Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.146
1.
J Clin Invest ; 134(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38690731

Herpesviruses establish latent infections, and most reactivate frequently, resulting in symptoms and virus shedding in healthy individuals. In immunocompromised patients, reactivating virus can cause severe disease. Persistent EBV has been associated with several malignancies in both immunocompromised and nonimmunocompromised persons. Reactivation and shedding occur with most herpesviruses, despite potent virus-specific antibodies and T cell immunity as measured in the blood. The licensure of therapeutic vaccines to reduce zoster indicates that effective therapeutic vaccines for other herpesviruses should be feasible. However, varicella-zoster virus is different from other human herpesviruses in that it is generally only shed during varicella and zoster. Unlike prophylactic vaccines, in which the correlate of immunity is antibody function, T cell immunity is the correlate of immunity for the only effective therapeutic herpesvirus vaccine-zoster vaccine. While most studies of therapeutic vaccines have measured immunity in the blood, cellular immunity at the site of reactivation is likely critical for an effective therapeutic vaccine for certain viruses. This Review summarizes the status of therapeutic vaccines for herpes simplex virus, cytomegalovirus, and Epstein-Barr virus and proposes approaches for future development.


Herpesvirus Vaccines , Humans , Herpesvirus Vaccines/immunology , Herpesvirus Vaccines/therapeutic use , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Herpesvirus 4, Human/immunology , Animals , Herpesviridae/immunology , Virus Activation/immunology , Cytomegalovirus/immunology
2.
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
3.
J Virol ; 98(5): e0198623, 2024 May 14.
Article En | MEDLINE | ID: mdl-38619272

Human cytomegalovirus (hCMV) is a ubiquitous facultative pathogen, which establishes a characteristic latent and reactivating lifelong infection in immunocompetent hosts. Murine CMV (mCMV) infection is widely used as an experimental model of hCMV infection, employed to investigate the causal nature and extent of CMV's contribution to inflammatory, immunological, and health disturbances in humans. Therefore, mimicking natural human infection in mice would be advantageous to hCMV research. To assess the role of route and age at infection in modeling hCMV in mice, we infected prepubescent and young sexually mature C57BL/6 (B6) mice intranasally (i.n., a likely physiological route in humans) and intraperitoneally (i.p., a frequently used experimental route, possibly akin to transplant-mediated infection). In our hands, both routes led to comparable early viral loads and tissue spreads. However, they yielded differential profiles of innate and adaptive systemic immune activation. Specifically, the younger, prepubescent mice exhibited the strongest natural killer cell activation in the blood in response to i.p. infection. Further, the i.p. infected animals (particularly those infected at 12 weeks) exhibited larger anti-mCMV IgG and greater expansion of circulating CD8+ T cells specific for both acute (non-inflationary) and latent phase (inflationary) mCMV epitopes. By contrast, tissue immune responses were comparable between i.n. and i.p. groups. Our results illustrate a distinction in the bloodborne immune response profiles across infection routes and ages and are discussed in light of physiological parameters of interaction between CMV, immunity, inflammation, and health over the lifespan. IMPORTANCE: The majority of experiments modeling human cytomegalovirus (hCMV) infection in mice have been carried out using intraperitoneal infection in sexually mature adult mice, which stands in contrast to the large number of humans being infected with human CMV at a young age, most likely via bodily fluids through the nasopharyngeal/oral route. This study examined the impact of the choice of age and route of infection in modeling CMV infection in mice. By comparing young, prepubescent to older sexually mature counterparts, infected either via the intranasal or intraperitoneal route, we discovered substantial differences in deployment and response intensity of different arms of the immune system in systemic control of the virus; tissue responses, by contrast, appeared similar between ages and infection routes.


Adaptive Immunity , Cytomegalovirus Infections , Disease Models, Animal , Immunity, Innate , Mice, Inbred C57BL , Muromegalovirus , Viral Load , Animals , Mice , Muromegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Age Factors , Killer Cells, Natural/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Female , Humans
4.
Viruses ; 16(4)2024 Apr 16.
Article En | MEDLINE | ID: mdl-38675960

Reactivation and infection with cytomegalovirus (CMV) are frequently observed in recipients of solid organ transplants, bone marrow transplants, and individuals with HIV infection. This presents an increasing risk of allograft rejection, opportunistic infection, graft failure, and patient mortality. Among immunocompromised hosts, interstitial pneumonia is the most critical clinical manifestation of CMV infection. Recent studies have demonstrated the potential therapeutic benefits of exosomes derived from mesenchymal stem cells (MSC-exos) in preclinical models of acute lung injury, including pneumonia, ARDS, and sepsis. However, the role of MSC-exos in the pathogenesis of infectious viral diseases, such as CMV pneumonia, remains unclear. In a mouse model of murine CMV-induced pneumonia, we observed that intravenous administration of mouse MSC (mMSC)-exos reduced lung damage, decreased the hyperinflammatory response, and shifted macrophage polarization from the M1 to the M2 phenotype. Treatment with mMSC-exos also significantly reduced the infiltration of inflammatory cells and pulmonary fibrosis. Furthermore, in vitro studies revealed that mMSC-exos reversed the hyperinflammatory phenotype of bone marrow-derived macrophages infected with murine CMV. Mechanistically, mMSC-exos treatment decreased activation of the NF-κB/NLRP3 signaling pathway both in vivo and in vitro. In summary, our findings indicate that mMSC-exo treatment is effective in severe CMV pneumonia by reducing lung inflammation and fibrosis through the NF-κB/NLRP3 signaling pathway, thus providing promising therapeutic potential for clinical CMV infection.


Disease Models, Animal , Exosomes , Mesenchymal Stem Cells , Muromegalovirus , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Animals , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Muromegalovirus/physiology , Mice, Inbred C57BL , Macrophages/immunology , Cytomegalovirus Infections/therapy , Cytomegalovirus Infections/virology , Lung/virology , Lung/pathology , Pneumonia, Viral/therapy , Pneumonia, Viral/virology , Herpesviridae Infections/therapy , Herpesviridae Infections/virology , Herpesviridae Infections/immunology , Pneumonia/therapy , Pneumonia/virology
5.
J Exp Med ; 221(7)2024 Jul 01.
Article En | MEDLINE | ID: mdl-38661718

Chemokines guide immune cells during their response against pathogens and tumors. Various techniques exist to determine chemokine production, but none to identify cells that directly sense chemokines in vivo. We have generated CCL3-EASER (ErAse, SEnd, Receive) mice that simultaneously report for Ccl3 transcription and translation, allow identifying Ccl3-sensing cells, and permit inducible deletion of Ccl3-producing cells. We infected these mice with murine cytomegalovirus (mCMV), where Ccl3 and NK cells are critical defense mediators. We found that NK cells transcribed Ccl3 already in homeostasis, but Ccl3 translation required type I interferon signaling in infected organs during early infection. NK cells were both the principal Ccl3 producers and sensors of Ccl3, indicating auto/paracrine communication that amplified NK cell response, and this was essential for the early defense against mCMV. CCL3-EASER mice represent the prototype of a new class of dual fluorescence reporter mice for analyzing cellular communication via chemokines, which may be applied also to other chemokines and disease models.


Cell Communication , Chemokine CCL3 , Killer Cells, Natural , Muromegalovirus , Protein Biosynthesis , Transcription, Genetic , Animals , Mice , Muromegalovirus/physiology , Chemokine CCL3/metabolism , Chemokine CCL3/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Genes, Reporter , Mice, Inbred C57BL , Herpesviridae Infections/immunology , Herpesviridae Infections/genetics , Mice, Transgenic , Interferon Type I/metabolism , Signal Transduction
6.
J Neurovirol ; 30(1): 86-99, 2024 Feb.
Article En | MEDLINE | ID: mdl-38453879

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.


Amyloid beta-Peptides , Cytokines , Macaca mulatta , Animals , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/blood , Cytokines/cerebrospinal fluid , Cytokines/blood , Virus Activation , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/blood , Varicellovirus/genetics , Varicellovirus/immunology , Herpesvirus 3, Human/pathogenicity , Herpesvirus 3, Human/immunology , Herpesviridae Infections/cerebrospinal fluid , Herpesviridae Infections/virology , Herpesviridae Infections/blood , Herpesviridae Infections/immunology , Male , Herpes Zoster/cerebrospinal fluid , Herpes Zoster/virology , Herpes Zoster/blood , Herpes Zoster/immunology , Monkey Diseases/virology , Monkey Diseases/cerebrospinal fluid , Monkey Diseases/blood
7.
Am J Vet Res ; 85(5)2024 May 01.
Article En | MEDLINE | ID: mdl-38457927

OBJECTIVE: Compare immune responses induced by 2 commercial intranasal (IN) modified-live viral (MLV) vaccines given individually or coadministered and evaluate prevention of infection and lung pathology following bovine herpesvirus-1 (BHV-1) challenge. ANIMALS: 36 male Holstein calves (ages, 5 to 12 days). METHODS: In a randomized complete block design, each calf received an IN injection of either vaccine diluent (Placebo), an MLV vaccine containing bovine herpesvirus-1 (BHV-1; N3), bovine coronavirus vaccine (BC), or both N3 and BC (BC + N3) with a booster 4 weeks later. Nasal secretions and blood were collected weekly. Three weeks after the booster, the calves were challenged with BHV-1, sampled for virus shedding, and euthanized 10 days later to quantify lung pathology. The study period was September 7, 2020, to April 6, 2021. RESULTS: Calves were seropositive for BHV-1 and BC before vaccination. No significant difference in BC-specific serum immunoglobin G and nasal immunoglobin A antibody responses in the BC versus BC + N3 group or BHV-1-specific serum immunoglobin G and nasal immunoglobin A antibody responses in the N3 versus BC + N3 group. Cytokine responses to BHV-1 and BC did not differ among groups. BHV-1 shedding after challenge was significantly reduced in N3 groups versus Placebo and BC. There was a significant reduction in lung pathology in the N3 + BC group versus Placebo. CLINICAL RELEVANCE: This study provides evidence an MLV vaccine containing BHV-1 and an MLV BC vaccine can be coadministered to neonatal calves without significantly altering immune responses to the 2 viruses or compromising the prevention of BHV-1 respiratory disease. Calves receiving the BC + N3 vaccine had a significant reduction in lung pathology after BHV-1 aerosol challenge.


Administration, Intranasal , Animals, Newborn , Cattle Diseases , Coronavirus Infections , Coronavirus, Bovine , Herpesviridae Infections , Herpesvirus 1, Bovine , Vaccines, Attenuated , Viral Vaccines , Animals , Cattle , Herpesvirus 1, Bovine/immunology , Administration, Intranasal/veterinary , Male , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Coronavirus, Bovine/immunology , Cattle Diseases/prevention & control , Cattle Diseases/virology , Cattle Diseases/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Coronavirus Infections/virology , Herpesviridae Infections/veterinary , Herpesviridae Infections/prevention & control , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Infectious Bovine Rhinotracheitis/prevention & control , Infectious Bovine Rhinotracheitis/immunology , Virus Shedding , Antibodies, Viral/blood , Random Allocation
8.
J Virol ; 97(11): e0132223, 2023 Nov 30.
Article En | MEDLINE | ID: mdl-37882519

IMPORTANCE: Chickens immunized with the infectious laryngotracheitis chicken embryo origin (CEO) vaccine (Medivac, PT Medion Farma Jaya) experience adverse reactions, hindering its safety and effective use in poultry flocks. To improve the effect of the vaccine, we sought to find a strategy to alleviate the respiratory reactions associated with the vaccine. Here, we confirmed that co-administering the CEO vaccine with chIL-2 by oral delivery led to significant alleviation of the vaccine reactions in chickens after immunization. Furthermore, we found that the co-administration of chIL-2 with the CEO vaccine reduced the clinical signs of the CEO vaccine while enhancing natural killer cells and cytotoxic T lymphocyte response to decrease viral loads in their tissues, particularly in the trachea and conjunctiva. Importantly, we demonstrated that the chIL-2 treatment can ameliorate the replication of the CEO vaccine without compromising its effectiveness. This study provides new insights into further applications of chIL-2 and a promising strategy for alleviating the adverse reaction of vaccines.


Chickens , Herpesviridae Infections , Herpesvirus 1, Gallid , Interleukin-2 , Killer Cells, Natural , T-Lymphocytes, Cytotoxic , Viral Vaccines , Animals , Administration, Oral , Chickens/immunology , Chickens/virology , Conjunctiva/virology , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/immunology , Interleukin-2/administration & dosage , Interleukin-2/immunology , Killer Cells, Natural/immunology , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/prevention & control , Respiratory Tract Diseases/veterinary , Respiratory Tract Diseases/virology , T-Lymphocytes, Cytotoxic/immunology , Trachea/virology , Viral Load , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Viral Vaccines/biosynthesis , Viral Vaccines/immunology
9.
Front Immunol ; 14: 1235590, 2023.
Article En | MEDLINE | ID: mdl-37600809

The incidence of human herpesvirus (HHVs) is gradually increasing and has affected a wide range of population. HHVs can result in serious consequences such as tumors, neonatal malformations, sexually transmitted diseases, as well as pose an immense threat to the human health. The cGAS-STING pathway is one of the innate immune pattern-recognition receptors discovered recently. This article discusses the role of the cGAS-STING pathway in human diseases, especially in human herpesvirus infections, as well as highlights how these viruses act on this pathway to evade the host immunity. Moreover, the author provides a comprehensive overview of modulators of the cGAS-STING pathway. By focusing on the small molecule compounds based on the cGAS-STING pathway, novel targets and concepts have been proposed for the development of antiviral drugs and vaccines, while also providing a reference for the investigation of disease models related to the cGAS-STING pathway. HHV is a double-stranded DNA virus that can trigger the activation of intracellular DNA sensor cGAS, after which the host cells initiate a cascade of reactions that culminate in the secretion of type I interferon to restrict the viral replication. Meanwhile, the viral protein can interact with various molecules in the cGAS-STING pathway. Viruses can evade immune surveillance and maintain their replication by inhibiting the enzyme activity of cGAS and reducing the phosphorylation levels of STING, TBK1 and IRF3 and suppressing the interferon gene activation. Activators and inhibitors of the cGAS-STING pathway have yielded numerous promising research findings in vitro and in vivo pertaining to cGAS/STING-related disease models. However, there remains a dearth of small molecule modulators that have been successfully translated into clinical applications, which serves as a hurdle to be overcome in the future.


Herpesviridae Infections , Immunity, Innate , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Humans , Chromogranin A , Herpesviridae Infections/immunology , Phosphorylation
10.
Virology ; 576: 134-140, 2022 Nov.
Article En | MEDLINE | ID: mdl-36244319

Gammaherpesviruses establish life-long infection in over 95% of adults and are associated with several cancers, including B cell lymphomas. Using the murine gammaherpesvirus 68 (MHV68) animal model, we previously showed a pro-viral role of Interleukin-1 (IL-1) signaling that supported viral reactivation during the establishment of chronic infection. Unexpectedly, in this study we found that the proviral effects of IL-1 signaling originally observed during the establishment of chronic gammaherpesvirus infection convert to antiviral effects during the long-term stage of infection. Specifically, IL-1 signaling promoted expansion of antiviral CD8+ T cells and control of viral reactivation in the peritoneal cavity of a long-term infected host. Using a novel mouse model of T cell-specific IL-1 signaling deficiency, we found that the antiviral effects of IL-1 signaling were T cell extrinsic. Our study highlights a dynamic nature of host factors that shape the parameters of chronic gammaherpesvirus infection.


Gammaherpesvirinae , Herpesviridae Infections , Interleukin-1 , Animals , Mice , Antiviral Agents , B-Lymphocytes , CD8-Positive T-Lymphocytes/pathology , Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Interleukin-1/immunology , Interleukin-1/metabolism , Mice, Inbred C57BL , Virus Latency
11.
Viruses ; 14(10)2022 10 19.
Article En | MEDLINE | ID: mdl-36298850

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with vascular endothelial cell tumor, Kaposi's sarcoma (KS) and lymphoproliferative disorder, multicentric Castleman's disease (MCD), primary effusion lymphoma (PEL) and KSHV inflammatory cytokine syndrome (KICS). Dysregulation of proinflammatory cytokines is found in most KSHV associated diseases. However, little is known about the role of host microenvironment in the regulation of KSHV establishment in B cells. In the present study, we demonstrated that IFN-γ has a strong inhibitory effect on KSHV infection but only in a subset of tonsil-derived lymphocyte samples that are intrinsically more susceptible to infection, contain higher proportions of naïve B cells, and display increased levels of IRF1 and STAT1-pY701. The effect of IFN-γ in responsive samples was associated with increased frequencies of germinal center B cells (GCB) and decreased infection of plasma cells, suggesting that IFN-γ-mediated modulation of viral dynamics in GC can inhibit the establishment of KSHV infection.


B-Lymphocytes , Herpesviridae Infections , Herpesvirus 8, Human , Interferon-gamma , Humans , Acquired Immunodeficiency Syndrome/immunology , B-Lymphocytes/immunology , Castleman Disease/immunology , Cytokines/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology , Interferon-gamma/immunology , Sarcoma, Kaposi/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Disease Susceptibility/immunology
12.
Proc Natl Acad Sci U S A ; 119(32): e2123362119, 2022 08 09.
Article En | MEDLINE | ID: mdl-35921433

The germinal center (GC) plays a central role in the generation of antigen-specific B cells and antibodies. Tight regulation of the GC is essential due to the inherent risks of tumorigenesis and autoimmunity posed by inappropriate GC B cell processes. Gammaherpesviruses such as Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) utilize numerous armaments to drive infected naïve B cells, independent of antigen, through GC reactions to expand the latently infected B cell population and establish a stable latency reservoir. We previously demonstrated that the MHV68 microRNA (miRNA) mghv-miR-M1-7-5p represses host EWSR1 (Ewing sarcoma breakpoint region 1) to promote B cell infection. EWSR1 is a transcription and splicing regulator that is recognized for its involvement as a fusion protein in Ewing sarcoma. A function for EWSR1 in B cell responses has not been previously reported. Here, we demonstrate that 1) B cell-specific deletion of EWSR1 had no effect on generation of mature B cell subsets or basal immunoglobulin levels in naïve mice, 2) repression or ablation of EWSR1 in B cells promoted expansion of MHV68 latently infected GC B cells, and 3) B cell-specific deletion of EWSR1 during a normal immune response to nonviral antigen resulted in significantly elevated numbers of antigen-specific GC B cells, plasma cells, and circulating antibodies. Notably, EWSR1 deficiency did not affect the proliferation or survival of GC B cells but instead resulted in the generation of increased numbers of precursor GC B cells. Cumulatively, these findings demonstrate that EWSR1 is a negative regulator of B cell responses.


B-Lymphocytes , Gammaherpesvirinae , Germinal Center , Herpesviridae Infections , MicroRNAs , RNA-Binding Protein EWS , Tumor Virus Infections , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , Gammaherpesvirinae/genetics , Gammaherpesvirinae/physiology , Gene Deletion , Germinal Center/immunology , Germinal Center/virology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Virus Latency
13.
Front Immunol ; 13: 787021, 2022.
Article En | MEDLINE | ID: mdl-35173716

Vaccination is the best form of protecting fish against viral diseases when the pathogen cannot be contained by biosecurity measures. Vaccines based on live attenuated viruses seem to be most effective for vaccination against challenging pathogens like Cyprinid herpesvirus 3. However, there are still knowledge gaps how these vaccines effectively protect fish from the deadly disease caused by the epitheliotropic CyHV-3, and which aspects of non-direct protection of skin or gill integrity and function are important in the aquatic environment. To elucidate some elements of protection, common carp were vaccinated against CyHV-3 using a double deletion vaccine virus KHV-T ΔDUT/TK in the absence or presence of a mix of common carp beta-defensins 1, 2 and 3 as adjuvants. Vaccination induced marginal clinical signs, low virus load and a minor upregulation of cd4, cd8 and igm gene expression in vaccinated fish, while neutralisation activity of blood serum rose from 14 days post vaccination (dpv). A challenge infection with CyHV-3 induced a severe disease with 80-100% mortality in non-vaccinated carp, while in vaccinated carp, no mortality was recorded and the virus load was >1,000-fold lower in the skin, gill and kidney. Histological analysis showed strongest pathological changes in the skin, with a complete destruction of the epidermis in non-vaccinated carp. In the skin of non-vaccinated fish, T and B cell responses were severely downregulated, inflammation and stress responses were increased upon challenge, whereas vaccinated fish had boosted neutrophil, T and B cell responses. A disruption of skin barrier elements (tight and adherence junction, desmosomes, mucins) led to an uncontrolled increase in skin bacteria load which most likely exacerbated the inflammation and the pathology. Using a live attenuated virus vaccine, we were able to show that increased neutrophil, T and B cell responses provide protection from CyHV-3 infection and lead to preservation of skin integrity, which supports successful protection against additional pathogens in the aquatic environment which foster disease development in non-vaccinated carp.


Fish Diseases/immunology , Fish Diseases/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae/immunology , Viral Vaccines/immunology , Animals , Carps , Herpesviridae/genetics , Herpesviridae Infections/immunology , Vaccination , Vaccines, Attenuated/immunology , Viral Vaccines/genetics
14.
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
15.
Immunohorizons ; 6(1): 8-15, 2022 01 14.
Article En | MEDLINE | ID: mdl-35031582

NK cells are important mediators of viral control with the capacity to form adaptive immune features following infection. However, studies of infection-induced adaptive NK cells require adoptive cell transfer to lower the precursor frequency of "Ag-specific" NK cells, potentially limiting the diversity of the NK cell response. In seeking an unmanipulated model to probe the adaptive NK cells, we interrogated a wide range of Collaborative Cross (CC) inbred mice, inbred mouse strains that exhibit broad genetic diversity across strains. Our assessment identified and validated a putative "ideal" CC strain, CC006, which does not require manipulation to generate and maintain adaptive NK cells. Critically, CC006 mice, in contrast to C57BL/6 mice, are capable of developing enhanced NK cell-mediated protective responses to murine CMV infection following m157-mediated vaccination. This work both furthers our understanding of adaptive NK cells and demonstrates the utility of CC mice in the development and interrogation of immunologic models.


Disease Models, Animal , Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , Adoptive Transfer , Animals , Female , Herpesviridae Infections/pathology , Killer Cells, Natural/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
16.
J Mol Biol ; 434(6): 167214, 2022 03 30.
Article En | MEDLINE | ID: mdl-34437888

Gammaherpesviruses are ubiquitous pathogens that establish lifelong infections in the vast majority of adults worldwide. Importantly, these viruses are associated with numerous malignancies and are responsible for significant human cancer burden. These virus-associated cancers are due, in part, to the ability of gammaherpesviruses to successfully evade the innate immune response throughout the course of infection. In this review, we will summarize the current understanding of how gammaherpesviruses are detected by innate immune sensors, how these viruses evade recognition by host cells, and how this knowledge can inform novel therapeutic approaches for these viruses and their associated diseases.


Gammaherpesvirinae , Herpesviridae Infections , Immune Evasion , Immunity, Innate , Gammaherpesvirinae/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Humans , Virus Latency
17.
Viruses ; 13(12)2021 11 26.
Article En | MEDLINE | ID: mdl-34960641

γδ T cells are activated in viral, bacterial and parasitic infections. Among viruses that promote γδ T cell mobilisation in humans, herpes viruses (HHVs) occupy a particular place since they infect the majority of the human population and persist indefinitely in the organism in a latent state. Thus, other infections should, in most instances, be considered co-infections, and the reactivation of HHV is a serious confounding factor in attributing γδ T cell alterations to a particular pathogen in human diseases. We review here the literature data on γδ T cell mobilisation in HHV infections and co-infections, and discuss the possible contribution of HHVs to γδ alterations observed in various infectious settings. As multiple infections seemingly mobilise overlapping γδ subsets, we also address the concept of possible cross-protection.


Coinfection , Herpesviridae Infections/immunology , Herpesviridae/immunology , Intraepithelial Lymphocytes/immunology , Malaria/complications , Mycobacterium Infections/complications , Virus Diseases/complications , Adaptive Immunity , Herpesviridae Infections/complications , Herpesviridae Infections/virology , Humans , Immunity, Innate , Intraepithelial Lymphocytes/virology , Lymphocytes/immunology , Virus Diseases/virology , Virus Latency
18.
mBio ; 12(6): e0211321, 2021 12 21.
Article En | MEDLINE | ID: mdl-34933450

The interferon-induced transmembrane proteins (IFITMs) are broad-spectrum antiviral proteins that inhibit the entry of enveloped viruses. We analyzed the effect of IFITMs on the gamma-2 herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related rhesus monkey rhadinovirus (RRV). We used CRISPR/Cas9-mediated gene knockout to generate A549 cells, human foreskin fibroblasts (HFF), and human umbilical vein endothelial cells (HUVEC) with combined IFITM1/2/3 knockout and identified IFITMs as cell-dependent inhibitors of KSHV and RRV infection in A549 cells and HFF but not HUVEC. IFITM overexpression revealed IFITM1 as the relevant IFITM that inhibits KSHV and RRV infection. Fluorescent KSHV particles did not pronouncedly colocalize with IFITM-positive compartments. However, we found that KSHV and RRV glycoprotein-mediated cell-cell fusion is enhanced upon IFITM1/2/3 knockout. Taken together, we identified IFITM1 as a cell-dependent restriction factor of KSHV and RRV that acts at the level of membrane fusion. Of note, our results indicate that recombinant IFITM overexpression may lead to results that are not representative for the situation at endogenous levels. Strikingly, we observed that the endotheliotropic KSHV circumvents IFITM-mediated restriction in HUVEC despite high IFITM expression, while influenza A virus (IAV) glycoprotein-driven entry into HUVEC is potently restricted by IFITMs even in the absence of interferon. Mechanistically, we found that KSHV colocalizes less with IFITM1 and IFITM2 in HUVEC than in A549 cells immediately after attachment, potentially contributing to the observed difference in restriction. IMPORTANCE IFITM proteins are the first line of defense against infection by many pathogens and may also have therapeutic importance, as they, among other effectors, mediate the antiviral effect of interferons. Neither their function against herpesviruses nor their mechanism of action is well understood. We report here that in some cells but not in, for example, primary umbilical vein endothelial cells, IFITM1 restricts KSHV and RRV and that, mechanistically, this is likely effected by reducing the fusogenicity of the cell membrane. Further, we demonstrate potent inhibition of IAV glycoprotein-driven infection of cells of extrapulmonary origin by high constitutive IFITM expression.


Antigens, Differentiation/immunology , Herpesviridae Infections/immunology , Herpesvirus 8, Human/physiology , Membrane Proteins/immunology , RNA-Binding Proteins/immunology , Rhadinovirus/physiology , Animals , Antigens, Differentiation/genetics , Coinfection/genetics , Coinfection/immunology , Coinfection/virology , Fibroblasts/immunology , Fibroblasts/virology , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/virology , Humans , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , Rhadinovirus/genetics , Species Specificity , Virus Internalization , Virus Replication
19.
Nat Commun ; 12(1): 7323, 2021 12 16.
Article En | MEDLINE | ID: mdl-34916520

T cell responses to Kaposi's sarcoma-associated herpesvirus (KSHV) are likely essential in the control of KSHV infection and protection from associated disease, but remain poorly characterised. KSHV prevalence in rural Uganda is high at >90%. Here we investigate IFN- γ T cell responses to the KSHV proteome in HIV-negative individuals from a rural Ugandan population. We use an ex-vivo IFN- γ ELISpot assay with overlapping peptide pools spanning 83 KSHV open reading frames (ORF) on peripheral blood mononuclear cells (PBMC) from 116 individuals. KSHV-specific T cell IFN- γ responses are of low intensity and heterogeneous, with no evidence of immune dominance; by contrast, IFN- γ responses to Epstein-Barr virus, Cytomegalovirus and influenza peptides are frequent and intense. Individuals with KSHV DNA in PBMC have higher IFN- γ responses to ORF73 (p = 0.02) and lower responses to K8.1 (p = 0.004) when compared with those without KSHV DNA. In summary, we demonstrate low intensity, heterogeneous T cell responses to KSHV in immune-competent individuals.


Herpesviridae Infections/immunology , Herpesvirus 8, Human/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Cohort Studies , Female , HIV Antibodies/blood , HIV Infections/blood , HIV Seronegativity , Herpesviridae Infections/blood , Herpesviridae Infections/virology , Herpesvirus 8, Human/genetics , Humans , Interferon-gamma/immunology , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Uganda , Young Adult
20.
Front Immunol ; 12: 757302, 2021.
Article En | MEDLINE | ID: mdl-34790199

Roles for viral infections and aberrant immune responses in driving localized neuroinflammation and neurodegeneration in multiple sclerosis (MS) are the focus of intense research. Epstein-Barr virus (EBV), as a persistent and frequently reactivating virus with major immunogenic influences and a near 100% epidemiological association with MS, is considered to play a leading role in MS pathogenesis, triggering localized inflammation near or within the central nervous system (CNS). This triggering may occur directly via viral products (RNA and protein) and/or indirectly via antigenic mimicry involving B-cells, T-cells and cytokine-activated astrocytes and microglia cells damaging the myelin sheath of neurons. The genetic MS-risk factor HLA-DR2b (DRB1*1501ß, DRA1*0101α) may contribute to aberrant EBV antigen-presentation and anti-EBV reactivity but also to mimicry-induced autoimmune responses characteristic of MS. A central role is proposed for inflammatory EBER1, EBV-miRNA and LMP1 containing exosomes secreted by viable reactivating EBV+ B-cells and repetitive release of EBNA1-DNA complexes from apoptotic EBV+ B-cells, forming reactive immune complexes with EBNA1-IgG and complement. This may be accompanied by cytokine- or EBV-induced expression of human endogenous retrovirus-W/-K (HERV-W/-K) elements and possibly by activation of human herpesvirus-6A (HHV-6A) in early-stage CNS lesions, each contributing to an inflammatory cascade causing the relapsing-remitting neuro-inflammatory and/or progressive features characteristic of MS. Elimination of EBV-carrying B-cells by antibody- and EBV-specific T-cell therapy may hold the promise of reducing EBV activity in the CNS, thereby limiting CNS inflammation, MS symptoms and possibly reversing disease. Other approaches targeting HHV-6 and HERV-W and limiting inflammatory kinase-signaling to treat MS are also being tested with promising results. This article presents an overview of the evidence that EBV, HHV-6, and HERV-W may have a pathogenic role in initiating and promoting MS and possible approaches to mitigate development of the disease.


Endogenous Retroviruses/pathogenicity , Herpesvirus 4, Human/pathogenicity , Herpesvirus 6, Human/pathogenicity , Multiple Sclerosis/etiology , Neuroinflammatory Diseases/virology , Antibodies, Viral/immunology , Antigen-Antibody Complex/immunology , Autoimmunity , B-Lymphocytes/immunology , Blood-Brain Barrier , Brain/virology , Coinfection , DNA, Viral/immunology , Endogenous Retroviruses/physiology , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/immunology , Gene Products, env/physiology , Genetic Predisposition to Disease , Herpesviridae Infections/complications , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 4, Human/immunology , Herpesvirus 6, Human/immunology , Humans , Lymph Nodes/virology , Models, Immunological , Molecular Mimicry , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Multiple Sclerosis/virology , Myelin Sheath/immunology , Myelin Sheath/pathology , Neuroinflammatory Diseases/etiology , Pregnancy Proteins/physiology , Transcriptional Activation , Virus Activation , Virus Latency
...