Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
PLoS Pathog ; 20(4): e1011939, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683861

ABSTRACT

Epstein-Barr virus (EBV) persistently infects 95% of adults worldwide and is associated with multiple human lymphomas that express characteristic EBV latency programs used by the virus to navigate the B-cell compartment. Upon primary infection, the EBV latency III program, comprised of six Epstein-Barr Nuclear Antigens (EBNA) and two Latent Membrane Protein (LMP) antigens, drives infected B-cells into germinal center (GC). By incompletely understood mechanisms, GC microenvironmental cues trigger the EBV genome to switch to the latency II program, comprised of EBNA1, LMP1 and LMP2A and observed in GC-derived Hodgkin lymphoma. To gain insights into pathways and epigenetic mechanisms that control EBV latency reprogramming as EBV-infected B-cells encounter microenvironmental cues, we characterized GC cytokine effects on EBV latency protein expression and on the EBV epigenome. We confirmed and extended prior studies highlighting GC cytokine effects in support of the latency II transition. The T-follicular helper cytokine interleukin 21 (IL-21), which is a major regulator of GC responses, and to a lesser extent IL-4 and IL-10, hyper-induced LMP1 expression, while repressing EBNA expression. However, follicular dendritic cell cytokines including IL-15 and IL-27 downmodulate EBNA but not LMP1 expression. CRISPR editing highlighted that STAT3 and STAT5 were necessary for cytokine mediated EBNA silencing via epigenetic effects at the EBV genomic C promoter. By contrast, STAT3 was instead necessary for LMP1 promoter epigenetic remodeling, including gain of activating histone chromatin marks and loss of repressive polycomb repressive complex silencing marks. Thus, EBV has evolved to coopt STAT signaling to oppositely regulate the epigenetic status of key viral genomic promoters in response to GC cytokine cues.


Subject(s)
Epigenesis, Genetic , Epstein-Barr Virus Infections , Gene Expression Regulation, Viral , Germinal Center , Herpesvirus 4, Human , Virus Latency , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Germinal Center/immunology , Germinal Center/virology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/immunology , Cytokines/metabolism , B-Lymphocytes/virology , B-Lymphocytes/metabolism
2.
J Virol ; 97(12): e0100823, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37962378

ABSTRACT

IMPORTANCE: The human gammaherpesviruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus are etiologic agents of numerous B cell lymphomas. A hallmark of gammaherpesvirus infection is their ability to establish lifelong latency in B cells. However, the specific mechanisms that mediate chronic infection in B cells in vivo remain elusive. Cellular E3 ubiquitin ligases regulate numerous biological processes by catalyzing ubiquitylation and modifying protein location, function, or half-life. Many viruses hijack host ubiquitin ligases to evade antiviral host defense and promote viral fitness. Here, we used the murine gammaherpesvirus 68 in vivo system to demonstrate that the E3 ligase Cul4b is essential for this virus to establish latency in germinal center B cells. These findings highlight an essential role for this E3 ligase in promoting chronic gammaherpesvirus infection in vivo and suggest that targeted inhibition of E3 ligases may provide a novel and effective intervention strategy against gammaherpesvirus-associated diseases.


Subject(s)
B-Lymphocytes , Gammaherpesvirinae , Herpesviridae Infections , Persistent Infection , Animals , Mice , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cullin Proteins/metabolism , Gammaherpesvirinae/physiology , Germinal Center/cytology , Germinal Center/virology , Herpesviridae Infections/enzymology , Herpesviridae Infections/virology , Persistent Infection/enzymology , Persistent Infection/virology , Ubiquitins/metabolism , Virus Latency
3.
Proc Natl Acad Sci U S A ; 119(32): e2123362119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35921433

ABSTRACT

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.


Subject(s)
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
4.
J Exp Med ; 218(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34665220

ABSTRACT

T follicular helper cell (TFH)-dependent antibody responses are critical for long-term immunity. Antibody responses are diminished in early life, limiting long-term protective immunity and allowing prolonged or recurrent infection, which may be important for viral lung infections that are highly prevalent in infancy. In a murine model using respiratory syncytial virus (RSV), we show that TFH and the high-affinity antibody production they promote are vital for preventing disease on RSV reinfection. Following a secondary RSV infection, TFH-deficient mice had significantly exacerbated disease characterized by delayed viral clearance, increased weight loss, and immunopathology. TFH generation in early life was compromised by heightened IL-2 and STAT5 signaling in differentiating naive T cells. Neutralization of IL-2 during early-life RSV infection resulted in a TFH-dependent increase in antibody-mediated immunity and was sufficient to limit disease severity upon reinfection. These data demonstrate the importance of TFH in protection against recurrent RSV infection and highlight a mechanism by which this is suppressed in early life.


Subject(s)
Interleukin-2/immunology , Respiratory Syncytial Virus Infections/immunology , T Follicular Helper Cells/immunology , T Follicular Helper Cells/virology , Age Factors , Animals , Antibodies, Viral , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/virology , Female , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/virology , Immunity, Humoral , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/physiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Pregnancy , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Reinfection/immunology , Reinfection/virology , Respiratory Syncytial Virus Infections/metabolism , STAT5 Transcription Factor/metabolism
5.
Sci Rep ; 11(1): 21220, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34707156

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic herpesvirus implicated in the pathogenesis of several malignant and non-malignant conditions. However, a number of fundamental aspects about the biology of EBV and the mechanism(s) by which this virus induces pathology remain unknown. One major obstacle has been the lack of a suitable animal model for EBV infection. In this study, using our recently established rabbit model of EBV infection, we examined the early events following primary EBV infection. We show that, both immunocompetent and immunosuppressed animals were readily susceptible to EBV infection. However, immunosuppressed animals showed marked splenomegaly and widespread infection. Following EBV infection, the virus primarily targeted naïve IgM+, CD20+, CD21+ and CD79a+ B cells. Infected cells expressed varying sets of viral latent/lytic gene products. Notably, co-expression of latent and lytic proteins in the same cell was not observed. Infected cells in type 0/1 latency (EBERs+), were small and proliferating (Ki67+). By contrast, cells in type 2/3 latency (LMP1+), were large, non-proliferating (Ki-67-) and p53+. Although infected B-cells were widely present in splenic follicles, they did not express germinal center marker, BCL-6. Taken together, this study shows for the first time, some of the early events following primary EBV infection.


Subject(s)
Epstein-Barr Virus Infections/immunology , Germinal Center/immunology , Spleen/immunology , Animals , Antigens, CD20/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/virology , CD79 Antigens/metabolism , Epstein-Barr Virus Infections/pathology , Germinal Center/virology , Immunoglobulin M/immunology , Ki-67 Antigen/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Rabbits , Receptors, Complement 3d/metabolism , Spleen/pathology , Spleen/virology , Tumor Suppressor Protein p53/metabolism
6.
Viruses ; 13(7)2021 07 19.
Article in English | MEDLINE | ID: mdl-34372605

ABSTRACT

The Epstein-Barr virus (EBV) is a well-adapted human virus, and its infection is exclusive to our species, generally beginning in the childhood and then persisting throughout the life of most of the affected adults. Although this infection generally remains asymptomatic, EBV can trigger life-threatening conditions under unclear circumstances. The EBV lifecycle is characterized by interactions with other viruses or bacteria, which increases the probability of awakening its pathobiont capacity. For instance, EBV infects B cells with the potential to alter the germinal center reaction (GCR)-an adaptive immune structure wherein mutagenic-driven processes take place. HIV- and Plasmodium falciparum-induced B cell hyperactivation also feeds the GCR. These agents, along with the B cell tropic KSHV, converge in the ontogeny of germinal center (GC) or post-GC lymphomas. EBV oral transmission facilitates interactions with local bacteria and HPV, thereby increasing the risk of periodontal diseases and head and neck carcinomas. It is less clear as to how EBV is localized in the stomach, but together with Helicobacter pylori, they are known to be responsible for gastric cancer. Perhaps this mechanism is reminiscent of the local inflammation that attracts different herpesviruses and enhances graft damage and chances of rejection in transplanted patients. In this review, we discussed the existing evidence suggestive of EBV possessing the potential to synergize or cooperate with these agents to trigger or worsen the disease.


Subject(s)
Coinfection/pathology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Host Microbial Interactions , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , Coinfection/microbiology , Coinfection/parasitology , Coinfection/virology , Epstein-Barr Virus Infections/immunology , Germinal Center/virology , Helicobacter pylori , Herpesvirus 4, Human/immunology , Humans , Mice , Stomach Neoplasms/virology
7.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34292314

ABSTRACT

Besides antigen-specific responses to viral antigens, humoral immune response in virus infection can generate polyreactive and autoreactive antibodies. Dengue and Zika virus infections have been linked to antibody-mediated autoimmune disorders, including Guillain-Barré syndrome. A unique feature of flaviviruses is the secretion of nonstructural protein 1 (NS1) by infected cells. NS1 is highly immunogenic, and antibodies targeting NS1 can have both protective and pathogenic roles. In the present study, we investigated the humoral immune response to Zika virus NS1 and found NS1 to be an immunodominant viral antigen associated with the presence of autoreactive antibodies. Through single B cell cultures, we coupled binding assays and BCR sequencing, confirming the immunodominance of NS1. We demonstrate the presence of self-reactive clones in germinal centers after both infection and immunization, some of which present cross-reactivity with NS1. Sequence analysis of anti-NS1 B cell clones showed sequence features associated with pathogenic autoreactive antibodies. Our findings demonstrate NS1 immunodominance at the cellular level as well as a potential role for NS1 in ZIKV-associated autoimmune manifestations.


Subject(s)
Cross Reactions/immunology , Viral Nonstructural Proteins/immunology , Zika Virus Infection/immunology , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , B-Lymphocytes/virology , Female , Germinal Center/pathology , Germinal Center/virology , Immunization , Immunoglobulin M/blood , Mice, Inbred BALB C , Viral Nonstructural Proteins/blood , Zika Virus Infection/virology
8.
STAR Protoc ; 2(3): 100499, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34195671

ABSTRACT

Location of immune cells that form the germinal center reaction within secondary lymphoid tissues can be characterized using confocal microscopy. Here, we present an optimized immunofluorescence staining protocol to image germinal center structures in fixed/frozen spleen sections from ChAdOx1 nCoV-19 immunized mice. This protocol can be adapted to identify other cell types within secondary lymphoid tissues. For complete information on the generation and use of this protocol to examine immune responses to the COVID vaccine ChAdOx1 nCoV-19, please refer to Silva-Cayetano et al. (2020).


Subject(s)
COVID-19/prevention & control , ChAdOx1 nCoV-19/administration & dosage , Fluorescent Antibody Technique/standards , Germinal Center/drug effects , Immunization, Secondary/methods , SARS-CoV-2/immunology , Spleen/drug effects , Animals , COVID-19/diagnostic imaging , COVID-19/immunology , COVID-19/virology , Fluorescent Antibody Technique/methods , Germinal Center/immunology , Germinal Center/pathology , Germinal Center/virology , Immunogenicity, Vaccine , Male , Mice , Spleen/immunology , Spleen/pathology , Spleen/virology
9.
Front Immunol ; 12: 683396, 2021.
Article in English | MEDLINE | ID: mdl-34177929

ABSTRACT

CD4 T cells are key mediators of adaptive immune responses during infection and vaccination. Within secondary lymphoid organs, helper CD4 T cells, particularly those residing in germinal centers known as follicular helper T cells (Tfh), provide critical help to B-cells to promote their survival, isotype switching and selection of high affinity memory B-cells. On the other hand, the important role of Tfh cells for the maintenance of HIV reservoir is well documented. Thus, interrogating and better understanding the tissue specific micro-environment and immune subsets that contribute to optimal Tfh cell differentiation and function is important for designing successful prevention and cure strategies. Here, we describe the development and optimization of eight multispectral confocal microscopy immunofluorescence panels designed for in depth characterization and immune-profiling of relevant immune cells in formalin-fixed paraffin-embedded human lymphoid tissue samples. We provide a comprehensive library of antibodies to use for the characterization of CD4+ T-cells -including Tfh and regulatory T-cells- as well as CD8 T-cells, B-cells, macrophages and dendritic cells and discuss how the resulting multispectral confocal datasets can be quantitatively dissected using the HistoCytometry pipeline to collect information about relative frequencies and immune cell spatial distributions. Cells harboring actively transcribed virus are analyzed using an in-situ hybridization assay for the characterization of HIV mRNA positive cells in combination with additional protein markers (multispectral RNAscope). The application of this methodology to lymphoid tissues offers a means to interrogate multiple relevant immune cell targets simultaneously at increased resolution in a reproducible manner to guide CD4 T-cell studies in infection and vaccination.


Subject(s)
Disease Reservoirs/virology , HIV Infections/virology , Image Processing, Computer-Assisted , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Microscopy, Confocal , Molecular Imaging , Biomarkers , Fluorescent Antibody Technique , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/virology , HIV Infections/diagnosis , HIV Infections/immunology , Host-Pathogen Interactions , Humans , Image Processing, Computer-Assisted/methods , Lymph Nodes/immunology , Lymph Nodes/virology , Lymphoid Tissue/virology , Microscopy, Confocal/methods , Molecular Imaging/methods
10.
PLoS Pathog ; 17(5): e1009575, 2021 05.
Article in English | MEDLINE | ID: mdl-33961680

ABSTRACT

HIV-infected infants are at an increased risk of progressing rapidly to AIDS in the first weeks of life. Here, we evaluated immunological and virological parameters in 25 SIV-infected infant rhesus macaques to understand the factors influencing a rapid disease outcome. Infant macaques were infected with SIVmac251 and monitored for 10 to 17 weeks post-infection. SIV-infected infants were divided into either typical (TypP) or rapid (RP) progressor groups based on levels of plasma anti-SIV antibody and viral load, with RP infants having low SIV-specific antibodies and high viral loads. Following SIV infection, 11 out of 25 infant macaques exhibited an RP phenotype. Interestingly, TypP had lower levels of total CD4 T cells, similar reductions in CD4/CD8 ratios and elevated activation of CD8 T cells, as measured by the levels of HLA-DR, compared to RP. Differences between the two groups were identified in other immune cell populations, including a failure to expand activated memory (CD21-CD27+) B cells in peripheral blood in RP infant macaques, as well as reduced levels of germinal center (GC) B cells and T follicular helper (Tfh) cells in spleens (4- and 10-weeks post-SIV). Reduced B cell proliferation in splenic germinal GCs was associated with increased SIV+ cell density and follicular type 1 interferon (IFN)-induced immune activation. Further analyses determined that at 2-weeks post SIV infection TypP infants exhibited elevated levels of the GC-inducing chemokine CXCL13 in plasma, as well as significantly lower levels of viral envelope diversity compared to RP infants. Our findings provide evidence that early viral and immunologic events following SIV infection contributes to impairment of B cells, Tfh cells and germinal center formation, ultimately impeding the development of SIV-specific antibody responses in rapidly progressing infant macaques.


Subject(s)
Disease Progression , Immunity, Humoral , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , Genetic Variation , Germinal Center/immunology , Germinal Center/virology , Humans , Interferon Type I/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/virology , Macaca mulatta , Phenotype , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Viral Load
11.
mBio ; 12(1)2021 02 02.
Article in English | MEDLINE | ID: mdl-33531399

ABSTRACT

Human bocavirus 1 (HBoV1), a nonenveloped single-stranded DNA parvovirus, causes mild to life-threatening respiratory tract infections, acute otitis media, and encephalitis in young children. HBoV1 often persists in nasopharyngeal secretions for months, hampering diagnosis. It has also been shown to persist in pediatric palatine and adenoid tonsils, which suggests that lymphoid organs are reservoirs for virus spread; however, the tissue site and host cells remain unknown. Our aim was to determine, in healthy nonviremic children with preexisting HBoV1 immunity, the adenotonsillar persistence site(s), host cell types, and virus activity. We discovered that HBoV1 DNA persists in lymphoid germinal centers (GCs), but not in the corresponding tonsillar epithelium, and that the cell types harboring the virus are mainly naive, activated, and memory B cells and monocytes. Both viral DNA strands and both sides of the genome were detected, as well as infrequent mRNA. Moreover, we showed, in B-cell and monocyte cultures and ex vivo tonsillar B cells, that the cellular uptake of HBoV1 occurs via the Fc receptor (FcγRII) through antibody-dependent enhancement (ADE). This resulted in viral mRNA transcription, known to occur exclusively from double-stranded DNA in the nucleus, however, with no detectable productive replication. Confocal imaging with fluorescent virus-like particles moreover disclosed endocytosis. To which extent the active HBoV1 GC persistence has a role in chronic inflammation or B-cell maturation disturbances, and whether the virus can be reactivated, will be interesting topics for forthcoming studies.IMPORTANCE Human bocavirus 1 (HBoV1), a common pediatric respiratory pathogen, can persist in airway secretions for months hampering diagnosis. It also persists in tonsils, providing potential reservoirs for airway shedding, with the exact location, host cell types, and virus activity unknown. Our study provides new insights into tonsillar HBoV1 persistence. We observed HBoV1 persistence exclusively in germinal centers where immune maturation occurs, and the main host cells were B cells and monocytes. In cultured cell lines and primary tonsillar B cells, we showed the virus uptake to be significantly enhanced by HBoV1-specific antibodies, mediated by the cellular IgG receptor, leading to viral mRNA synthesis, but without detectable productive replication. Possible implications of such active viral persistence could be tonsillar inflammation, disturbances in immune maturation, reactivation, or cell death with release of virus DNA, explaining the long-lasting HBoV1 airway shedding.


Subject(s)
Antibody-Dependent Enhancement , Germinal Center/virology , Human bocavirus/immunology , Palatine Tonsil/virology , Parvoviridae Infections/virology , Adolescent , Adult , Aged , B-Lymphocytes/virology , Child , Child, Preschool , DNA, Viral/analysis , Endosomes/virology , Humans , Infant , Infant, Newborn , Middle Aged , Monocytes/virology , Parvoviridae Infections/immunology , Young Adult
12.
JCI Insight ; 6(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33427210

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) with CCR5- donor cells is the only treatment known to cure HIV-1 in patients with underlying malignancy. This is likely due to a donor cell-mediated graft-versus-host effect targeting HIV reservoirs. Allo-HSCT would not be an acceptable therapy for most people living with HIV due to the transplant-related side effects. Chimeric antigen receptor (CAR) immunotherapies specifically traffic to malignant lymphoid tissues (lymphomas) and, in some settings, are able to replace allo-HSCT. Here, we quantified the engraftment of HSC-derived, virus-directed CAR T cells within HIV reservoirs in a macaque model of HIV infection, using potentially novel IHC assays. HSC-derived CAR cells trafficked to and displayed multilineage engraftment within tissue-associated viral reservoirs, persisting for nearly 2 years in lymphoid germinal centers, the brain, and the gastrointestinal tract. Our findings demonstrate that HSC-derived CAR+ cells reside long-term and proliferate in numerous tissues relevant for HIV infection and cancer.


Subject(s)
HIV Infections/immunology , HIV Infections/therapy , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Animals , Cell Lineage/immunology , Disease Models, Animal , Disease Reservoirs/virology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/virology , Germinal Center/immunology , Germinal Center/pathology , Germinal Center/virology , HIV Infections/virology , HIV-1 , Humans , Immunohistochemistry , Macaca nemestrina , Male , Receptors, Chimeric Antigen/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Acquired Immunodeficiency Syndrome/virology , Transplantation, Homologous
13.
J Virol ; 95(1)2020 12 09.
Article in English | MEDLINE | ID: mdl-33028711

ABSTRACT

Gammaherpesviruses (GHVs) are DNA tumor viruses that establish lifelong, chronic infections in lymphocytes of humans and other mammals. GHV infections are associated with numerous cancers, especially in immunocompromised hosts. While it is known that GHVs utilize host germinal center (GC) B cell responses during latency establishment, an understanding of how viral gene products function in specific B cell subsets to regulate this process is incomplete. Using murine gammaherpesvirus 68 (MHV68) as a small-animal model to define mechanisms of GHV pathogenesis in vivo, we generated a virus in which the M2 gene was flanked by loxP sites (M2.loxP), enabling the use of Cre-lox technology to define M2 function in specific cell types in infection and disease. The M2 gene encodes a protein that is highly expressed in GC B cells that promotes plasma cell differentiation and viral reactivation. M2 was efficiently deleted in Cre-expressing cells, and the presence of loxP sites flanking M2 did not alter viral replication or latency in mice that do not express Cre. In contrast, M2.loxP MHV68 exhibited a deficit in latency establishment and reactivation that resembled M2-null virus, following intranasal (IN) infection of mice that express Cre in all B cells (CD19-Cre). Nearly identical phenotypes were observed for M2.loxP MHV68 in mice that express Cre in germinal center (GC) B cells (AID-Cre). However, colonization of neither draining lymph nodes after IN infection nor the spleen after intraperitoneal (IP) infection required M2, although the reactivation defect was retained. Together, these data confirm that M2 function is B cell-specific and demonstrate that M2 primarily functions in AID-expressing cells to facilitate MHV68 dissemination to distal latency reservoirs within the host and reactivation from latency. Our study reveals that a viral latency gene functions within a distinct subset of cells to facilitate host colonization.IMPORTANCE Gammaherpesviruses establish lifelong chronic infections in cells of the immune system that can lead to lymphomas and other diseases. To facilitate colonization of a host, gammaherpesviruses encode gene products that manipulate processes involved in cellular proliferation and differentiation. Whether and how these viral gene products function in specific cells of the immune system is poorly defined. We report here the use of a viral genetic system that allows for deletion of specific viral genes in discrete populations of cells. We employ this system in an in vivo model to demonstrate cell-type-specific requirements for a particular viral gene. Our findings reveal that a viral gene product can function in distinct cellular subsets to direct gammaherpesvirus pathogenesis.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/immunology , Herpesviridae Infections/virology , Rhadinovirus/physiology , Viral Proteins/immunology , Virus Activation , Animals , Antigens, CD19/metabolism , B-Lymphocytes/virology , Cell Differentiation , Cell Proliferation , Germinal Center/immunology , Germinal Center/virology , Herpesviridae Infections/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/virology , Mice , Rhadinovirus/genetics , Rhadinovirus/metabolism , Viral Proteins/genetics , Virus Latency
14.
BMJ Case Rep ; 13(9)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32878845

ABSTRACT

We present this case of human herpes virus 8-positive germinotropic lymphoproliferative disorder in a 20-year-old woman seen in the surgical oncology clinic for localised lymphadenopathy. This is the first case to be reported in the UK, and we discuss it along with a literature review including investigations and treatment options. This will demonstrate the importance of preoperative workup and multidisciplinary teamwork in deciding management plans and serve as a guide for future encounters of this rare condition in clinical practice.


Subject(s)
Germinal Center/pathology , Herpesviridae Infections/diagnosis , Herpesvirus 8, Human/isolation & purification , Lymphoproliferative Disorders/diagnosis , Biopsy, Large-Core Needle , Female , Germinal Center/virology , Groin/diagnostic imaging , Groin/surgery , Herpesviridae Infections/pathology , Herpesviridae Infections/therapy , Herpesviridae Infections/virology , Humans , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/therapy , Lymphoproliferative Disorders/virology , Positron Emission Tomography Computed Tomography , Radiotherapy, Adjuvant , Treatment Outcome , United Kingdom , Young Adult
15.
J Vis Exp ; (160)2020 06 27.
Article in English | MEDLINE | ID: mdl-32658200

ABSTRACT

T Follicular Helper (Tfh) cells are an independent CD4+ T cell subset specialized in providing help for germinal center (GC) development and generation of high-affinity antibodies. In influenza virus infection, robust Tfh and GC B cell responses are induced to facilitate effective virus eradication, which confers a qualified mouse model for Tfh-associated study. In this paper, we described protocols in detection of basic Tfh-associated immune response during influenza virus infection in mice. These protocols include: intranasal inoculation of influenza virus; flow cytometry staining and analysis of polyclonal and antigen-specific Tfh cells, GC B cells and plasma cells; immunofluorescence detection of GCs; enzyme-linked immunosorbent assay (ELISA) of influenza virus-specific antibody in serum. These assays basically quantify the differentiation and function of Tfh cells in influenza virus infection, thus providing help for studies in elucidating differentiation mechanism and manipulation strategy.


Subject(s)
Germinal Center/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , Germinal Center/virology , Mice , T-Lymphocyte Subsets/virology , T-Lymphocytes, Helper-Inducer/virology
16.
PLoS Pathog ; 16(4): e1008438, 2020 04.
Article in English | MEDLINE | ID: mdl-32353066

ABSTRACT

One of the defining characteristics of the B cell receptor (BCR) is the extensive diversity in the repertoire of immunoglobulin genes that make up the BCR, resulting in broad range of specificity. Gammaherpesviruses are B lymphotropic viruses that establish life-long infection in B cells, and although the B cell receptor plays a central role in B cell biology, very little is known about the immunoglobulin repertoire of gammaherpesvirus infected cells. To begin to characterize the Ig genes expressed by murine gammaherpesvirus 68 (MHV68) infected cells, we utilized single cell sorting to sequence and clone the Ig variable regions of infected germinal center (GC) B cells and plasma cells. We show that MHV68 infection is biased towards cells that express the Igλ light chain along with a single heavy chain variable gene, IGHV10-1*01. This population arises through clonal expansion but is not viral antigen specific. Furthermore, we show that class-switching in MHV68 infected cells differs from that of uninfected cells. Fewer infected GC B cells are class-switched compared to uninfected GC B cells, while more infected plasma cells are class-switched compared to uninfected plasma cells. Additionally, although they are germinal center derived, the majority of class switched plasma cells display no somatic hypermutation regardless of infection status. Taken together, these data indicate that selection of infected B cells with a specific BCR, as well as virus mediated manipulation of class switching and somatic hypermutation, are critical aspects in establishing life-long gammaherpesvirus infection.


Subject(s)
B-Lymphocytes/immunology , Gammaherpesvirinae/physiology , Herpesviridae Infections/veterinary , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Immunoglobulin lambda-Chains/immunology , Rodent Diseases/immunology , Animals , B-Lymphocytes/virology , Female , Gammaherpesvirinae/genetics , Germinal Center/immunology , Germinal Center/virology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin lambda-Chains/genetics , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Plasma Cells/virology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Rodent Diseases/genetics , Rodent Diseases/virology
17.
Life Sci Alliance ; 3(3)2020 03.
Article in English | MEDLINE | ID: mdl-32029571

ABSTRACT

The gammaherpesviruses (γHVs), human Kaposi sarcoma-associated herpesvirus (KSHV), EBV, and murine γHV68 are prevalent infections associated with lymphocyte pathologies. After primary infection, EBV and γHV68 undergo latent expansion in germinal center (GC) B cells and persists in memory cells. The GC reaction evolves and selects antigen-specific B cells for memory development but whether γHV passively transients or manipulates this process in vivo is unknown. Using the γHV68 infection model, we analyzed the Ig repertoire of infected and uninfected GC cells from individual mice. We found that infected cells displayed the hallmarks of affinity maturation, hypermutation, and isotype switching but underwent clonal expansion. Strikingly, infected cells displayed distinct repertoire, not found in uninfected cells, with recurrent utilization of certain Ig heavy V segments including Ighv10-1 In a manner observed with KSHV, γHV68 infected cells also displayed lambda light chain bias. Thus, γHV68 subverts GC selection to expand in a specific B cell subset during the process that develops long-lived immunologic memory.


Subject(s)
Gammaherpesvirinae/metabolism , Germinal Center/immunology , Herpesviridae Infections/immunology , Animals , B-Lymphocytes/immunology , Female , Gammaherpesvirinae/pathogenicity , Germinal Center/virology , Immunologic Memory , Male , Mice , Mice, Inbred C57BL
18.
Front Immunol ; 11: 614743, 2020.
Article in English | MEDLINE | ID: mdl-33679702

ABSTRACT

Enterovirus 71 (EV71) is a positive single-stranded RNA (ssRNA) virus from the enterovirus genus of Picornaviridae family and causes diseases ranged from the mild disease of hand, foot and mouth disease (HFMD) to the severe disease of neurological involvement in young children. TLR7 is an intracellular pattern recognition receptor (PRR) recognizing viral ssRNA. In this study, we investigated the role of TLR7 in EV71 infection in mouse pups (10-12 days old) and found that wild-type (WT) and TLR7 knock-out (TLR7KO) mice infected with EV71 showed similar limb paralysis at the onset and peak of the disease, comparable loss of motor neurons, and similar levels of antiviral molecules in the spinal cord. These results suggest that TLR7 is not the absolute PRR for EV71 in the spinal cord. Interestingly, TLR7KO mice infected with EV71 exhibited significantly delayed recovery from limb paralysis compared with WT mice. TLR7KO mice infected with EV71 showed significantly decreased levels of IgM and IgG2, important antibodies for antiviral humoral immunity. Furthermore, TLR7KO mice infected with EV71 showed a decrease of germinal center B cells in the spleen compared with WT mice. Altogether, our study suggests that TLR7 plays a critical role in anti-viral humoral immunity rather than in being a PRR in the spinal cord during EV71 infection in young mice.


Subject(s)
Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Immunity, Humoral , Motor Neurons/metabolism , Spinal Cord/metabolism , Toll-Like Receptor 7/metabolism , Animals , Astrocytes/metabolism , B-Lymphocytes/immunology , Cytokines/metabolism , Enterovirus Infections/genetics , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Germinal Center/immunology , Germinal Center/virology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Motor Neurons/pathology , Motor Neurons/virology , Oligodendroglia/metabolism , Proteome/genetics , Proteome/metabolism , Receptors, IgG/metabolism , Spinal Cord/virology , Spleen/immunology , Spleen/virology , Toll-Like Receptor 7/genetics
19.
Front Immunol ; 10: 2362, 2019.
Article in English | MEDLINE | ID: mdl-31649673

ABSTRACT

As we learn more about the HIV latent reservoir, we continue to discover that the viral reservoir is more complicated than just a pool of infected resting memory CD4+ T cells in peripheral blood. Evidence increasingly points to both certain tissues and certain types of cells as potential viral reservoirs. T follicular helper cells (TFH) are prime targets of HIV infection-this creates a sanctuary for infected cells because CD8+ T cells generally do not enter lymph node follicles unless they express CXCR5, and are not as effective at killing infected CD4+ T cells as peripheral CD8+ T cells. In this review, we summarize the current state of research on TFH cell infection in peripheral lymphoid tissues and focus on the question of whether CD8+ T cell exclusion from B cell follicles is responsible, at least in part, for establishing secondary lymphoid tissue B cell follicles as an anatomic site of HIV transcription and replication.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Germinal Center/immunology , HIV Infections/immunology , HIV-1/physiology , Virus Replication/immunology , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Germinal Center/pathology , Germinal Center/virology , HIV Infections/pathology , Humans
20.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597758

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...