Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 203(4): 813-824, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31270149

ABSTRACT

Tick-borne allergies are a growing public health concern and have been associated with the induction of IgE-mediated food allergy to red meat. However, despite the increasing prevalence of tick bite-induced allergies, the mechanisms by which cutaneous exposure to ticks leads to sensitization and the production of IgE Abs are poorly understood. To address this question, an in vivo approach was used to characterize the IgE response to lone star tick proteins administered through the skin of mice. The results demonstrated that tick sensitization and challenge induced a robust production of IgE Abs and supported a role for IgE-mediated hypersensitivity reactions in sensitized animals following oral administration of meat. The induction of IgE responses was dependent on cognate CD4+ T cell help during both the sensitization phase and challenge phase with cutaneous tick exposure. In addition, IgE production was dependent on B cell-intrinsic MyD88 expression, suggesting an important role for TLR signaling in B cells to induce IgE responses to tick proteins. This model of tick-induced IgE responses could be used to study the factors within tick bites that cause allergies and to investigate how sensitization to food Ags occurs through the skin that leads to IgE production.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Food Hypersensitivity/immunology , Insect Proteins/immunology , Myeloid Differentiation Factor 88/immunology , Tick Bites/immunology , Allergens/immunology , Animals , Immunoglobulin E/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/immunology , Ticks/immunology
2.
J Infect Dis ; 214(1): 130-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26917573

ABSTRACT

BACKGROUND: The elderly host is highly susceptible to severe disease and treatment failure in Clostridium difficile infection (CDI). We investigated how treatment with vancomycin in the aged host influences systemic and intestinal humoral responses and select intestinal microbiota. METHODS: Young (age, 2 months) and aged (age, 18 months) C57BL/6 mice were infected with VPI 10463 after exposure to broad-spectrum antibiotics. Vancomycin was given 24 hours after infection, and treatment was continued for 5 days. At select time points, specimens of serum and intestinal tissue and contents were collected for histopathologic analysis, to measure antibody levels and the pathogen burden, and to determine the presence and levels of select intestinal microbiota and C. difficile toxin. RESULTS: Levels of disease severity, relapse, and mortality were increased, and recovery from infection was slower in aged mice compared to young mice. Serum levels of immunoglobulin M, immunoglobulin A, and immunoglobulin G against C. difficile toxin A were depressed in aged mice, and vancomycin treatment reduced antibody responses in both age groups. While baseline levels of total bacterial load, Bacteroidetes, Firmicutes, and Enterobacteriaceae were mostly similar, aged mice had a significant change in the Firmicutes to Bacteroidetes ratio with vancomycin treatment. CONCLUSIONS: Vancomycin treatment decreases the systemic humoral response to CDI. Increased mortality from and recurrence of CDI in the aged host are associated with an impaired humoral response and a greater susceptibility to vancomycin-induced alteration of intestinal microbiota.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/drug effects , Clostridium Infections/drug therapy , Gastrointestinal Microbiome/drug effects , Immunity, Humoral/drug effects , Vancomycin/therapeutic use , Age Factors , Animals , Humans , Mice, Inbred C57BL , Models, Animal
3.
Arthritis Rheumatol ; 67(3): 773-84, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25385309

ABSTRACT

OBJECTIVE: Follicular helper T (Tfh) cells are critical for the development of protective antibodies via germinal center (GC) B cell responses; however, uncontrolled Tfh cell expansion activates autoreactive B cells to produce antibodies that cause autoimmunity. The mechanisms that control Tfh cell homeostasis remain largely unknown. The aim of this study was to determine the contribution of BAFF to Tfh cell responses in autoimmunity. METHODS: We analyzed the properties of Tfh cells in lupus-prone mice sufficient or deficient in BCMA. Adoptive transfer studies and mixed bone marrow chimeras were used to test BCMA signaling in T cells. We assessed BAFF stimulation of Tfh cells through in vitro cell cocultures and in vivo depletion studies using flow cytometry. RESULTS: In Nba2 mice, Tfh cells expressed the BAFF receptors BCMA and B lymphocyte stimulator receptor 3 (BR-3) and accumulated in the spleen when BCMA was absent. BCMA deficiency in T cells promoted the expansion of Tfh cells, GC formation, autoantibody production, and interferon-γ (IFNγ) production by Tfh cells through BR-3. IFNγ-producing Tfh cells increased BAFF expression in dendritic cells. Blocking BAFF or IFNγ in vivo reduced Tfh cell accumulation and reduced autoimmunity in BCMA-deficient animals. Moreover, circulating Tfh-like cells that expressed BR-3 (but not BCMA) were elevated in patients with systemic lupus erythematosus, and this correlated with serum BAFF and IFNγ levels. CONCLUSION: In Nba2 mice, BCMA negatively regulates Tfh cell expansion, while BAFF signaling through BR-3 promotes Tfh cell accumulation. Our findings suggest that the balance between BCMA and BR-3 signaling in Tfh cells serves as a checkpoint of immune tolerance.


Subject(s)
Autoimmunity/immunology , B-Cell Activating Factor/physiology , Interferon-gamma/metabolism , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Animals , B-Cell Activation Factor Receptor/metabolism , B-Cell Maturation Antigen/metabolism , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Real-Time Polymerase Chain Reaction , Terpenes/pharmacology
4.
PLoS One ; 9(7): e102284, 2014.
Article in English | MEDLINE | ID: mdl-25010693

ABSTRACT

Despite increased frequencies of neutrophils found in autoimmune diseases such as systemic lupus erythematosus (SLE), how they contribute to disease pathogenesis and the mechanisms that affect the accumulation of neutrophils are poorly understood. The aim of this study was to identify factors in autoantibody-mediated autoimmunity that controls the accumulation of spleen resident neutrophils and to determine whether neutrophils contribute to abnormal B cell responses. Increased levels of the cytokine BAFF have been linked to loss of B cell tolerance in autoimmunity, but the cellular source responsible for excess BAFF is unknown. B cell maturation antigen (BCMA) is a receptor for BAFF and is critical for the survival of bone marrow plasma cells. Paradoxically, BCMA deficiency exacerbates the formation of autoantibody-secreting plasma cells in spleens of lupus-prone mice and the reasons for this effect are not understood. Here we analyzed the phenotype, localization and function of neutrophils in spleens of healthy mice and congenic lupus-prone mice, and compared mice sufficient or deficient in BCMA expression. Neutrophils were found to be significantly increased in frequency and activation status in spleens of lupus-prone mice when BCMA was absent. Furthermore, neutrophils localized within T cell zones and enhanced CD4(+) T cell proliferation and IFNγ production through the production of BAFF. Reduced BAFF and IFNγ serum levels, decreased frequencies of IFNγ-producing T cells, germinal center B cells, and autoantibody production after neutrophil depletion indicated the involvement of neutrophils in these autoimmune traits. Thus, we have identified a novel role for BCMA to control excess BAFF production in murine lupus through restraining the accumulation of BAFF-producing neutrophils. Our data suggests that devising therapeutic strategies to reduce neutrophils in autoimmunity may decrease BAFF levels and ameliorate disease.


Subject(s)
Autoimmunity/immunology , B-Cell Activating Factor/metabolism , B-Cell Maturation Antigen/metabolism , Lupus Erythematosus, Systemic/genetics , Animals , Autoimmunity/genetics , B-Cell Activating Factor/immunology , B-Cell Maturation Antigen/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Neutrophils/immunology , Neutrophils/pathology , Spleen/immunology , Spleen/pathology , T-Lymphocytes/immunology
5.
J Immunol ; 186(11): 6136-47, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21536804

ABSTRACT

Systemic lupus erythematosus and its preclinical lupus-prone mouse models are autoimmune disorders involving the production of pathogenic autoantibodies. Genetic predisposition to systemic lupus erythematosus results in B cell hyperactivity, survival of self-reactive B cells, and differentiation to autoantibody-secreting plasma cells (PCs). These corrupt B cell responses are, in part, controlled by excess levels of the cytokine BAFF that normally maintains B cell homeostasis and self-tolerance through limited production. B cell maturation Ag (BCMA) is a receptor for BAFF that, under nonautoimmune conditions, is important for sustaining enduring Ab protection by mediating survival of long-lived PCs but is not required for B cell maturation and homeostasis. Through analysis of two different lupus-prone mouse models deficient in BCMA, we identify BCMA as an important factor in regulating peripheral B cell expansion, differentiation, and survival. We demonstrate that a BCMA deficiency combined with the lpr mutation or the murine lupus susceptibility locus Nba2 causes dramatic B cell and PC lymphoproliferation, accelerated autoantibody production, and early lethality. This study unexpectedly reveals that BCMA works to control B cell homeostasis and self-tolerance in systemic autoimmunity.


Subject(s)
Autoimmunity/immunology , B-Cell Maturation Antigen/deficiency , Lupus Erythematosus, Systemic/immunology , Lymphoproliferative Disorders/immunology , Animals , B-Cell Activating Factor/immunology , B-Cell Activating Factor/metabolism , B-Cell Maturation Antigen/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Female , Flow Cytometry , Immunoblotting , Kaplan-Meier Estimate , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/immunology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mutation , Plasma Cells/immunology , Plasma Cells/metabolism , Self Tolerance/immunology , Spleen/immunology , Spleen/metabolism , Spleen/pathology
6.
J Immunol ; 184(2): 775-86, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20018631

ABSTRACT

Autoantibodies are of central importance in the pathogenesis of Ab-mediated autoimmune disorders. The murine lupus susceptibility locus Nba2 on chromosome 1 and the syntenic human locus are associated with a loss of immune tolerance that leads to antinuclear Ab production. To identify gene intervals within Nba2 that control the development of autoantibody-producing B cells and to determine the cellular components through which Nba2 genes accomplish this, we generated congenic mice expressing various Nba2 intervals where genes for the FcgammaR, SLAM, and IFN-inducible families are encoded. Analysis of congenic strains demonstrated that the FcgammaR and SLAM intervals independently controlled the severity of autoantibody production and renal disease, yet are both required for lupus susceptibility. Deregulated homeostasis of terminally differentiated B cells was found to be controlled by the FcgammaR interval where FcgammaRIIb-mediated apoptosis of germinal center B cells and plasma cells was impaired. Increased numbers of activated plasmacytoid dendritic cells that were distinctly CD19+ and promoted plasma cell differentiation via the proinflammatory cytokines IL-10 and IFNalpha were linked to the SLAM interval. These findings suggest that SLAM and FcgammaR intervals act cooperatively to influence the clinical course of disease through supporting the differentiation and survival of autoantibody-producing cells.


Subject(s)
Antigens, CD/genetics , Lupus Erythematosus, Systemic/genetics , Receptors, Cell Surface/genetics , Receptors, IgG/genetics , Animals , Apoptosis , Autoantibodies/biosynthesis , B-Lymphocytes/pathology , Cell Differentiation , Cytokines/physiology , Disease Progression , Genetic Predisposition to Disease/genetics , Kidney Diseases , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Congenic , Plasma Cells/pathology , Signaling Lymphocytic Activation Molecule Family Member 1
7.
J Immunol ; 181(11): 7537-49, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017943

ABSTRACT

Deregulation of the c-Myc oncogene is tightly associated with human and murine plasma cell (PC) neoplasms. Through the analysis of Ag-specific B cell responses in mice where Myc is targeted to the Igh Calpha locus, we show here that c-Myc dramatically impairs the primary and secondary Ab response. This impairment is differentiation stage specific, since germinal center B cell formation, affinity maturation, and class switch recombination were intact. Examination of PC viability revealed that c-Myc triggered apoptosis only upon final maturation when Ab is secreted and is resistant to the survival factor BAFF (B cell-activating factor belonging to the TNF family). In contrast, PC precursors (PC(pre)) that ultimately give rise to mature PCs survived normally and vigorously expanded with BAFF signaling. We further show that c-Myc also facilitates the apoptosis of memory B cells. Thus, Calpha-Myc controls both cellular arms of long-lived B cell immunity than previously anticipated. Only when deregulation of c-Myc was combined with enforced Bcl-x(L) expression were mature PCs able to survive in response to BAFF. These data indicate that the survival requirements for tumor-susceptible PC(pre) and PCs are distinct and that tumor progression likely develops as PC(pre) transition to functional PCs when apoptotic pathways such as members of the Bcl-2 family are disabled.


Subject(s)
Apoptosis/immunology , Immunologic Memory , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology , Proto-Oncogene Proteins c-myc/immunology , Animals , Antibody Formation/genetics , Antibody Formation/immunology , Apoptosis/genetics , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , Cell Survival/genetics , Cell Survival/immunology , Gene Targeting/methods , Germinal Center/immunology , Humans , Immunologic Memory/genetics , Mice , Mice, Transgenic , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Plasmacytoma/genetics , Plasmacytoma/immunology , Proto-Oncogene Proteins c-myc/genetics , Quantitative Trait Loci/genetics , Quantitative Trait Loci/immunology , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology , bcl-X Protein/genetics , bcl-X Protein/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...