Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 450
Filter
Add more filters

Publication year range
1.
Biochem Soc Trans ; 48(3): 1109-1119, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32453419

ABSTRACT

The development of B lymphocytes into antibody-secreting plasma cells is central to the adaptive immune system in that it confers protective and specific antibody response against invading pathogen. This developmental process involves extensive morphological and functional alterations that begin early after antigenic stimulation. These include chromatin restructuring that is critical in regulating gene expression, DNA rearrangement and other cellular processes. Here we outline the recent understanding of the three-dimensional architecture of the genome, specifically focused on its contribution to the process of B cell activation and terminal differentiation into antibody-secreting cells.


Subject(s)
Antibodies/metabolism , B-Lymphocytes/metabolism , Genome , Plasma Cells/metabolism , Adaptive Immunity , Animals , Antibody Formation , Antibody-Producing Cells/cytology , Cell Differentiation , Cell Division , DNA/metabolism , Gene Expression Regulation , Humans , Lymphocyte Activation , Recombination, Genetic , Transcription, Genetic
2.
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
3.
Immunol Rev ; 270(1): 65-77, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26864105

ABSTRACT

Monoclonal antibodies are essential therapeutics and diagnostics in a large number of diseases. Moreover, they are essential tools in all sectors of life sciences. Although the great majority of monoclonal antibodies currently in use are of mouse origin, the use of human B cells to generate monoclonal antibodies is increasing as new techniques to tap the human B cell repertoire are rapidly emerging. Cloned lines of immortalized human B cells are ideal sources of monoclonal antibodies. In this review, we summarize our studies to the regulation of the replicative life span, differentiation, and maturation of B cells that led to the development of a platform that uses immortalization of human B cells by in vitro genetic modification for antibody development. We describe a number of human antibodies that were isolated using this platform and the application of the technique in other species. We also discuss the use of immortalized B cells as antigen-presenting cells for the discovery of tumor neoantigens.


Subject(s)
Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Self Renewal , Animals , Antibodies, Monoclonal/biosynthesis , Antibody Formation/immunology , Antibody-Producing Cells/cytology , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B-Lymphocytes/cytology , Cell Culture Techniques , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Drug Discovery , Gene Expression Regulation , Germinal Center/cytology , Germinal Center/physiology , Humans , Immunologic Memory , Interleukins/metabolism , Phenotype , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Vaccines/immunology , bcl-X Protein/genetics , bcl-X Protein/metabolism
4.
J Immunol ; 196(7): 2986-94, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26895835

ABSTRACT

Fanconi anemia (FA) is characterized by a progressive bone marrow failure and an increased incidence of cancer. FA patients have high susceptibility to immune-related complications such as infection and posttransplant graft-versus-host disease. In this study, we investigated the effect of FA deficiency in B cell function using the Fancc mouse model. Fancc(-/-) B cells show a specific defect in IgG2a switch and impaired Ab-secreting cell (ASC) differentiation. Global transcriptome analysis of naive B cells by mRNA sequencing demonstrates that FA deficiency deregulates a network of genes involved in immune function. Significantly, many genes implicated in Wnt signaling were aberrantly expressed in Fancc(-/-) B cells. Consistently, Fancc(-/-) B cells accumulate high levels of ß-catenin under both resting and stimulated conditions, suggesting hyperactive Wnt signaling. Using an in vivo Wnt GFP reporter assay, we verified the upregulation of Wnt signaling as a potential mechanism responsible for the impaired Fancc(-/-) B cell differentiation. Furthermore, we showed that Wnt signaling inhibits ASC differentiation possibly through repression of Blimp1 and that Fancc(-/-) B cells are hypersensitive to Wnt activation during ASC differentiation. Our findings identify Wnt signaling as a physiological regulator of ASC differentiation and establish a role for the Wnt pathway in normal B cell function and FA immune deficiency.


Subject(s)
Antibody-Producing Cells/cytology , Antibody-Producing Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Fanconi Anemia Complementation Group C Protein/deficiency , Wnt Signaling Pathway , Animals , Antibody-Producing Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cluster Analysis , Gene Expression Profiling , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Knockout , Transcriptome , Wnt Proteins/metabolism
5.
J Proteome Res ; 16(1): 45-54, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27779884

ABSTRACT

One direct route for the discovery of therapeutic human monoclonal antibodies (mAbs) involves the isolation of peripheral B cells from survivors/sero-positive individuals after exposure to an infectious reagent or disease etiology, followed by single-cell sequencing or hybridoma generation. Peripheral B cells, however, are not always easy to obtain and represent only a small percentage of the total B-cell population across all bodily tissues. Although it has been demonstrated that tandem mass spectrometry (MS/MS) techniques can interrogate the full polyclonal antibody (pAb) response to an antigen in vivo, all current approaches identify MS/MS spectra against databases derived from genetic sequencing of B cells from the same patient. In this proof-of-concept study, we demonstrate the feasibility of a novel MS/MS antibody discovery approach in which only serum antibodies are required without the need for sequencing of genetic material. Peripheral pAbs from a cytomegalovirus-exposed individual were purified by glycoprotein B antigen affinity and de novo sequenced from MS/MS data. Purely MS-derived mAbs were then manufactured in mammalian cells to validate potency via antigen-binding ELISA. Interestingly, we found that these mAbs accounted for 1 to 2% of total donor IgG but were not detected in parallel sequencing of memory B cells from the same patient.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , B-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Immunoglobulin G/chemistry , Sequence Analysis, Protein , Amino Acid Sequence , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/biosynthesis , Antibodies, Viral/isolation & purification , Antibody Formation , Antibody-Producing Cells/cytology , Antibody-Producing Cells/immunology , B-Lymphocytes/virology , Chromatography, Affinity/methods , Cytomegalovirus/growth & development , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/virology , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immune Sera/chemistry , Immunoglobulin G/biosynthesis , Immunoglobulin G/isolation & purification , Tandem Mass Spectrometry , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
6.
J Immunol ; 194(1): 231-42, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25429072

ABSTRACT

Natural IgM is constitutively present in the serum, where it aids in the early control of viral and bacterial expansions. Natural IgM also plays a significant role in the prevention of autoimmune disease by promoting the clearance of cellular debris. Nevertheless, the origins of natural IgM have not been precisely defined. Previous studies focused on the role of CD5(+) B1 cells in the production of natural IgM, but we show in this article that a discrete population of CD5(-) IgM plasmablasts and plasma cells in the bone marrow (BM) produces the majority of serum IgM in resting mice. These Ab-secreting cells (ASC) originate from peritoneal cavity-resident cells, because transfer of peritoneal cells completely restores serum IgM and the specific compartment of BM ASC in Rag1-deficient mice. We show that BM natural IgM ASC arise from a fetal-lineage progenitor that is neither B1a nor B1b, and that this IgM ASC compartment contains a substantial fraction of long-lived plasma cells that do not occupy the IgG plasma cell survival niche in the BM; instead, they are supported by IL-5. In summary, we identified the primary source of natural IgM and showed that these ASC are maintained long-term in a unique survival niche within the BM.


Subject(s)
Antibody-Producing Cells/immunology , Bone Marrow Cells/immunology , Immunoglobulin M/immunology , Plasma Cells/immunology , Animals , Antibody-Producing Cells/cytology , Base Sequence , CD40 Ligand/immunology , CD5 Antigens/genetics , Cell Count , Cell Differentiation/immunology , Female , Homeodomain Proteins/genetics , Immunoglobulin G/immunology , Immunoglobulin M/blood , Interleukin-5/immunology , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Peritoneal Cavity/cytology , Sequence Analysis, DNA , V(D)J Recombination/genetics , V(D)J Recombination/immunology
7.
J Immunol ; 188(3): 1036-48, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22198952

ABSTRACT

Regulatory B cells that are functionally defined by their capacity to express IL-10 (B10 cells) downregulate inflammation and autoimmunity. In studies using well-defined IL-10 reporter mice, this rare B10 cell subset was also found to maintain a capacity for plasma cell differentiation. During a transient period of il10 transcription, the blimp1 and irf4 transcription factors were induced in B10 cells, whereas pax5 and bcl6 were downregulated as a significant fraction of B10 cells completed the genetic and phenotypic program leading to Ab-secreting cell differentiation in vitro and in vivo. B10 cell-derived IgM reacted with both self- and foreign Ags, whereas B10 cells generated Ag-specific IgG in response to immunizations. Moreover, B10 cells represented a significant source of serum IgM and IgG during adoptive-transfer experiments and produced Ag-specific, polyreactive and autoreactive Ab specificities that were consistent with their expression of a diverse AgR repertoire. Thereby, B10 cells limit inflammation and immune responses by the transient production of IL-10, and may facilitate clearance of their eliciting Ags through an inherent capacity to quickly generate polyreactive and/or Ag-specific Abs during humoral immune responses.


Subject(s)
Antibody-Producing Cells/cytology , B-Lymphocyte Subsets/cytology , B-Lymphocytes, Regulatory/immunology , Cell Differentiation/immunology , Interleukin-10/biosynthesis , Animals , Antigens , Autoimmunity , B-Lymphocyte Subsets/immunology , B-Lymphocytes, Regulatory/cytology , Gene Expression Regulation , Immunity, Humoral , Immunoglobulin G , Immunoglobulin M , Inflammation , Mice
8.
J Immunol ; 189(12): 5729-44, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23162132

ABSTRACT

Targeting CD14(+) dermal-derived dendritic cells (DDCs) is a rational approach for vaccination strategies aimed at improving humoral immune responses, because of their natural ability to stimulate naive B cells. In this study, we show that CD14(+) DDCs express mRNA for TLRs 1-9, but respond differentially to single or paired TLR ligands. Compared to single ligands, some combinations were particularly effective at activating CD14(+) DDCs, as shown by enhanced expression of B cell stimulatory cytokines (IL-6, IL-10, and TNF-α) and more pronounced phenotypic maturation. These combinations were resiquimod (R-848) plus polyinosinic-polycytidylic acid [Poly(I:C)], R-848 plus LPS, Pam3CSK4 plus Poly(I:C), and LPS plus Poly(I:C). We also found that selected TLR ligand pairs [R-848 plus either LPS or Poly(I:C)] were superior to individual agents at boosting the inherent capacity of CD14(+) DDCs to induce naive B cells to proliferate and differentiate into CD27(+) CD38(+) B cells that secrete high levels of IgG and IgA. When treated with the same TLR ligand combinations, CD14(+) DDCs also promoted the differentiation of Th1 (IFN-γ-secreting) CD4(+) T cells, but not of Th2 or Th17 CD4(+) T cells. These observations may help to identify adjuvant strategies aimed at inducing protective immune responses to various pathogens, including but not limited to HIV-1.


Subject(s)
Antibody-Producing Cells/immunology , B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Dermis/immunology , Lipopolysaccharide Receptors/biosynthesis , Lymphocyte Activation/immunology , Toll-Like Receptors/metabolism , Adolescent , Adult , Antibody-Producing Cells/cytology , Antibody-Producing Cells/metabolism , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dermis/cytology , Dermis/metabolism , Humans , Immunophenotyping , Ligands , Male , Middle Aged , Toll-Like Receptors/genetics , Young Adult
9.
J Immunol ; 189(2): 1036-42, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22711890

ABSTRACT

The resolution of inflammation is an active and dynamic process critical in maintaining homeostasis. Newly identified lipid mediators have been recognized as key players during the resolution phase. These specialized proresolving mediators (SPM) constitute separate families that include lipoxins, resolvins, protectins, and maresins, each derived from essential polyunsaturated fatty acids. New results demonstrate that SPM regulate aspects of the immune response, including reduction of neutrophil infiltration, decreased T cell cytokine production, and stimulation of macrophage phagocytic activity. The actions of SPM on B lymphocytes remain unknown. Our study shows that the novel SPM 17-hydroxydosahexaenoic acid (17-HDHA), resolvin D1, and protectin D1 are present in the spleen. Interestingly, 17-HDHA and resolvin D1, but not protectin D1, strongly increase activated human B cell IgM and IgG production. Furthermore, increased Ab production by 17-HDHA is due to augmented B cell differentiation toward a CD27(+)CD38(+) Ab-secreting cell phenotype. The 17-HDHA did not affect proliferation and was nontoxic to cells. Increase of plasma cell differentiation and Ab production supports the involvement of SPM during the late stages of inflammation and pathogen clearance. The present study provides new evidence for SPM activity in the humoral response. These new findings highlight the potential applications of SPM as endogenous and nontoxic adjuvants, and as anti-inflammatory therapeutic molecules.


Subject(s)
Antibody-Producing Cells/cytology , Antibody-Producing Cells/immunology , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Docosahexaenoic Acids/physiology , Up-Regulation/immunology , Animals , Antibody-Producing Cells/metabolism , B-Lymphocyte Subsets/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chromatography, Liquid , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Metabolomics/methods , Mice , Mice, Inbred Strains , Tandem Mass Spectrometry , Up-Regulation/drug effects
10.
J Exp Med ; 203(12): 2683-90, 2006 Nov 27.
Article in English | MEDLINE | ID: mdl-17101733

ABSTRACT

After induction in secondary lymphoid organs, a subset of antibody-secreting cells (ASCs) homes to the bone marrow (BM) and contributes to long-term antibody production. The factors determining secondary lymphoid organ residence versus BM tropism have been unclear. Here we demonstrate that in mice treated with FTY720 or that lack sphingosine-1-phosphate (S1P) receptor-1 (S1P1) in B cells, IgG ASCs are induced and localize normally in secondary lymphoid organs but they are reduced in numbers in blood and BM. Many IgG ASCs home to BM on day 3 of the secondary response and day 3 splenic ASCs exhibit S1P responsiveness, whereas the cells remaining at day 5 are unable to respond. S1P1 mRNA abundance is higher in ASCs isolated from blood compared to spleen, whereas CXCR4 expression is lower. Blood ASCs also express higher amounts of Kruppel-like factor (KLF)2, a regulator of S1P1 gene expression. These findings establish an essential role for S1P1 in IgG plasma cell homing and they suggest that differential regulation of S1P1 expression in differentiating plasma cells may determine whether they remain in secondary lymphoid organs or home to BM.


Subject(s)
Bone Marrow Cells/cytology , Cell Movement/immunology , Lymphoid Tissue/metabolism , Lysophospholipids/biosynthesis , Plasma Cells/metabolism , Receptors, Lysosphingolipid/biosynthesis , Sphingosine/analogs & derivatives , Animals , Antibody-Producing Cells/cytology , Antibody-Producing Cells/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Immunoglobulin G/biosynthesis , Lymphoid Tissue/cytology , Lysophospholipids/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/cytology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/physiology , Sphingosine/biosynthesis , Sphingosine/physiology
11.
J Autoimmun ; 39(3): 173-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22695187

ABSTRACT

Maturation of B cells depends on environmental stimuli. Peripheral immature B cells develop into follicular pathway when antigenic stimulation is combined with T cell signals. Here, we wished to identify stimuli contributing to the development into marginal zone B cells known to be involved in autoimmune response. We found that TLR9 stimulation of transitional B cells induces proliferation and specific maturation into CD24(-) CD38(+) CD21(high) CD23(low) IgM(high) IgD(low) and Notch2(high) B cells characteristics of marginal zone B cells. Terminal differentiation into antibody-secreting cell associated with isotype switch commitment is also triggered which leads to a striking production of autoantibodies. Interestingly, mature B cells do not differentiate into marginal zone pathway following TLR9 stimulation, nor do transitional B cells under antigenic and T cell combined signals. These results suggest that transitional B cells are specifically sensitive to TLR9 stimulation to produce autoreactive marginal zone B cells.


Subject(s)
Antibody-Producing Cells/immunology , Autoantibodies/biosynthesis , Autoimmunity , Precursor Cells, B-Lymphoid/immunology , Toll-Like Receptor 9/metabolism , Adjuvants, Immunologic/pharmacology , Antibody-Producing Cells/cytology , Antibody-Producing Cells/drug effects , Antigens, CD/immunology , Autoantibodies/immunology , Cell Differentiation , Cell Proliferation/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/immunology , Flow Cytometry , Humans , Lymphocyte Activation/drug effects , Oligodeoxyribonucleotides/pharmacology , Palatine Tonsil/cytology , Palatine Tonsil/drug effects , Palatine Tonsil/immunology , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/drug effects , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Toll-Like Receptor 9/immunology
12.
Radiats Biol Radioecol ; 52(3): 229-33, 2012.
Article in Russian | MEDLINE | ID: mdl-22891544

ABSTRACT

The ability of post-radiation (4 Gy) bystander chemosignals (the volatile components of mouse urine) to distantly modulate the humoral immune response to the sheep red blood cells in the spleen and popliteal lymph nodes of intact recipients has been investigated. It was shown that the exposure of animals to chemosignals before antigen injection resulted in the decrease and increase of the immune response in the spleen and lymph nodes, respectively. When animals were exposed to chemosignals after the antigenic stimulus, an increased immune response was observed in both spleen and lymph nodes. The contribution of radiation-induced bystander signaling in the response of socially organized animals to the effect of ionizing irradiation is discussed.


Subject(s)
Bystander Effect/radiation effects , Gamma Rays , Immunity, Humoral , Lymph Nodes/immunology , Spleen/immunology , Urine , Animals , Antibody-Producing Cells/cytology , Antibody-Producing Cells/drug effects , Antibody-Producing Cells/immunology , Antibody-Producing Cells/radiation effects , Antigens/immunology , Cell Count , Erythrocytes/immunology , Immunity, Humoral/drug effects , Immunity, Humoral/radiation effects , Lymph Nodes/drug effects , Lymph Nodes/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Radiation Injuries, Experimental/immunology , Radiation Injuries, Experimental/urine , Sheep , Spleen/drug effects , Spleen/radiation effects , Urine/chemistry , Volatilization , Whole-Body Irradiation
13.
Cell Immunol ; 268(1): 9-15, 2011.
Article in English | MEDLINE | ID: mdl-21296339

ABSTRACT

Common variable immunodeficiency (CVID) is a primary immunodeficiency characterised by hypogammaglobulinaemia and antibody deficiency to T dependent and independent antigens. Patients suffer from recurrent respiratory infections and poor response to vaccination. Although the underlying molecular defect is unknown, most CVID patients show impaired late B cell differentiation. We investigated B cell differentiation and immunoglobulin secretion induced by two different stimuli: TLR9 specific ligand (CpG-ODN) and anti-CD40 combined with IL21. The contribution of BCR signalling (anti-IgM stimulation) was also evaluated. B cells from CVID patients produced low levels of IgG and IgA in response to both kinds of stimuli that was not restored by anti-IgM. Production of IgM was conserved when cells were stimulated with anti-CD40 and IL21. These results point to a wide signalling defect in B lymphocytes from CVID patients that may be related to their hypogammaglobulinaemia and poor response to vaccination.


Subject(s)
Antibody-Producing Cells , B-Lymphocytes , CD40 Ligand/immunology , Cell Differentiation/drug effects , Common Variable Immunodeficiency/immunology , Interleukins/pharmacology , Toll-Like Receptor 9/immunology , Adult , Aged , Antibodies/immunology , Antibodies/pharmacology , Antibody-Producing Cells/cytology , Antibody-Producing Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Female , Humans , Immunoglobulins/immunology , Male , Middle Aged , Oligodeoxyribonucleotides/pharmacology
14.
Br J Nutr ; 105(1): 24-30, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20727240

ABSTRACT

Mortality of neonates continues to be a major problem in humans and animals. IgA provides protection against microbial antigens at mucosal surfaces. Although ß-carotene supplementation has been expected to enhance retinoic acid-mediated immune response in neonates, the exact mechanism by which ß-carotene enhances IgA production is still unclear. We investigated the effect of supplemental ß-carotene for maternal mice during pregnancy and lactation on IgA antibody-secreting cells (ASC) in mammary gland and guts and on IgA transfer from milk to neonatal mice. Pregnant mice were fed untreated or 50 mg/kg ß-carotene-supplemented diets from 6·5 d postcoitus (dpc) to 14 d postpartum (dpp). Supplemental ß-carotene increased the numbers of IgA ASC in mammary gland (P < 0·05) and ileum (P < 0·001), and also mRNA expression of IgA C-region in ileum (P < 0·05) of maternal mice at 14 dpp, but few IgA ASC were detected in mammary gland at 17·5 dpc. IgA concentration in stomach contents, which represents milk IgA level, was significantly higher (P < 0·01) in neonatal mice born to ß-carotene-supplemented mothers at 7 and 14 dpp, and IgA concentration in serum, stomach contents and faeces increased (P < 0·001) drastically with age. These results suggest that ß-carotene supplementation for maternal mice during pregnancy and lactation is useful for enhancing IgA transfer from maternal milk to neonates owing to the increase in IgA ASC in mammary gland and ileum during lactation.


Subject(s)
Antibody-Producing Cells/drug effects , Immunoglobulin A/metabolism , Lactation/immunology , Mammary Glands, Animal/immunology , Maternal Nutritional Physiological Phenomena/immunology , Milk/immunology , beta Carotene/pharmacology , Animals , Animals, Newborn , Animals, Suckling , Antibody-Producing Cells/cytology , Antibody-Producing Cells/immunology , Dietary Supplements , Feces/chemistry , Female , Gastrointestinal Contents/chemistry , Ileum/immunology , Immunoglobulin A/genetics , Male , Mice , Mice, Inbred ICR , Pregnancy , RNA, Messenger/metabolism , beta Carotene/blood , beta Carotene/metabolism
16.
J Immunol ; 183(12): 7851-9, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19933861

ABSTRACT

We have recently reported that the sublingual (s.l.) mucosa is an efficient site for inducing systemic and mucosal immune responses. In this study, the potential of s.l. immunization to induce remote Ab responses and CD8(+) cytotoxic responses in the female genital tract was examined in mice by using a nonreplicating Ag, OVA, and cholera toxin (CT) as an adjuvant. Sublingual administration of OVA and CT induced Ag-specific IgA and IgG Abs in blood and in cervicovaginal secretions. These responses were associated with large numbers of IgA Ab-secreting cells (ASCs) in the genital mucosa. Genital ASC responses were similar in magnitude and isotype distribution after s.l., intranasal, or vaginal immunization and were superior to those seen after intragastric immunization. Genital, but not blood or spleen, IgA ASC responses were inhibited by treatment with anti-CCL28 Abs, suggesting that the chemokine CCL28 plays a major role in the migration of IgA ASC progenitors to the reproductive tract mucosa. Furthermore, s.l. immunization with OVA induced OVA-specific effector CD8(+) cytolytic T cells in the genital mucosa, and these responses required coadministration of the CT adjuvant. Furthermore, s.l. administration of human papillomavirus virus-like particles with or without the CT adjuvant conferred protection against genital challenge with human papillomavirus pseudovirions. Taken together, these findings underscore the potential of s.l. immunization as an efficient vaccination strategy for inducing genital immune responses and should impact on the development of vaccines against sexually transmitted diseases.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antibody-Producing Cells/immunology , Cholera Toxin/immunology , Human papillomavirus 16/immunology , Ovalbumin/immunology , Papillomavirus Infections/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Uterine Cervical Neoplasms/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Sublingual , Animals , Antibodies, Viral/biosynthesis , Antibody-Producing Cells/cytology , Antibody-Producing Cells/virology , Cell Differentiation/immunology , Cells, Cultured , Cholera Toxin/administration & dosage , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucous Membrane/immunology , Mucous Membrane/virology , Ovalbumin/administration & dosage , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , T-Lymphocytes, Cytotoxic/virology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Virion/immunology
17.
PLoS One ; 16(11): e0259644, 2021.
Article in English | MEDLINE | ID: mdl-34767590

ABSTRACT

BACKGROUND: Streptococcus pneumoniae infections cause morbidity and mortality worldwide. A rapid, simple diagnostic method could reduce the time needed to introduce definitive therapy potentially improving patient outcomes. METHODS: We introduce two new methods for diagnosing S. pneumoniae infections by measuring the presence of newly activated, pathogen-specific, circulating Antibody Secreting Cells (ASC). First, ASC were detected by ELISpot assays that measure cells secreting antibodies specific for signature antigens. Second, the antibodies secreted by isolated ASC were collected in vitro in a novel matrix, MENSA (media enriched with newly synthesized antibodies) and antibodies against S. pneumoniae antigens were measured using Luminex immunoassays. Each assay was evaluated using blood from S. pneumoniae and non-S. pneumoniae-infected adult patients. RESULTS: We enrolled 23 patients with culture-confirmed S. pneumoniae infections and 24 controls consisting of 12 non-S. pneumoniae infections, 10 healthy donors and two colonized with S. pneumoniae. By ELISpot assays, twenty-one of 23 infected patients were positive, and all 24 controls were negative. Using MENSA samples, four of five S. pneumoniae-infected patients were positive by Luminex immunoassays while all five non-S. pneumoniae-infected patients were negative. CONCLUSION: Specific antibodies produced by activated ASC may provide a simple diagnostic for ongoing S. pneumoniae infections. This method has the potential to diagnose acute bacterial infections.


Subject(s)
Antibodies, Bacterial/blood , Antibody-Producing Cells , Diagnostic Tests, Routine/methods , Immunoassay/methods , Pneumococcal Infections , Streptococcus pneumoniae/immunology , Adult , Aged , Aged, 80 and over , Antibody-Producing Cells/cytology , Antibody-Producing Cells/immunology , Female , Humans , Male , Middle Aged , Pneumococcal Infections/diagnosis , Pneumococcal Infections/immunology , Young Adult
18.
J Exp Med ; 144(1): 288-92, 1976 Jul 01.
Article in English | MEDLINE | ID: mdl-819612

ABSTRACT

Neonatally initiated injection of anti-mu antiserum in mice has been shown to suppress the formation of reaginic antibodies in response to infection with the intestinal nematode, Nippostrongylus brasiliensis. This observation supports the hypothesis that IgE-producing cells arise from IgM-bearing precursors.


Subject(s)
Antibodies , Antibody Formation , Immunoglobulin E/pharmacology , Immunoglobulin Heavy Chains , Immunoglobulin mu-Chains , Antibody-Producing Cells/cytology , Antibody-Producing Cells/immunology , Cell Differentiation , Immunoglobulin M/biosynthesis , Immunosuppression Therapy , Nippostrongylus/immunology
19.
J Exp Med ; 155(3): 914-23, 1982 Mar 01.
Article in English | MEDLINE | ID: mdl-6977612

ABSTRACT

We report here a factor (B cell growth factor) found in induced supernatants of the mouse thymoma EL4 that co-stimulates with anti-IgM antibodies in short-term cultures of purified B lymphocytes to induce polyclonal B cell proliferation but not antibody-forming cell production. The factor is not mitogenic for resting B cells and interacts with anti-IgM-activated B cells in a non-H-2-restricted manner. Absorption studies and molecular weight analysis reveal the factor is distinct from interleukin 2. This factor synergises with antigen, interleukin 2, and an interleukin 2-free, B cell growth factor-free T cell supernatant that contains T cell-replacing factor to produce erythrocyte-specific plaque-forming cells in cultures of highly purified B cells.


Subject(s)
B-Lymphocytes/cytology , Interleukin-2/pharmacology , Lymphocyte Activation , Lymphokines/pharmacology , Animals , Antibodies, Anti-Idiotypic/immunology , Antibody-Producing Cells/cytology , Immunoglobulin M/immunology , Mice , Mice, Inbred BALB C , Molecular Weight
20.
J Exp Med ; 135(3): 491-502, 1972 Mar 01.
Article in English | MEDLINE | ID: mdl-4550768

ABSTRACT

Efferent lymph of the popliteal lymph nodes of rabbits was collected 4 days after a single footpad injection of SRBC. Thin-layer agar plating was done to isolate plaque-forming cells of the lymph for electron microscope examination, and the numbers of plaque-forming cells (PFC) in cells from the lymph and lymph nodes were determined. Of 71 PFC of lymph isolated and examined, 93% were lymphocytes, most of them with signs of substantial levels of physiologic activity. The cytoplasm showed an abundance of free ribosomes and many finger-like projections. The endoplasmic reticulum (ER) was barely detectable in most of the active lymphocytic PFC, and in some, a few short narrow channels of ER could be seen. Approximately one-fifth of the lymphocytic PFC presented an appearance of senescence, with signs of degeneration: rounded cells, with amorphous nuclear chromatin, and very few microvilli. The remaining 7% of the PFC of the lymph showed an unusual combination of features: small round cells with a narrow ring of cytoplasm which, however, contained well-organized channels of ER. Such cells had been found only among PFC of peripheral blood of the rabbit. The number of PFC per million cells was higher in the lymph than in the suspensions of lymph node cells. In both the contralateral lymph node and its efferent lymph, the number of PFC was less than 1% that of the injected side.


Subject(s)
Antibody-Producing Cells/cytology , Lymph/cytology , Animals , Cell Count , Erythrocytes/immunology , Hemolytic Plaque Technique , In Vitro Techniques , Lymph/immunology , Lymph Nodes/cytology , Microscopy, Electron , Plasma Cells/cytology , Rabbits , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL