs presented mainly an activated phenotype and, upon stimulation with LPS, its levels increased rapidly on antigen-presenting cells. Syncytin-1 ligation promoted the activation of monocytes, as demonstrated by the upregulation of CD80 and the nonclassical subset CD14low CD16+ . Our results suggest an important role for syncytin-1 in the activation of leukocytes. Given that the expression of syncytin-1 is upregulated in MS patients, this protein might be contributing to the autoimmune cascade in the disease.


Subject(s)
Endogenous Retroviruses/immunology , Gene Products, env/genetics , Monocytes/virology , Multiple Sclerosis/genetics , Multiple Sclerosis/virology , Pregnancy Proteins/genetics , Adult , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/virology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Case-Control Studies , Endogenous Retroviruses/genetics , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation , Gene Products, env/immunology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , Male , Middle Aged , Monocytes/drug effects , Monocytes/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Pregnancy Proteins/immunology , Primary Cell Culture , Receptors, IgG/genetics , Receptors, IgG/immunology , Recurrence , Remission Induction , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology
20.
PLoS Pathog ; 15(12): e1008248, 2019 12.
Article in English | MEDLINE | ID: mdl-31869393

ABSTRACT

Viral and/or host factors that are directly responsible for the acute versus chronic outcome of hepatitis B virus (HBV) infection have not been identified yet. Information on immune response during the early stages of HBV infection in humans is mainly derived from blood samples of patients with acute hepatitis B (AHB), which are usually obtained after the onset of clinical symptoms. Features of intrahepatic immune response in these patients are less studied due to the difficulty of obtaining multiple liver biopsies. Woodchuck hepatitis virus (WHV) infection in woodchucks is a model for HBV infection in humans. In the present study, five adult woodchucks were experimentally infected with WHV and then followed for 18 weeks. Blood and liver tissues were frequently collected for assaying markers of WHV replication and innate and adaptive immune responses. Liver tissues were further analyzed for pathological changes and stained for important immune cell subsets and cytokines. The increase and subsequent decline of viral replication markers in serum and liver, the elicitation of antibodies against viral proteins, and the induction of virus-specific T-cell responses indicated eventual resolution of acute WHV infection in all animals. Intrahepatic innate immune makers stayed unchanged immediately after the infection, but increased markedly during resolution, as determined by changes in transcript levels. The presence of interferon-gamma and expression of natural killer (NK) cell markers suggested that a non-cytolytic response mechanism is involved in the initial viral control in liver. This was followed by the expression of T-cell markers and cytolytic effector molecules, indicating the induction of a cytolytic response mechanism. Parallel increases in regulatory T-cell markers suggested that this cell subset participates in the overall immune cell infiltration in liver and/or has a role in regulating AHB induced by the cytolytic response mechanism. Since the transcript levels of immune cell markers in blood, when detectable, were lower than in liver, and the kinetics, except for NK-cells and interferon-gamma, did not correlate well with their intrahepatic expression, this further indicated enrichment of immune cells within liver. Conclusion: The coordinated interplay of innate and adaptive immunity mediates viral clearance in the woodchuck animal model of HBV infection. The initial presence of NK-cell associated interferon-gamma response points to an important role of this cytokine in HBV resolution.


Subject(s)
Adaptive Immunity , Hepatitis B Virus, Woodchuck/pathogenicity , Hepatitis B/virology , Immunity, Innate , Killer Cells, Natural/virology , Marmota/virology , Aging , Animals , Hepatitis B Virus, Woodchuck/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Liver/pathology , Liver/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL