Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.560
Filter
1.
Front Immunol ; 15: 1374838, 2024.
Article in English | MEDLINE | ID: mdl-39281683

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells, which are key components of the immune system and involved in early immune responses. DCs are specialized in capturing, processing, and presenting antigens to facilitate immune interactions. Chickens infected with avian influenza virus (AIV) demonstrate a wide range of clinical symptoms, based on pathogenicity of the virus. Low pathogenic avian influenza (LPAI) viruses typically induce mild clinical signs, whereas high pathogenic avian influenza (HPAI) induce more severe disease, which can lead to death. For this study, chicken bone marrow-derived DC (ckBM-DC)s were produced and infected with high and low pathogenic avian influenza viruses of H5N2 or H7N3 subtypes to characterize innate immune responses, study effect on cell morphologies, and evaluate virus replication. A strong proinflammatory response was observed at 8 hours post infection, via upregulation of chicken interleukin-1ß and stimulation of the interferon response pathway. Microscopically, the DCs underwent morphological changes from classic elongated dendrites to a more general rounded shape that eventually led to cell death with the presence of scattered cellular debris. Differences in onset of morphologic changes were observed between H5 and H7 subtypes. Increases in viral titers demonstrated that both HPAI and LPAI are capable of infecting and replicating in DCs. The increase in activation of infected DCs may be indicative of a dysregulated immune response typically seen with HPAI infections.


Subject(s)
Chickens , Cytokines , Dendritic Cells , Influenza in Birds , Animals , Dendritic Cells/immunology , Dendritic Cells/virology , Chickens/virology , Influenza in Birds/immunology , Influenza in Birds/virology , Influenza in Birds/pathology , Cytokines/metabolism , Cytokines/immunology , Influenza A virus/immunology , Virus Replication , Bone Marrow Cells/immunology , Bone Marrow Cells/virology
2.
PLoS Pathog ; 20(9): e1012555, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39283919

ABSTRACT

Manipulation of immune cell functions, independently of direct infection of these cells, emerges as a key process in viral pathophysiology. Chronic infection by Human T-cell Leukemia Virus type 1 (HTLV-1) is associated with immune dysfunctions, including misdirected responses of dendritic cells (DCs). Here, we interrogate the ability of transformed HTLV-1-infected T cells to manipulate human DC functions. We show that exposure to transformed HTLV-1-infected T cells induces a biased and peculiar transcriptional signature in monocyte-derived DCs, associated with an inefficient maturation and a poor responsiveness to subsequent stimulation by a TLR4 agonist. This poor responsiveness is also associated with a unique transcriptional landscape characterized by a set of genes whose expression is either conferred, impaired or abolished by HTLV-1 pre-exposure. Induction of this functional impairment requires several hours of coculture with transformed HTLV-1-infected cells, and associated mechanisms driven by viral capture, cell-cell contacts, and soluble mediators. Altogether, this cross-talk between infected T cells and DCs illustrate how HTLV-1 might co-opt communications between cells to induce a unique local tolerogenic immune microenvironment suitable for its own persistence.


Subject(s)
Dendritic Cells , HTLV-I Infections , Human T-lymphotropic virus 1 , Humans , Dendritic Cells/immunology , Dendritic Cells/virology , Dendritic Cells/metabolism , Human T-lymphotropic virus 1/immunology , HTLV-I Infections/immunology , HTLV-I Infections/virology , HTLV-I Infections/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology , Cellular Reprogramming
3.
Int J Mol Med ; 54(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-39027993

ABSTRACT

CD150, also termed signaling lymphocyte activation molecule family member 1, is a cell surface receptor expressed on T cells, B cells, dendritic cells (DCs) and some tumors. Stimulation of CD150 on immune cells induces cell proliferation and cytokine production. However, the function of CD150 in Epstein­Barr virus (EBV)­infected B cells is still not fully understood. In the present study, CD150 expression on B cells increased rapidly following EBV infection, and various CD150 antibodies, measles viral proteins and recombinant CD150 proteins induced the secretion of multiple cytokines in both CD150+ EBV­transformed B cells and EBV+ lymphoma cells. Notably, the IL­1α protein level showed the greatest increase among all cytokines measured. The culture supernatant containing these cytokines induced the rapid differentiation of monocytes to DCs after only 2 days in vitro, which was faster than the established DC maturation time. Furthermore, knockdown of CD150 expression led to a reduction in the secretion of multiple cytokines, and monocyte differentiation was partially inhibited by anti­IL­1α and anti­granulocyte­macrophage colony­stimulating factor neutralizing antibodies. Collectively, the results of the present study suggest that CD150 activation triggers cytokine production in EBV­transformed B cells, and that measles virus coinfection might affect immune responses through the production of various cytokines in EBV+ lymphoma cells.


Subject(s)
B-Lymphocytes , Cell Differentiation , Cytokines , Herpesvirus 4, Human , Monocytes , Signaling Lymphocytic Activation Molecule Family Member 1 , Humans , Herpesvirus 4, Human/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/virology , Cytokines/metabolism , Monocytes/metabolism , Monocytes/immunology , Monocytes/virology , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Dendritic Cells/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , Lymphocyte Activation/immunology
4.
Cell Rep ; 43(7): 114478, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38985668

ABSTRACT

Lyssavirus is a kind of neurotropic pathogen that needs to evade peripheral host immunity to enter the central nervous system to accomplish infection. NLRP3 inflammasome activation is essential for the host to defend against pathogen invasion. This study demonstrates that the matrix protein (M) of lyssavirus can inhibit both the priming step and the activation step of NLRP3 inflammasome activation. Specifically, M of lyssavirus can compete with NEK7 for binding to NLRP3, which restricts downstream apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization. The serine amino acid at the 158th site of M among lyssavirus is critical for restricting ASC oligomerization. Moreover, recombinant lab-attenuated lyssavirus rabies (rabies lyssavirus [RABV]) with G158S mutation at M decreases interleukin-1ß (IL-1ß) production in bone-marrow-derived dendritic cells (BMDCs) to facilitate lyssavirus invasion into the brain thereby elevating pathogenicity in mice. Taken together, this study reveals a common mechanism by which lyssavirus inhibits NLRP3 inflammasome activation to evade host defenses.


Subject(s)
Dendritic Cells , Inflammasomes , Interleukin-1beta , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Viral Matrix Proteins , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Inflammasomes/metabolism , Inflammasomes/immunology , Mice , Viral Matrix Proteins/metabolism , Humans , Interleukin-1beta/metabolism , Dendritic Cells/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , Lyssavirus/metabolism , Lyssavirus/immunology , NIMA-Related Kinases/metabolism , Protein Binding , CARD Signaling Adaptor Proteins/metabolism , HEK293 Cells
5.
Proc Natl Acad Sci U S A ; 121(29): e2404349121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38985764

ABSTRACT

Intron-containing RNA expressed from the HIV-1 provirus activates type 1 interferon in primary human blood cells, including CD4+ T cells, macrophages, and dendritic cells. To identify the innate immune receptor required for detection of intron-containing RNA expressed from the HIV-1 provirus, a loss-of-function screen was performed with short hairpin RNA-expressing lentivectors targeting twenty-one candidate genes in human monocyte-derived dendritic cells. Among the candidate genes tested, only knockdown of XPO1 (CRM1), IFIH1 (MDA5), or MAVS prevented activation of the interferon-stimulated gene ISG15. The importance of IFIH1 protein was demonstrated by rescue of the knockdown with nontargetable IFIH1 coding sequence. Inhibition of HIV-1-induced ISG15 by the IFIH1-specific Nipah virus V protein, and by IFIH1-transdominant 2-CARD domain-deletion or phosphomimetic point mutations, indicates that IFIH1 (MDA5) filament formation, dephosphorylation, and association with MAVS are all required for innate immune activation in response to HIV-1 transduction. Since both IFIH1 (MDA5) and DDX58 (RIG-I) signal via MAVS, the specificity of HIV-1 RNA detection by IFIH1 was demonstrated by the fact that DDX58 knockdown had no effect on activation. RNA-Seq showed that IFIH1 knockdown in dendritic cells globally disrupted the induction of IFN-stimulated genes by HIV-1. Finally, specific enrichment of unspliced HIV-1 RNA by IFIH1 (MDA5), over two orders of magnitude, was revealed by formaldehyde cross-linking immunoprecipitation (f-CLIP). These results demonstrate that IFIH1 is the innate immune receptor for intron-containing RNA from the HIV-1 provirus and that IFIH1 potentially contributes to chronic inflammation in people living with HIV-1, even in the presence of effective antiretroviral therapy.


Subject(s)
Dendritic Cells , HIV-1 , Immunity, Innate , Interferon-Induced Helicase, IFIH1 , Introns , Proviruses , RNA, Viral , Humans , HIV-1/genetics , HIV-1/immunology , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Proviruses/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Dendritic Cells/metabolism , Introns/genetics , RNA, Viral/genetics , RNA, Viral/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/immunology , HIV Infections/immunology , HIV Infections/virology , HIV Infections/genetics , Karyopherins/genetics , Karyopherins/metabolism
6.
Gene ; 927: 148649, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38852697

ABSTRACT

During the birch pollen season an enhanced incidence of virus infections is noticed, raising the question whether pollen can affect anti-viral responses independent of allergic reactions. We previously showed that birch pollen-treatment of monocyte-derived dendritic cells (moDC) enhances human cytomegalovirus (HCMV) infection. Here we addressed how in moDC the relatively weak pollen response can affect the comparably strong response to HCMV. To this end, moDC were stimulated with aqueous birch pollen extract (APE), HCMV, and APE with HCMV, and transcriptomic signatures were determined after 6 and 24 h of incubation. Infection was monitored upon exposure of moDC to GFP expressing HCMV by flow cytometric analysis of GFP expressing cells. Principle component analysis of RNA sequencing data revealed close clustering of mock and APE treated moDC, whereas HCMV as well as APE with HCMV treated moDC clustered separately after 6 and 24 h of incubation, respectively. Communally induced genes were detected in APE, HCMV and APE with HCMV treated moDC. In APE with HCMV treated moDC, the comparably weak APE induced signatures were maintained after HCMV exposure. In particular, NF-κB/RELA and PI3K/AKT/MAPK signaling were altered upon APE with HCMV exposure. Earlier, we discovered that NF-κB inhibition alleviated APE induced enhancement of HCMV infection. Here we additionally found that impairment of PI3K signaling reduced HCMV infection in HCMV and APE with HCMV treated moDC. APE treated moDC that were exposed to HCMV show a unique host gene signature, which to a large extent is regulated by NF-κB activation and PI3K/AKT/MAPK signaling.


Subject(s)
Betula , Cytomegalovirus , Dendritic Cells , Pollen , Dendritic Cells/virology , Dendritic Cells/metabolism , Dendritic Cells/immunology , Pollen/genetics , Pollen/immunology , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Humans , Phosphatidylinositol 3-Kinases/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Transcriptome , Signal Transduction , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Cells, Cultured
7.
Cell Mol Immunol ; 21(9): 999-1007, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38918577

ABSTRACT

The interstitial fluids in tissues are constantly drained into the lymph nodes (LNs) as lymph through afferent lymphatic vessels and from LNs into the blood through efferent lymphatics. LNs are strategically positioned and have the appropriate cellular composition to serve as sites of adaptive immune initiation against invading pathogens. However, for lymph-borne viruses, which disseminate from the entry site to other tissues through the lymphatic system, immune cells in the draining LN (dLN) also play critical roles in curbing systemic viral dissemination during primary and secondary infections. Lymph-borne viruses in tissues can be transported to dLNs as free virions in the lymph or within infected cells. Regardless of the entry mechanism, infected myeloid antigen-presenting cells, including various subtypes of dendritic cells, inflammatory monocytes, and macrophages, play a critical role in initiating the innate immune response within the dLN. This innate immune response involves cellular crosstalk between infected and bystander innate immune cells that ultimately produce type I interferons (IFN-Is) and other cytokines and recruit inflammatory monocytes and natural killer (NK) cells. IFN-I and NK cell cytotoxicity can restrict systemic viral spread during primary infections and prevent serious disease. Additionally, the memory CD8+ T-cells that reside or rapidly migrate to the dLN can contribute to disease prevention during secondary viral infections. This review explores the intricate innate immune responses orchestrated within dLNs that contain primary viral infections and the role of memory CD8+ T-cells following secondary infection or CD8+ T-cell vaccination.


Subject(s)
Adaptive Immunity , Immunity, Innate , Lymph Nodes , Lymph Nodes/immunology , Lymph Nodes/virology , Animals , Humans , Virus Diseases/immunology , Virus Diseases/virology , Viruses/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Killer Cells, Natural/immunology
8.
Front Immunol ; 15: 1260439, 2024.
Article in English | MEDLINE | ID: mdl-38863700

ABSTRACT

Dengue virus (DENV), transmitted by infected mosquitoes, is a major public health concern, with approximately half the world's population at risk for infection. Recent decades have increasing incidence of dengue-associated disease alongside growing frequency of outbreaks. Although promising progress has been made in anti-DENV immunizations, post-infection treatment remains limited to non-specific supportive treatments. Development of antiviral therapeutics is thus required to limit DENV dissemination in humans and to help control the severity of outbreaks. Dendritic cells (DCs) are amongst the first cells to encounter DENV upon injection into the human skin mucosa, and thereafter promote systemic viral dissemination to additional human target cells. Autophagy is a vesicle trafficking pathway involving the formation of cytosolic autophagosomes, and recent reports have highlighted the extensive manipulation of autophagy by flaviviruses, including DENV, for viral replication. However, the temporal profiling and function of autophagy activity in DENV infection and transmission by human primary DCs remains poorly understood. Herein, we demonstrate that mechanisms of autophagosome formation and extracellular vesicle (EV) release have a pro-viral role in DC-mediated DENV transmission. We show that DENV exploits early-stage canonical autophagy to establish infection in primary human DCs. DENV replication enhanced autophagosome formation in primary human DCs, and intrinsically-heightened autophagosome biogenesis correlated with relatively higher rates of DC susceptibility to DENV. Furthermore, our data suggest that viral replication intermediates co-localize with autophagosomes, while productive DENV infection introduces a block at the late degradative stages of autophagy in infected DCs but not in uninfected bystander cells. Notably, we identify for the first time that approximately one-fourth of DC-derived CD9/CD81/CD63+ EVs co-express canonical autophagy marker LC3, and demonstrate that DC-derived EV populations are an alternative, cell-free mechanism by which DCs promote DENV transmission to additional target sites. Taken together, our study highlights intersections between autophagy and secretory pathways during viral infection, and puts forward autophagosome accumulation and viral RNA-laden EVs as host determinants of DC-mediated DENV infection in humans. Host-directed therapeutics targeting autophagy and exocytosis pathways thus have potential to enhance DC-driven resistance to DENV acquisition and thereby limit viral dissemination by initial human target cells following mosquito-to-human transmission of DENV.


Subject(s)
Autophagosomes , Autophagy , Dendritic Cells , Dengue Virus , Dengue , Secretory Pathway , Virus Replication , Humans , Dengue Virus/physiology , Dendritic Cells/immunology , Dendritic Cells/virology , Dendritic Cells/metabolism , Dengue/transmission , Dengue/virology , Dengue/immunology , Autophagosomes/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/virology , Cells, Cultured
9.
Eur Respir Rev ; 33(172)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38811032

ABSTRACT

Respiratory viral infections represent one of the major causes of death worldwide. The recent coronavirus disease 2019 pandemic alone claimed the lives of over 6 million people around the globe. It is therefore crucial to understand how the immune system responds to these threats and how respiratory infection can be controlled and constrained. Dendritic cells (DCs) are one of the key players in antiviral immunity because of their ability to detect pathogens. They can orchestrate an immune response that will, in most cases, lead to viral clearance. Different subsets of DCs are present in the lung and each subset can contribute to antiviral responses through various mechanisms. In this review, we discuss the role of the different lung DC subsets in response to common respiratory viruses, with a focus on respiratory syncytial virus, influenza A virus and severe acute respiratory syndrome coronavirus 2. We also review how lung DC-mediated responses to respiratory viruses can lead to the worsening of an existing chronic pulmonary disease such as asthma. Throughout the review, we discuss results obtained from animal studies as well as results generated from infected patients.


Subject(s)
Dendritic Cells , Respiratory Tract Infections , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Animals , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Lung/immunology , Lung/virology , Host-Pathogen Interactions , COVID-19/immunology , Virus Diseases/immunology , Virus Diseases/virology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
10.
J Immunol ; 212(11): 1782-1790, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38629901

ABSTRACT

Enterovirus 71 (EV71) is a significant causative agent of hand, foot, and mouth disease, with potential serious neurologic complications or fatal outcomes. The lack of effective treatments for EV71 infection is attributed to its elusive pathogenicity. Our study reveals that human plasmacytoid dendritic cells (pDCs), the main type I IFN-producing cells, selectively express scavenger receptor class B, member 2 (SCARB2) and P-selectin glycoprotein ligand 1 (PSGL-1), crucial cellular receptors for EV71. Some strains of EV71 can replicate within pDCs and stimulate IFN-α production. The activation of pDCs by EV71 is hindered by Abs to PSGL-1 and soluble PSGL-1, whereas Abs to SCARB2 and soluble SCARB2 have a less pronounced effect. Our data suggest that only strains binding to PSGL-1, more commonly found in severe cases, can replicate in pDCs and induce IFN-α secretion, highlighting the importance of PSGL-1 in these processes. Furthermore, IFN-α secretion by pDCs can be triggered by EV71 or UV-inactivated EV71 virions, indicating that productive infection is not necessary for pDC activation. These findings provide new insights into the interaction between EV71 and pDCs, suggesting that pDC activation could potentially mitigate the severity of EV71-related diseases.


Subject(s)
Dendritic Cells , Enterovirus A, Human , Interferon-alpha , Lysosomal Membrane Proteins , Membrane Glycoproteins , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Enterovirus A, Human/immunology , Enterovirus A, Human/physiology , Membrane Glycoproteins/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/immunology , Interferon-alpha/metabolism , Interferon-alpha/immunology , Receptors, Scavenger/metabolism , Enterovirus Infections/immunology , Enterovirus Infections/virology , Virus Replication
11.
Poult Sci ; 103(6): 103640, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38688195

ABSTRACT

The transmission electron microscopy revealed a dendritic cell in the medulla of the chicken bursal follicle. This dendritic cell has a classical secretory machinery; therefore, it has been named a bursal secretory dendritic cell (BSDC). The corticomedullary epithelial arch (CMEA) encloses lymphoid-like cells, which can proliferate and after entering the medulla, begin to differentiate to immature, then mature BSDC, which discharges glycoprotein (gp). With the exhaustion of gp production, the BSDC rapidly transforms into a macrophage-like cell (Mal), which is an activated endocytic cell of innate immunity. The Mal drifts through the follicle-associated epithelium (FAE)-supporting cells into the FAE, and via FAE, the Mal is eliminated in the bursal lumen. The infectious bursal disease virus (IBDV) infection accelerates the maturation process of BSDC precursors, which results in acute emptying of CMEA and subsequently, numerous immature BSDC(s) emerge. The IBDV infection stops the gp discharge, and the gp appears in the virus-containing Mal. The Movat pentachrome staining recognizes the gp in the extracellular spaces of the medulla and after infection in the Mal. The BSDC is the primary target of the IBDV. During IBDV infection, a large number of suddenly formed Mal actively migrate into the cortex, initiating cytokine storm and recruiting heterophil granulocytes. During embryogenesis, the vimentin-positive, possibly embryonic dendritic cells provide a microenvironment for carbohydrate switch. Around hatching, these embryonic, temporary dendritic cells get the Fc receptor, which bind maternal IgY. The posthatched forms of BSDC(s) gradually replace the embryonic ones and bind their own IgY.


Subject(s)
Bursa of Fabricius , Chickens , Dendritic Cells , Infectious bursal disease virus , Animals , Bursa of Fabricius/virology , Dendritic Cells/physiology , Dendritic Cells/virology , Infectious bursal disease virus/physiology , Birnaviridae Infections/veterinary , Birnaviridae Infections/virology , Poultry Diseases/virology , Poultry Diseases/immunology
12.
J Virol ; 98(5): e0019424, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38567950

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne flavivirus that caused an epidemic in the Americas in 2016 and is linked to severe neonatal birth defects, including microcephaly and spontaneous abortion. To better understand the host response to ZIKV infection, we adapted the 10× Genomics Chromium single-cell RNA sequencing (scRNA-seq) assay to simultaneously capture viral RNA and host mRNA. Using this assay, we profiled the antiviral landscape in a population of human monocyte-derived dendritic cells infected with ZIKV at the single-cell level. The bystander cells, which lacked detectable viral RNA, expressed an antiviral state that was enriched for genes coinciding predominantly with a type I interferon (IFN) response. Within the infected cells, viral RNA negatively correlated with type I IFN-dependent and -independent genes (the antiviral module). We modeled the ZIKV-specific antiviral state at the protein level, leveraging experimentally derived protein interaction data. We identified a highly interconnected network between the antiviral module and other host proteins. In this work, we propose a new paradigm for evaluating the antiviral response to a specific virus, combining an unbiased list of genes that highly correlate with viral RNA on a per-cell basis with experimental protein interaction data. IMPORTANCE: Zika virus (ZIKV) remains a public health threat given its potential for re-emergence and the detrimental fetal outcomes associated with infection during pregnancy. Understanding the dynamics between ZIKV and its host is critical to understanding ZIKV pathogenesis. Through ZIKV-inclusive single-cell RNA sequencing (scRNA-seq), we demonstrate on the single-cell level the dynamic interplay between ZIKV and the host: the transcriptional program that restricts viral infection and ZIKV-mediated inhibition of that response. Our ZIKV-inclusive scRNA-seq assay will serve as a useful tool for gaining greater insight into the host response to ZIKV and can be applied more broadly to the flavivirus field.


Subject(s)
Dendritic Cells , Single-Cell Analysis , Zika Virus Infection , Zika Virus , Humans , Zika Virus/physiology , Zika Virus Infection/virology , Zika Virus Infection/immunology , Dendritic Cells/virology , Dendritic Cells/immunology , RNA, Viral/metabolism , RNA, Viral/genetics , Interferon Type I/metabolism , Host-Pathogen Interactions , Sequence Analysis, RNA
13.
J Infect Dis ; 229(6): 1781-1785, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38385222

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) disease manifestations differ between cisgender women and men, including better control of viral replication during primary infection and less frequent residual HIV-1 replication on antiretroviral therapy (ART) in cisgender women with HIV-1 (WWH). Investigating plasmacytoid dendritic cell (pDC) functions and HIV-1 reservoir sizes in 20 WWH on stable ART, we observed inverse correlations between interferon-α and tumor necrosis factor responses of pDCs to Toll-like receptor 7/8 stimulation and intact/total proviral HIV-1 DNA levels. Additionally, ISG15 mRNA levels in peripheral blood mononuclear cells correlated with cytokine responses of pDCs. These findings demonstrate an association between higher type I interferon responses and lower HIV-1 reservoir sizes in WWH on ART, warranting studies to identify the underlying mechanisms.


Subject(s)
Dendritic Cells , HIV Infections , HIV-1 , Interferon Type I , Toll-Like Receptor 7 , Humans , Dendritic Cells/immunology , Dendritic Cells/virology , Female , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , Adult , Middle Aged , Virus Replication/drug effects , Viral Load , Anti-Retroviral Agents/therapeutic use , Leukocytes, Mononuclear/virology , Leukocytes, Mononuclear/immunology
14.
J Infect Dis ; 230(2): 467-479, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-38135285

ABSTRACT

Respiratory syncytial virus (RSV) primarily infects the respiratory epithelium, but growing evidence suggests that it may also be responsible for neurologic sequelae. In 3-dimensional microphysiologic peripheral nerve cultures, RSV infected neurons, macrophages, and dendritic cells along 2 distinct trajectories depending on the initial viral load. Low-level infection was transient, primarily involved macrophages, and induced moderate chemokine release with transient neural hypersensitivity. Infection with higher viral loads was persistent, infected neuronal cells in addition to monocytes, and induced robust chemokine release followed by progressive neurotoxicity. In spinal cord cultures, RSV infected microglia and dendritic cells but not neurons, producing a moderate chemokine expression pattern. The persistence of infection was variable but could be identified in dendritic cells as long as 30 days postinoculation. This study suggests that RSV can disrupt neuronal function directly through infection of peripheral neurons and indirectly through infection of resident monocytes and that inflammatory chemokines likely mediate both mechanisms.


Subject(s)
Chemokines , Respiratory Syncytial Virus Infections , Spinal Cord , Chemokines/metabolism , Spinal Cord/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Animals , Neurons/virology , Neurons/metabolism , Humans , Peripheral Nerves/virology , Macrophages/virology , Macrophages/immunology , Macrophages/metabolism , Viral Load , Dendritic Cells/virology , Dendritic Cells/immunology , Monocytes/virology , Monocytes/immunology , Monocytes/metabolism , Cells, Cultured , Respiratory Syncytial Viruses/immunology , Microglia/virology , Microglia/metabolism
15.
Nat Commun ; 14(1): 2898, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37217469

ABSTRACT

The DNA sensor cyclic GMP-AMP synthase (cGAS) is critical in host antiviral immunity. Vaccinia virus (VACV) is a large cytoplasmic DNA virus that belongs to the poxvirus family. How vaccinia virus antagonizes the cGAS-mediated cytosolic DNA-sensing pathway is not well understood. In this study, we screened 80 vaccinia genes to identify potential viral inhibitors of the cGAS/Stimulator of interferon gene (STING) pathway. We discovered that vaccinia E5 is a virulence factor and a major inhibitor of cGAS. E5 is responsible for abolishing cGAMP production during vaccinia virus (Western Reserve strain) infection of dendritic cells. E5 localizes to the cytoplasm and nucleus of infected cells. Cytosolic E5 triggers ubiquitination of cGAS and proteasome-dependent degradation via interacting with cGAS. Deleting the E5R gene from the Modified vaccinia virus Ankara (MVA) genome strongly induces type I IFN production by dendritic cells (DCs) and promotes DC maturation, and thereby improves antigen-specific T cell responses.


Subject(s)
Dendritic Cells , Nucleotidyltransferases , Vaccinia virus , Viral Proteins , Mice, Inbred C57BL , Animals , Mice , Mice, Knockout , Female , Nucleotidyltransferases/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Vaccinia virus/pathogenicity , Virulence Factors/immunology , Ubiquitination , Viral Proteins/genetics , Viral Proteins/immunology , Proteasome Endopeptidase Complex , Interferon Type I/immunology , HEK293 Cells , Humans , Membrane Proteins/immunology , T-Lymphocytes/immunology
16.
J Virol ; 96(18): e0124022, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36094317

ABSTRACT

Viruses have evolved numerous strategies to impair immunity so that they can replicate more efficiently. Among those, the immunosuppressive effects of morbillivirus infection can be particularly problematic, as they allow secondary infections to take hold in the host, worsening disease prognosis. In the present work, we hypothesized that the highly contagious morbillivirus peste des petits ruminants virus (PPRV) could target monocytes and dendritic cells (DC) to contribute to the immunosuppressive effects produced by the infection. Monocytes isolated from healthy sheep, a natural host of the disease, were able be infected by PPRV and this impaired the differentiation and phagocytic ability of immature monocyte-derived DC (MoDC). We also assessed PPRV capacity to infect differentiated MoDC. Ovine MoDC could be productively infected by PPRV, and this drastically reduced MoDC capacity to activate allogeneic T cell responses. Transcriptomic analysis of infected MoDC indicated that several tolerogenic DC signature genes were upregulated upon PPRV infection. Furthermore, PPRV-infected MoDC could impair the proliferative response of autologous CD4+ and CD8+ T cell to the mitogen concanavalin A (ConA), which indicated that DC targeting by the virus could promote immunosuppression. These results shed new light on the mechanisms employed by morbillivirus to suppress the host immune responses. IMPORTANCE Morbilliviruses pose a threat to global health given their high infectivity. The morbillivirus peste des petits ruminants virus (PPRV) severely affects small-ruminant-productivity and leads to important economic losses in communities that rely on these animals for subsistence. PPRV produces in the infected host a period of severe immunosuppression that opportunistic pathogens exploit, which worsens the course of the infection. The mechanisms of PPRV immunosuppression are not fully understood. In the present work, we demonstrate that PPRV can infect professional antigen-presenting cells called dendritic cells (DC) and disrupt their capacity to elicit an immune response. PPRV infection promoted a DC activation profile that favored the induction of tolerance instead of the activation of an antiviral immune response. These results shed new light on the mechanisms employed by morbilliviruses to suppress the immune responses.


Subject(s)
Dendritic Cells , Lymphocyte Activation , Peste-des-Petits-Ruminants , Peste-des-petits-ruminants virus , Animals , Antiviral Agents , Cell Differentiation , Concanavalin A/genetics , Concanavalin A/immunology , Dendritic Cells/cytology , Dendritic Cells/virology , Goats , Immunosuppression Therapy , Lymphocyte Activation/immunology , Mitogens/immunology , Peste-des-Petits-Ruminants/immunology , Peste-des-Petits-Ruminants/virology , Phenotype , Sheep , T-Lymphocytes/immunology , T-Lymphocytes/virology
17.
Viruses ; 14(1)2022 01 05.
Article in English | MEDLINE | ID: mdl-35062296

ABSTRACT

Due to their ability to trigger strong immune responses, adenoviruses (HAdVs) in general and the serotype5 (HAdV-5) in particular are amongst the most popular viral vectors in research and clinical application. However, efficient transduction using HAdV-5 is predominantly achieved in coxsackie and adenovirus receptor (CAR)-positive cells. In the present study, we used the transduction enhancer LentiBOOST® comprising the polycationic Polybrene to overcome these limitations. Using LentiBOOST®/Polybrene, we yielded transduction rates higher than 50% in murine bone marrow-derived dendritic cells (BMDCs), while maintaining their cytokine expression profile and their capability to induce T-cell proliferation. In human dendritic cells (DCs), we increased the transduction rate from 22% in immature (i)DCs or 43% in mature (m)DCs to more than 80%, without inducing cytotoxicity. While expression of specific maturation markers was slightly upregulated using LentiBOOST®/Polybrene on iDCs, no effect on mDC phenotype or function was observed. Moreover, we achieved efficient HAdV5 transduction also in human monocytes and were able to subsequently differentiate them into proper iDCs and functional mDCs. In summary, we introduce LentiBOOST® comprising Polybrene as a highly potent adenoviral transduction agent for new in-vitro applications in a set of different immune cells in both mice and humans.


Subject(s)
Adenoviruses, Human/genetics , Dendritic Cells/virology , Monocytes/virology , Transduction, Genetic , Adenoviruses, Human/physiology , Animals , Cell Differentiation , Cell Proliferation , Dendritic Cells/immunology , Electroporation , Genetic Vectors , Hexadimethrine Bromide , Host Specificity , Humans , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Phenotype , Virus Internalization
18.
Sci Rep ; 12(1): 135, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997007

ABSTRACT

Interleukin-7 (IL-7) is a cytokine known for its importance in T cell development and survival. How IL-7 shapes CD8 T cell responses during an acute viral infection is less understood. We had previously shown that IL-7 signaling deficient mice have reduced accumulation of influenza-specific CD8 T cells following influenza infection. We sought to determine whether IL-7 affects early CD8 T cell expansion in the mediastinal lymph node and effector function in the lungs. Using IL-7Rα signaling deficient mice, we show that IL-7 is required for a normal sized mediastinal lymph node and the early clonal expansion of influenza-specific CD8 T cells therein. We show that IL-7 plays a cell-intrinsic role in the accumulation of NP366-374 and PA224-233-specific CD8 T cells in the lymph node. We also found that IL-7 shapes terminal differentiation, degranulation and cytokine production to a greater extent in PA224-233-specific than NP366-374-specific CD8 T cells. We further demonstrate that IL-7 is induced in the lung tissue by viral infection and we characterize multiple cellular sources that contribute to IL-7 production. Our findings on IL-7 and its effects on lower respiratory diseases will be important for expanding the utility of therapeutics that are currently available.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Interleukin-7/metabolism , Lung/metabolism , Lymph Nodes/metabolism , Orthomyxoviridae Infections/metabolism , A549 Cells , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Disease Models, Animal , Female , Host-Pathogen Interactions , Humans , Alphainfluenzavirus/immunology , Alphainfluenzavirus/pathogenicity , Interleukin-7/genetics , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Lung/immunology , Lung/virology , Lymph Nodes/immunology , Lymph Nodes/virology , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction
19.
J Virol ; 96(4): e0186721, 2022 02 23.
Article in English | MEDLINE | ID: mdl-34878888

ABSTRACT

Common to all cytomegalovirus (CMV) genomes analyzed to date is the presence of G protein-coupled receptors (GPCR). Animal models of CMV provide insights into their role in viral fitness. The mouse cytomegalovirus (MCMV) GPCR, M33, facilitates dendritic cell (DC)-dependent viremia, the extravasation of blood-borne infected DCs to the salivary gland, and the frequency of reactivation events from latently infected tissue explants. Constitutive G protein-coupled M33 signaling is required for these phenotypes, although the contribution of distinct biochemical pathways activated by M33 is unknown. M33 engages Gq/11 to constitutively activate phospholipase C ß (PLCß) and downstream cyclic AMP response-element binding protein (CREB) in vitro. Identification of a MCMV M33 mutant (M33ΔC38) for which CREB signaling was disabled but PLCß activation was preserved provided the opportunity to investigate their relevance in vivo. Following intranasal infection with MCMV M33ΔC38, the absence of M33 CREB Gq/11-dependent signaling correlated with reduced mobilization of lytically-infected DCs to the draining lymph node high endothelial venules (HEVs) and reduced viremia compared with wild type MCMV. In contrast, M33ΔC38-infected DCs within the vascular compartment extravasated to the salivary glands via a pertussis toxin-sensitive, Gi/o-dependent, and CREB-independent mechanism. In the context of MCMV latency, spleen explants from M33ΔC38-infected mice were markedly attenuated for reactivation. Taken together, these data demonstrate that key features of the MCMV life cycle are coordinated in diverse tissues by distinct pathways of the M33 signaling repertoire. IMPORTANCE G protein-coupled receptors (GPCRs) act as cell surface molecular "switches" that regulate the cellular response to environmental stimuli. All cytomegalovirus (CMV) genomes analyzed to date possess GPCR homologs with phylogenetic evidence for independent gene capture events, signifying important in vivo roles. The mouse CMV (MCMV) GPCR homolog, designated M33, is important for cell-associated virus spread and the establishment and/or reactivation of latent MCMV infection. The signaling repertoire of M33 is distinct from cellular GPCRs and little is known of the relevance of component signaling pathways for in vivo M33 function. In this report, we showed that temporal and tissue-specific M33 signaling was required to facilitate in vivo infection. Understanding the relevance of the viral GPCR signaling profiles for in vivo function will provide opportunities for future targeted interventions.


Subject(s)
Herpesviridae Infections/virology , Muromegalovirus/physiology , Receptors, G-Protein-Coupled/metabolism , Viral Proteins/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Cells/virology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Herpesviridae Infections/metabolism , Lymph Nodes/virology , Mice , Mice, Inbred BALB C , Muromegalovirus/genetics , Muromegalovirus/metabolism , Mutation , Phospholipase C beta/metabolism , Receptors, G-Protein-Coupled/genetics , Salivary Glands/virology , Signal Transduction , Viral Proteins/genetics , Viremia/metabolism , Viremia/virology , Virus Activation/genetics
20.
J Mol Biol ; 434(6): 167389, 2022 03 30.
Article in English | MEDLINE | ID: mdl-34883114

ABSTRACT

Dendritic cells (DCs) play a pivotal role in the functional differentiation of CD4+ T cells in response to pathogens. In CD4+ T cells, HIV-1 replicates efficiently, while HIV-2, a related virus of reduced pathogenicity, is better controlled. How the DC response to HIV-1 vs HIV-2 contributes to programming an antiviral state in CD4+ T cells is not known. Here, we identify a transcriptional signature associated with progressive resistance to HIV infection in CD4+ T cells. We developed a model of naïve CD4+ T cell priming by DCs stimulated with a panel of seven viruses or synthetic ligands for the viral nucleic acid sensors cGAS and TLRs. DCs produced a cytokine response to HIV-2 infection more similar to the response to cGAS ligands than TLR ligands. In response to these signals, naive CD4+ T cells acquired a gradual antiviral resistance to subsequent HIV infection. The antiviral state was concomitant with the induction of the TH1 cytokine IFNγ and the type I interferon-stimulated gene (ISG) MX1, while the TFH cytokine IL-21 was not increased. By performing a transcriptional network analysis in T cells, we identified five distinct gene modules with characteristic ISG, TH1, TFH, IFN-I and proliferative signatures. Finally, we leverage this module to assemble a T antiviral signature of 404 genes that correlate with the antiviral state in T cells. Altogether, the study illuminates the programming of the antiviral state in T cells. The T antiviral gene signature in human CD4+ lymphocytes constitutes a resource for genetic screens and genomics analysis.


Subject(s)
CD4-Positive T-Lymphocytes , Dendritic Cells , HIV Infections , HIV-2 , Transcriptome , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/genetics , HIV Infections/immunology , HIV-2/genetics , HIV-2/physiology , Humans , Ligands , Nucleotidyltransferases/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL