Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Tuberculosis (Edinb) ; 124: 101978, 2020 09.
Article in English | MEDLINE | ID: mdl-32781412

ABSTRACT

BACKGROUND: While T cell responses to Mycobacterium tuberculosis (Mtb) have been extensively studied, the role of B-cells and antibodies are less well characterised. The aim of this study was to assess levels of Mtb-specific IgG + plasmablasts across the Mtb infection spectrum. METHODS: Patients with active TB were analysed at baseline and 6 months of therapy (n = 20).Their exposed household contacts (HHC) included individuals with latent TB infection (LTBI; n = 20); evident at baseline; individuals with a negative Tuberculin Skin Test (TST) at baseline who became; positive at 6 months (converters; n = 11) and those who remained negative (non-converters; n = 10). An e x-vivo B-cell ELISPOT was performed to analyse plasmablast responses. RESULTS: Frequencies of ESAT-6/CFP-10 (EC)- but not Whole Cell Lysate (WCL)-specific plasmablasts were significantly higher in patients with active TB pre-treatment compared to post-treatment (p = 0.002) and compared to HHC with LTBI (p < 0.0001). Conversely, total IgG + plasmablasts were significantly decreased in TB patients at baseline. No difference was seen in levels of plasmablasts between TST converters and non-converters at baseline. CONCLUSIONS: We show that EC-specific plasmablast levels are differentially modulated during TB infection and disease, with levels highest during active TB. These data provide new insights into TB biomarker development and avenues for novel immune interventions.


Subject(s)
B-Lymphocyte Subsets/microbiology , Mycobacterium tuberculosis/immunology , Tuberculosis/microbiology , Adolescent , Adult , Antigens, Bacterial/immunology , Antitubercular Agents/therapeutic use , B-Lymphocyte Subsets/immunology , Bacterial Proteins/immunology , Enzyme-Linked Immunospot Assay , Female , Gambia , Host-Pathogen Interactions , Humans , Male , Middle Aged , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Pilot Projects , Predictive Value of Tests , Time Factors , Treatment Outcome , Tuberculosis/diagnosis , Tuberculosis/drug therapy , Tuberculosis/immunology , Young Adult
2.
Int J Immunopathol Pharmacol ; 32: 2058738418792310, 2018.
Article in English | MEDLINE | ID: mdl-30114952

ABSTRACT

A better knowledge of factors predicting the development of sepsis in patients hospitalized in intensive care unit (ICU) might help deploy more targeted preventive and therapeutic strategies. In addition to the known clinical and demographic predictors of septic syndromes, in this study, we investigated whether measuring T and B lymphocyte subsets upon admission in the ICU may help individualize the prediction of ensuing sepsis during ICU stay. Between May 2015 and December 2016, we performed a prospective cohort study evaluating peripheral blood lymphocyte T-CD4+ (T-helper cells), T-CD8+ (cytotoxic T-cells), T-CD56 + (natural killer cells), and T-CD19+ (B-lymphocytes), using flow cytometry on blood samples collected 2 days after admission in the ICU. We enrolled 176 patients, 65.3% males, with mean age of 61.1 ± 15.4 years. At univariate analyses, higher percentages of CD19 B-cells were significantly associated with ensuing sepsis (20.5% (15.7-27.7)% vs 16.9% (11.3-22)%, P = 0.0001), whereas median interquartile range (IQR) proportions of CD4 T-cells (41.2% (33.4-50.6)% vs 40% (35-47)%, P = 0.5), CD8 T-cells (21.1% (15.8-28.2)% vs 19.6% (14.6-25.1)%, P = 0.2) and CD56 T-cells (1.7% (0.9-3.1)% vs 1.45% (0.7-2.3)%, P = 0.4) did not reveal any significant association. An unexpected, highly significant inverse correlation of CD8 T-cells and CD19 B-cells proportions, however, was observed, suggesting that patients with lower CD19 and higher CD8 proportions might be somehow protected from ensuing sepsis. We therefore studied the ability of the CD8/CD19 ratio to predict ensuing sepsis in our sample. In final models of multivariate logistic regression, the following independent associations were found: previous antibiotic exposure (odds ratio (OR): 3.8 (95% confidence interval (CI): 1.35-10.87), P = 0.01), isolation of at least one multi-drug resistant organism at any time during ICU stay (OR: 8.4 (95% CI: 3.47-20.6), P < 0.0001), decreasing age (OR: 0.9 (95% CI: 0.93-0.99), P = 0.02) and a CD8/CD19 ratio >2.2 (OR: 10.3 (95% CI: 1.91-55.36), P = 0.007). Our data provide preliminary evidence that immune characterization of critically ill patients on ICU admission may help personalize the prediction of ensuing sepsis during their ICU stay. Further polycentric evaluation of the true potential of this new tool is warranted.


Subject(s)
B-Lymphocyte Subsets/immunology , Intensive Care Units , Patient Admission , Sepsis/immunology , T-Lymphocyte Subsets/immunology , APACHE , Age Factors , Aged , Anti-Bacterial Agents/adverse effects , B-Lymphocyte Subsets/microbiology , Biomarkers/blood , Female , Flow Cytometry , Host-Pathogen Interactions , Humans , Immunophenotyping/methods , Lymphocyte Count , Male , Middle Aged , Phenotype , Predictive Value of Tests , Preliminary Data , Prognosis , Prospective Studies , Risk Factors , Sepsis/blood , Sepsis/diagnosis , Sepsis/microbiology , Sex Factors , T-Lymphocyte Subsets/microbiology
3.
Infect Immun ; 83(12): 4826-36, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26438792

ABSTRACT

Despite Coxiella burnetii being an obligate intracellular bacterial pathogen, our recent study demonstrated that B cells play a critical role in vaccine-induced immunity to C. burnetii infection by producing protective antibodies. However, the role of B cells in host defense against primary C. burnetii infection remains unclear. In this study, we investigated whether B cells play an important role in host defense against primary C. burnetii infection. The results showed that peritoneal B cells were able to phagocytose virulent C. burnetii bacteria and form Coxiella-containing vacuoles (CCVs) and that C. burnetii can infect and replicate in peritoneal B1a subset B cells in vitro, demonstrating a potential role for peritoneal B cells in host defense against C. burnetii infection in vivo. In addition, the results showing that B1a cells secreted a high level of interleukin-10 (IL-10) in response to C. burnetii infection in vitro suggest that B1a cells may play an important role in inhibiting the C. burnetii infection-induced inflammatory response. The observation that adoptive transfer of peritoneal B cells did not significantly affect the severity of C. burnetii infection-induced diseases in both severe combined immunity-deficient (SCID) and µMT mice indicates that peritoneal B cells alone may not be able to control C. burnetii infection. In contrast, our finding that C. burnetii infection induced more-severe splenomegaly and a higher bacterial burden in the spleens of B1a cell-deficient Bruton's tyrosine kinase x-linked immunity-deficient (BTK(xid)) mice than in their wild-type counterparts further suggests that B1a cells play an important role in host defense against primary C. burnetii infection.


Subject(s)
B-Lymphocyte Subsets/immunology , Coxiella burnetii/genetics , Host-Pathogen Interactions/immunology , Q Fever/immunology , Spleen/immunology , Adoptive Transfer , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocyte Subsets/microbiology , B-Lymphocyte Subsets/pathology , B-Lymphocyte Subsets/transplantation , Coxiella burnetii/pathogenicity , Female , Gene Expression , Immunity, Humoral , Interleukin-10/genetics , Interleukin-10/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Phagocytosis , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Q Fever/microbiology , Q Fever/pathology , Spleen/microbiology , Spleen/pathology
4.
Immunol Lett ; 167(2): 131-40, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26292028

ABSTRACT

Salmonella persists for a long time in B cells; however, the mechanism(s) through which infected B cells avoid effector CD8 T cell responses has not been characterized. In this study, we show that Salmonella infects and survives within all B1 and B2 cell subpopulations. B cells are infected with a Salmonella typhimurium strain expressing an ovalbumin (OVA) peptide (SIINFEKL) to evaluate whether B cells process and present Salmonella antigens in the context of MHC-I molecules. Our data showed that OVA peptides are presented by MHC class I K(b)-restricted molecules and the presented antigen is generated through proteasomal degradation and vacuolar processing. In addition, Salmonella-infected B cells express co-stimulatory molecules such as CD40, CD80, and CD86 as well as inhibitory molecules such as PD-L1. Thus, the cross-presentation of Salmonella antigens and the expression of activation molecules suggest that infected B cells are able to prime and activate specific CD8(+) T cells. However, the Salmonella infection-stimulated expression of PD-L1 suggests that the PD-1/PD-L1 pathway may be involved in turning off the cytotoxic effector response during Salmonella persistent infection, thereby allowing B cells to become a reservoir for the bacteria.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-H1 Antigen/genetics , Gene Expression Regulation , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella/immunology , Animals , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/microbiology , B-Lymphocytes/microbiology , B7-H1 Antigen/metabolism , Biological Transport , Cross-Priming/immunology , Disease Models, Animal , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Lymphocyte Activation/immunology , Mice , Salmonella typhimurium/immunology , Vacuoles/immunology , Vacuoles/metabolism
5.
Immunology ; 146(2): 243-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26095067

ABSTRACT

The immune system plays an important role in the pathogenesis of pulmonary tuberculosis-type 2 diabetes mellitus (PTB-DM) co-morbidity. However, the phenotypic profile of leucocyte subsets at homeostasis in individuals with active or latent tuberculosis (LTB) with coincident diabetes is not known. To characterize the influence of diabetes on leucocyte phenotypes in PTB or LTB, we examined the frequency (Fo ) of leucocyte subsets in individuals with TB with (PTB-DM) or without (PTB) diabetes; individuals with latent TB with (LTB-DM) or without (LTB) diabetes and non-TB-infected individuals with (NTB-DM) or without (NTB) diabetes. Coincident DM is characterized by significantly lower Fo of effector memory CD4(+) T cells in LTB individuals. In contrast, DM is characterized by significantly lower Fo of effector memory CD8(+) T cells and significantly higher Fo of central memory CD8(+) T cells in PTB individuals. Coincident DM resulted in significantly higher Fo of classical memory B cells in PTB and significantly higher Fo of activated memory and atypical B cells in LTB individuals. Coincident DM resulted in significantly lower Fo of classical and intermediate monocytes in PTB, LTB and NTB individuals. Finally, DM resulted in significantly lower Fo of myeloid and plasmacytoid dendritic cells in PTB, LTB and NTB individuals. Our data reveal that coincident diabetes alters the cellular subset distribution of T cells, B cells, dendritic cells and monocytes in both individuals with active TB and those with latent TB, thus potentially impacting the pathogenesis of this co-morbid condition.


Subject(s)
B-Lymphocyte Subsets/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Type 2/immunology , Latent Tuberculosis/immunology , Monocytes/immunology , T-Lymphocyte Subsets/immunology , Tuberculosis/immunology , Adult , Aged , B-Lymphocyte Subsets/microbiology , Biomarkers/analysis , Comorbidity , Dendritic Cells/microbiology , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Female , Humans , Immunophenotyping , India/epidemiology , Latent Tuberculosis/diagnosis , Latent Tuberculosis/epidemiology , Latent Tuberculosis/microbiology , Lymphocyte Count , Male , Middle Aged , Monocytes/microbiology , Phenotype , T-Lymphocyte Subsets/microbiology , Tuberculosis/diagnosis , Tuberculosis/epidemiology , Tuberculosis/microbiology , Young Adult
6.
Neurología (Barc., Ed. impr.) ; 30(5): 276-282, jun. 2015. ilus, tab, graf
Article in Spanish | IBECS | ID: ibc-139066

ABSTRACT

Antecedentes: La neuromielitis óptica (NMO) es una enfermedad predominantemente humoral mediada por anticuerpos IgG-NMO/AQP-4. Sin embargo, no se conoce bien la contribución de las diferentes subpoblaciones de células B y T en su patogenia o en la respuesta a los tratamientos. Objetivos: Describir los cambios clínicos e inmunológicos asociados al tratamiento con inmunoglobulinas intravenosas (Ig-IV) y rituximab (RTX) en una paciente con un brote grave de NMO y síntesis intratecal de IgG-NMO/AQP-4 que no había respondido a metilprednisolona y recambio plasmático. Métodos: Se analizaron, de forma secuencial en el LCR y en la sangre periférica (SP), las subpoblaciones linfocitarias mediante citometría de flujo multiparamétrica y los IgG-NMO/AQP-4, antes y después del tratamiento con Ig-IV y RTX. Resultados: En el LCR antes del tratamiento, y comparado con la SP, predominaban las células T CD4+ y estaban menos representadas las T CD8+ y las B CD19+. Tras el tratamiento, el porcentaje de células T CD4+ se mantuvo alto, el de T CD8+ aumentó y el de B CD19+ disminuyó, aunque menos que en la SP. Al comparar los LCR se vio que tras la terapia el porcentaje de células T CD8+ memoria efectoras y efectoras había aumentado, y el de células B memoria IgM y el de células B con cambio de isotipos, disminuido. Los cambios observados fueron paralelos a la negativización de los IgG-NMO/AQP-4 y a la mejoría clínica. Conclusiones: Nuestros hallazgos confirman que, además de una respuesta inmunitaria humoral intratecal durante el brote de NMO, subpoblaciones específicas de células B y T participan en la modulación de la inflamación dentro y fuera del sistema nervioso central


Background: There is increasing evidence supporting that neuromyelitis optica (NMO) is an inflammatory humoral mediated disorder associated with NMO-IgG/AQP-4 antibodies. However, little is known about the subsets of B cells and T cells that contribute to the pathogenesis or therapy response. Objectives: To describe the clinical and immunological changes associated with intravenous immunoglobulins (IV-Igs) plus rituximab (RTX) in a patient with a severe acute attack of NMO and intrathecal synthesis of NMO-IgG/AQP-4, who previously did not respond to intravenous methylprednisolone and plasma exchange. Methods: We sequentially analysed the levels of NMO-IgG/AQP-4 by immunohistochemistry, and B and T cells subsets by multiparametric flow-cytometry, in the CSF and peripheral blood (PB), before and alter IV-Igs plus RTX therapy. Results: In the CSF before treatment, and compared with PB, there was a higher percentage of CD4+ T cells and a lower percentage of CD8+ T cells and CD19+ B cells. After therapy, the percentage of CD4+ T cells remained high, and that of CD8+ T cells increased. The observed decrease in the percentage of CD19+ B cells was lower than in the PB. When the CSF was compared, it was found that the percentage of effector-memory and effector CD8+ T cells had increased after therapy, and that of IgM memory B cells and switched-memory B cells decreased. The observed changes paralleled the decrease of NMO-IgG/AQP-4 results to negative and the clinical improvement. Conclusions: Our findings confirm that, besides intrathecal humoral immune response against AQP4, B and T cell subsets are involved in the modulation of inflammation within and outside the central nervous system


Subject(s)
Female , Humans , Lymphocyte Activation/genetics , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Pharmaceutical Preparations/administration & dosage , Neuromyelitis Optica/diagnosis , Lymphocyte Activation/physiology , B-Lymphocyte Subsets/classification , B-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/pathology , Pharmaceutical Preparations , Neuromyelitis Optica/metabolism , Epidemiology, Descriptive
7.
Int Immunol ; 27(10): 537-41, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25957266

ABSTRACT

Innate response activator (IRA) B cells are a subset of B-1a derived B cells that produce the growth factors granulocyte macrophage colony stimulating factor and IL-3. In mouse models of sepsis and pneumonia, B-1a B cells residing in serosal sites recognize bacteria, migrate to the spleen or lung, and differentiate to IRA B cells that then contribute to the host response by amplifying inflammation and producing polyreactive IgM. In atherosclerosis, IRA B cells accumulate in the spleen, where they promote extramedullary hematopoiesis and activate classical dendritic cells. In this review, we focus on the ontogeny and function of IRA B cells in acute and chronic inflammation.


Subject(s)
Atherosclerosis/immunology , B-Lymphocyte Subsets/immunology , Cell Lineage/immunology , Immunity, Innate , Pneumonia/immunology , Sepsis/immunology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , B-Lymphocyte Subsets/microbiology , B-Lymphocyte Subsets/pathology , Disease Models, Animal , Gene Expression Regulation/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunoglobulin M/biosynthesis , Immunoglobulin M/genetics , Interleukin-3/genetics , Interleukin-3/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Pneumonia/genetics , Pneumonia/microbiology , Pneumonia/pathology , Sepsis/genetics , Sepsis/microbiology , Sepsis/pathology , Signal Transduction , Spleen/immunology , Spleen/microbiology , Spleen/pathology
8.
Ann N Y Acad Sci ; 1362: 57-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25962322

ABSTRACT

Although evidence of the protective immunity conferred by B-1b cells (CD19(+) B220(+) IgM(hi) Mac1(+) CD5(-)) has been established, the mechanisms governing the maintenance and activation of B-1b cells following pathogen encounter remain unclear. B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) mediate their function in mature B cells through the BAFF receptor (BAFFR) and transmembrane activator and CAML interactor (TACI). BAFFR-deficient mice have lower numbers of B-1b cells, and this reduction is directly proportional to BAFFR levels. The generation of B-1b cells is also dependent on the strength of B cell receptor (BCR) signaling. Mice with impaired BCR signaling, such as X-linked immunodeficient (xid) mice, have B-1b cell deficiency, indicating that both BCR- and BAFFR-mediated signaling are critical for B-1b cell homeostasis. Borrelia hermsii induces expansion and persistence of B-1b cells in xid mice, and these B-1b cells provide a heightened protective response. Toll-like receptor (TLR)-mediated stimulation of xid B cells results in a significant increase in TACI expression and restoration of TACI-mediated functions. The activation of TLR signaling by B. hermsii and BCR/TLR costimulation-mediated upregulation of BAFFR and TACI on B-1b cells suggests that B-1b cell maintenance and function following bacterial exposure may depend on BAFFR- and TACI-mediated signaling. In fact, the loss of both BAFFR and TACI results in a greater impairment in anti-B. hermsii responses compared to deficiency of BAFFR or TACI alone.


Subject(s)
B-Cell Activation Factor Receptor/metabolism , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/microbiology , Transmembrane Activator and CAML Interactor Protein/metabolism , Animals , B-Cell Activating Factor/metabolism , Humans , Immunity, Humoral/physiology
9.
BMC Immunol ; 16: 15, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25884828

ABSTRACT

Recent studies on HIV infection have identified new human B-cell subsets with a potentially important impact on anti-viral immunity. Current work highlights the occurrence of similar B-cell alterations in other viral, bacterial, and parasitic infections, suggesting that common strategies have been developed by pathogens to counteract protective immunity. For this review, we have selected key examples of human infections for which B-cell alterations have been described, to highlight the similarities and differences in the immune responses to a variety of pathogens. We believe that further comparisons between these models will lead to critical progress in the understanding of B-cell mechanisms and will open new target avenues for therapeutic interventions.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Infections/immunology , Animals , B-Lymphocyte Subsets/microbiology , B-Lymphocyte Subsets/parasitology , B-Lymphocyte Subsets/virology , B-Lymphocytes/microbiology , B-Lymphocytes/parasitology , B-Lymphocytes/virology , Biological Therapy , Host-Parasite Interactions , Humans , Immune Evasion , Infections/therapy
10.
Dev Comp Immunol ; 51(1): 99-107, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25743381

ABSTRACT

Porcine ileum is populated with a high proportion of B cells but previous studies have shown that they are not developed there. While B cells prevail in the ileum even in germ-free animals, microbial colonization is a major factor that causes even a greater prevalence of B cells in the ileum and further differential representation of lymphoid cells throughout small intestine. Analysis of lymphoid subpopulations showed that the effector cells appear only after colonization. These include class-switched IgM(+)IgA(+) B cells, primed CD2(-)CD21(+) B cells, antibody-producing/memory CD2(+)CD21(-) B cells, and effector/memory CD4(+)CD8(+) αß Th cells. While colonization resulted in a uniform distribution of effector cells throughout the gut, it caused a decrease in the frequency of cytotoxic αß and CD2(+)CD8(+) γδ T cells. These results suggest that the ileum is a site where naive B cells expand presumably to increase antibody repertoire but the entire small intestine is immunofunctionally comparable.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Ileum/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Sus scrofa/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens, CD/metabolism , B-Lymphocyte Subsets/microbiology , B-Lymphocytes/microbiology , Bacteria/growth & development , Bacteria/immunology , Germ-Free Life/immunology , Ileum/microbiology , Immunoglobulin Class Switching , Immunologic Memory , Immunophenotyping , Lymphocyte Activation , Microbiota/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes, Cytotoxic/microbiology , T-Lymphocytes, Helper-Inducer/microbiology
11.
Comp Immunol Microbiol Infect Dis ; 37(3): 143-52, 2014 May.
Article in English | MEDLINE | ID: mdl-24766724

ABSTRACT

The present paper is an overview of the primary events that are associated with the histoplasmosis immune response in the murine model. Valuable data that have been recorded in the scientific literature have contributed to an improved understanding of the clinical course of this systemic mycosis, which is caused by the dimorphic fungus Histoplasma capsulatum. Data must be analyzed carefully, given that misinterpretation could be generated because most of the available information is based on experimental host-parasite interactions that used inappropriate proceedings, i.e., the non-natural route of infection with the parasitic and virulent fungal yeast-phase, which is not the usual infective phase of the etiological agent of this mycosis. Thus, due to their versatility, complexity, and similarities with humans, several murine models have played a fundamental role in exploring the host-parasite interaction during H. capsulatum infection.


Subject(s)
Histoplasma/immunology , Histoplasmosis/immunology , Immunity, Innate , Adaptive Immunity , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/microbiology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Cell Wall/chemistry , Cell Wall/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Disease Models, Animal , Histoplasma/growth & development , Histoplasma/pathogenicity , Histoplasmosis/microbiology , Histoplasmosis/pathology , Host Specificity , Humans , Mice , Species Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology
12.
J Immunol ; 190(6): 2756-66, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23378429

ABSTRACT

B1a cells are an important source of natural Abs, Abs directed against T-independent Ags, and are a primary source of IL-10. Bruton's tyrosine kinase (btk) is a cytoplasmic kinase that is essential for mediating signals from the BCR and is critical for development of B1a cells. Consequentially, animals lacking btk have few B1a cells, minimal Ab responses, and can preferentially generate Th1-type immune responses following infection. B1a cells have been shown to aid in protection against infection with attenuated Francisella tularensis, but their role in infection mediated by fully virulent F. tularensis is not known. Therefore, we used mice with defective btk (CBA/CaHN-Btk(XID)/J [XID mice]) to determine the contribution of B1a cells in defense against the virulent F. tularensis ssp. tularensis strain SchuS4. Surprisingly, XID mice displayed increased resistance to pulmonary infection with F. tularensis. Specifically, XID mice had enhanced clearance of bacteria from the lung and spleen and significantly greater survival of infection compared with wild-type controls. We revealed that resistance to infection in XID mice was associated with decreased numbers of IL-10-producing B1a cells and concomitant increased numbers of IL-12-producing macrophages and IFN-γ-producing NK/NKT cells. Adoptive transfer of wild-type B1a cells into XID mice reversed the control of bacterial replication. Similarly, depletion of NK/NKT cells also increased bacterial burdens in XID mice. Together, our data suggest B cell-NK/NKT cell cross-talk is a critical pivot controlling survival of infection with virulent F. tularensis.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Francisella tularensis/immunology , Genetic Predisposition to Disease , Killer Cells, Natural/immunology , Natural Killer T-Cells/immunology , Animals , Antibodies, Bacterial/biosynthesis , B-Lymphocyte Subsets/transplantation , Cell Communication/genetics , Cell Communication/immunology , Cells, Cultured , Exudates and Transudates/microbiology , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Natural Killer T-Cells/microbiology , Natural Killer T-Cells/pathology , Peritoneum/immunology , Peritoneum/microbiology , Peritoneum/pathology , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Survival Analysis , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/microbiology , Tularemia/immunology , Tularemia/mortality , Tularemia/pathology , Virulence/genetics , Virulence/immunology
13.
J Immunol ; 190(6): 2692-701, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23401589

ABSTRACT

Streptococcus pyogenes (group A Streptococcus [GAS]) is a leading human pathogen associated with a diverse array of mucosal and systemic infections. Vaccination with J8, a conserved region synthetic peptide derived from the M-protein of GAS and containing only 12 aa from GAS, when conjugated to diphtheria toxoid, has been shown to protect mice against a lethal GAS challenge. Protection has been previously shown to be Ab-mediated. J8 does not contain a dominant GAS-specific T cell epitope. The current study examined long-term Ab memory and dissected the role of B and T cells. Our results demonstrated that vaccination generates specific memory B cells (MBC) and long-lasting Ab responses. The MBC response can be activated following boost with Ag or limiting numbers of whole bacteria. We further show that these memory responses protect against systemic infection with GAS. T cell help is required for activation of MBC but can be provided by naive T cells responding directly to GAS at the time of infection. Thus, individuals whose T cells do not recognize the short synthetic peptide in the vaccine will be able to generate a protective and rapid memory Ab response at the time of infection. These studies significantly strengthen previous findings, which showed that protection by the J8-diphtheria toxoid vaccine is Ab-mediated and suggest that in vaccine design for other organisms the source of T cell help for Ab responses need not be limited to sequences from the organism itself.


Subject(s)
Antibodies, Bacterial/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Immunologic Memory/immunology , Streptococcal Infections/prevention & control , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Vaccines, Subunit/administration & dosage , Animals , Antibodies, Bacterial/administration & dosage , B-Lymphocyte Subsets/metabolism , Immunization, Secondary/methods , Mice , Mice, Inbred BALB C , Mice, SCID , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Time Factors , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
14.
J Immunol ; 190(4): 1591-602, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23325892

ABSTRACT

Induction of polyclonal B cell activation is a phenomenon observed in many types of infection, but its immunological relevance is unclear. In this study we show that staphylococcal protein A induces T cell-independent human B cell proliferation by enabling uptake of TLR-stimulating nucleic acids via the V(H)3(+) BCR. We further demonstrate that Staphylococcus aureus strains with high surface protein A expression concomitantly trigger activation of human plasmacytoid dendritic cells (pDC). Sensitivity to chloroquine, cathepsin B inhibition, and a G-rich inhibitory oligodeoxynucleotide supports the involvement of TLR9 in this context. We then identify pDC as essential cellular mediators of B cell proliferation and Ig production in response to surface protein A-bearing S. aureus. The in vivo relevancy of these findings is confirmed in a human PBMC Nod/scid(Prkdc)/γc(-/-) mouse model. Finally, we demonstrate that co-operation of pDC and B cells enhances B cell-derived IL-10 production, a cytokine associated with immunosuppression and induction of IgG4, an isotype frequently dominating the IgG response to S. aureus. IL-10 release is partially dependent on TLR2-active lipoproteins, a hallmark of the Staphylococcus species. Collectively, our data suggest that S. aureus exploits pDC and TLR to establish B cell-mediated immune tolerance.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Interleukin-10/biosynthesis , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Protein A/pharmacology , Animals , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic , Dendritic Cells/metabolism , HEK293 Cells , Humans , Immunoglobulin G/blood , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Staphylococcal Infections/metabolism , Up-Regulation/immunology
15.
J Immunol ; 189(12): 5527-32, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23162127

ABSTRACT

Vaccination with purified capsular polysaccharide Vi Ag from Salmonella typhi can protect against typhoid fever, although the mechanism for its efficacy is not clearly established. In this study, we have characterized the B cell response to this vaccine in wild-type and T cell-deficient mice. We show that immunization with typhoid Vi polysaccharide vaccine rapidly induces proliferation in B1b peritoneal cells, but not in B1a cells or marginal zone B cells. This induction of B1b proliferation is concomitant with the detection of splenic Vi-specific Ab-secreting cells and protective Ab in Rag1-deficient B1b cell chimeras generated by adoptive transfer-induced specific Ab after Vi immunization. Furthermore, Ab derived from peritoneal B cells is sufficient to confer protection against Salmonella that express Vi Ag. Expression of Vi by Salmonella during infection did not inhibit the development of early Ab responses to non-Vi Ags. Despite this, the protection conferred by immunization of mice with porin proteins from Salmonella, which induce Ab-mediated protection, was reduced postinfection with Vi-expressing Salmonella, although protection was not totally abrogated. This work therefore suggests that, in mice, B1b cells contribute to the protection induced by Vi Ag, and targeting non-Vi Ags as subunit vaccines may offer an attractive strategy to augment current Vi-based vaccine strategies.


Subject(s)
Antigens, Bacterial/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Polysaccharides, Bacterial/biosynthesis , Salmonella typhi/immunology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/physiology , Antigens, Bacterial/biosynthesis , B-Lymphocyte Subsets/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Cavity/cytology , Peritoneal Cavity/microbiology , Peritoneum/cytology , Peritoneum/immunology , Peritoneum/metabolism , Polysaccharides, Bacterial/immunology , Porins , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/biosynthesis , Salmonella Vaccines/immunology , Typhoid Fever/immunology , Typhoid Fever/metabolism , Typhoid Fever/prevention & control
16.
J Immunol ; 189(12): 5820-30, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23175699

ABSTRACT

The role of B cells in host defense against fungi has been difficult to establish. We quantified and determined the molecular derivation of B-1a, B-1b, and B-2 B cell populations in C57BL/6 mice after pulmonary infection with Cryptococcus neoformans. Total B-1 and B-2 cell numbers increased in lungs and peritoneal cavity as early as day 1 postinfection, but lacked signs of clonal expansion. Labeled capsular (24067) and acapsular (Cap67) C. neoformans strains were used to identify C. neoformans-binding B cell subsets by flow cytometry. Peritoneal cavity B-1a B cells exhibited the most acapsular and capsular C. neoformans binding in C. neoformans-infected mice, and C. neoformans-selected B-1 B cells secreted laminarin- and C. neoformans-binding IgM. Single-cell PCR-based sequence analysis of B-1a, B-1b, and B-2 cell IgH V region H chain (V(H)) genes revealed increased usage of V(H)11 and V(H)12, respectively, in acapsular and capsular C. neoformans-selected B-1a cells. Germline V(H) segments were used, with capsular C. neoformans-selected cells having less junctional diversity than acapsular C. neoformans-selected cells. Further studies in B-1 B cell-depleted mice showed that these mice had higher brain and lung fungal burdens and less alveolar macrophage phagocytosis of C. neoformans than did control and B-1a B cell-reconstituted mice. Taken together, these results establish a mechanistic role for B-1 B cells in the innate B cell response to pulmonary infection with C. neoformans and reveal that IgM-producing B-1a cells, which express germline V(H) genes, bind C. neoformans and contribute to early fungal clearance. Thus, B-1a B cells provide a first line of defense during pulmonary C. neoformans infection in mice.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcus neoformans/immunology , Lung Diseases, Fungal/immunology , Lung Diseases, Fungal/microbiology , Animals , B-Lymphocyte Subsets/pathology , Cryptococcosis/pathology , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Variable Region/biosynthesis , Lung Diseases, Fungal/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peritoneal Cavity/microbiology , Peritoneal Cavity/pathology
17.
J Immunol ; 189(12): 5503-7, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23150714

ABSTRACT

Typhoid fever and nontyphoidal bacteremia caused by Salmonella remain critical human health problems. B cells are required for protective immunity to Salmonella, but the mechanism of protection remains unclear. In this study, we immunized wild-type, B cell-deficient, Ab-deficient, and class-switched Ab-deficient mice with attenuated Salmonella and examined protection against secondary infection. As expected, wild-type mice were protected and B cell-deficient mice succumbed to secondary infection. Interestingly, mice with B cells but lacking secreted Ab or class-switched Ab had little deficiency in resistance to Salmonella infection. The susceptibility of B cell-deficient mice correlated with marked reductions in CD4 T cell IFN-γ production after secondary infection. Taken together, these data suggest that the primary role of B cells in acquired immunity to Salmonella is via the development of protective T cell immunity.


Subject(s)
Antibodies, Bacterial/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Salmonella Infections/prevention & control , Salmonella typhimurium/immunology , Animals , B-Lymphocyte Subsets/metabolism , Immunity, Cellular , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Salmonella Infections/immunology , Salmonella Infections/pathology , Salmonella typhimurium/pathogenicity , Th1 Cells/immunology , Th1 Cells/microbiology , Virulence/immunology
18.
J Immunol ; 189(7): 3319-27, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22933629

ABSTRACT

Rag-1-knockout (KO) mice are highly resistant to Listeria monocytogenes infection. The role played by the many Rag-1-dependent lymphocyte lineages was studied using a genetic approach in which each Rag-1-dependent lymphocyte lineage was eliminated one at a time. Only B cell-deficient Igh-KO mice displayed reduced bacterial load and improved survival upon Listeria infection. Listeria infection of Rag-1-KO and Il-10-KO hosts that had been adoptively transferred with wild-type marginal zone B (MZB) cells, but not follicular B cells, resulted in heightened bacterial load and increased Il-10 production in the spleen, but not the liver. This MZB cell-dependent increase in bacterial load was eliminated by anti-Il-10 mAb. In addition, Listeria infection of MZB cell-deficient Rbpj-cKO mice showed decreased bacterial load and increased survival. Whereas multiple cell types have been shown to be capable of Il-10 production, our results indicate that the MZB cell is the most dominant and relevant Il-10 source in the context of Listeria susceptibility. In marked contrast to the generally protective nature of MZB cells in defending against pathogenic infection, our results demonstrate that MZB cells play a detrimental role in Listeria infection and possibly other infections as well.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Interleukin-10/metabolism , Listeriosis/immunology , Spleen/immunology , Spleen/microbiology , Animals , B-Lymphocyte Subsets/transplantation , Bacterial Load/genetics , Bacterial Load/immunology , Cell Survival/immunology , Interleukin-10/biosynthesis , Interleukin-10/physiology , Listeriosis/genetics , Listeriosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Spleen/transplantation , Up-Regulation/genetics , Up-Regulation/immunology
19.
J Immunol ; 189(5): 2673-81, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855707

ABSTRACT

The maintenance of adequate serum Ab levels following immunization has been identified as the most important mechanism for individual long-term protection against rapidly invading encapsulated bacteria. The mechanisms for maintaining adequate serum Ab levels and the relationship between Ag-specific memory B cells and Ab at steady state are poorly understood. We measured the frequency of circulating serogroup C meningococcal (MenC)-specific memory B cells in 250 healthy 6- to 12-y-old children 6 y following MenC conjugate vaccine priming, before a booster of a combined Haemophilus influenzae type b-MenC conjugate vaccine and then 1 wk, 1 mo, and 1 y after the booster. We investigated the relationship between circulating MenC-specific memory B cell frequencies and Ab at baseline and following the booster vaccine. We found very low frequencies of circulating MenC-specific memory B cells at steady state in primary school-aged children and little association with MenC IgG Ab levels. Following vaccination, there were robust memory B cell booster responses that, unlike Ab levels, were not dependent on age at priming with MenC. Measurement of B cell memory in peripheral blood does not predict steady state Ab levels nor the capacity to respond to a booster dose of MenC Ag.


Subject(s)
Antibodies, Bacterial/blood , B-Lymphocyte Subsets/immunology , Immunization, Secondary/methods , Immunoglobulin G/blood , Immunologic Memory , Meningococcal Vaccines/administration & dosage , Neisseria meningitidis, Serogroup C/immunology , Age Factors , Antibodies, Bacterial/biosynthesis , B-Lymphocyte Subsets/microbiology , Child , Clinical Trials, Phase IV as Topic/methods , Haemophilus Vaccines/administration & dosage , Haemophilus Vaccines/immunology , Humans , Immunoglobulin G/biosynthesis , Meningococcal Vaccines/immunology , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
20.
J Immunol ; 189(2): 841-9, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22696443

ABSTRACT

Abs confer protection from secondary infection with Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaires' disease. In this study, we demonstrate that Ab-mediated protection is effective across L. pneumophila serogroups, suggesting that Abs specific for conserved protein Ags are sufficient to mediate this protective effect. We used two independent methods to identify immunogenic L. pneumophila protein Ags, namely, the screening of a λ phage library representing the complete L. pneumophila genome and two-dimensional gel electrophoresis combined with Western blot analysis and protein spot identification by mass spectrometry. A total of 30 novel L. pneumophila B cell Ags were identified, the majority of which are located in or associated with the bacterial membrane, where they are accessible for Abs and, therefore, likely to be relevant for Ab-mediated protection against L. pneumophila. Selected B cell Ags were recombinantly expressed and tested in a vaccination protocol. Mice immunized with either single-protein Ags or an Ag combination showed reduced bacterial titers in bronchoalveolar lavage and lung after L. pneumophila challenge. To determine the clinical relevance of these findings, we tested Legionnaires' disease patient sera for reactivity with the identified L. pneumophila Ags. The recognized Ags were indeed conserved across host species, because Abs specific for all three selected Ags could be detected in patient sera, rendering the identified protein Ags potential vaccine candidates.


Subject(s)
Antigens, Bacterial/isolation & purification , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Legionella pneumophila/immunology , Legionnaires' Disease/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Antibodies, Bacterial/therapeutic use , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , B-Lymphocyte Subsets/metabolism , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/chemical synthesis , Bacterial Vaccines/immunology , Bacteriophage lambda/genetics , Bacteriophage lambda/immunology , Conserved Sequence/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Legionella pneumophila/pathogenicity , Legionnaires' Disease/blood , Legionnaires' Disease/prevention & control , Mice , Mice, Inbred A , Mice, Inbred C57BL , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...