Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 151
1.
J Virol ; 98(5): e0049324, 2024 May 14.
Article En | MEDLINE | ID: mdl-38578092

CD4+ T cells play a key role in γ-herpesvirus infection control. However, the mechanisms involved are unclear. Murine herpesvirus type 4 (MuHV-4) allows relevant immune pathways to be dissected experimentally in mice. In the lungs, it colonizes myeloid cells, which can express MHC class II (MHCII), and type 1 alveolar epithelial cells (AEC1), which lack it. Nevertheless, CD4+ T cells can control AEC1 infection, and this control depends on MHCII expression in myeloid cells. Interferon-gamma (IFNγ) is a major component of CD4+ T cell-dependent MuHV-4 control. Here, we show that the action of IFNγ is also indirect, as CD4+ T cell-mediated control of AEC1 infection depended on IFNγ receptor (IFNγR1) expression in CD11c+ cells. Indirect control also depended on natural killer (NK) cells. Together, the data suggest that the activation of MHCII+ CD11c+ antigen-presenting cells is key to the CD4+ T cell/NK cell protection axis. By contrast, CD8+ T cell control of AEC1 infection appeared to operate independently. IMPORTANCE: CD4+ T cells are critical for the control of gamma-herpesvirus infection; they act indirectly, by recruiting natural killer (NK) cells to attack infected target cells. Here, we report that the CD4+ T cell/NK cell axis of gamma-herpesvirus control requires interferon-γ engagement of CD11c+ dendritic cells. This mechanism of CD4+ T cell control releases the need for the direct engagement of CD4+ T cells with virus-infected cells and may be a common strategy for host control of immune-evasive pathogens.


CD4-Positive T-Lymphocytes , Herpesviridae Infections , Interferon-gamma , Killer Cells, Natural , Receptors, Interferon , Rhadinovirus , Animals , CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Killer Cells, Natural/immunology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Rhadinovirus/immunology , Mice, Inbred C57BL , Interferon gamma Receptor , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/virology , CD8-Positive T-Lymphocytes/immunology , CD11c Antigen/metabolism , CD11c Antigen/immunology , Lung/immunology , Lung/virology
2.
Viruses ; 14(1)2022 01 06.
Article En | MEDLINE | ID: mdl-35062301

Human respiratory syncytial virus (hRSV) infection brings a wide spectrum of clinical outcomes, from a mild cold to severe bronchiolitis or even acute interstitial pneumonia. Among the known factors influencing this clinical diversity, genetic background has often been mentioned. In parallel, recent evidence has also pointed out that an early infectious experience affects heterologous infections severity. Here, we analyzed the importance of these two host-related factors in shaping the immune response in pneumoviral disease. We show that a prior gammaherpesvirus infection improves, in a genetic background-dependent manner, the immune system response against a subsequent lethal dose of pneumovirus primary infection notably by inducing a systematic expansion of the CD8+ bystander cell pool and by modifying the resident alveolar macrophages (AMs) phenotype to induce immediate cyto/chemokinic responses upon pneumovirus exposure, thereby drastically attenuating the host inflammatory response without affecting viral replication. Moreover, we show that these AMs present similar rapid and increased production of neutrophil chemokines both in front of pneumoviral or bacterial challenge, confirming recent studies attributing a critical antibacterial role of primed AMs. These results corroborate other recent studies suggesting that the innate immunity cells are themselves capable of memory, a capacity hitherto reserved for acquired immunity.


Genetic Background , Herpesviridae Infections/immunology , Macrophages, Alveolar/immunology , Pneumovirus Infections/immunology , Pneumovirus/immunology , Rhadinovirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Immunity, Innate , Inflammation/immunology , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Pneumococcal Infections/immunology , Pneumovirus/physiology , Pneumovirus Infections/genetics , Pneumovirus Infections/pathology , Pneumovirus Infections/virology , Rhadinovirus/physiology
3.
J Virol ; 95(14): e0033021, 2021 06 24.
Article En | MEDLINE | ID: mdl-33910957

A prophylactic vaccine that confers durable protection against human immunodeficiency virus (HIV) would provide a valuable tool to prevent new HIV/AIDS cases. As herpesviruses establish lifelong infections that remain largely subclinical, the use of persistent herpesvirus vectors to deliver HIV antigens may facilitate the induction of long-term anti-HIV immunity. We previously developed recombinant (r) forms of the gamma-herpesvirus rhesus monkey rhadinovirus (rRRV) expressing a replication-incompetent, near-full-length simian immunodeficiency virus (SIVnfl) genome. We recently showed that 8/16 rhesus macaques (RMs) vaccinated with a rDNA/rRRV-SIVnfl regimen were significantly protected against intrarectal (i.r.) challenge with SIVmac239. Here we investigated the longevity of this vaccine-mediated protection. Despite receiving no additional booster immunizations, the protected rDNA/rRRV-SIVnfl vaccinees maintained detectable cellular and humoral anti-SIV immune responses for more than 1.5 years after the rRRV boost. To assess if these responses were still protective, the rDNA/rRRV-SIVnfl vaccinees were subjected to a second round of marginal-dose i.r. SIVmac239 challenges, with eight SIV-naive RMs serving as concurrent controls. After three SIV exposures, 8/8 control animals became infected, compared to 3/8 vaccinees. This difference in SIV acquisition was statistically significant (P = 0.0035). The three vaccinated monkeys that became infected exhibited significantly lower viral loads than those in unvaccinated controls. Collectively, these data illustrate the ability of rDNA/rRRV-SIVnfl vaccination to provide long-term immunity against stringent mucosal challenges with SIVmac239. Future work is needed to identify the critical components of this vaccine-mediated protection and the extent to which it can tolerate sequence mismatches in the challenge virus. IMPORTANCE We report on the long-term follow-up of a group of rhesus macaques (RMs) that received an AIDS vaccine regimen and were subsequently protected against rectal acquisition of simian immunodeficiency virus (SIV) infection. The vaccination regimen employed included a live recombinant herpesvirus vector that establishes persistent infection in RMs. Consistent with the recurrent SIV antigen expression afforded by this herpesvirus vector, vaccinees maintained detectable SIV-specific immune responses for more than 1.5 years after the last vaccination. Importantly, these vaccinated RMs were significantly protected against a second round of rectal SIV exposures performed 1 year after the first SIV challenge phase. These results are relevant for HIV vaccine development because they show the potential of herpesvirus-based vectors to maintain functional antiretroviral immunity without the need for repeated boosting.


Genetic Vectors , Rhadinovirus/genetics , SAIDS Vaccines/genetics , Simian Immunodeficiency Virus/genetics , Animals , Antibodies, Viral/immunology , Female , Follow-Up Studies , Immunogenicity, Vaccine , Immunologic Memory , Macaca mulatta , Male , Rhadinovirus/immunology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/immunology , Time Factors
4.
Ann Clin Transl Neurol ; 8(2): 456-470, 2021 02.
Article En | MEDLINE | ID: mdl-33440071

OBJECTIVE: To determine whether animals with Japanese macaque encephalomyelitis (JME), a spontaneous demyelinating disease similar to multiple sclerosis (MS), harbor myelin-specific T cells in their central nervous system (CNS) and periphery. METHODS: Mononuclear cells (MNCs) from CNS lesions, cervical lymph nodes (LNs) and peripheral blood of Japanese macaques (JMs) with JME, and cervical LN and blood MNCs from healthy controls or animals with non-JME conditions were analyzed for the presence of myelin-specific T cells and changes in interleukin 17 (IL-17) and interferon gamma (IFNγ) expression. RESULTS: Demyelinating JME lesions contained CD4+ T cells and CD8+ T cells specific to myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), and/or proteolipid protein (PLP). CD8+ T-cell responses were absent in JME peripheral blood, and in age- and sex-matched controls. However, CD4+ Th1 and Th17 responses were detected in JME peripheral blood versus controls. Cervical LN MNCs from eight of nine JME animals had CD3+ T cells specific for MOG, MBP, and PLP that were not detected in controls. Mapping myelin epitopes revealed a heterogeneity in responses among JME animals. Comparison of myelin antigen sequences with those of JM rhadinovirus (JMRV), which is found in JME lesions, identified six viral open reading frames (ORFs) with similarities to myelin antigen sequences. Overlapping peptides to these JMRV ORFs did not induce IFNγ responses. INTERPRETATIONS: JME possesses an immune-mediated component that involves both CD4+ and CD8+ T cells specific for myelin antigens. JME may shed new light on inflammatory demyelinating disease pathogenesis linked to gamma-herpesvirus infection.


Demyelinating Diseases/diagnostic imaging , Demyelinating Diseases/pathology , Encephalomyelitis/diagnostic imaging , Encephalomyelitis/pathology , Myelin Sheath/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/immunology , Demyelinating Diseases/virology , Encephalomyelitis/virology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , Female , Herpesviridae Infections/immunology , Interferon-gamma/analysis , Interleukin-17/analysis , Macaca fuscata , Male , Monkey Diseases , Myelin Basic Protein/genetics , Myelin Basic Protein/immunology , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/immunology , Myelin Sheath/pathology , Myelin-Oligodendrocyte Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein/immunology , Rhadinovirus/genetics , Rhadinovirus/immunology
5.
JCI Insight ; 6(2)2021 01 25.
Article En | MEDLINE | ID: mdl-33491663

The aryl-hydrocarbon receptor (AHR) is an intracellular sensor of aromatic hydrocarbons that sits at the top of various immunomodulatory pathways. Here, we present evidence that AHR plays a role in controlling IL-17 responses and the development of pulmonary fibrosis in response to respiratory pathogens following bone marrow transplant (BMT). Mice infected intranasally with gamma-herpesvirus 68 (γHV-68) following BMT displayed elevated levels of the AHR ligand, kynurenine (kyn), in comparison with control mice. Inhibition or genetic ablation of AHR signaling resulted in a significant decrease in IL-17 expression as well as a reduction in lung pathology. Lung CD103+ DCs expressed AHR following BMT, and treatment of induced CD103+ DCs with kyn resulted in altered cytokine production in response to γHV-68. Interestingly, mice deficient in the kyn-producing enzyme indolamine 2-3 dioxygenase showed no differences in cytokine responses to γHV-68 following BMT; however, isolated pulmonary fibroblasts infected with γHV-68 expressed the kyn-producing enzyme tryptophan dioxygenase (TDO2). Our data indicate that alterations in the production of AHR ligands in response to respiratory pathogens following BMT results in a pro-Th17 phenotype that drives lung pathology. We have further identified the TDO2/AHR axis as a potentially novel form of intercellular communication between fibroblasts and DCs that shapes immune responses to respiratory pathogens.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Transplantation/adverse effects , Pulmonary Fibrosis/etiology , Receptors, Aryl Hydrocarbon/metabolism , Rhadinovirus/pathogenicity , Tryptophan Oxygenase/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Dendritic Cells/pathology , Dendritic Cells/physiology , Disease Models, Animal , Hematopoietic Stem Cell Transplantation/adverse effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency , Interleukin-17/biosynthesis , Kynurenine/metabolism , Ligands , Lung/immunology , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Rhadinovirus/immunology , Signal Transduction , Th17 Cells/immunology
6.
PLoS Pathog ; 16(7): e1008701, 2020 07.
Article En | MEDLINE | ID: mdl-32735617

Gammaherpesviruses have evolved various strategies to take advantage of host cellular factors or signaling pathways to establish a lifelong latent infection. Like the human gammaherpesvirus Epstein-Barr virus, murine gammaherpesvirus 68 (MHV68) establishes and maintains latency in the memory B cells during infection of laboratory mice. We have previously shown that MHV68 can immortalize fetal liver-derived B cells that induce lymphomas when injected into immunodeficient mice. Here we identify interleukin 16 (IL16) as a most abundantly expressed cytokine in MHV68-immortalized B cells and show that MHV68 infection elevates IL16 expression. IL16 is not important for MHV68 lytic infection but plays a critical role in MHV68 reactivation from latency. IL16 deficiency increases MHV68 lytic gene expression in MHV68-immortalized B cells and enhances reactivation from splenic latency. Correlatively, IL16 deficiency increases the frequency of MHV68-infected plasma cells that can be attributed to enhanced MHV68 reactivation. Furthermore, similar to TPA-mediated lytic replication of Kaposi's sarcoma-associated herpesvirus, IL16 deficiency markedly induces Tyr705 STAT3 de-phosphorylation and elevates p21 expression, which can be counteracted by the tyrosine phosphatase inhibitor orthovanadate. Importantly, orthovanadate strongly blocks MHV68 lytic gene expression mediated by IL16 deficiency. These data demonstrate that virus-induced IL16 does not directly participate in MHV68 lytic replication, but rather inhibits virus reactivation to facilitate latent infection, in part through the STAT3-p21 axis.


Herpesviridae Infections/metabolism , Interleukin-16/metabolism , Tumor Virus Infections/metabolism , Virus Activation/physiology , Virus Latency/physiology , Animals , B-Lymphocytes/virology , Herpesviridae Infections/immunology , Interleukin-16/immunology , Lymphoma/virology , Mice , Rhadinovirus/immunology , Rhadinovirus/metabolism
7.
Immunol Cell Biol ; 98(4): 332-343, 2020 04.
Article En | MEDLINE | ID: mdl-31997396

Vaccination against γ-herpesviruses has proved difficult. CD4+ T cells are essential to contain infection, but how best to prime them and whether this can reduce viral loads remain unclear. To address these questions, we used ovalbumin (OVA) as a model antigen, delivering it with murine cytomegalovirus (MCMV) to protect mice against OVA-expressing murine herpesvirus-4 (MuHV-4). Membrane-associated OVA (mOVA) was more effective than soluble OVA, both to prime CD4+ T cells and as an effector target. It was also a better target than an OVA epitope limited to infected cells, suggesting that protective CD4+ T cells recognize infected cell debris rather than infected cells themselves. While MCMV-mOVA protected acutely against MuHV-4-mOVA, long-term protection was incomplete, even when OVA-specific CD8+ T cells and B cells were also primed. Thus, even optimized single-target vaccines may poorly reduce long-term γ-herpesvirus infections.


CD4-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Herpesvirus Vaccines/immunology , Immunogenicity, Vaccine/immunology , Ovalbumin/immunology , Rhadinovirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/prevention & control , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells , Rhadinovirus/genetics , Time Factors , Vaccination
8.
J Gen Virol ; 101(4): 420-425, 2020 04.
Article En | MEDLINE | ID: mdl-31985394

The γ-herpesviruses have proved hard to vaccination against, with no convincing protection against long-term latent infection by recombinant viral subunits. In experimental settings, whole-virus vaccines have proved more effective, even when the vaccine virus itself establishes latent infection poorly. The main alternative is replication-deficient virus particles. Here high-dose, replication-deficient murid herpesvirus-4 only protected mice partially against wild-type infection. By contrast, latency-deficient but replication-competent vaccine protected mice strongly, even when delivered non-invasively to the olfactory epithelium. Thus, this approach seems to provide the best chance of a safe and effective γ-herpesvirus vaccine.


Herpesviridae Infections/prevention & control , Rhadinovirus/immunology , Viral Vaccines , Animals , Antibodies, Viral/blood , Gammaherpesvirinae/immunology , Herpesviridae Infections/immunology , Immediate-Early Proteins/genetics , Mice , Mice, Inbred C57BL , Neutralization Tests , Trans-Activators/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Virion/immunology , Virus Latency/immunology , Virus Replication/genetics
9.
J Virol ; 94(2)2020 01 06.
Article En | MEDLINE | ID: mdl-31645449

A replication-competent, recombinant strain of rhesus monkey rhadinovirus (RRV) expressing the Gag protein of SIVmac239 was constructed in the context of a glycoprotein L (gL) deletion mutation. Deletion of gL detargets the virus from Eph family receptors. The ability of this gL-minus Gag recombinant RRV to infect, persist, and elicit immune responses was evaluated after intravenous inoculation of two Mamu-A*01+ RRV-naive rhesus monkeys. Both monkeys responded with an anti-RRV antibody response, and quantitation of RRV DNA in peripheral blood mononuclear cells (PBMC) by real-time PCR revealed levels similar to those in monkeys infected with recombinant gL+ RRV. Comparison of RRV DNA levels in sorted CD3+ versus CD20+ versus CD14+ PBMC subpopulations indicated infection of the CD20+ subpopulation by the gL-minus RRV. This contrasts with results obtained with transformed B cell lines in vitro, in which deletion of gL resulted in markedly reduced infectivity. Over a period of 20 weeks, Gag-specific CD8+ T cell responses were documented by major histocompatibility complex class I (MHC-I) tetramer staining. Vaccine-induced CD8+ T cell responses, which were predominantly directed against the Mamu-A*01-restricted Gag181-189CM9 epitope, could be inhibited by blockade of MHC-I presentation. Our results indicate that gL and the interaction with Eph family receptors are dispensable for the colonization of the B cell compartment following high-dose infection by the intravenous route, which suggests the existence of alternative receptors. Further, gL-minus RRV elicits cellular immune responses that are predominantly canonical in nature.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with a substantial disease burden in sub-Saharan Africa, often in the context of human immunodeficiency virus (HIV) infection. The related rhesus monkey rhadinovirus (RRV) has shown potential as a vector to immunize monkeys with antigens from simian immunodeficiency virus (SIV), the macaque model for HIV. KSHV and RRV engage cellular receptors from the Eph family via the viral gH/gL glycoprotein complex. We have now generated a recombinant RRV that expresses the SIV Gag antigen and does not express gL. This recombinant RRV was infectious by the intravenous route, established persistent infection in the B cell compartment, and elicited strong immune responses to the SIV Gag antigen. These results argue against a role for gL and Eph family receptors in B cell infection by RRV in vivo and have implications for the development of a live-attenuated KSHV vaccine or vaccine vector.


Gene Deletion , Gene Products, gag , Genetic Vectors , Herpesviridae Infections , Rhadinovirus , SAIDS Vaccines , Simian Immunodeficiency Virus , Animals , Antigens, CD/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Gene Products, gag/genetics , Gene Products, gag/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Humans , Macaca mulatta , Rhadinovirus/genetics , Rhadinovirus/immunology , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
10.
J Virol ; 94(1)2019 12 12.
Article En | MEDLINE | ID: mdl-31597758

Gammaherpesviruses are ubiquitous pathogens that establish lifelong infections in the majority of adults worldwide. Chronic gammaherpesvirus infection has been implicated in both lymphomagenesis and, somewhat controversially, autoimmune disease development. Pathogenesis is largely associated with the unique ability of gammaherpesviruses to usurp B cell differentiation, specifically, the germinal center response, to establish long-term latency in memory B cells. The host tyrosine phosphatase SHP1 is known as a brake on immune cell activation and is downregulated in several gammaherpesvirus-driven malignancies. However, here we demonstrate that B cell- but not T cell-intrinsic SHP1 expression supports the gammaherpesvirus-driven germinal center response and the establishment of viral latency. Furthermore, B cell-intrinsic SHP1 deficiency cooperated with gammaherpesvirus infection to increase the levels of double-stranded DNA-reactive antibodies at the peak of viral latency. Thus, in spite of decreased SHP1 levels in gammaherpesvirus-driven B cell lymphomas, B cell-intrinsic SHP1 expression plays a proviral role during the establishment of chronic infection, suggesting that the gammaherpesvirus-SHP1 interaction is more nuanced and is modified by the stage of infection and pathogenesis.IMPORTANCE Gammaherpesviruses establish lifelong infection in a majority of adults worldwide and are associated with a number of malignancies, including B cell lymphomas. These viruses infect naive B cells and manipulate B cell differentiation to achieve a lifelong infection of memory B cells. The germinal center stage of B cell differentiation is important as both an amplifier of the viral latent reservoir and the target of malignant transformation. In this study, we demonstrate that expression of tyrosine phosphatase SHP1, a negative regulator that normally limits the activation and proliferation of hematopoietic cells, enhances the gammaherpesvirus-driven germinal center response and the establishment of chronic infection. The results of this study uncover an intriguing beneficial interaction between gammaherpesviruses that are presumed to profit from B cell activation and a cellular phosphatase that is traditionally perceived to be a negative regulator of the same processes.


B-Lymphocytes/immunology , Germinal Center/immunology , Herpesviridae Infections/genetics , Host-Pathogen Interactions/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Rhadinovirus/genetics , Tumor Virus Infections/genetics , Animals , Antibodies, Antinuclear/biosynthesis , B-Lymphocytes/virology , Chronic Disease , DNA/genetics , DNA/immunology , Female , Germinal Center/virology , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Host-Pathogen Interactions/immunology , Humans , Immunologic Memory , Lymphocyte Activation , Male , Mice , Mice, Transgenic , Primary Cell Culture , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Rhadinovirus/immunology , Rhadinovirus/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Virus Latency/genetics , Virus Latency/immunology
11.
J Virol ; 93(22)2019 11 15.
Article En | MEDLINE | ID: mdl-31462575

Equine herpesvirus type 1 (EHV-1) outbreaks continue to occur despite widely used vaccination. Therefore, development of EHV-1 vaccines providing improved immunity and protection is ongoing. Here, an open reading frame 2 deletion mutant of the neuropathogenic EHV-1 strain Ab4 (Ab4ΔORF2) was tested as a vaccine candidate. Three groups of horses (n = 8 each) were infected intranasally with Ab4ΔORF2 or the parent Ab4 virus or were kept as noninfected controls. Horses infected with Ab4ΔORF2 had reduced fever and nasal virus shedding compared to those infected with Ab4 but mounted similar adaptive immunity dominated by antibody responses. Nine months after the initial infection, all horses were challenged intranasally with Ab4. Previously noninfected horses (control/Ab4) displayed clinical signs, shed large amounts of virus, and developed cell-associated viremia. In contrast, 5/8 or 3/8 horses previously infected with Ab4ΔORF2 or Ab4, respectively, were fully protected from challenge infection as indicated by the absence of fever, clinical disease, nasal virus shedding, and viremia. All of these outcomes were significantly reduced in the remaining, partially protected 3/8 (Ab4ΔORF2/Ab4) and 5/8 (Ab4/Ab4) horses. Protected horses had EHV-1-specific IgG4/7 antibodies prior to challenge infection, and intranasal antibodies increased rapidly postchallenge. Intranasal inflammatory markers were not detectable in protected horses but quickly increased in control/Ab4 horses during the first week after infection. Overall, our data suggest that preexisting nasal IgG4/7 antibodies neutralize EHV-1, prevent viral entry, and thereby protect from disease, viral shedding, and cell-associated viremia. In conclusion, improved protection from challenge infection emphasizes further evaluation of Ab4ΔORF2 as a vaccine candidate.IMPORTANCE Nasal equine herpesvirus type 1 (EHV-1) shedding is essential for virus transmission during outbreaks. Cell-associated viremia is a prerequisite for the most severe disease outcomes, abortion and equine herpesvirus myeloencephalopathy (EHM). Thus, protection from viremia is considered essential for preventing EHM. Ab4ΔORF2 vaccination prevented EHV-1 challenge virus replication in the upper respiratory tract in fully protected horses. Consequently, these neither shed virus nor developed cell-associated viremia. Protection from virus shedding and viremia during challenge infection in combination with reduced virulence at the time of vaccination emphasizes ORF2 deletion as a promising modification for generating an improved EHV-1 vaccine. During this challenge infection, full protection was linked to preexisting local and systemic EHV-1-specific antibodies combined with rapidly increasing intranasal IgG4/7 antibodies and lack of nasal type I interferon and chemokine induction. These host immune parameters may constitute markers of protection against EHV-1 and be utilized as indicators for improved vaccine development and informed vaccination strategies.


Herpesvirus 1, Equid/genetics , Herpesvirus 1, Equid/immunology , Herpesvirus Vaccines/immunology , Horse Diseases/virology , Administration, Intranasal/methods , Animals , Antibodies, Viral , Female , Herpesviridae Infections/virology , Herpesvirus 1, Equid/metabolism , Horses , Male , Nasal Mucosa/virology , Open Reading Frames , Rhadinovirus/immunology , Vaccination/veterinary , Viremia/immunology , Virulence , Virus Shedding/immunology
12.
J Virol ; 93(19)2019 10 01.
Article En | MEDLINE | ID: mdl-31315996

Autosomal dominant STAT1 mutations in humans have been associated with chronic mucocutaneous candidiasis (CMC), as well as with increased susceptibility to herpesvirus infections. Prior studies have focused on mucosal and Th17-mediated immunity against Candida, but mechanisms of impaired antiviral immunity have not previously been examined. To begin to explore the mechanisms of STAT1-associated immunodeficiency against herpesviruses, we generated heterozygous STAT1 R274W knock-in mice that have a frequently reported STAT1 mutation associated in humans with susceptibility to herpesvirus infections. In primary macrophages and fibroblasts, we found that STAT1 R274W had no appreciable effect on cell-intrinsic immunity against herpes simplex virus 1 (HSV-1) or gammaherpesvirus 68 (γHV68) infection. However, intraperitoneal inoculation of mice with γHV68 was associated with impaired control of infection at day 14 in STAT1 R274W mice compared with that in wild-type (WT) littermate control animals. Infection of STAT1 R274W mice was associated with paradoxically decreased expression of IFN-stimulated genes (ISGs) and gamma interferon (IFN-γ), likely secondary to defective CD4+ and CD8+ T cell responses, including diminished numbers of antigen-specific CD8+ T cells. Viral pathogenesis studies in WT and STAT1 R274W mixed bone marrow chimeric mice revealed that the presence of WT leukocytes was sufficient to limit infection and that antigen-specific STAT1 R274W CD8+ T cell responses were impaired even in the presence of WT leukocytes. Thus, in addition to regulating Th17 responses against Candida, a STAT1 gain-of-function mutant impedes antigen-specific T cell responses against a common gammaherpesvirus in mice.IMPORTANCE Mechanisms of immunodeficiency related to STAT1 gain of function have not been previously studied in an animal model of viral pathogenesis. Using virological and immunological techniques, we examined the immune response to γHV68 in heterozygous mice that have an autosomal dominant mutation in the STAT1 coiled-coil domain (STAT1 R274W). We observed impaired control of infection, which was associated with diminished production of gamma interferon (IFN-γ), fewer effector CD4+ and CD8+ T cells, and a reduction in the number of antigen-specific CD8+ T cells. These findings indicate that a STAT1 gain-of-function mutation limits production of antiviral T cells, likely contributing to immunodeficiency against herpesviruses.


CD8-Positive T-Lymphocytes/immunology , Gain of Function Mutation , Herpesviridae Infections/immunology , Mutation, Missense , Rhadinovirus/immunology , STAT1 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Fibroblasts/immunology , Fibroblasts/virology , Gene Knock-In Techniques , Interferon-gamma/metabolism , Macrophages/immunology , Macrophages/virology , Mice , STAT1 Transcription Factor/genetics
13.
Eur J Immunol ; 49(2): 351-352, 2019 02.
Article En | MEDLINE | ID: mdl-30488950

We show that latently gammaherpesvirus-infected B cells are present in the thymus. This could result in a functional T-cell tolerance against certain viral epitopes. It is conceivable that also antigens from other viruses or pathogens may be conveyed to the thymus for their immune evasion.


B-Lymphocytes/immunology , Herpesviridae Infections/immunology , Immune Evasion , Immune Tolerance , Rhadinovirus/immunology , Thymus Gland/immunology , Animals , B-Lymphocytes/pathology , Herpesviridae Infections/pathology , Mice , Thymus Gland/pathology
14.
Methods Mol Biol ; 1826: 157-182, 2018.
Article En | MEDLINE | ID: mdl-30194600

Serpins have a wide range of functions in regulation of serine proteases in the thrombotic cascade and in immune responses, representing up to 2-10% of circulating proteins in the blood. Selected serpins also have cross-class inhibitory actions for cysteine proteases in inflammasome and apoptosis pathways. The arterial and venous systems transport blood throughout the mammalian body representing a central site for interactions between coagulation proteases and circulating blood cells (immune cells) and target tissues, a very extensive and complex interaction. While analysis of serpin functions in vitro in kinetics or gel shift assays or in tissue culture provides very necessary information on molecular mechanisms, the penultimate assessment of biological or physiological functions and efficacy for serpins as therapeutics requires study in vivo in whole animal models (some also consider cell culture to be an in vivo approach).Mouse models of arterial transplant with immune rejection as well as models of inflammatory vasculitis induced by infection have been used to study the interplay between the coagulation and immune response pathways. We describe here three in vivo vasculitis models that are used to study the roles of serpins in disease and as therapeutics. The models described include (1) mouse aortic allograft transplantation, (2) human temporal artery (TA) xenograft into immunodeficient mouse aorta, and (3) mouse herpes virus (MHV68)-induced inflammatory vasculitis in interferon-gamma receptor (IFNγR) knockout mice.


Aortitis , Arteritis , Herpesviridae Infections , Rhadinovirus , Serpins , Allografts , Animals , Aorta/immunology , Aorta/pathology , Aorta/transplantation , Aortitis/genetics , Aortitis/immunology , Aortitis/pathology , Arteritis/genetics , Arteritis/immunology , Arteritis/pathology , Disease Models, Animal , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Transplantation , Rhadinovirus/genetics , Rhadinovirus/immunology , Serpins/genetics , Serpins/immunology , Temporal Arteries/immunology , Temporal Arteries/pathology , Temporal Arteries/transplantation
15.
J Virol ; 92(19)2018 10 01.
Article En | MEDLINE | ID: mdl-29997213

Like many other large double-stranded DNA (dsDNA) viruses, herpesviruses are known to capture host genes to evade host defenses. Little is known about the detailed natural history of such genes, nor do we fully understand their evolutionary dynamics. A major obstacle is that they are often highly divergent, maintaining very low sequence similarity to host homologs. Here we use the herpesvirus genus Rhadinovirus as a model system to develop an analytical approach that combines complementary evolutionary and bioinformatic techniques, offering results that are both detailed and robust for a range of genes. Using a systematic phylogenetic strategy, we identify the original host lineage of viral genes with high confidence. We show that although host immunomodulatory genes evolve rapidly compared to other host genes, they undergo a clear increase in purifying selection once captured by a virus. To characterize this shift in detail, we developed a novel technique to identify changes in selection pressure that can be attributable to particular domains. These findings will inform us on how viruses develop strategies to evade the immune system, and our synthesis of techniques can be reapplied to other viruses or biological systems with similar analytical challenges.IMPORTANCE Viruses and hosts have been shown to capture genes from one another as part of the evolutionary arms race. Such genes offer a natural experiment on the effects of evolutionary pressure, since the same gene exists in vastly different selective environments. However, sequences of viral homologs often bear little similarity to the original sequence, complicating the reconstruction of their shared evolutionary history with host counterparts. In this study, we use a genus of herpesviruses as a model system to comprehensively investigate the evolution of host-derived viral genes, using a synthesis of genomics, phylogenetics, selection analysis, and nucleotide and amino acid modeling.


Genes, Viral/immunology , Histocompatibility Antigens Class I/genetics , Host-Pathogen Interactions , Rhadinovirus/genetics , Selection, Genetic , Viral Proteins/genetics , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/immunology , Atelinae/virology , Biological Evolution , CD59 Antigens/chemistry , CD59 Antigens/genetics , CD59 Antigens/immunology , Callithrix/virology , Chemokine CCL3/chemistry , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Computational Biology , Gene Expression Regulation , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Interleukin-17/chemistry , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Models, Molecular , Phylogeny , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Rats , Rhadinovirus/chemistry , Rhadinovirus/immunology , Saimiri/virology , Viral Proteins/chemistry , Viral Proteins/immunology
16.
J Immunol ; 200(8): 2703-2713, 2018 04 15.
Article En | MEDLINE | ID: mdl-29500240

IL-27 is an immunoregulatory cytokine consisting of p28 and EBI3. Its receptor also has two subunits, WSX1 and gp130. Although IL-27 promotes Th1 differentiation in naive T cells, it also induces IL-10 expression in effector Th1 cells to curtail excessive immune responses. By using p28-deficient mice and WSX1-deficient mice (collectively called IL-27-deficient mice), we examined the role of IL-27 in primary infection by murine γ-herpesvirus 68 (MHV68), a murine model of EBV. Upon airway infection with MHV68, IL-27-deficient mice had more aggravated lung inflammation than wild-type mice, although MHV68 infection per se was better controlled in IL-27-deficient mice. Although epithelial cells and alveolar macrophages were primarily infected by MHV68, interstitial macrophages and dendritic cells were the major producers of IL-27. The lung inflammation of IL-27-deficient mice was characterized by more IFN-γ-producing CD8+ T cells and fewer IL-10-producing CD8+ T cells than that of wild-type mice. An infectious mononucleosis-like disease was also aggravated in IL-27-deficient mice, with prominent splenomegaly and severe hepatitis. Infiltration of IFN-γ-producing effector cells and upregulation of the CXCR3 ligand chemokines CXCL9, CXCL10, and CXCL11 were noted in the liver of MHV68-infected mice. Oral neomycin effectively ameliorated hepatitis, with decreased production of these chemokines in the liver, suggesting that the intestinal microbiota plays a role in liver inflammation through upregulation of these chemokines. Collectively, IL-27 is essential for the generation of IL-10-producing effector cells in primary infection by MHV68. Our findings may also provide new insight into the mechanism of hepatitis associated with infectious mononucleosis.


Interleukins/immunology , Liver Diseases/drug therapy , Neomycin/pharmacology , Pneumonia/immunology , Pneumonia/virology , Rhadinovirus/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chemokines/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Interferon-gamma/immunology , Liver Diseases/immunology , Liver Diseases/virology , Mice , Mice, Inbred C57BL , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
17.
Mucosal Immunol ; 11(3): 881-893, 2018 05.
Article En | MEDLINE | ID: mdl-29044226

Idiopathic pneumonia syndrome (IPS) is a common, often fatal, complication following hematopoietic stem cell transplantation (HSCT) characterized by severe pneumonitis and interstitial fibrosis. Fully reconstituted syngeneic bone marrow transplant (BMT) mice infected with murine γ-herpesvirus-68 develop interleukin-17 (IL-17)-driven pneumonitis and fibrosis, which mimics clinical manifestations of IPS. We found CD103+ and CD11b+ dendritic cells (DCs) are selectively deficient for the Notch ligand, DLL4, following BMT and CD4+ T cells isolated from lungs and spleens of infected BMT mice display Notch signaling defects. Mice transplanted with CD4-Cre-driven dominant-negative Notch transcriptional regulator Mastermind-Like (CD4-Cre-DNMAML (CCD) mice) bone marrow displayed elevated IL-17 and transforming growth factor-ß (TGF ß) in the lungs, a further expansion of T-helper type 17 (Th17) cells, and developed more fibrosis than wild-type (WT)-BMT mice. Culture of BMT lung leukocytes with recombinant Notch ligand, DLL4, restored Notch signaling and decreased production of IL-17. Adoptive transfer of CD11c+ DCs could restore Th1 and limit Th17 in WT-BMT but not CCD-BMT mice, indicating that a specific DC/CD4+ T-cell Notch interaction modulates IL-17 production following reconstitution in syngeneic BMT mice. Given recent clinical observations showing that patients with pulmonary complications post-transplant harbor occult herpesvirus infections, these data provide mechanistic insight and suggest potential therapies for these devastating conditions.


Dendritic Cells/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Herpesviridae Infections/immunology , Interleukin-17/metabolism , Lung/pathology , Pneumonia/immunology , Postoperative Complications/immunology , Rhadinovirus/immunology , Th17 Cells/immunology , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cells, Cultured , Fibrosis , Herpesviridae Infections/etiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/virology , Lymphocyte Activation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/etiology , Pneumonia/virology , Postoperative Complications/virology , Receptors, Notch/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
18.
Nat Immunol ; 18(12): 1310-1320, 2017 Dec.
Article En | MEDLINE | ID: mdl-29035391

The hygiene hypothesis postulates that the recent increase in allergic diseases such as asthma and hay fever observed in Western countries is linked to reduced exposure to childhood infections. Here we investigated how infection with a gammaherpesvirus affected the subsequent development of allergic asthma. We found that murid herpesvirus 4 (MuHV-4) inhibited the development of house dust mite (HDM)-induced experimental asthma by modulating lung innate immune cells. Specifically, infection with MuHV-4 caused the replacement of resident alveolar macrophages (AMs) by monocytes with regulatory functions. Monocyte-derived AMs blocked the ability of dendritic cells to trigger a HDM-specific response by the TH2 subset of helper T cells. Our results indicate that replacement of embryonic AMs by regulatory monocytes is a major mechanism underlying the long-term training of lung immunity after infection.


Asthma/therapy , Macrophages, Alveolar/immunology , Monocytes/immunology , Pyroglyphidae/immunology , Rhadinovirus/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Asthma/immunology , Cell Line , Cricetinae , Dendritic Cells/immunology , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Macrophages, Alveolar/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Th2 Cells/transplantation
19.
J Virol ; 91(23)2017 12 01.
Article En | MEDLINE | ID: mdl-28904198

Murid herpesvirus 4 (MuHV-4) is a B cell-tropic gammaherpesvirus that can be studied in vivo Despite viral evasion, type I interferons (IFN-I) limit its spread. After MuHV-4 inoculation into footpads, IFN-I protect lymph node subcapsular sinus macrophages (SSM) against productive infection; after peritoneal inoculation, they protect splenic marginal zone macrophages, and they limit MuHV-4 replication in the lungs. While invasive infections can be used to test specific aspects of host colonization, it is also important to understand natural infection. MuHV-4 taken up spontaneously by alert mice enters them via olfactory neurons. We determined how IFN-I act in this context. Blocking IFN-I signaling did not increase neuronal infection but allowed the virus to spread to the adjacent respiratory epithelium. In lymph nodes, a complete IFN-I signaling block increased MuHV-4 lytic infection in SSM and increased the number of dendritic cells (DC) expressing viral green fluorescent protein (GFP) independently of lytic infection. A CD11c+ cell-directed signaling block increased infection of DC only. However, this was sufficient to increase downstream infection, consistent with DC providing the main viral route to B cells. The capacity of IFN-I to limit DC infection indicated that viral IFN-I evasion was only partly effective. Therefore, DC are a possible target for IFN-I-based interventions to reduce host colonization.IMPORTANCE Human gammaherpesviruses infect B cells and cause B cell cancers. Interventions to block virus binding to B cells have not stopped their infection. Therefore, we must identify other control points that are relevant to natural infection. Human infections are difficult to analyze. However, gammaherpesviruses colonize all mammals. A related gammaherpesvirus of mice reaches B cells not directly but via infected dendritic cells. We show that type I interferons, an important general antiviral defense, limit gammaherpesvirus B cell infection by acting on dendritic cells. Therefore, dendritic cell infection is a potential point of interferon-based therapeutic intervention.


Dendritic Cells/immunology , Interferon Type I/metabolism , Olfactory Mucosa/virology , Rhadinovirus/physiology , Signal Transduction , Animals , B-Lymphocytes/virology , Dendritic Cells/virology , Green Fluorescent Proteins , Herpesviridae Infections/virology , Immune Evasion , Immunity, Innate , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Lymph Nodes/virology , Macrophages/virology , Mice , Respiratory System/immunology , Respiratory System/virology , Rhadinovirus/immunology
20.
PLoS Pathog ; 13(8): e1006543, 2017 Aug.
Article En | MEDLINE | ID: mdl-28767707

Murine gammaherpesvirus 68 (MHV68) infection of laboratory strains of mice has provided a tractable small animal model for dissecting gammaherpesvirus pathogenesis. The MHV68 latency associated antigen M2 promotes viral latency establishment in germinal center (GC) B cells and plays an important role in virus infection of plasma cells (PCs), which is linked to virus reactivation. More recently, M2 has been highlighted as a potent immunomodulatory molecule capable of hindering both cell-mediated and humoral immunity to MHV68 infection and subsequent challenges. M2 expression in B cells results in activation of B cell receptor signaling pathways that promote proliferation, differentiation, and cytokine production-a hallmark of gammaherpesviruses. In this study, we utilized an adoptive transfer model to explore the biological consequence of M2 expression in activated B cells in vivo. Secondly, we engineered and validated two independent MHV68 M2 reporter viruses that track M2 protein expression in latently infected B cells during infection. Here we demonstrate that upon adoptive transfer into naive mice, M2 expression promotes activated primary B cells to competitively establish residency in the spleen as either a GC B cell or a PC, most notably in the absence of an ongoing GC reaction. Moreover, M2 antigen drives robust PC differentiation and IL10 production in vivo in the absence of other viral factors. Lastly, we confirm that M2 expression during MHV68 infection is localized to the GC compartment, which is a long term latency reservoir for gammaherpesviruses. Overall, these observations are consistent with, and extend upon previous reports of M2 function in B cells and within the context of MHV68 infection. Moreover, this work provides support for a model by which M2-driven dysregulation of B cell function compromises multiple aspects of antiviral immunity to achieve persistence within the infected host.


B-Lymphocytes/immunology , B-Lymphocytes/virology , Herpesviridae Infections/immunology , Lymphocyte Activation/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Disease Models, Animal , Flow Cytometry , Germinal Center/immunology , Mice , Mice, Inbred C57BL , Rhadinovirus/immunology , Spleen/immunology , Viral Proteins , Virus Latency/immunology
...