Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Methods Mol Med ; 136: 3-18, 2007.
Article in English | MEDLINE | ID: mdl-17983136

ABSTRACT

B-cells and antibody-secreting plasma cells are key players in protective immunity, but also in autoimmune disease. To understand their various functions in the initiation and maintenance of autoimmune pathology, a detailed dissection of their functional diversity is mandatory. This requires a detailed phenotypic classification of the diversity of B-cells. Here, technologies of immunocytometry and ELISpot are described in detail, and their value for phenotypic characterization of cells of the B lineage, as well as for preparative cell sorting, to further characterize them functionally and on the molecular level are described.


Subject(s)
B-Lymphocytes/physiology , Phenotype , Plasma Cells/physiology , Animals , Cell Differentiation , Cell Separation/methods , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism
2.
Infect Immun ; 75(3): 1413-23, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17220318

ABSTRACT

Cholera toxin (CT) and the type II heat-labile enterotoxins (LT-IIa and LT-IIb) are potent immunological adjuvants which are hypothesized to enhance the production of antibody (Ab)-secreting cells, although their mechanisms of action are not fully understood. The treatment of splenic cells with concanavalin A (ConA) plus CT enhanced the production of immunoglobulin A (IgA) and IgM by dividing cells that expressed high levels of major histocompatibility complex class II (MHC-II), CD19, and CD138 and low levels of B220 a phenotype characteristic of plasma blasts. LT-IIa or LT-IIb moderately enhanced IgA and IgM production without enhancing plasma blast differentiation. CT up-regulated CD25, CD69, CD80, CD86, and MHC-II in isolated B cells but failed to induce proliferation or differentiation. The treatment of unfractionated splenic cells with ConA plus CT induced B-cell proliferation and differentiation, but the elimination of CD4(+) T cells inhibited this effect. CT treatment of ConA-activated CD4(+) T cells up-regulated CD134 and CD154, whereas the blockage of CD40-CD154 interactions inhibited the induction of plasma blasts and Ig synthesis. The treatment of unfractionated splenic cells with CT, LT-IIa, or LT-IIb enhanced the production of interleukin-6 (IL-6) and IL-10, whereas the production of gamma interferon was inhibited in both CD4(+) and CD8(+) T cells mostly by CT. Thus, major regulatory effects of CT on lymphocytes are likely exerted early during the induction of immune responses when B and T cells initially encounter antigen. Neither LT-IIa or LT-IIb had these effects, indicating that type II enterotoxins augment Ab responses by other mechanisms.


Subject(s)
CD40 Ligand/biosynthesis , Cholera Toxin/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin M/biosynthesis , Interferon-gamma/antagonists & inhibitors , Plasma Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , Up-Regulation/immunology , Animals , CD40 Ligand/genetics , CD40 Ligand/physiology , Cells, Cultured , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Plasma Cells/metabolism
3.
Nat Rev Immunol ; 6(10): 741-50, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16977339

ABSTRACT

Plasma cells provide humoral immunity. They have traditionally been viewed mainly as short-lived end-stage products of B-cell differentiation that deserve little interest. This view is changing, however, because we now know that plasma cells can survive for long periods in the appropriate survival niches and that they are an independent cellular component of immunological memory. Studies of the biology of plasma cells reveal a mechanism of intriguing simplicity and elegance that focuses memory provided by plasma cells on recently encountered pathogens while minimizing the 'fading' of memory for pathogens encountered in the distant past. This mechanism is based on competition for survival niches between newly generated plasmablasts and older plasma cells.


Subject(s)
Cell Differentiation/immunology , Immunologic Memory , Plasma Cells/cytology , Plasma Cells/immunology , Animals , Humans
4.
Immunol Rev ; 211: 295-302, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16824136

ABSTRACT

Immunological memory, as provided by antibodies, depends on the continued presence of antibody-secreting cells, such as long-lived plasma cells of the bone marrow. Survival niches for these memory plasma cells are limited in number. In an established immune system, acquisition of new plasma cells, generated in response to recent pathogenic challenges, requires elimination of old memory plasma cells. Here, we review the adaptation of plasma cell memory to new pathogens. This adaptation is dependent upon the influx of plasmablasts, generated in a secondary systemic immune reaction, into the pool of memory plasma cells, the efficiency of competition of new plasmablasts with old plasma cells, and the frequency of infection with novel pathogens. To maintain old plasma cells at frequencies high enough to provide protection and to accommodate as many specificities as possible, an optimal influx rate per infection exists. This optimal rate is approximately three times higher than the minimal number of plasma cells providing protection. Influx rates of plasmablasts generated by vaccination approximately match this optimum level. Furthermore, the observed stability of serum concentrations of vaccine-specific antibodies implies that the influxing plasmablasts mobilize a similar number of plasma cells and that competitive infectious challenges are not more frequent than once per month.


Subject(s)
Adaptation, Biological/immunology , Immunologic Memory/immunology , Plasma Cells/immunology , Animals , Antibody Formation , Humans , Infections/immunology , Mice , Models, Immunological
5.
Immunol Lett ; 103(2): 83-5, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16280169

ABSTRACT

Following tetanus vaccination, a wave of antibody-secreting cells appear in the peripheral blood composed of vaccine-specific, migratory plasmablasts and plasma cells secreting antibodies specific for other antigens. The latter probably were tissue resident plasma cells formed in earlier immune responses that are mobilized due to competition with the newly formed tetanus-specific plasmablasts. Newly formed plasma cells secreting antibodies specific for a particular antigen/vaccine are accommodated in the bone marrow likely at the global expense of the pre-existing long-lived plasma cell population providing humoral memory for other antigens. Plasmablasts but not mature plasma cells are attracted by the ligands for the chemokine receptors CXCR4 and CXCR3. While CXCR4 and its cognate ligand is important for plasma cell homing to the bone marrow, CXCR3 and its ligand IP10 are likely to be involved in attracting them to inflamed tissue. In NZB/W mice, a model for systemic lupus, long-lived autoreactive plasma cells are present not only in bone marrow, but also in inflamed tissues and spleen. Autoreactive plasma cells in the spleen are present long before the onset of the disease, suggesting that these cells contribute to induction of immunopathology.


Subject(s)
Autoimmunity/physiology , Immunity/physiology , Plasma Cells/immunology , Receptors, Chemokine/metabolism , Animals , Humans , Plasma Cells/metabolism
6.
Blood ; 105(10): 3965-71, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15687242

ABSTRACT

C-X-C motif chemokine receptor 3 (CXCR3) and CXCR4 expressed on immunoglobulin G (IgG)-plasma-cell precursors formed in memory immune responses are crucial modulators of the homing of these cells. Here, we studied the regulation of the expression of these chemokine receptors during the differentiation of human memory B cells into plasma cells. We show that CXCR3 is absent on CD27- naive B cells but is expressed on a fraction of memory B cells, preferentially on those coexpressing IgG1. On differentiation into plasma-cell precursors, CXCR3+ memory B cells maintain the expression of this chemokine receptor. CXCR3- memory B cells up-regulate CXCR3 and migrate toward concentration gradients of its ligands only when costimulated with interferon gamma (IFN-gamma), but not interleukin 4 (IL-4), IL-1beta, IL-6, IFN-alpha, IFN-beta, or tumor necrosis factor alpha (TNF-alpha). In contrast, the differentiation of CXCR4- B cells into plasma cells is generally accompanied by the induction of CXCR4 expression. These results show that lack of CXCR4 expression on plasma-cell precursors is not a limiting factor for plasma-cell homing and that the expression of CXCR3 on memory B cells and plasma-cell precursors is induced by IFN-gamma, provided in human T helper type 1 (Th1)-biased immune responses. Once induced in memory B cells, CXCR3 expression remains part of the individual cellular memory.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation , Gene Expression Regulation , Immunologic Memory , Receptors, CXCR4/metabolism , Receptors, Chemokine/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Cycle , Cell Differentiation/drug effects , Cell Movement , Cells, Cultured , Chemokine CXCL9 , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunologic Memory/drug effects , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Receptors, CXCR3 , Receptors, CXCR4/immunology , Receptors, Chemokine/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Up-Regulation/drug effects
7.
Blood ; 105(4): 1614-21, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15507523

ABSTRACT

Maintenance of protective humoral immunity depends on the generation and survival of antibody-secreting cells. The bone marrow provides niches for long-term survival of plasma cells generated in the course of systemic immune responses in secondary lymphoid organs. Here, we have analyzed migratory human plasma blasts and plasma cells after secondary vaccination with tetanus toxin. On days 6 and 7 after immunization, CD19(+)/CD27(high)/intracellular immunoglobulin G(high) (IgG(high))/HLA-DR(high)/CD38(high)/CD20(-)/CD95(+) tetanus toxin-specific antibody-secreting plasma blasts were released in large numbers from the secondary lymphoid organs into the blood. These cells show chemotactic responsiveness toward ligands for CXCR3 and CXCR4, probably guiding them to the bone marrow or inflamed tissue. At the same time, a population of CD19(+)/CD27(high)/intracellular IgG(high)/HLA-DR(low)/CD38(+)/CD20(-)/CD95(+) cells appeared in the blood in large numbers. These cells, with the phenotype of long-lived plasma cells, secreted antibodies of unknown specificity, not tetanus toxoid. The appearance of these plasma cells in the blood indicates successful competition for survival niches in the bone marrow between newly generated plasma blasts and resident plasma cells as a fundamental mechanism for the establishment of humoral memory and its plasticity.


Subject(s)
Chemotaxis, Leukocyte/immunology , Epitopes, B-Lymphocyte/immunology , Immunization, Secondary , Lymphocyte Activation , Plasma Cells/cytology , Plasma Cells/immunology , Adult , Antibodies, Bacterial/biosynthesis , Antibody Specificity , Antibody-Producing Cells/immunology , Antigens, CD19/biosynthesis , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Chemokine CXCL12 , Chemokine CXCL9 , Chemokines, CXC/metabolism , Female , HLA-DR Antigens/biosynthesis , Humans , Immunologic Memory , Immunophenotyping , Intercellular Signaling Peptides and Proteins/metabolism , Kinetics , Lymphocyte Count , Male , Plasma Cells/metabolism , Receptors, CXCR3 , Receptors, CXCR4/biosynthesis , Receptors, Chemokine/biosynthesis , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Vaccines, Synthetic/immunology
8.
J Leukoc Biol ; 75(6): 1022-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15020647

ABSTRACT

Despite the important role immunoglobulin G (IgG)-secreting plasma cells play in memory immune responses, the differentiation and homeostasis of these cells are not completely understood. Here, we studied the differentiation of human IgG-secreting cells ex vivo and in vitro, identifying these cells by the cellular affinity matrix technology. Several subpopulations of IgG-secreting cells were identified among the cells isolated from tonsils and bone marrow, particularly differing in the expression levels of CD9, CD19, and CD38. CD38 low IgG-secreting cells were present exclusively in the tonsils. A major fraction of these cells appeared to be early plasma cell precursors, as upon activation of B cells in vitro, IgG secretion preceded up-regulation of CD38, and on tonsillar sections, IgG-containing, CD38 low cells with a plasmacytoid phenotype were found in follicles, where plasma cell differentiation starts. A unitary phenotype of migratory peripheral blood IgG-secreting cells suggests that all bone marrow plasma cell populations share a common precursor cell. These data are compatible with a multistep model for plasma cell differentiation and imply that a common CD38 low IgG-secreting precursor gives rise to a diverse plasma cell compartment.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Bone Marrow/immunology , Immunoglobulin G/metabolism , Palatine Tonsil/immunology , Plasma Cells/immunology , ADP-ribosyl Cyclase 1 , Antigens, CD19/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation , Cell Movement , Humans , In Vitro Techniques , Membrane Glycoproteins/metabolism , Palatine Tonsil/cytology , Phenotype , Plasma Cells/metabolism , Tetraspanin 29
9.
Blood ; 103(9): 3511-5, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14726396

ABSTRACT

Notch receptors expressed on hematopoietic stem cells interact with their ligands on bone marrow stromal cells and thereby control cell fate decisions and survival. We recently demonstrated that Notch signaling is involved in proliferation and survival of B cell-derived tumor cells of classic Hodgkin disease and described a novel mechanism for the oncogenic capacity of Notch. In this study we investigated whether Notch signaling is involved in the tight interactions between neoplastic plasma cells and their bone marrow microenvironment, which are essential for tumor cell growth in multiple myeloma (MM). Here we demonstrate that Notch receptors and their ligand Jagged1 are highly expressed in cultured and primary MM cells, whereas nonneoplastic counterparts show low to undetectable levels of Notch. Functional data indicate that ligand-induced Notch signaling is a growth factor for MM cells and suggest that these interactions contribute to myelomagenesis in vivo.


Subject(s)
Membrane Proteins/physiology , Multiple Myeloma/pathology , Receptors, Cell Surface/physiology , Transcription Factors , B-Lymphocytes/chemistry , Bone Marrow Cells/chemistry , Calcium-Binding Proteins , Cell Communication , Cell Division , Cell Line, Tumor , Cells, Cultured , Humans , Intercellular Signaling Peptides and Proteins , Jagged-1 Protein , Membrane Proteins/analysis , Multiple Myeloma/etiology , Proteins/analysis , Proteins/physiology , Receptor, Notch1 , Receptor, Notch2 , Receptors, Cell Surface/analysis , Receptors, Notch , Serrate-Jagged Proteins , Signal Transduction
10.
J Immunol ; 171(4): 1684-90, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12902466

ABSTRACT

Recent results suggest that plasma cell longevity is not an intrinsic capacity, but depends on yet unknown factors produced in their environment. In this study, we show that the cytokines IL-5, IL-6, TNF-alpha, and stromal cell-derived factor-1alpha as well as signaling via CD44 support the survival of isolated bone marrow plasma cells. The cytokines IL-7 and stem cell factor, crucially important for early B cell development, do not mediate plasma cell survival, indicating that plasma cells and early B cells have different survival requirements. As shown in IL-6-deficient mice, IL-6 is required for a normal induction, but not for the maintenance of plasma cell responses in vivo, indicating that the effects of individual survival factors are redundant. Optimal survival of isolated plasma cells requires stimulation by a combination of factors acting synergistically. These results strongly support the concept that plasma cell survival depends on niches in which a combination of specific signals, including IL-5, IL-6, stromal cell-derived factor-1alpha, TNF-alpha, and ligands for CD44, provides an environment required to mediate plasma cell longevity.


Subject(s)
Cytokines/physiology , Plasma Cells/cytology , Plasma Cells/physiology , Signal Transduction/physiology , Animals , Antibodies, Monoclonal/pharmacology , Antibody Formation/genetics , Apoptosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Adhesion/physiology , Cell Separation , Cell Survival/physiology , Cells, Cultured , Drug Synergism , Gene Expression Regulation/physiology , Interleukin-5/deficiency , Interleukin-5/genetics , Interleukin-6/deficiency , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/physiology , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...