Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Med ; 28(6): 1167-1177, 2022 06.
Article in English | MEDLINE | ID: mdl-35662283

ABSTRACT

Chemotherapy combined with immunotherapy has improved the treatment of certain solid tumors, but effective regimens remain elusive for pancreatic ductal adenocarcinoma (PDAC). We conducted a randomized phase 2 trial evaluating the efficacy of nivolumab (nivo; anti-PD-1) and/or sotigalimab (sotiga; CD40 agonistic antibody) with gemcitabine/nab-paclitaxel (chemotherapy) in patients with first-line metastatic PDAC ( NCT03214250 ). In 105 patients analyzed for efficacy, the primary endpoint of 1-year overall survival (OS) was met for nivo/chemo (57.7%, P = 0.006 compared to historical 1-year OS of 35%, n = 34) but was not met for sotiga/chemo (48.1%, P = 0.062, n = 36) or sotiga/nivo/chemo (41.3%, P = 0.223, n = 35). Secondary endpoints were progression-free survival, objective response rate, disease control rate, duration of response and safety. Treatment-related adverse event rates were similar across arms. Multi-omic circulating and tumor biomarker analyses identified distinct immune signatures associated with survival for nivo/chemo and sotiga/chemo. Survival after nivo/chemo correlated with a less suppressive tumor microenvironment and higher numbers of activated, antigen-experienced circulating T cells at baseline. Survival after sotiga/chemo correlated with greater intratumoral CD4 T cell infiltration and circulating differentiated CD4 T cells and antigen-presenting cells. A patient subset benefitting from sotiga/nivo/chemo was not identified. Collectively, these analyses suggest potential treatment-specific correlates of efficacy and may enable biomarker-selected patient populations in subsequent PDAC chemoimmunotherapy trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Albumins , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Humans , Nivolumab/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Pancreatic Neoplasms
2.
J Clin Invest ; 131(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34473651

ABSTRACT

Little is known about how cells regulate and integrate distinct biosynthetic pathways governing differentiation and cell division. For B lineage cells it is widely accepted that activated cells must complete several rounds of mitosis before yielding antibody-secreting plasma cells. However, we report that marginal zone (MZ) B cells, innate-like naive B cells known to generate plasma cells rapidly in response to blood-borne bacteria, generate functional plasma cells despite cell-cycle arrest. Further, short-term Notch2 blockade in vivo reversed division-independent differentiation potential and decreased transcript abundance for numerous mTORC1- and Myc-regulated genes. Myc loss compromised plasma cell differentiation for MZ B cells, and reciprocally induced ectopic mTORC1 signaling in follicular B cells enabled division-independent differentiation and plasma cell-affiliated gene expression. We conclude that ongoing in situ Notch2/mTORC1 signaling in MZ B cells establishes a unique cellular state that enables rapid division-independent plasma cell differentiation.


Subject(s)
B-Lymphocytes/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Plasma Cells/cytology , Receptor, Notch2/physiology , Animals , Cell Differentiation , Cells, Cultured , Memory B Cells/physiology , Mice , Mice, Inbred C57BL , Mitosis , Signal Transduction/physiology
4.
Nat Commun ; 11(1): 723, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024827

ABSTRACT

How activated B cells build biosynthetic pathways and organelle structures necessary for subsequent robust antibody secretion is still unclear. The dominant model holds that nascent plasma cells adapt to increased antibody synthesis by activating the unfolded protein response (UPR) under the control of the transcription factor Xbp1. Here, by analyzing gene expression in activated B cells with or without plasma cell-inductive signals, we find that follicular B cells up-regulate a wide array of UPR-affiliated genes before initiating antibody secretion; furthermore, initial transcription of these loci requires the mTORC1 kinase adaptor, Raptor, but not Xbp1. Transcriptomic analyses of resting marginal zone B cells, which generate plasma cells with exceptionally rapid kinetics, reinforce these results by revealing the basal expression of UPR-affiliated mRNA networks without detectable Xbp1 activity. We thus conclude that B cells utilize mTORC1 to prepare for subsequent plasma cell function, before the onset of antibody synthesis.


Subject(s)
Antibodies/metabolism , B-Lymphocytes/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Unfolded Protein Response/physiology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Differentiation , Gene Expression Regulation , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Spleen/cytology , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
5.
Cell Host Microbe ; 23(3): 302-311.e3, 2018 Mar 14.
Article in English | MEDLINE | ID: mdl-29478774

ABSTRACT

Serum immunoglobulin A (IgA) antibodies are readily detected in mice and people, but the mechanisms underlying the induction of serum IgA and its role in host protection remain uncertain. We report that select commensal bacteria induce several facets of systemic IgA-mediated immunity. Exposing conventional mice to a unique but natural microflora that included several members of the Proteobacteria phylum led to T cell-dependent increases in serum IgA levels and the induction of large numbers of IgA-secreting plasma cells in the bone marrow. The resulting serum IgA bound to a restricted collection of bacterial taxa, and antigen-specific serum IgA antibodies were readily induced after intestinal colonization with the commensal bacterium Helicobacter muridarum. Finally, movement to a Proteobacteria-rich microbiota led to serum IgA-mediated resistance to polymicrobial sepsis. We conclude that commensal microbes overtly influence the serum IgA repertoire, resulting in constitutive protection against bacterial sepsis.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulin A/blood , Immunoglobulin A/immunology , Microbiota/immunology , Sepsis/immunology , Sepsis/microbiology , Symbiosis , Animals , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , Bone Marrow , Female , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Proteobacteria/classification , Proteobacteria/immunology , Proteobacteria/physiology , RNA, Ribosomal, 16S/genetics , T-Lymphocytes/immunology
6.
Eur J Immunol ; 47(8): 1386-1388, 2017 08.
Article in English | MEDLINE | ID: mdl-28654161

ABSTRACT

Plasma cells are rare cells that have been notoriously difficult to detect by flow cytometry. New advances have described B220+ CD138+ plasma cells in the bone marrow that are particularly difficult to distinguish between CD138 intermediate B220+ developing B cells. Herein we describe a novel method for detecting plasma cells in the bone marrow using a combination of CD138 and Sca-1 staining.


Subject(s)
Antigens, Ly/analysis , Flow Cytometry/methods , Immunophenotyping/methods , Membrane Proteins/analysis , Plasma Cells/classification , Plasma Cells/immunology , Syndecan-1/analysis , Animals , Bone Marrow Cells/immunology , Leukocyte Common Antigens/analysis , Mice , Peyer's Patches/cytology , Peyer's Patches/immunology , Positive Regulatory Domain I-Binding Factor 1 , Spleen/cytology , Spleen/immunology , Transcription Factors/analysis
7.
J Clin Invest ; 126(11): 4250-4261, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27760048

ABSTRACT

Little is known about the role of mTOR signaling in plasma cell differentiation and function. Furthermore, for reasons not understood, mTOR inhibition reverses antibody-associated disease in a murine model of systemic lupus erythematosus. Here, we have demonstrated that induced B lineage-specific deletion of the gene encoding RAPTOR, an essential signaling adaptor for rapamycin-sensitive mTOR complex 1 (mTORC1), abrogated the generation of antibody-secreting plasma cells in mice. Acute treatment with rapamycin recapitulated the effects of RAPTOR deficiency, and both strategies led to the ablation of newly formed plasma cells in the spleen and bone marrow while also obliterating preexisting germinal centers. Surprisingly, although perturbing mTOR activity caused a profound decline in serum antibodies that were specific for exogenous antigen or DNA, frequencies of long-lived bone marrow plasma cells were unaffected. Instead, mTORC1 inhibition led to decreased expression of immunoglobulin-binding protein (BiP) and other factors needed for robust protein synthesis. Consequently, blockade of antibody synthesis was rapidly reversed after termination of rapamycin treatment. We conclude that mTOR signaling plays critical but diverse roles in early and late phases of antibody responses and plasma cell differentiation.


Subject(s)
Antibody Formation/physiology , Cell Differentiation/physiology , Immunity, Humoral/physiology , Plasma Cells/immunology , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Regulatory-Associated Protein of mTOR , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
8.
Dev Comp Immunol ; 65: 114-123, 2016 12.
Article in English | MEDLINE | ID: mdl-27377583

ABSTRACT

Bat immunity has received increasing attention because some bat species are being decimated by the fungal disease, White Nose Syndrome, while other species are potential reservoirs of zoonotic viruses. Identifying specific immune processes requires new specific tools and reagents. In this study, we describe a new mouse monoclonal antibody (mAb) reactive with Eptesicus fuscus immunoglobulins. The epitope recognized by mAb BT1-4F10 was localized to immunoglobulin light (lambda) chains; hence, the mAb recognized serum immunoglobulins and B lymphocytes. The BT1-4F10 epitope appeared to be restricted to Microchiropteran immunoglobulins and absent from Megachiropteran immunoglobulins. Analyses of sera and other E. fuscus fluids showed that most, if not all, secreted immunoglobulins utilized lambda light chains. Finally, mAb BT1-4F10 permitted the identification of B cell follicles in splenic white pulp. This Microchiropteran-specific mAb has potential utility in seroassays; hence, this reagent may have both basic and practical applications for studying immune process.


Subject(s)
Antibodies, Fungal/isolation & purification , Antibodies, Monoclonal/isolation & purification , B-Lymphocytes/immunology , Chiroptera/immunology , Mycoses/immunology , Zoonoses/immunology , Animals , Cell Line , Cell Separation , Epitope Mapping , Flow Cytometry , Immunoglobulin lambda-Chains/immunology , Immunophenotyping , Mice , Microscopy
9.
J Immunol ; 195(10): 4753-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26438523

ABSTRACT

Despite their critical role in long-term immunity, the life span of individual memory B cells remains poorly defined. Using a tetracycline-regulated pulse-chase system, we measured population turnover rates and individual t1/2 of pre-established Ag-induced Ig class-switched and IgM-positive memory B cells over 402 d. Our results indicate that, once established, both IgG-positive and less frequent IgM-positive memory populations are exceptionally stable, with little evidence of attrition or cellular turnover. Indeed, the vast majority of cells in both pools exhibited t1/2 that appear to exceed the life span of the mouse, contrasting dramatically with mature naive B cells. These results indicate that recall Ab responses are mediated by stable pools of extremely long-lived cells, and suggest that Ag-experienced B cells employ remarkably efficient survival mechanisms.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Immunologic Memory/physiology , Animals , B-Lymphocytes/cytology , Cell Survival , Immunoglobulin G/genetics , Immunoglobulin M/genetics , Mice , Mice, Transgenic , Time Factors
10.
Infect Immun ; 83(5): 2139-47, 2015 May.
Article in English | MEDLINE | ID: mdl-25776744

ABSTRACT

Infection of mice with the bacterium Ehrlichia muris elicits a protective T cell-independent (TI) IgM response mediated primarily by a population of CD11c-expressing plasmablasts in the spleen. Although splenic marginal zone (MZ) B cells are considered to be important for TI responses to blood-borne pathogens, MZ B cells were not responsible for generating plasmablasts in response to Ehrlichia muris. Moreover, antigen-specific serum IgM was decreased only modestly in splenectomized mice and in mice that lacked spleen, lymph nodes, and Peyer's patches (SLP mice). Both splenectomized and SLP mice were protected against lethal ehrlichial challenge infection. Moreover, we found a high frequency of Ehrlichia-specific plasmablasts in the omentum of both conventional and SLP mice. Omental plasmablasts elicited during Ehrlichia infection lacked expression of CD138 but expressed CD11c in a manner similar to that of their splenic counterparts. Selective ablation of CD11c-expressing B cells nearly eliminated the omental Ehrlichia-specific plasmablasts and reduced antigen-specific serum IgM, identifying the omental B cells as a source of IgM production in the SLP mice. Generation of the omental plasmablasts was route dependent, as they were detected following peritoneal infection but not following intravenous infection. Our data identify the omentum as an important auxiliary site of IgM production during intracellular bacterial infection.


Subject(s)
Antibodies, Bacterial/metabolism , Ehrlichiosis/immunology , Immunoglobulin M/metabolism , Omentum/immunology , Plasma Cells/immunology , Animals , CD11c Antigen/analysis , Ehrlichia/immunology , Female , Male , Mice , Mice, Inbred C57BL , Omentum/pathology , Syndecan-1/analysis
11.
J Immunol ; 193(10): 4971-9, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25326027

ABSTRACT

Current models hold that serum Ab titers are maintained chiefly by long-lived bone marrow (BM) plasma cells (PCs). In this study, we characterize the role of subpopulations of BM PCs in long-term humoral responses to T cell-dependent Ag. Surprisingly, our results indicate that 40-50% of BM PCs are recently formed cells, defined, in part, by rapid steady-state turnover kinetics and secretion of low-affinity IgM Abs. Further, for months after immunization with a hapten-protein conjugate, newly formed Ag-induced, IgM-secreting BM PCs were detected in parallel with longer-lived IgG-secreting cells, suggesting ongoing and parallel input to the BM PC pool from two distinct pools of activated B cells. Consistent with this interpretation, IgM and IgG Abs secreted by cells within distinct PC subsets exhibited distinct L chain usage. We conclude that long-term Ab responses are maintained by a dynamic BM PC pool composed of both recently formed and long-lived PCs drawn from clonally disparate precursors.


Subject(s)
B-Lymphocyte Subsets/immunology , Bone Marrow Cells/immunology , Immunity, Humoral , Immunoglobulin E/biosynthesis , Immunoglobulin M/biosynthesis , Plasma Cells/immunology , Animals , B-Lymphocyte Subsets/cytology , Bone Marrow Cells/cytology , CD4 Antigens/metabolism , Cell Lineage/immunology , Female , Immunologic Memory , Immunophenotyping , Mice , Mice, Inbred C57BL , Models, Immunological , Plasma Cells/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Infect Immun ; 81(12): 4490-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24082070

ABSTRACT

B cell activating factor of the tumor necrosis factor family (BAFF) is an essential survival factor for B cells and has been shown to regulate T cell-independent (TI) IgM production. During Ehrlichia muris infection, TI IgM secretion in the spleen was BAFF dependent, and antibody-mediated BAFF neutralization led to an impairment of IgM-mediated host defense. The failure of TI plasmablasts to secrete IgM was not a consequence of alterations in their generation, survival, or early differentiation, since all occurred normally in infected mice following BAFF neutralization. Gene expression characteristic of plasma cell differentiation was also unaffected by BAFF neutralization in vivo, and except for CD138, plasmablast cell surface marker expression was unaffected. IgM was produced, since it was detected intracellularly, and impaired secretion was not due to a failure to express the IgM secretory exon. Addition of BAFF to plasmablasts in vitro rescued IgM secretion, suggesting that BAFF signaling can directly regulate secretory processes. Our findings indicate that BAFF signaling can modulate TI host defense by acting at a late stage in B cell differentiation, via its regulation of terminal plasmablast differentiation and/or IgM secretion.


Subject(s)
B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Ehrlichia/immunology , Ehrlichiosis/immunology , Immunoglobulin M/immunology , Animals , B-Cell Activating Factor/antagonists & inhibitors , B-Cell Activating Factor/metabolism , Cell Differentiation , Immunoglobulin M/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Plasma Cells/metabolism , Signal Transduction/immunology , Syndecan-1/immunology , T-Lymphocytes/immunology
13.
J Immunol ; 189(3): 1440-7, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22730531

ABSTRACT

Polyreactivity is well known as a property of natural IgM produced by B-1 cells. We demonstrate that polyreactive IgM is also generated during infection of mice with Ehrlichia muris, a tick-borne intracellular bacterial pathogen. The polyreactive IgM bound self and foreign Ags, including single-stranded and double-stranded DNA, insulin, thyroglobulin, LPS, influenza virus, and Borrelia burgdorferi. Production of polyreactive IgM during infection was Ag driven, not due to polyclonal B cell activation, as the majority of polyreactive IgM recognized ehrlichial Ag(s), including an immunodominant outer membrane protein. Monoclonal polyreactive IgM derived from T cell-independent spleen plasmablasts, which was germline-encoded, also bound cytoplasmic and nuclear Ags in HEp-2 cells. Polyreactive IgM protected immunocompromised mice against lethal bacterial challenge infection. Serum from human ehrlichiosis patients also contained polyreactive and self-reactive IgM. We propose that polyreactivity increases IgM efficacy during infection but may also exacerbate or mollify the response to foreign and self Ags.


Subject(s)
Antigens, Bacterial/immunology , Immunoglobulin M/biosynthesis , Intracellular Fluid/immunology , Intracellular Fluid/microbiology , Animals , Antigens, T-Independent/immunology , Bacterial Outer Membrane Proteins/administration & dosage , Ehrlichia/immunology , Ehrlichiosis/blood , Ehrlichiosis/immunology , Ehrlichiosis/metabolism , Immunodominant Epitopes/immunology , Intracellular Fluid/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plasma Cells/immunology , Plasma Cells/metabolism , Spleen/immunology , Spleen/metabolism , Spleen/pathology
14.
J Immunol ; 186(2): 1032-43, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21149601

ABSTRACT

Although microbial infections can alter steady-state hematopoiesis, the mechanisms that drive such changes are not well understood. We addressed a role for IFN-γ signaling in infection-induced bone marrow suppression and anemia in a murine model of human monocytic ehrlichiosis, an emerging tick-borne disease. Within the bone marrow of Ehrlichia muris-infected C57BL/6 mice, we observed a reduction in myeloid progenitor cells, as defined both phenotypically and functionally. Infected mice exhibited a concomitant increase in developing myeloid cells within the bone marrow, an increase in the frequency of circulating monocytes, and an increase in splenic myeloid cells. The infection-induced changes in progenitor cell phenotype were critically dependent on IFN-γ, but not IFN-α, signaling. In mice deficient in the IFN-γ signaling pathway, we observed an increase in myeloid progenitor cells and CDllb(lo)Gr1(lo) promyelocytic cells within the bone marrow, as well as reduced frequencies of mature granulocytes and monocytes. Furthermore, E. muris-infected IFN-γR-deficient mice did not exhibit anemia or an increase in circulating monocytes, and they succumbed to infection. Gene transcription studies revealed that IFN-γR-deficient CDllb(lo)Gr1(lo) promyelocytes from E. muris-infected mice exhibited significantly reduced expression of irf-1 and irf-8, both key transcription factors that regulate the differentiation of granulocytes and monocytes. Finally, using mixed bone marrow chimeric mice, we show that IFN-γ-dependent infection-induced myelopoiesis occurs via the direct effect of the cytokine on developing myeloid cells. We propose that, in addition to its many other known roles, IFN-γ acts to control infection by directly promoting the differentiation of myeloid cells that contribute to host defense.


Subject(s)
Ehrlichiosis/immunology , Ehrlichiosis/metabolism , Interferon-gamma/physiology , Intracellular Fluid/microbiology , Myeloid Cells/immunology , Myeloid Cells/microbiology , Myelopoiesis/immunology , Signal Transduction/immunology , Animals , Blood Cell Count , Cell Differentiation/immunology , Cells, Cultured , Ehrlichia/immunology , Ehrlichia/pathogenicity , Ehrlichiosis/pathology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Hematopoietic Stem Cells/pathology , Humans , Immunophenotyping , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/pathology
15.
J Immunol ; 186(2): 1011-21, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21148037

ABSTRACT

IgM responses are well known to occur early postinfection and tend to be short-lived, which has suggested that this Ig does not significantly contribute to long-term immunity. In this study, we demonstrate that chronic infection with the intracellular bacterium Ehrlichia muris elicits a protective, long-term IgM response. Moreover, we identified a population of CD138(high)IgM(high) B cells responsible for Ag-specific IgM production in the bone marrow. The IgM-secreting cells, which exhibited characteristics of both plasmablasts and plasma cells, contributed to protection against fatal ehrlichial challenge. Mice deficient in activation-induced cytidine deaminase, which produce only IgM, were protected against fatal ehrlichial challenge infection. The IgM-secreting cells that we have identified were maintained in the bone marrow in the absence of chronic infection, as antibiotic-treated mice remained protected against challenge infection. Our studies identify a cell population that is responsible for the IgM production in the bone marrow, and they highlight a novel role for IgM in the maintenance of long-term immunity during intracellular bacterial infection.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Cells/microbiology , Ehrlichiosis/immunology , Ehrlichiosis/prevention & control , Immunoglobulin M/biosynthesis , Intracellular Fluid/immunology , Plasma Cells/immunology , Plasma Cells/microbiology , Animals , Bone Marrow Cells/metabolism , Chronic Disease , Ehrlichia/immunology , Ehrlichiosis/microbiology , Immunoglobulin M/physiology , Intracellular Fluid/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plasma Cells/metabolism , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/microbiology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Time Factors
16.
J Immunol ; 184(9): 5085-93, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20351185

ABSTRACT

Germinal centers (GCs) are specialized microenvironments in secondary lymphoid organs that facilitate the development of high-affinity, isotype-switched Abs, and immunological memory; consequently, many infections require GC-derived IgG for pathogen clearance. Although Ehrlichia muris infection elicits a robust expansion of splenic, IgM-secreting plasmablasts, we detected only very low frequencies of isotype-switched IgG-secreting cells in mouse spleens, until at least 3 wk postinfection. Instead, Ag-specific IgG was produced in lymph nodes, where it required CD4 T cell help. Consistent with these findings, organized GCs and phenotypically defined splenic GC B cells were found in lymph nodes, but not spleens. Ehrlichial infection also inhibited spleen IgG responses against a coadministered T cell-dependent Ag, hapten 4-hydroxy-3-nitrophenyl acetyl (NP)-conjugated chicken gamma globulin in alum. NP-specific B cells failed to undergo expansion and differentiation into GC B cells in the spleen, Ab titers were reduced, and splenic IgG production was inhibited nearly 10-fold when the Ag was administered during infection. Our data provide a mechanism whereby an intracellular bacterial infection can compromise local immunity to coinfecting pathogens or antigenic challenge.


Subject(s)
Antibodies, Bacterial/metabolism , Ehrlichia/immunology , Ehrlichiosis/immunology , Germinal Center/immunology , Immunoglobulin G/metabolism , Immunosuppression Therapy , Intracellular Fluid/immunology , Intracellular Fluid/microbiology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , Ehrlichiosis/microbiology , Ehrlichiosis/pathology , Epitopes, T-Lymphocyte/immunology , Germinal Center/microbiology , Germinal Center/pathology , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin M/biosynthesis , Intracellular Fluid/metabolism , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Spleen/immunology , Spleen/microbiology , Spleen/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...