Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 959
Filter
1.
J Exp Med ; 218(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33306108

ABSTRACT

Cell migration relies on coordinated activity of chemotactic and guidance receptors. Here, we report a specific role for the RNA-binding protein ZFP36L1 in limiting the abundance of molecules involved in the homing of antibody-secreting cells (ASCs) to the bone marrow (BM). In the absence of ZFP36L1, ASCs build up in the spleen and the liver and show diminished accumulation in the BM. ZFP36L1 facilitates migration by directly regulating G protein-coupled receptor kinase 2 (GRK2) and the integrin chains α4 and ß1 in splenic ASCs. Expression of CXCR4 and of the integrins α4 and ß1 is differentially regulated on ASCs produced at the early and late stages of the immune response. Consequently, deletion of the Zfp36l1 gene has a stronger effect on BM accumulation of high-affinity ASCs formed late in the response. Thus, ZFP36L1 is an integral part of the regulatory network controlling gene expression during ASC homing.


Subject(s)
Antibody-Producing Cells/metabolism , Bone Marrow/metabolism , Butyrate Response Factor 1/metabolism , Animals , Antibody-Producing Cells/drug effects , Antigens/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Base Sequence , Butyrate Response Factor 1/genetics , Cell Count , Cell Death/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Expression Regulation/drug effects , Germinal Center/cytology , Immunization , Integrins/metabolism , Lysophospholipids/pharmacology , Mice, Inbred C57BL , Receptors, CXCR4/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors/metabolism , Spleen/metabolism
2.
Front Immunol ; 10: 2214, 2019.
Article in English | MEDLINE | ID: mdl-31616417

ABSTRACT

Immaturity of the immune system contributes to poor vaccine responses in early life. Germinal center (GC) activation is limited due to poorly developed follicular dendritic cells (FDC), causing generation of few antibody-secreting cells (ASCs) with limited survival and transient antibody responses. Herein, we compared the potential of five adjuvants, namely LT-K63, mmCT, MF59, IC31, and alum to overcome limitations of the neonatal immune system and to enhance and prolong responses of neonatal mice to a pneumococcal conjugate vaccine Pnc1-TT. The adjuvants LT-K63, mmCT, MF59, and IC31 significantly enhanced GC formation and FDC maturation in neonatal mice when co-administered with Pnc1-TT. This enhanced GC induction correlated with significantly enhanced vaccine-specific ASCs by LT-K63, mmCT, and MF59 in spleen 14 days after immunization. Furthermore, mmCT, MF59, and IC31 prolonged the induction of vaccine-specific ASCs in spleen and increased their persistence in bone marrow up to 9 weeks after immunization, as previously shown for LT-K63. Accordingly, serum Abs persisted above protective levels against pneumococcal bacteremia and pneumonia. In contrast, alum only enhanced the primary induction of vaccine-specific IgG Abs, which was transient. Our comparative study demonstrated that, in contrast to alum, LT-K63, mmCT, MF59, and IC31 can overcome limitations of the neonatal immune system and enhance both induction and persistence of protective immune response when administered with Pnc1-TT. These adjuvants are promising candidates for early life vaccination.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibody-Producing Cells/drug effects , Bone Marrow/drug effects , Germinal Center/drug effects , Spleen/drug effects , Alum Compounds/pharmacology , Animals , Animals, Newborn , Antibodies, Bacterial/blood , Bacterial Toxins/pharmacology , Bone Marrow/immunology , Cholera Toxin/pharmacology , Drug Combinations , Enterotoxins/pharmacology , Escherichia coli Proteins/pharmacology , Immunoglobulin G/blood , Mice , Oligodeoxyribonucleotides/pharmacology , Oligopeptides/pharmacology , Pneumococcal Vaccines/administration & dosage , Polysorbates/pharmacology , Spleen/immunology , Squalene/pharmacology
3.
Curr Opin Gastroenterol ; 35(6): 499-506, 2019 11.
Article in English | MEDLINE | ID: mdl-31567498

ABSTRACT

PURPOSE OF REVIEW: To describe the latest developments in the field of anti-trafficking agents (ATAs), a class of therapeutics with growing importance in the field of inflammatory bowel diseases (IBDs) that specifically inhibit steps of immune cell trafficking. RECENT FINDINGS: Several translational and clinical studies have further shaped the knowledge about the mechanisms and effects of the anti-α4ß7 integrin antibody vedolizumab. In parallel, new ATAs like the anti-ß7 integrin antibody etrolizumab and the anti-MAdCAM-1 antibody ontamalimab are investigated in phase III clinical trials and might soon increase the therapeutic armamentarium in IBD. SUMMARY: ATAs have unique mechanisms of action and can meanwhile be considered an indispensable column of IBD therapy. Further efforts are necessary to elucidate complex mechanistic aspects, to exactly define their role in relation to other therapeutic approaches and to identify novel treatment targets as well as biomarkers for personalized medicine.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibody-Producing Cells/drug effects , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Antibody-Producing Cells/immunology , Cell Movement/drug effects , Female , Humans , Male , Patient Selection , Prognosis , Risk Assessment , Treatment Outcome
4.
Exp Parasitol ; 195: 1-7, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30266573

ABSTRACT

L-arginine (L-Arg), the precursor of nitric oxide (NO), plays multiple, important roles in nutrient metabolism and immune regulation. Hypoargininemia is one of the distinctive features of malaria patients in endemic areas. To understand the immunoregulatory function of L-Arg in malaria, we investigated the effects of L-Arg, pre- or/and post-treatment, on the cellular/humoral immune response during Plasmodium yoelii 17XL (P.y17XL) infection in DBA/2 mice. Populations of splenic CD4+T-bet+IFN-γ+ T cells (Th1), F4/80+ macrophages, CD4+GATA-3+IL-4+ T cells (Th2), B220+CD138+ plasmacytes and antibody-producing cells (IgG+/IgG1+-plasma cells) were assessed by flow cytometry. Pro-inflammatory cytokines and antibodies (IgG and IgG1) were quantified by immunoassays. We found that treatment with L-Arg significantly decreased parasitemia and shortened disease duration. Prophylactic treatment with L-Arg promotes an enhanced Th1 cell response during the early stages of P.y17XL infection, and treatment with L-Arg in the course of infection facilitates the later humoral immune response. Our findings suggest that treatment with L-Arg may decrease parasite burden and control the host's susceptibility to parasite synchronously by regulating host immune responses against P.y17XL, producing better outcomes for malaria infection. This implies that the supplementation of L-Arg may be a promising adjunctive therapy to reduce malaria-associated mortality in endemic areas.


Subject(s)
Arginine/therapeutic use , Malaria/drug therapy , Plasmodium yoelii/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/immunology , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/immunology , Arginine/blood , Flow Cytometry , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Interferon-gamma/metabolism , Malaria/immunology , Malaria/prevention & control , Mice , Mice, Inbred DBA , Nitric Oxide/metabolism , Parasitemia/drug therapy , Parasitemia/prevention & control , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/drug effects , Th2 Cells/immunology
5.
Eur J Immunol ; 47(7): 1211-1219, 2017 07.
Article in English | MEDLINE | ID: mdl-28463395

ABSTRACT

Disturbances of plasma cell homeostasis and auto-antibody production are hallmarks of systemic lupus erythematosus. The aim of this study was to explore the presence of circulating anti-ENA and anti-dsDNA antibody-secreting cells, to determine their dependence on plasma cell-niche cytokines and to analyze their clinical value. The study was performed in SLE patients with serum anti-ENA and/or anti-dsDNA antibodies (n = 57). Enriched B-cell fractions and sorted antibody-secreting cells (CD19low CD38high ) were obtained from blood. dsDNA- and ENA-specific antibody-secreting cells were identified as cells capable of active auto-antibody production in culture. The addition of a combination of IL-6, IL-21, BAFF, APRIL, and CXCL12 to the cultures significantly augmented auto-antibody production and antibody-secreting cell proliferation, whereas it diminished apoptosis. The effect on auto-antibody production was dependent on STAT-3 activation as it was abrogated in the presence of the JAK/STAT-3 pathway inhibitors ruxolitinib and stattic. Among patients with serum anti-dsDNA antibodies, the detection of circulating anti-dsDNA-antibody-secreting cells was associated with higher disease activity markers. In conclusion, auto-antibody production in response to plasma cell-niche cytokines that are usually at high levels in SLE patients is dependent on JAK/STAT-3 activation. Thus, patients with circulating anti-dsDNA antibody-secreting cells and active disease could potentially benefit from therapies targeting the JAK/STAT3 pathway.


Subject(s)
Antibodies, Antinuclear/blood , Antibody-Producing Cells/immunology , DNA/immunology , Lupus Erythematosus, Systemic/immunology , STAT3 Transcription Factor/metabolism , Adolescent , Adult , Aged , Antibodies, Antinuclear/immunology , Antibody-Producing Cells/drug effects , Apoptosis/drug effects , B-Cell Activating Factor/pharmacology , Cell Proliferation , Chemokine CXCL2/pharmacology , Cyclic S-Oxides/pharmacology , DNA/blood , Female , Humans , Interleukin-6/pharmacology , Interleukins/pharmacology , Lupus Erythematosus, Systemic/blood , Male , Middle Aged , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Young Adult
6.
J Neurosci Methods ; 285: 58-68, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28495370

ABSTRACT

BACKGROUND: CNS inflammation resulting from infection, injury, or neurodegeneration leads to accumulation of diverse B cell subsets. Although antibody secreting cells (ASC) within the inflamed CNS have been extensively examined, memory B cell (Bmem) characterization has been limited as they do not secrete antibody without stimulation. Moreover, unlike human Bmem, reliable surface markers for murine Bmem remain elusive. NEW METHOD: Using a viral encephalomyelitis model we developed a modified limiting dilution in vitro stimulation assay to convert CNS-derived virus specific Bmem into ASC. COMPARISON WITH EXISTING METHODS: Stimulation methods established for lymphoid tissue cells using prolonged stimulation with viral lysate resulted in substantial ASC loss and minimal Bmem to ASC conversion of CNS-derived cells. By varying stimulation duration, TLR activators, and culture supplements, we achieved optimal conversion by culturing cells with TLR7/8 agonist R848 in the presence of feeder cells for 2days. RESULTS: Flow cytometry markers CD38 and CD73 characterizing murine Bmem from lymphoid tissue showed more diverse expression patterns on corresponding CNS-derived B cell subsets. Using the optimized TLR7/8 stimulation protocol, we compared virus-specific IgG Bmem versus pre-existing ASC within the brain and spinal cord. Increasing Bmem frequencies during chronic infection mirrored kinetics of ASC. However, despite initially similar Bmem and ASC accumulation, Bmem prevailed in the brain, but were lower than ASC in the spinal cord during persistence. CONCLUSION: Simultaneous enumeration of antigen-specific Bmem and ASC using the Bmem assay optimized for CNS-derived cells enables characterization of temporal changes during microbial or auto-antigen induced neuroinflammation.


Subject(s)
Antibody-Producing Cells/physiology , B-Lymphocytes/cytology , Central Nervous System/pathology , Hepatitis, Viral, Animal/complications , Inflammation/etiology , Inflammation/pathology , Animals , Antibody-Producing Cells/drug effects , B-Lymphocytes/drug effects , Cell Differentiation , Cell Movement , Central Nervous System/drug effects , Central Nervous System/virology , Cyclopropanes/pharmacology , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry , Guanosine/analogs & derivatives , Guanosine/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Murine hepatitis virus/pathogenicity , Spinal Cord/pathology , Spinal Cord/virology , Spleen/cytology , Time Factors , Toll-Like Receptor 1/antagonists & inhibitors , Toll-Like Receptor 1/metabolism
7.
PLoS One ; 12(1): e0169646, 2017.
Article in English | MEDLINE | ID: mdl-28052118

ABSTRACT

TGF-ß is a pleotropic cytokine involved in various biological processes. Of the three isoforms of TGF-ß, TGF-ß1 has long been recognized as an important inhibitory cytokine in the immune system and has been reported to inhibit B cell function in both mice and humans. Recently, it has been suggested that TGF-ß3 may play an important role in the regulation of immune system in mice. Murine CD4+CD25-LAG3+ regulatory T cells suppress B cell function through the production of TGF-ß3, and it has been reported that TGF-ß3 is therapeutic in a mouse model of systemic lupus erythematosus. The effect of TGF-ß3 on human B cells has not been reported, and we herein examined the effect of TGF-ß3 on human B cells. TGF-ß3 suppressed B cell survival, proliferation, differentiation into plasmablasts, and antibody secretion. Although the suppression of human B cells by TGF-ß1 has long been recognized, the precise mechanism for the suppression of B cell function by TGF-ß1 remains elusive; therefore, we examined the effect of TGF-ß1 and ß3 on pathways important in B cell activation and differentiation. TGF-ß1 and TGF-ß3 inhibited some of the key molecules of the cell cycle, as well as transcription factors important in B cell differentiation into antibody secreting cells such as IRF4, Blimp-1, and XBP1. TGF-ß1 and ß3 also inhibited B cell receptor signaling. Our results suggest that TGF-ß3 modifying therapy might be therapeutic in autoimmune diseases with B cell dysregulation in humans.


Subject(s)
Antibody Formation/drug effects , B-Lymphocytes/immunology , Transforming Growth Factor beta3/pharmacology , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/metabolism , B-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Humans , Immunoglobulin A/genetics , Immunoglobulin A/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad Proteins/metabolism , Syk Kinase/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology
8.
Immunol Rev ; 270(1): 78-94, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26864106

ABSTRACT

Antibody-secreting cells (ASCs) are critical for a functional and effective adaptive immune system. In a number of illnesses, however, these same cells contribute to the underlying disease state leading to significant morbidity and mortality. While therapeutic targeting of antibody-secreting cells has progressed significantly over the last two decades, many of these conditions remain major health problems. In this review, we will discuss current and potential therapeutic targeting of ASCs in the context of the known biology of these cells.


Subject(s)
Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Animals , Antibody Formation , Antibody-Producing Cells/cytology , Antibody-Producing Cells/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Disease Susceptibility , Gene Expression Regulation , Humans , Molecular Targeted Therapy , Phenotype , Plasma Cells/cytology , Plasma Cells/drug effects , Transcription, Genetic
9.
Arthritis Rheumatol ; 68(4): 965-76, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26606525

ABSTRACT

OBJECTIVE: To evaluate treatment with MEDI-551, a humanized anti-human CD19 monoclonal antibody, in a model of autoimmunity involving mice transgenic (Tg) for Sle1 and human CD19 (hCD19). METHODS: Sle1.hCD19-Tg mice were given either a single intravenous dose of MEDI-551 or repeated doses of MEDI-551 biweekly for up to 12 weeks. The numbers of B cells in the blood, spleen, and bone marrow were determined by flow cytometry assay. In the spleen and bone marrow, the number of IgM- and IgG-specific antibody-secreting cells (ASCs) and the number of ASCs specific for anti-double-stranded DNA (anti-dsDNA) were determined by enzyme-linked immunospot assay. Serum autoantibody and total immunoglobulin levels were determined by enzyme-linked immunosorbent assay, and levels of inflammatory proteins were tested using a multianalyte profiling platform. RESULTS: MEDI-551 treatment of Sle1.hCD19-Tg mice resulted in effective and sustained B cell depletion throughout the duration of the experiment. The frequency of IgM and IgG ASCs in the spleen was reduced by ≥90%, whereas in the bone marrow, the total ASC frequency was not changed. Levels of autoantibodies specific for dsDNA as well as antihistone and antinuclear antibodies were each reduced by 40-80%, but total serum immunoglobulin levels were largely unchanged at the end of 12 weeks of treatment. CONCLUSION: These findings highlight the ability of MEDI-551 to deplete B cells and ASCs in autoimmune Sle1.hCD19-Tg mice. MEDI-551 treatment resulted in a robust reduction of autoantibodies but had minimal effect on total serum immunoglobulins. Thus, the novel ability of MEDI-551 to remove a broad range of B cells as well as to lower most disease-driving autoantibodies in an autoimmune disease mouse model warrants continued research. Several clinical studies to explore the safety and activity of MEDI-551 in autoantibody-associated autoimmune diseases are ongoing.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD19/genetics , Autoantibodies/drug effects , B-Lymphocytes/drug effects , Lupus Erythematosus, Systemic/genetics , Animals , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/immunology , Antigens, CD19/immunology , Autoantibodies/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , DNA/immunology , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Flow Cytometry , Genetic Loci/genetics , Humans , Immunoglobulin G , Immunoglobulin M , Lupus Erythematosus, Systemic/immunology , Lymphocyte Count , Mice , Mice, Transgenic , Spleen/cytology , Spleen/drug effects
10.
Oncotarget ; 6(21): 18484-503, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26050196

ABSTRACT

Recent research has shown that chronic lymphocytic leukemia (CLL) B-cells display a strong tendency to differentiate into antibody-secreting cells (ASCs) and thus may be amenable to differentiation therapy. However, the effect of this differentiation on factors associated with CLL pathogenesis has not been reported. In the present study, purified CLL B-cells were stimulated to differentiate into ASCs by phorbol myristate acetate or CpG oligodeoxynucleotide, in combination with CD40 ligand and cytokines in a two-step, seven-day culture system. We investigated (i) changes in the immunophenotypic, molecular, functional, morphological features associated with terminal differentiation into ASCs, (ii) the expression of factors involved in CLL pathogenesis, and (iii) the expression of pro- and anti-apoptotic proteins in the differentiated cells. Our results show that differentiated CLL B-cells are able to display the transcriptional program of ASCs. Differentiation leads to depletion of the malignant program and deregulation of the apoptosis/survival balance. Analysis of apoptosis and the cell cycle showed that differentiation is associated with low cell viability and a low rate of cell cycle entry. Our findings shed new light on the potential for differentiation therapy as a part of treatment strategies for CLL.


Subject(s)
Antibody-Producing Cells/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , CD40 Ligand/pharmacology , Cell Culture Techniques , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle/immunology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Cytokines/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression/immunology , Humans , Immunoblotting , Immunoglobulin Isotypes/immunology , Immunoglobulin Isotypes/metabolism , Immunophenotyping , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Inhibitor of Apoptosis Proteins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/immunology , Lymphoid Enhancer-Binding Factor 1/metabolism , Oligodeoxyribonucleotides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Survivin , Tetradecanoylphorbol Acetate/pharmacology
11.
PLoS One ; 9(7): e102885, 2014.
Article in English | MEDLINE | ID: mdl-25050555

ABSTRACT

The memory B-cell (MBC) ELISpot assay is the main technique used to measure antigen-specific MBCs as a readout of humoral immune memory. This assay relies on the ability of MBCs to differentiate into antibody-secreting cells (ASC) upon polyclonal stimulation. The total number of IgG+ ASCs generated by mitogen-stimulation is often used as a reference point; alternatively antigen-specific MBCs are expressed as a frequency of post-culture peripheral blood mononuclear cells (PBMC) as a surrogate for absolute frequencies. Therefore, it is important to know whether IgG+ B-cells are uniformly expanded during the preceding mitogen-culture as a true reflection of MBC frequencies ex vivo. We systematically compared B-cell phenotype and proportions before and after mitogen stimulation in cultures of 269 peripheral blood mononuclear cell samples from 62 volunteers by flow cytometry and analyzed the number of resulting ASCs. Our data show that the number of total IgG+ ASCs detected by ELISpot after mitogen stimulation correlates with the proportion of IgG+ MBCs ex vivo, highlighting its general robustness for comparisons of study cohorts at group level. The expansion of total and IgG+ B-cells during mitogen-stimulation, however, was not identical in all cultures, but influenced by size and composition of the ex vivo B-cell compartment. The uncorrected readout of antigen-specific MBCs per million post-culture PBMCs therefore only preserves the quality, but not the magnitude of differences in the ex vivo MBC response between groups or time points, particularly when comparing samples where the B-cell compartment substantially differs between cohorts or over time. Therefore, expressing antigen-specific cells per total IgG+ ASCs is currently the best measure to correct for mitogen-culture effects. Additionally, baseline information on the size and composition of the ex vivo B-cell compartment should be supplied to additionally inform about differences or changes in the size and composition of the ex vivo MBC compartment.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin G/immunology , Immunologic Memory/immunology , Mitogens/immunology , Adolescent , Adult , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Enzyme-Linked Immunospot Assay , Flow Cytometry , Humans , Immunoglobulin G/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Count , Mitogens/pharmacology , Young Adult
12.
J Allergy Clin Immunol ; 134(3): 679-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24815737

ABSTRACT

BACKGROUND: IgG4-related disease (IgG4-RD) is a poorly understood, multiorgan, chronic inflammatory disease characterized by tumefactive lesions, storiform fibrosis, obliterative phlebitis, and accumulation of IgG4-expressing plasma cells at disease sites. OBJECTIVE: The role of B cells and IgG4 antibodies in IgG4-RD pathogenesis is not well defined. We evaluated patients with IgG4-RD for activated B cells in both disease lesions and peripheral blood and investigated their role in disease pathogenesis. METHODS: B-cell populations from the peripheral blood of 84 patients with active IgG4-RD were analyzed by using flow cytometry. The repertoire of B-cell populations was analyzed in a subset of patients by using next-generation sequencing. Fourteen of these patients were longitudinally followed for 9 to 15 months after rituximab therapy. RESULTS: Numbers of CD19(+)CD27(+)CD20(-)CD38(hi) plasmablasts, which are largely IgG4(+), are increased in patients with active IgG4-RD. These expanded plasmablasts are oligoclonal and exhibit extensive somatic hypermutation, and their numbers decrease after rituximab-mediated B-cell depletion therapy; this loss correlates with disease remission. A subset of patients relapse after rituximab therapy, and circulating plasmablasts that re-emerge in these subjects are clonally distinct and exhibit enhanced somatic hypermutation. Cloning and expression of immunoglobulin heavy and light chain genes from expanded plasmablasts at the peak of disease reveals that disease-associated IgG4 antibodies are self-reactive. CONCLUSIONS: Clonally expanded CD19(+)CD27(+)CD20(-)CD38(hi) plasmablasts are a hallmark of active IgG4-RD. Enhanced somatic mutation in activated B cells and plasmablasts and emergence of distinct plasmablast clones on relapse indicate that the disease pathogenesis is linked to de novo recruitment of naive B cells into T cell-dependent responses by CD4(+) T cells, likely driving a self-reactive disease process.


Subject(s)
Antibody-Producing Cells/physiology , B-Lymphocytes/physiology , Immune System Diseases/immunology , Immunoglobulin G/metabolism , Lymphocytosis/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/adverse effects , Antibody Diversity/genetics , Antibody-Producing Cells/drug effects , Antigens, CD/metabolism , Autoantigens/immunology , B-Lymphocytes/drug effects , Cell Proliferation , Clone Cells , Disease Progression , Female , Follow-Up Studies , Humans , Immune System Diseases/therapy , Lymphocyte Activation , Lymphocytosis/therapy , Male , Middle Aged , Recurrence , Rituximab , Somatic Hypermutation, Immunoglobulin/genetics , Young Adult
13.
Regul Toxicol Pharmacol ; 70(2 Suppl): S43-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24793263

ABSTRACT

Female Sprague Dawley rats were exposed via inhalation to vapor condensates of either gasoline or gasoline combined with various fuel oxygenates to assess potential immunotoxicity of evaporative emissions. Test articles included vapor condensates prepared from "baseline gasoline" (BGVC), or gasoline combined with methyl tertiary butyl ether (G/MTBE), ethyl t-butyl ether (G/ETBE), t-amyl methyl ether (G/TAME), diisopropyl ether (G/DIPE), ethanol (G/EtOH), or t-butyl alcohol (G/TBA). Target concentrations were 0, 2000, 10,000 or 20,000mg/mg(3) administered for 6h/day, 5days/week for 4weeks. The antibody-forming cell (AFC) response to the T-dependent antigen, sheep erythrocyte (sRBC), was used to determine the effects of the gasoline vapor condensates on the humoral components of the immune system. Exposure to BGVC, G/MTBE, G/TAME, and G/TBA did not result in significant changes in the IgM AFC response to sRBC, when evaluated as either specific activity (AFC/10(6) spleen cells) or as total spleen activity (AFC/spleen). Exposure to G/EtOH and G/DIPE resulted in a dose-dependent decrease in the AFC response, reaching the level of statistical significance only at the high 20,000mg/m(3) level. Exposure to G/ETBE resulted in a statistically significant decrease in the AFC response at the middle (10,000mg/m(3)) and high (20,000mg/m(3)) exposure concentrations.


Subject(s)
Air Pollutants/toxicity , Antibody-Producing Cells/drug effects , Gasoline/toxicity , Animals , Antibody-Producing Cells/immunology , Female , Immunoglobulin M/immunology , Inhalation , Rats , Rats, Sprague-Dawley , Risk Assessment
14.
Am J Trop Med Hyg ; 90(5): 873-81, 2014 May.
Article in English | MEDLINE | ID: mdl-24686738

ABSTRACT

Protective immunity to cholera is serogroup specific, and serogrouping is defined by the O-specific polysaccharide (OSP) of lipopolysaccharide (LPS). We characterized OSP-specific immune responses in adult recipients of an oral killed cholera vaccine (OCV WC-rBS) and compared these with responses in patients with cholera caused by Vibrio cholerae O1 Ogawa. Although vaccinees developed plasma immunoglobulin G (IgG), IgM, IgA antibody and antibody secreting cell (ASC, marker of mucosal response) to Ogawa OSP and LPS 7 days after vaccination, responses were significantly lower than that which occurred after cholera. Similarly, patients recovering from cholera had detectable IgA, IgM, and IgG memory B cell (MBC) responses against OSP and LPS on Day 30 and Day 90, whereas vaccinees only developed IgG responses to OSP 30 days after the second immunization. The markedly lower ASC and MBC responses to OSP and LPS observed among vaccinees might explain, in part, the lower protection of an OCV compared with natural infection.


Subject(s)
Antibodies, Bacterial/blood , Cholera Vaccines/therapeutic use , Cholera/immunology , Lipopolysaccharides/immunology , O Antigens/immunology , Vibrio cholerae O1/immunology , Administration, Oral , Adult , Antibodies, Bacterial/immunology , Antibody-Producing Cells/cytology , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bangladesh/epidemiology , Cholera/prevention & control , Cohort Studies , Humans , Immunization, Secondary , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Vaccination , Vaccines, Inactivated/therapeutic use , Vibrio cholerae O1/isolation & purification , Young Adult
15.
Mol Cell Biol ; 34(3): 522-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24277931

ABSTRACT

B cell differentiation into antibody-secreting cells (ASCs) is a tightly regulated process under the control of multiple transcription factors. One such transcription factor, Ets1, blocks the transition of B cells to ASCs via two separate activities: (i) stimulating the expression of target genes that promote B cell identity and (ii) interfering with the functional activity of the transcription factor Blimp1. Ets1 is a member of a multigene family, several members of which are expressed within the B cell lineage, including the closely related protein Ets2. In this report, we demonstrate that Ets1, but not Ets2, can block ASC formation despite the fact that Ets1 and Ets2 bind to apparently identical DNA sequence motifs and are thought to regulate overlapping sets of target genes. The DNA binding domain of Ets1 is required, but not sufficient by itself, to block ASC formation. In addition, less conserved regions within the N terminus of Ets1 play an important role in inhibiting B cell differentiation. Differences between the N termini of Ets1 and Ets2, rather than differences in the DNA binding domains, determine whether the proteins are capable of blocking ASC formation or not.


Subject(s)
Antibody-Producing Cells/metabolism , Cell Differentiation , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Protein c-ets-2/metabolism , Amino Acid Sequence , Animals , Antibody-Producing Cells/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Blotting, Western , COS Cells , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Gene Expression , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Nucleotide Motifs/genetics , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Protein Binding , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Bull Exp Biol Med ; 155(2): 221-3, 2013 Jun.
Article in English | MEDLINE | ID: mdl-24130995

ABSTRACT

We observed immunorehabilitation effects of ultrahigh frequency electromagnetic fields (microwaves) in immunocompromised animals. It was shown that microwave irradiation of the thyroid gland area could abolish actinomycin D- and colchicine-induced immunosuppression and did not affect immunosuppression caused by 5-fluorouracil. These findings suggest that changes in the hormonal profile of the organism during microwave exposure can stimulate the processes of transcription and mitotic activity of lymphoid cells.


Subject(s)
Adrenal Cortex/radiation effects , Electromagnetic Fields , Immunocompromised Host/radiation effects , Magnetic Field Therapy/methods , Thyroid Gland/radiation effects , 11-Hydroxycorticosteroids/blood , Adrenal Cortex/immunology , Adrenal Cortex/metabolism , Adrenal Glands/metabolism , Adrenal Glands/radiation effects , Animals , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/radiation effects , Colchicine , Dactinomycin , Erythrocytes/immunology , Erythrocytes/radiation effects , Immunocompromised Host/immunology , Immunosuppression Therapy , Male , Microwaves/therapeutic use , Rabbits , Spleen/cytology , Thyroid Gland/immunology , Thyroid Gland/metabolism
17.
PLoS One ; 8(9): e74566, 2013.
Article in English | MEDLINE | ID: mdl-24058589

ABSTRACT

Switched CD19-positive memory B cells purified from mice with chronic immune response against Thalassophrynenattereri venom proteins were cultured with venom or cytokines. Our results confirm the existence of a hierarchic process of differentiation: activated memory B cells progressively acquire increasing levels of CD138 and decreasing levels of CD45R/B220 to finally arrive at ASC with B220(neg) phenotype, which are IgG1-secreting cells. Only Bmem from peritoneal cavity or bone marrow of VTn immunized mice presented the capacity to generate ASC functionally active. IL-17A or IL-21/IL-23/IL-33 improves the ability of venom to induce intracellular IgG of peritoneal derived-ASC. Cognate stimulation with venom and IL-17A is sufficient to down-regulate the expression of CD45R/B220. BAFF-R is up-regulated in splenic or medullar derived-ASC stimulated by venom, CpG or cytokines. Only splenic derived-ASC up-regulate Bcl-2 expression after CpG or the combination of IL-21/IL-23/IL-33 stimulation. Finally, the activation of ASC for IgG1 secretion is triggered by venom proteins in peritoneal cavity and by IL-17A in medullar niche. These results show the importance of the integration of signals downstream of BCR and IL17-A receptors in modulating ASC differentiation, focusing in the microenvironment niche of their generation.


Subject(s)
Antibody-Producing Cells/cytology , Antigens/immunology , Cell Differentiation/immunology , Interleukin-17/metabolism , Signal Transduction , Animals , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/metabolism , Antigens, CD19/metabolism , Apoptosis/drug effects , Apoptosis/immunology , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Fish Venoms/immunology , Immunoglobulin G/biosynthesis , Immunologic Memory/drug effects , Interleukin-23/pharmacology , Interleukins/pharmacology , Leukocyte Common Antigens/metabolism , Lymphocyte Count , Male , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Spleen/cytology , Toll-Like Receptor 9/metabolism
18.
Anim Sci J ; 84(4): 322-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23590506

ABSTRACT

Mortality and morbidity of neonates continue to be major problems in humans and animals, and immunoblogulin A (IgA) provides protection against microbial antigens at mucosal surfaces. The present study was conducted to clarify the effects of coumestrol administration to maternal mice during pregnancy and lactation on IgA antibody-secreting cells (ASC) in mammary glands in lactating mice. From 6.5 to 16.5 days post coitus and 1 to 13 days post partum (dpp), maternal mice were administered coumestrol at 200 µg/kg body weight/day. Coumestrol administration increased the number of IgA ASC and the messenger RNA expression of IgA C-region and vascular cell adhesion molecule-1 in mammary glands of maternal mice at 14 dpp, but coumestrol administration had no effect on the number of IgA ASC in the ileum. Coumestrol administration increased serum IgA concentration in maternal mice at 14 dpp, but IgA concentrations in serum, stomach contents, intestine and feces of neonatal mice were not affected by treatment. These results imply that coumestrol administration to maternal mice during pregnancy and lactation is effective in increasing the numbers of IgA ASC in mammary glands during lactation owing to the activated messenger RNA expressions of IgA C-region and vascular cell adhesion molecule-1 in mammary gland.


Subject(s)
Antibody-Producing Cells/drug effects , Coumestrol/pharmacology , Immunoglobulin A/physiology , Lactation , Mammary Glands, Animal/cytology , Mice/physiology , Phytoestrogens/pharmacology , Pregnancy, Animal/drug effects , Animals , Antibody-Producing Cells/cytology , Coumestrol/administration & dosage , Female , Ileum/cytology , Immunoassay , Immunoglobulin A/blood , Immunohistochemistry , Mice, Inbred ICR , Phytoestrogens/administration & dosage , Pregnancy , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
19.
J Virol ; 87(6): 3382-92, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23302888

ABSTRACT

Microbial infections of the central nervous system (CNS) are often associated with local accumulation of antibody (Ab)-secreting cells (ASC). By providing a source of Ab at the site of infection, CNS-localized ASC play a critical role in acute viral control and in preventing viral recrudescence. Following coronavirus-induced encephalomyelitis, the CNS accumulation of ASC is chemokine (C-X-C motif) receptor 3 (CXCR3) dependent. This study demonstrates that CNS-expressed CXCR3 ligand CXCL10 is the critical chemokine regulating ASC accumulation. Impaired ASC recruitment in CXCL10(-/-) but not CXCL9(-/-) mice was consistent with reduced CNS IgG and κ-light chain mRNA and virus-specific Ab. Moreover, the few ASC recruited to the CNS in CXCL10(-/-) mice were confined to the vasculature, distinct from the parenchymal localization in wild-type and CXCL9(-/-) mice. However, neither CXCL9 nor CXCL10 deficiency diminished neutralizing serum Ab, supporting a direct role for CXCL10 in ASC migration. T cell accumulation, localization, and effector functions were also not affected in either CXCL9(-/-) or CXCL10(-/-) mice, consistent with similar control of infectious virus. There was also no evidence for dysregulation of chemokines or cytokines involved in ASC regulation. The distinct roles of CXCL9 and CXCL10 in ASC accumulation rather coincided with their differential localization. While CXCL10 was predominantly expressed by astrocytes, CXCL9 expression was confined to the vasculature/perivascular spaces. These results suggest that CXCL10 is critical for two phases: recruitment of ASC to the CNS vasculature and ASC entry into the CNS parenchyma.


Subject(s)
Antibody-Producing Cells/immunology , Astrocytes/metabolism , Cell Movement , Chemokine CXCL10/metabolism , Coronavirus Infections/immunology , Coronavirus/immunology , Encephalomyelitis/immunology , Animals , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/physiology , Astrocytes/immunology , Chemokine CXCL10/deficiency , Chemokine CXCL10/immunology , Coronavirus/pathogenicity , Coronavirus Infections/virology , Encephalomyelitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
J Immunol Methods ; 387(1-2): 167-72, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23098839

ABSTRACT

The generation of antibodies with designated specificity requires cost-intensive and time-consuming screening procedures. Here we present a new method by which hybridoma cells can be selected based on the specificity of the produced antibody by the use of antigen-toxin-conjugates thus eliminating the need of a screening procedure. Initial experiments were done with methotrexate as low molecular weight toxin and fluorescein as model antigen. Methotrexate and a methotrexate-fluorescein conjugate were characterized regarding their toxicity. Afterwards the effect of the fluorescein-specific antibody B13-DE1 on the toxicity of the methotrexate-fluorescein conjugate was determined. Finally, first results showed that hybridoma cells that produce fluorescein specific antibodies are able to grow in the presence of fluorescein-toxin-conjugates.


Subject(s)
Antibody-Producing Cells/immunology , Antigens/immunology , Immunotoxins/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/metabolism , Antigens/chemistry , Antigens/pharmacology , Cell Survival/drug effects , Cell Survival/immunology , Dose-Response Relationship, Drug , Fluoresceins/pharmacology , Hybridomas/drug effects , Hybridomas/immunology , Hybridomas/metabolism , Immunologic Techniques/methods , Immunotoxins/chemistry , Immunotoxins/pharmacology , Methotrexate/analogs & derivatives , Methotrexate/immunology , Methotrexate/pharmacology , Mice , Mice, Inbred BALB C , Ovalbumin/chemistry , Ovalbumin/immunology , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...