Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Exp Med ; 220(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37314481

ABSTRACT

Chronic viral infections are known to lead to T cell exhaustion or dysfunction. However, it remains unclear if antigen exposure episodes from periodic viral reactivation, such as herpes simplex virus type-2 (HSV-2) recrudescence, are sufficient to induce T cell dysfunction, particularly in the context of a tissue-specific localized, rather than a systemic, infection. We designed and implemented a stringent clinical surveillance protocol to longitudinally track both viral shedding and in situ tissue immune responses in a cohort of HSV+ volunteers that agreed to avoid using anti-viral therapy for the course of this study. Comparing lesion to control skin biopsies, we found that tissue T cells expanded immediately after reactivation, and then returned numerically and phenotypically to steady state. T cell responses appeared to be driven at least in part by migration of circulating T cells to the infected tissue. Our data indicate that tissue T cells are stably maintained in response to HSV reactivation, resembling a series of acute recall responses.


Subject(s)
Reinfection , Sepsis , Humans , T-Lymphocytes , Biopsy , Homeostasis
2.
J Clin Invest ; 133(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-36951943

ABSTRACT

Mucosal infections pose a significant global health burden. Antigen-specific tissue-resident T cells are critical to maintaining barrier immunity. Previous studies in the context of systemic infection suggest that memory CD8+ T cells may also provide innate-like protection against antigenically unrelated pathogens independent of T cell receptor engagement. Whether bystander T cell activation is also an important defense mechanism in the mucosa is poorly understood. Here, we investigated whether innate-like memory CD8+ T cells could protect against a model mucosal virus infection, herpes simplex virus 2 (HSV-2). We found that immunization with an irrelevant antigen delayed disease progression from lethal HSV-2 challenge, suggesting that memory CD8+ T cells may mediate protection despite the lack of antigen specificity. Upon HSV-2 infection, we observed an early infiltration, rather than substantial local proliferation, of antigen-nonspecific CD8+ T cells, which became bystander-activated only within the infected mucosal tissue. Critically, we show that bystander-activated CD8+ T cells are sufficient to reduce early viral burden after HSV-2 infection. Finally, local cytokine cues within the tissue microenvironment after infection were sufficient for bystander activation of mucosal tissue memory CD8+ T cells from mice and humans. Altogether, our findings suggest that local bystander activation of CD8+ memory T cells contributes a fast and effective innate-like response to infection in mucosal tissue.


Subject(s)
Herpes Simplex , Memory T Cells , Humans , Mice , Animals , Herpesvirus 2, Human , CD8-Positive T-Lymphocytes , Immunization , Immunologic Memory
3.
J Immunol ; 207(9): 2217-2222, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34588220

ABSTRACT

Cognate interactions between autoreactive B and T cells promote systemic lupus erythematosus pathogenesis by inter alia facilitating spontaneous germinal center (GC) formation. Whereas both myeloid and B cell APCs express B7 ligands (CD80 and CD86), the prevailing model holds that dendritic cell costimulation is sufficient for CD28-dependent T cell activation. In this study, we report that B cell-intrinsic CD80/CD86 deletion unexpectedly abrogates GCs in murine lupus. Interestingly, absent GCs differentially impacted serum autoantibodies. In keeping with distinct extrafollicular and GC activation pathways driving lupus autoantibodies, lack of GCs correlated with loss of RNA-associated autoantibodies but preserved anti-dsDNA and connective tissue autoantibody titers. Strikingly, even heterozygous B cell CD80/CD86 deletion was sufficient to prevent autoimmune GCs and RNA-associated autoantibodies. Together, these findings identify a key mechanism whereby B cells promote lupus pathogenesis by providing a threshold of costimulatory signals required for autoreactive T cell activation.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoantibodies/metabolism , Autoimmunity , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Lymphocyte Activation , Mice , Mice, Knockout , Receptor Cross-Talk
4.
Eur J Immunol ; 51(9): 2225-2236, 2021 09.
Article in English | MEDLINE | ID: mdl-34146342

ABSTRACT

Polymorphisms in TACI, a BAFF family cytokine receptor, are linked to diverse human immune disorders including common variable immunodeficiency (CVID) and systemic lupus erythematosus (SLE). Functional studies of individual variants show modest impacts on surface TACI expression and/or downstream signal transduction, indicating that relatively subtle variation in TACI activity can impact human B-cell biology. However, significant complexity underlies TACI biology, including both positive and negative regulation of physiologic and pathogenic B-cell responses. To model these contradictory events, we compared the functional impact of TACI deletion on separate models of murine SLE driven by T cell-independent and -dependent breaks in B-cell tolerance. First, we studied whether reduced surface TACI expression was sufficient to protect against progressive BAFF-mediated systemic autoimmunity. Strikingly, despite a relatively modest impact on surface TACI levels, TACI haploinsufficiency markedly reduced pathogenic RNA-associated autoantibody titers and conferred long-term protection from BAFF-driven lupus nephritis. In contrast, B cell-intrinsic TACI deletion exerted a limited impact of autoantibody generation in murine lupus characterized by spontaneous germinal center formation and T cell-dependent humoral autoimmunity. Together, these combined data provide new insights into TACI biology and highlight how TACI signals must be tightly regulated during protective and pathogenic B-cell responses.


Subject(s)
Autoimmunity/genetics , B-Cell Activating Factor/immunology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Transmembrane Activator and CAML Interactor Protein/genetics , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Autoimmunity/immunology , B-Cell Activating Factor/antagonists & inhibitors , B-Cell Activating Factor/genetics , B-Cell Activation Factor Receptor/genetics , B-Lymphocytes/immunology , Chimera , Female , Haploinsufficiency/genetics , Immunosuppressive Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Transmembrane Activator and CAML Interactor Protein/immunology
5.
J Immunol Methods ; 488: 112901, 2021 01.
Article in English | MEDLINE | ID: mdl-33069767

ABSTRACT

The isolation of human monoclonal antibodies (mAbs) arising from natural infection with human pathogens has proven to be a powerful technology, facilitating the understanding of the host response to infection at a molecular level. mAbs can reveal sites of vulnerability on pathogens and illuminate the biological function of the antigenic targets. Moreover, mAbs have the potential to be used directly for therapeutic applications such as passive delivery to prevent infection in susceptible target populations, and as treatment of established infection. The isolation of antigen-specific B cells from vaccine trials can also assist in deciphering whether the desired B cells are being targeted by a given vaccine. Several different processes have been developed to isolate mAbs, but all are generally labor-intensive and result in varying degrees of efficiency. Here, we describe the development of a cost-effective feeder cell line that stably expresses CD40-ligand, interleukin-2 and interleukin-21. Sorting of single B cells onto a layer of irradiated feeder cells sustained antibody production that permits functional screening of secreted antibodies in a manner that enables subsequent recovery of B cells for recombinant antibody cloning. As a proof of concept, we show that this approach can be used to isolate B cells that secrete antibodies that neutralize human papilloma virus (HPV) from participants of an HPV vaccine study.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , B-Lymphocytes/metabolism , Cell Separation , High-Throughput Screening Assays , Immunoglobulin G/metabolism , Papillomavirus Vaccines/administration & dosage , 3T3 Cells , Adolescent , Adult , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Feeder Cells , Female , Humans , Immunogenicity, Vaccine , Immunoglobulin G/immunology , Mice , Papillomavirus Vaccines/immunology , Proof of Concept Study , Time Factors , Vaccination , Young Adult
6.
J Immunol ; 203(11): 2817-2826, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31636237

ABSTRACT

Age-associated B cells (ABCs) are a unique subset of B cells defined by surface CD11b and CD11c expression. Although ABC expansion has been observed in both human and animal studies in the setting of advanced age, during humoral autoimmunity and following viral infection, the functional properties of this cellular subset remain incompletely defined. In the current study, we demonstrate that ABCs fulfill the criteria for memory B cells (MBCs), based on evidence of Ag-dependent expansion and persistence in a state poised for rapid differentiation into Ab-secreting plasma cells during secondary responses. First, we show that a majority of ABCs are not actively cycling but exhibit an extensive replication history consistent with prior Ag engagement. Second, despite unswitched surface IgM expression, ABCs show evidence of activation-induced cytidine deaminase (AID)-dependent somatic hypermutation. Third, BCRs cloned from sorted ABCs exhibit broad autoreactivity and polyreactivity. Although the overall level of ABC self-reactivity was not increased relative to naive B cells, ABCs lacked features of functional anergy characteristic of autoreactive B cells. Fourth, ABCs express MBC surface markers consistent with being poised for rapid plasma cell differentiation during recall responses. Finally, in a murine model of viral infection, adoptively transferred CD11c+ B cells rapidly differentiated into class-switched Ab-secreting cells upon Ag rechallenge. In summary, we phenotypically and functionally characterize ABCs as IgM-expressing MBCs, findings that together implicate ABCs in the pathogenesis of systemic autoimmunity.


Subject(s)
Aging/immunology , B-Lymphocytes/immunology , CD11c Antigen/immunology , Animals , Immunologic Memory/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Front Immunol ; 10: 44, 2019.
Article in English | MEDLINE | ID: mdl-30740104

ABSTRACT

TYK2 is a JAK family member that functions downstream of multiple cytokine receptors. Genome wide association studies have linked a SNP (rs34536443) within TYK2 encoding a Proline to Alanine substitution at amino acid 1104, to protection from multiple autoimmune diseases including systemic lupus erythematosus (SLE) and multiple sclerosis (MS). The protective role of this SNP in autoimmune pathogenesis, however, remains incompletely understood. Here we found that T follicular helper (Tfh) cells, switched memory B cells, and IFNAR signaling were decreased in healthy individuals that expressed the protective variant TYK2A1104 (TYK2P ). To study this variant in vivo, we developed a knock-in murine model of this allele. Murine Tyk2P expressing T cells homozygous for the protective allele, but not cells heterozygous for this change, manifest decreased IL-12 receptor signaling, important for Tfh lineage commitment. Further, homozygous Tyk2P T cells exhibited diminished in vitro Th1 skewing. Surprisingly, despite these signaling changes, in vivo formation of Tfh and GC B cells was unaffected in two models of T cell dependent immune responses and in two alternative SLE models. TYK2 is also activated downstream of IL-23 receptor engagement. Here, we found that Tyk2P expressing T cells had reduced IL-23 dependent signaling as well as a diminished ability to skew toward Th17 in vitro. Consistent with these findings, homozygous, but not heterozygous, Tyk2P mice were fully protected in a murine model of MS. Homozygous Tyk2P mice had fewer infiltrating CD4+ T cells within the CNS. Most strikingly, homozygous mice had a decreased proportion of IL-17+/IFNγ+, double positive, pathogenic CD4+ T cells in both the draining lymph nodes (LN) and CNS. Thus, in an autoimmune model, such as EAE, impacted by both altered Th1 and Th17 signaling, the Tyk2P allele can effectively shield animals from disease. Taken together, our findings suggest that TYK2P diminishes IL-12, IL-23, and IFN I signaling and that its protective effect is most likely manifest in the setting of autoimmune triggers that concurrently dysregulate at least two of these important signaling cascades.


Subject(s)
Autoimmunity/immunology , TYK2 Kinase/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Adult , Animals , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Knock-In Techniques , Humans , Interferon Type I/metabolism , Interleukin-12/metabolism , Interleukin-23/metabolism , Lupus Erythematosus, Systemic/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Multiple Sclerosis/immunology , Polymorphism, Single Nucleotide , Receptors, Interleukin-12/metabolism , TYK2 Kinase/genetics , Young Adult
8.
Eur J Immunol ; 49(1): 170-178, 2019 01.
Article in English | MEDLINE | ID: mdl-30353919

ABSTRACT

Age-associated B cells (ABC), a novel subset of activated B cells defined by CD11b and CD11c expression, have been linked with both protective anti-viral responses and the pathogenesis of systemic autoimmunity. Expression of the TH 1 lineage transcription factor T-bet has been identified as a defining feature of ABC biology, with B cell-intrinsic expression of this transcription factor proposed to be required for ABC formation. In contrast to this model, we report that Tbx21 (encoding T-bet)-deficient B cells upregulate CD11b and CD11c surface expression in vitro in response to integrated TLR and cytokine signals. Moreover, B cell-intrinsic T-bet deletion in a murine lupus model exerted no impact of ABC generation in vivo, with Tbx21-/- ABCs exhibiting an identical surface phenotype to wild-type (WT) ABCs. Importantly, WT and Tbx21-/- ABCs sorted from autoimmune mice produced equivalent amounts of IgM and IgG ex vivo following TLR stimulation, indicating that T-bet-deficient ABCs are likely functional in vivo. In summary, our data contradict the established literature by demonstrating that T-bet expression is not uniformly required for ABC generation.


Subject(s)
Aging/immunology , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , T-Box Domain Proteins/metabolism , Animals , Autoimmunity , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/genetics
9.
J Immunol ; 201(11): 3258-3268, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30373855

ABSTRACT

The B cell survival cytokine BAFF has been linked with the pathogenesis of systemic lupus erythematosus (SLE). BAFF binds distinct BAFF-family surface receptors, including the BAFF-R and transmembrane activator and CAML interactor (TACI). Although originally characterized as a negative regulator of B cell activation, TACI signals are critical for class-switched autoantibody (autoAb) production in BAFF transgenic mice. Consistent with this finding, a subset of transitional splenic B cells upregulate surface TACI expression and contribute to BAFF-driven autoAb. In the current study, we interrogated the B cell signals required for transitional B cell TACI expression and Ab production. Surprisingly, despite established roles for dual BCR and TLR signals in autoAb production in SLE, signals downstream of these receptors exerted distinct impacts on transitional B cell TACI expression and autoAb titers. Whereas loss of BCR signals prevented transitional B cell TACI expression and resulted in loss of serum autoAb across all Ig isotypes, lack of TLR signals exerted a more limited impact restricted to autoAb class-switch recombination without altering transitional B cell TACI expression. Finally, in parallel with the protective effect of TACI deletion, loss of BAFF-R signaling also protected against BAFF-driven autoimmunity. Together, these findings highlight how multiple signaling pathways integrate to promote class-switched autoAb production by transitional B cells, events that likely impact the pathogenesis of SLE and other BAFF-dependent autoimmune diseases.


Subject(s)
Autoantibodies/metabolism , Glomerulonephritis, IGA/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/metabolism , Precursor Cells, B-Lymphoid/physiology , Receptors, Antigen, B-Cell/metabolism , Toll-Like Receptor 7/metabolism , Animals , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/genetics , Disease Models, Animal , Humans , Immunoglobulin Class Switching , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor Cross-Talk , Receptors, Antigen, B-Cell/genetics , Signal Transduction , Toll-Like Receptor 7/genetics , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism
10.
J Exp Med ; 215(9): 2445-2461, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30127060

ABSTRACT

Activating mutations in the adapter protein CARD11 associated with diffuse large B cell lymphomas (DLBCLs) are predicted to arise during germinal center (GC) responses, leading to inappropriate activation of NF-κB signaling. Here, we modeled the B cell-intrinsic impact of the L251P activating mutation in CARD11 (aCARD11) on the GC response. Global B cell aCARD11 expression led to a modest increase in splenic B cells and a severe reduction in B1 B cell numbers, respectively. Following T cell-dependent immunization, aCARD11 cells exhibited increased rates of GC formation, resolution, and differentiation. Restriction of aCARD11 to GC B cells similarly altered the GC response and B cell differentiation. In this model, aCARD11 promoted dark zone skewing along with increased cycling, AID levels, and class switch recombination. Furthermore, aCard11 GC B cells displayed increased biomass and mTORC1 signaling, suggesting a novel strategy for targeting aCARD11-driven DLBCL. While aCARD11 potently impacts GC responses, the rapid GC contraction suggests it requires collaboration with events that limit terminal differentiation to promote lymphoma.


Subject(s)
B-Lymphocytes/immunology , CARD Signaling Adaptor Proteins/immunology , Cell Differentiation/immunology , Germinal Center/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Models, Immunological , Neoplasm Proteins/immunology , Signal Transduction/immunology , Animals , B-Lymphocytes/pathology , CARD Signaling Adaptor Proteins/genetics , Cell Differentiation/genetics , Germinal Center/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Signal Transduction/genetics
11.
J Clin Invest ; 128(9): 4163-4178, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29999501

ABSTRACT

Germinal centers (GCs) are major sites of clonal B cell expansion and generation of long-lived, high-affinity antibody responses to pathogens. Signaling through TLRs on B cells promotes many aspects of GC B cell responses, including affinity maturation, class switching, and differentiation into long-lived memory and plasma cells. A major challenge for effective vaccination is identifying strategies to specifically promote GC B cell responses. Here, we have identified a mechanism of regulation of GC B cell TLR signaling, mediated by αv integrins and noncanonical autophagy. Using B cell-specific αv-KO mice, we show that loss of αv-mediated TLR regulation increased GC B cell expansion, somatic hypermutation, class switching, and generation of long-lived plasma cells after immunization with virus-like particles (VLPs) or antigens associated with TLR ligand adjuvants. Furthermore, targeting αv-mediated regulation increased the magnitude and breadth of antibody responses to influenza virus vaccination. These data therefore identify a mechanism of regulation of GC B cells that can be targeted to enhance antibody responses to vaccination.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Integrin alphaV/immunology , Animals , Autophagy/immunology , Female , Germinal Center/cytology , Immunization , Immunoglobulin Class Switching , Immunoglobulin G/blood , Immunologic Memory , Influenza A virus/immunology , Integrin alphaV/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Signal Transduction/immunology , Somatic Hypermutation, Immunoglobulin , Toll-Like Receptors/immunology , Vaccines, Virus-Like Particle/immunology
12.
Kidney Int ; 94(4): 728-740, 2018 10.
Article in English | MEDLINE | ID: mdl-29907458

ABSTRACT

B cells are known to promote the pathogenesis of systemic lupus erythematosus (SLE) via the production of pathogenic anti-nuclear antibodies. However, the signals required for autoreactive B cell activation and the immune mechanisms whereby B cells impact lupus nephritis pathology remain poorly understood. The B cell survival cytokine B cell activating factor of the TNF Family (BAFF) has been implicated in the pathogenesis of SLE and lupus nephritis in both animal models and human clinical studies. Although the BAFF receptor has been predicted to be the primary BAFF family receptor responsible for BAFF-driven humoral autoimmunity, in the current study we identify a critical role for signals downstream of Transmembrane Activator and CAML Interactor (TACI) in BAFF-dependent lupus nephritis. Whereas transgenic mice overexpressing BAFF develop progressive membranoproliferative glomerulonephritis, albuminuria and renal dysfunction, TACI deletion in BAFF-transgenic mice provided long-term (about 1 year) protection from renal disease. Surprisingly, disease protection in this context was not explained by complete loss of glomerular immune complex deposits. Rather, TACI deletion specifically reduced endocapillary, but not mesangial, immune deposits. Notably, although excess BAFF promoted widespread breaks in B cell tolerance, BAFF-transgenic antibodies were enriched for RNA- relative to DNA-associated autoantigen reactivity. These RNA-associated autoantibody specificities were specifically reduced by TACI or Toll-like receptor 7 deletion. Thus, our study provides important insights into the autoantibody specificities driving proliferative lupus nephritis, and suggests that TACI inhibition may be novel and effective treatment strategy in lupus nephritis.


Subject(s)
Autoantibodies/blood , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Lupus Nephritis/genetics , Ribonucleoproteins/immunology , Transmembrane Activator and CAML Interactor Protein/genetics , Albuminuria/genetics , Albuminuria/urine , Animals , B-Cell Activating Factor/blood , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Creatinine/urine , Disease Progression , Female , Hypergammaglobulinemia/genetics , Immunoglobulins/blood , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
13.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978795

ABSTRACT

Foxp3+ Tregs possess potent immunosuppressive activity, which is critical for maintaining immune homeostasis and self-tolerance. Defects in Treg development or function result in inadvertent immune activation and autoimmunity. Despite recent advances in Treg biology, we still do not completely understand the molecular and cellular mechanisms governing the development and suppressive function of these cells. Here, we have demonstrated an essential role of the dedicator of cytokinesis 8 (DOCK8), guanine nucleotide exchange factors required for cytoskeleton rearrangement, cell migration, and immune cell survival in controlling Treg fitness and their function. Treg-specific DOCK8 deletion led to spontaneous multiorgan inflammation in mice due to uncontrolled T cell activation and production of proinflammatory cytokines. In addition, we show that DOCK8-deficient Tregs are defective in competitive fitness and in vivo suppressive function. Furthermore, DOCK8 controls IL-2 signaling, crucial for maintenance and competitive fitness of Tregs, via a STAT5-dependent manner. Our study provides potentially novel insights into the essential function of DOCK8 in Tregs and immune regulation, and it explains the autoimmune manifestations associated with DOCK8 deficiency.


Subject(s)
Guanine Nucleotide Exchange Factors/immunology , Interleukin-2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Guanine Nucleotide Exchange Factors/deficiency , Immune Tolerance/immunology , Inflammation/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation/immunology , Mice, Knockout , STAT5 Transcription Factor/immunology , Signal Transduction/immunology
14.
J Exp Med ; 214(11): 3207-3217, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-28899868

ABSTRACT

Recent studies have identified critical roles for B cells in triggering autoimmune germinal centers (GCs) in systemic lupus erythematosus (SLE) and other disorders. The mechanisms whereby B cells facilitate loss of T cell tolerance, however, remain incompletely defined. Activated B cells produce interleukin 6 (IL-6), a proinflammatory cytokine that promotes T follicular helper (TFH) cell differentiation. Although B cell IL-6 production correlates with disease severity in humoral autoimmunity, whether B cell-derived IL-6 is required to trigger autoimmune GCs has not, to our knowledge, been addressed. Here, we report the unexpected finding that a lack of B cell-derived IL-6 abrogates spontaneous GC formation in mouse SLE, resulting in loss of class-switched autoantibodies and protection from systemic autoimmunity. Mechanistically, B cell IL-6 production was enhanced by IFN-γ, consistent with the critical roles for B cell-intrinsic IFN-γ receptor signals in driving autoimmune GC formation. Together, these findings identify a key mechanism whereby B cells drive autoimmunity via local IL-6 production required for TFH differentiation and autoimmune GC formation.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Interleukin-6/immunology , Animals , Autoantibodies/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cells, Cultured , Flow Cytometry , Germinal Center/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
15.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553952

ABSTRACT

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Subject(s)
Autoimmunity/genetics , Cardiovirus Infections/genetics , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/genetics , Adolescent , Adult , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/immunology , Blotting, Southern , Cardiovirus Infections/immunology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Encephalomyocarditis virus/immunology , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Immunoblotting , Interferon-Induced Helicase, IFIH1/immunology , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Diseases/genetics , Virus Diseases/immunology , Young Adult
16.
Oncotarget ; 7(40): 64726-64742, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27556515

ABSTRACT

Anti-chlamydial immunity involves efficient presentation of antigens (Ag) to effector cells resulting in Ag-specific immune responses. There is limited information on inherent underlying mechanisms regulating these events. Previous studies from our laboratory have established that select microRNAs (miRs) function as molecular regulators of immunity in Chlamydia muridarum (Cm) genital infection. In this report, we investigated immune cell type-specific miRs, i.e. miR-155 and -182, and the role in Ag-specific immunity. We observed significant up-regulation of miR-155 in C57BL/6 bone marrow derived dendritic cells (BMDC), and miR-182 in splenic Ag-specific CD4+ T-cells. Using mimics and inhibitors, we determined that miR-155 contributed to BMDC activation following Cm infection. Co-cultures of miR-155 over-expressed in BMDC and miR-182 over-expressed in Ag-specific CD4+ T-cells, or miR-155-/- BMDC with miR-182 inhibitor treated Ag-specific CD4+ T-cells, resulted in IFN-γ production comparable to Ag-specific CD4+ T-cells isolated from Cm infected mice. Additionally, miR-182 was significantly up-regulated in intranasally vaccinated mice protected against Cm infection. In vivo depletion of miR-182 resulted in reduction in Ag-specific IFN-γ and genital pathology in Cm infected mice. To the best of our knowledge, this is the first study to report an interaction of miR-155 (in Cm infected DC) and miR-182 (in CD4+ T-cell) resulting in Ag specific immune responses against genital Cm.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Genitalia/immunology , MicroRNAs/genetics , Animals , Antigen Presentation , Cells, Cultured , Female , Genitalia/microbiology , Humans , Immunity , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Up-Regulation
17.
J Exp Med ; 213(5): 733-50, 2016 05 02.
Article in English | MEDLINE | ID: mdl-27069113

ABSTRACT

Dysregulated germinal center (GC) responses are implicated in the pathogenesis of human autoimmune diseases, including systemic lupus erythematosus (SLE). Although both type 1 and type 2 interferons (IFNs) are involved in lupus pathogenesis, their respective impacts on the establishment of autoimmune GCs has not been addressed. In this study, using a chimeric model of B cell-driven autoimmunity, we demonstrate that B cell type 1 IFN receptor signals accelerate, but are not required for, lupus development. In contrast, B cells functioning as antigen-presenting cells initiate CD4(+) T cell activation and IFN-γ production, and strikingly, B cell-intrinsic deletion of the IFN-γ receptor (IFN-γR) abrogates autoimmune GCs, class-switched autoantibodies (auto-Abs), and systemic autoimmunity. Mechanistically, although IFN-γR signals increase B cell T-bet expression, B cell-intrinsic deletion of T-bet exerts an isolated impact on class-switch recombination to pathogenic auto-Ab subclasses without impacting GC development. Rather, in both mouse and human B cells, IFN-γ synergized with B cell receptor, toll-like receptor, and/or CD40 activation signals to promote cell-intrinsic expression of the GC master transcription factor, B cell lymphoma 6 protein. Our combined findings identify a novel B cell-intrinsic mechanism whereby IFN signals promote lupus pathogenesis, implicating this pathway as a potential therapeutic target in SLE.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Lupus Erythematosus, Systemic/immunology , Proto-Oncogene Proteins c-bcl-6/immunology , Receptors, Interferon/immunology , Signal Transduction/immunology , Animals , Autoantibodies/immunology , B-Lymphocytes/pathology , Germinal Center/pathology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/genetics , Receptors, Interferon/genetics , Signal Transduction/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Interferon gamma Receptor
18.
J Immunol ; 196(9): 3525-31, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27022196

ABSTRACT

Mice overexpressing B cell activating factor of the TNF family (BAFF) develop systemic autoimmunity characterized by class-switched anti-nuclear Abs. Transmembrane activator and CAML interactor (TACI) signals are critical for BAFF-mediated autoimmunity, but the B cell developmental subsets undergoing TACI-dependent activation in settings of excess BAFF remain unclear. We report that, although surface TACI expression is usually limited to mature B cells, excess BAFF promotes the expansion of TACI-expressing transitional B cells. TACI(+) transitional cells from BAFF-transgenic mice are characterized by an activated, cycling phenotype, and the TACI(+) cell subset is specifically enriched for autoreactivity, expresses activation-induced cytidine deaminase and T-bet, and exhibits evidence of somatic hypermutation. Consistent with a potential contribution to BAFF-mediated humoral autoimmunity, TACI(+) transitional B cells from BAFF-transgenic mice spontaneously produce class-switched autoantibodies ex vivo. These combined findings highlight a novel mechanism through which BAFF promotes humoral autoimmunity via direct, TACI-dependent activation of transitional B cells.


Subject(s)
Autoantibodies/biosynthesis , B-Cell Activating Factor/metabolism , Precursor Cells, B-Lymphoid/immunology , Transmembrane Activator and CAML Interactor Protein/metabolism , Animals , Autoantibodies/immunology , Autoimmunity , B-Cell Activating Factor/genetics , B-Lymphocyte Subsets/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Precursor Cells, B-Lymphoid/physiology , Transmembrane Activator and CAML Interactor Protein/genetics
19.
Immunology ; 145(4): 534-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25865776

ABSTRACT

The hallmark of chlamydial infection is the development of upper genital pathology in the form of hydrosalpinx and oviduct and/or tubal dilatation. Although molecular events leading to genital tissue presentation and cellular architectural remodelling are unclear, early-stage host immune responses are believed to contribute to these long-term sequelae. Recently, we reported the contribution of selected infection-associated microRNAs (miRs) in the generation of host immunity at early-stage infection (day 6 after intravaginal Chlamydia muridarum challenge in C57BL/6 mice). In this report, we describe the contribution of an infection-associated microRNA, i.e. miR-214, to host immunity. Chlamydia muridarum infection in the C57BL/6 mouse genital tract significantly down-regulated miR-214 while up-regulating intracellular adhesion molecule 1 (ICAM1) gene expression. These in vivo observations were confirmed by establishing direct regulation of ICAM-1 by miR-214 in ex vivo genital cell cultures in the presence of miR-214 mimic and inhibitor. Because, ICAM-1 contributes to recruitment of neutrophils following infection, we also demonstrated that alteration of ICAM1 by miR-214 in interleukin-17A-deficient (IL-17A(-/-) ) mice correlated with reduction of neutrophils infiltrating genital tissue at day 6 after challenge. Additionally, these early-stage events resulted in significantly decreased genital pathology in IL-17A(-/-) mice compared with C57BL/6 mice. This report provides evidence for early-stage regulation of ICAM1 by microRNAs, resulting in reduction of genital pathology associated with chlamydial infection.


Subject(s)
Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Down-Regulation/immunology , Intercellular Adhesion Molecule-1/immunology , MicroRNAs/immunology , Reproductive Tract Infections/immunology , Up-Regulation/immunology , Animals , Chlamydia Infections/genetics , Chlamydia Infections/pathology , Chlamydia muridarum/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Reproductive Tract Infections/genetics , Reproductive Tract Infections/pathology
20.
Am J Reprod Immunol ; 73(2): 126-40, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24976530

ABSTRACT

PROBLEM: Chlamydia trachomatis (CT) is the leading sexually transmitted bacterial infection in humans and is associated with reproductive tract damage. However, little is known about the involvement and regulation of microRNAs (miRs) in genital CT. METHODS: We analyzed miRs in the genital tract (GT) following C. muridarum (murine strain of CT) challenge of wild type (WT) and CD4(+) T-cell deficient (CD4(-/-)) C57BL/6 mice at days 6 and 12 post-challenge. RESULTS: At day 6, miRs significantly downregulated in the lower GT were miR-125b-5p, -16, -214, -23b, -135a, -182, -183, -30c, and -30e while -146 and -451 were significantly upregulated, profiles not exhibited at day 12 post-bacterial challenge. Significant differences in miR-125b-5p (+5.06-fold change), -135a (+4.9), -183 (+7.9), and -182 (+3.2) were observed in C. muridarum-infected CD4(-/-) compared to WT mice. In silico prediction and mass spectrometry revealed regulation of miR-135a and -182 and associated proteins, that is, heat-shock protein B1 and alpha-2HS-glycoprotein. CONCLUSION: This study provides evidence on regulation of miRs following genital chlamydial infection suggesting a role in pathogenesis and host immunity.


Subject(s)
Chlamydia Infections/genetics , Genitalia, Female/metabolism , MicroRNAs/metabolism , Animals , Antigens, Bacterial/immunology , Bacterial Load , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Chlamydia muridarum , Endopeptidases/immunology , Female , Genitalia, Female/immunology , Genitalia, Female/microbiology , HeLa Cells , Humans , Immunization , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...