Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Lupus ; 22(4): 350-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23553778

ABSTRACT

B lymphocyte stimulator (BLyS; also known as B cell activating factor (BAFF)) plays a key role in peripheral B cell tolerance. Mounting evidence indicates that B cell tolerance can be either broken or modulated by deliberately manipulating BLyS levels, and belimumab, a BLyS-neutralizing antibody, was recently approved for the treatment of systemic lupus erythematosus (SLE). Thus, intense investigation has focused on understanding how therapeutics targeting BLyS may work, and accumulating evidence suggests multiple points of action. BLyS signaling, in conjunction with B cell receptor (BCR) signaling, determines the size and quality of the mature primary B cell compartment. Moreover, BLyS family members play roles in antigen-experienced B cell selection and differentiation. Together, these findings have implications for the continued development of novel therapeutics that target BLyS.


Subject(s)
B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Differentiation , Drug Approval , Humans , Immune Tolerance/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Signal Transduction/immunology
2.
Ann Rheum Dis ; 65 Suppl 3: iii34-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17038469

ABSTRACT

The B lymphocyte stimulator (BLyS; also termed BAFF) family of ligands and receptors plays a central role in B lymphocyte development, selection, and homoeostasis. Members of this family can independently influence different B cell subsets, because the interactions between the two ligands and three receptors vary, and the receptors themselves are differentially expressed among developing, naive, and antigen experienced B cell subsets. These properties prompt careful assessment of how ablative therapies may influence the behaviour of upstream or downstream B lineage populations, as well as how the implementation and expectations of therapeutics targeting BLyS family members must be guided by knowledge of the B cell subsets contributing to pathogenesis.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , B-Cell Activating Factor/immunology , Animals , Autoimmunity , B-Cell Activation Factor Receptor/immunology , B-Lymphocytes/immunology , Homeostasis/immunology , Humans , Ligands , Lymphocyte Activation/immunology
3.
Curr Biol ; 11(24): 1986-9, 2001 Dec 11.
Article in English | MEDLINE | ID: mdl-11747827

ABSTRACT

Striking cell losses occur during late B lymphocyte maturation, reflecting BcR-mediated selection coupled with requisites for viability promoting signals. How selection and survival cues are integrated remains unclear, but a key role for B lymphocyte stimulator (BLyS(TM); trademark of Human Genome Sciences, Inc.) is suggested by its marked effects on B cell numbers and autoantibody formation as well as the B lineage-specific expression of BLyS receptors. Our analyses of the B cell-deficient A/WySnJ mouse have established Bcmd as a gene controlling follicular B cell life span, and recent reports show Bcmd encodes a novel BLyS receptor. Here we show that A/WySnJ B cells are unresponsive to BLyS, affording interrogation of how Bcmd influences B cell homeostasis. Mixed marrow chimeras indicate A/WySnJ peripheral B cells compete poorly for peripheral survival. Moreover, in vivo BrdU labeling shows that (A/WySnJ x BALB/c)F(1) B cells have an intermediate but uniform life span, indicating viability requires continuous signaling via this pathway. Together, these findings establish the BLyS/Bcmd pathway as a dominant mediator of B cell survival, suggesting competition for BLyS/Bcmd signals regulates follicular B cell numbers.


Subject(s)
B-Lymphocytes/cytology , Lymphocyte Count , Membrane Proteins , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Animals , B-Cell Activation Factor Receptor , Cell Survival/physiology , Heterozygote , Mice , Mice, Inbred BALB C , Receptors, Tumor Necrosis Factor/genetics
4.
Int Immunol ; 13(12): 1501-14, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11717191

ABSTRACT

Human X-linked agammaglobulinemia (XLA) and murine X-linked immune defect (XID) are both immunodeficiencies mediated by mutations in Bruton's tyrosine kinase (Btk), yet the developmental stage(s) affected remain controversial. To further refine the placement of the XID defect(s), we used bromodeoxyuridine labeling to determine turnover, production and transition rates of developing B cell subsets in normal, xid and xid mice expressing a human Bcl-2 transgene (xid/bcl-2). We find the xid mutation manifest at two stages of B cell development. The first is early, reducing pre-B cell production by restricting pro-B to pre-B cell transit. Surprisingly, this impairment is offset by increased survival of cells progressing from the pre- to immature B cell pool, suggesting that Btk-independent homeostatic mechanisms act to maintain this compartment. The second point of action is late, substantially reducing mature B cell production. Together, these findings reconcile apparent discrepancies in the developmental stage affected by the murine versus human lesions and suggest previously unappreciated homeostatic processes that act at the pre-B to immature B cell transition. Finally, Btk likely functions differently at these two checkpoints, since ectopic Bcl-2 expression fails to directly complement the early xid lesion, yet reverses the defect impeding final B cell maturation.


Subject(s)
B-Lymphocyte Subsets/immunology , Homeostasis/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Protein-Tyrosine Kinases/genetics , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/pathology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Cycle/genetics , Cell Cycle/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Female , Flow Cytometry , Gene Expression Regulation/immunology , Homeostasis/genetics , Humans , Immunologic Deficiency Syndromes/enzymology , Immunologic Deficiency Syndromes/pathology , Lymphocyte Count , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Spleen/cytology , Spleen/immunology , Spleen/pathology , Transgenes/immunology , X Chromosome
5.
Curr Biol ; 11(19): 1547-52, 2001 Oct 02.
Article in English | MEDLINE | ID: mdl-11591325

ABSTRACT

BLyS (also called BAFF, TALL-1, THANK, and zTNF4), a TNF superfamily member, binds two receptors, TACI and BCMA, and regulates humoral immune responses [1-7]. These two receptors also bind APRIL [7-10], another TNF superfamily member. The results from TACI(-/-) and BCMA(-/-) mice suggest the existence of additional receptor(s) for BLyS. The TACI knockout gives the paradoxical result of B cells being hyperresponsive, suggesting an inhibitory role for this receptor [11, 12], while BCMA null mice have no discernable phenotype [13]. Here we report the identification of a third BLyS receptor (BR3; BLyS receptor 3). This receptor is unique in that, in contrast to TACI and BCMA, BR3 only binds BLyS. Treatment of antigen-challenged mice with BR3-Fc inhibited antibody production, indicating an essential role for BLyS, but not APRIL, in this response. A critical role for BR3 in B cell ontogeny is underscored by our data showing that the BR3 gene had been inactivated by a discrete, approximately 4.7 kb gene insertion event that disrupted the 3' end of the BR3 gene in A/WySnJ mice, which lack peripheral B cells.


Subject(s)
B-Lymphocytes/physiology , Membrane Proteins/metabolism , Mutagenesis , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , B-Cell Activating Factor , B-Cell Activation Factor Receptor , B-Cell Maturation Antigen , B-Lymphocytes/metabolism , Base Sequence , COS Cells , Chlorocebus aethiops , DNA, Complementary , Humans , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/classification , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spleen/metabolism , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor-alpha/genetics
6.
Curr Top Microbiol Immunol ; 252: 31-8, 2000.
Article in English | MEDLINE | ID: mdl-11125490

ABSTRACT

The CBA/N strain carries xid, a murine btk missense mutation that reduces peripheral B cell numbers. Using in vivo BrdU labeling and cytofluorimetry, we have compared the magnitude, production rates, and turnover rates of each B lineage subset in the marrow and periphery of CBA/Ca and CBA/N mice. Our results show the pro-B compartment is largely unaffected by xid. In contrast, the pre-B cell pool is markedly reduced, reflecting a diminished production rate and unaltered turnover time. Despite diminished pre-B cell formation, the size of the immature B cell pool is relatively normal in CBA/N mice, due to increased proportional survival of pre-B cells. In addition, we have assessed the marrow and peripheral B cell subsets of CBA/N mice transgenic for bcl-2. These results indicate that while the bcl-2 transgene promotes lengthened survival in most B cell subsets, the pro/pre-B cell losses mediated by xid are not abrogated by bcl-2 overexpression. Taken together, these findings suggest that the initial [not readable: see text] from the pro- to pre-B cell pools, and that anomalies in subsequent compartments likely reflects the action of homeostatic mechanisms compensating for compromised pre-B cell production.


Subject(s)
B-Lymphocyte Subsets/pathology , Genes, bcl-2 , Hematopoietic Stem Cells/pathology , Immunologic Deficiency Syndromes/pathology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocyte Subsets/immunology , Bone Marrow/pathology , Cell Death , Cell Differentiation , Cell Survival , Female , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Hematopoietic Stem Cells/immunology , Homeostasis , Humans , Immunologic Deficiency Syndromes/enzymology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphocyte Activation , Lymphocyte Count , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Mutation, Missense , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Spleen/pathology
7.
Immunogenetics ; 51(11): 924-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11003386

ABSTRACT

Peripheral B-lymphocyte homeostasis is determined through incompletely defined positive and negative regulatory processes. The A/WySnJ mouse, but not the related A/J strain, has disturbed homeostasis leading to peripheral B-lymphocyte deficiency. B lymphopoeisis is normal in A/WySnJ mice, but the B cells apoptose rapidly in the periphery. This B cell-intrinsic defect segregated as a single locus, Bcmd, in (A/WySnJxA/J)F2 mice. Here we mapped a quantitative-trait locus (QTL) that contributes to the A/WySnJ B-cell deficiency by examining the F2 progeny of a cross between strains A/WySnJ and CAST/Ei. In this cross, minimally 1.9 QTLs controlling peripheral B lymphocyte deficiency segregated. The (A/WySnJxCAST/Ei)F2 mice were phenotyped for splenic B-cell percentage and the DNA from progeny with extreme phenotypes was used to map the QTL by the simple-sequence length polymorphism method. A genome scan showed linkage between peripheral B-cell deficiency and Chromosome (Chr) 15 markers. When closely spaced Chr 15 markers were analyzed, the 99% confidence interval for the QTL map position extended along the entire chromosome length. The peak lod scores >17 occurred between 30 and 45 cM. We conclude that a significant QTL segregating in (A/WySnJxCAST/Ei)F2 mice resides in this middle region of Chr 15.


Subject(s)
B-Lymphocytes/cytology , Quantitative Trait, Heritable , Animals , Cell Count , Chromosome Mapping , Female , Genetic Linkage , Male , Mice , Mice, Inbred A , Rats
8.
Int Immunol ; 12(6): 915-26, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10837419

ABSTRACT

Aging is characterized by a decline in humoral immunity and a concommitant increased incidence of anti-DNA and other autoantibodies. To define how the regulation of autoreactive B cells is altered with age, we have used BALB/c mice with an Ig heavy H chain transgene to track the fate of anti-double-stranded (ds) DNA B cells in vivo. In young adult mice, anti-dsDNA B cells are developmentally arrested and excluded from the splenic B cell follicle, whereas in most aged mice they are mature and localize within the B cell follicle. Furthermore, we have detailed global changes in lymphoid architecture that accompany aging: CD4(+) T cells are found not only in the periarteriolar lymphoid sheath, but also in the B cell follicles. Strikingly, these disruptions are similar to those that precede serum anti-dsDNA antibody expression in autoimmune MRL-lpr/lpr mice.


Subject(s)
Aging/immunology , Antibodies, Antinuclear/analysis , B-Lymphocytes/physiology , DNA/immunology , Spleen/pathology , Aging/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Nephritis/etiology , fas Receptor/analysis
9.
Proc Natl Acad Sci U S A ; 97(6): 2743-8, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10688906

ABSTRACT

Seventy percent of peripheral immature conventional (B2) B cells fail to develop into mature B cells. The nature of this cell loss has not been characterized; the process that governs which immature B cells develop into long-lived peripheral B cells could be either stochastic or selective. Here, we demonstrate that this step is in fact selective, in that the fate of an immature B cell is highly dependent on its Ig receptor specificity. A significant skewing of the B cell receptor repertoire occurs by the time cells enter the mature B cell fraction, which indicates that there is selection of only a minority of immature B cells to become mature B cells. Because only a few heavy-light chain pairs are enhanced of the diverse available repertoire, we favor the idea that selection is positive for these few heavy-light chain pairs rather than negative against nearly all others. Because most immature B cells are lost at this transition, this putative positive selection event is likely to be a major force shaping the mature B cell receptor repertoire available for adaptive immune responses.


Subject(s)
B-Lymphocytes/physiology , Receptors, Antigen, B-Cell/physiology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Death , Cell Differentiation , Cloning, Molecular , Flow Cytometry , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/immunology , Immunoglobulin M/immunology , Immunologic Memory , Mice , Mice, Transgenic , Molecular Sequence Data , Spleen/cytology , Stochastic Processes
10.
Int Immunol ; 11(11): 1753-61, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545479

ABSTRACT

During T cell development, cells that fail to meet stringent selection criteria undergo programmed cell death. Thymocyte and peripheral T cell susceptibility to apoptosis is influenced by expression of Bcl-2 family members, some of which are expressed in a developmentally patterned manner. We previously showed developmentally regulated expression of A1, an anti-apoptotic Bcl-2 family member, among B cell developmental subsets. Here we show that cells of the T lineage also express A1 in a developmentally regulated manner. Both A1 mRNA and A1 protein are readily detectable in the thymus, and while present among DN cells, A1 mRNA is up-regulated to very high levels among double-positive (DP) thymocytes. It is then down-regulated to moderate levels among single-positive (SP) thymocytes, and finally expressed at approximately 25-fold lower levels among mature SP CD4(+) and CD8(+) lymph node T cells than among DP thymocytes. Furthermore, we find that in vitro TCR ligation up-regulates A1 expression among both DP and SP thymocytes. Together, these data show that A1 expression is developmentally regulated in T lymphocytes and is responsive to TCR signaling, suggesting that A1 may play a role in maintaining the viability of DP thymocytes.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/genetics , Gene Expression Regulation , Homeodomain Proteins , Repressor Proteins , Saccharomyces cerevisiae Proteins , T-Lymphocytes/immunology , Actins/metabolism , Animals , Apoptosis/genetics , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA, Complementary/genetics , DNA-Binding Proteins/biosynthesis , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Replication Protein C , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , T-Lymphocytes/cytology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
11.
J Immunol ; 160(1): 107-11, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9551962

ABSTRACT

Only 5% of the 15 million B cells formed daily reach the long-lived peripheral B cell pool, presumably reflecting both negative and positive selection. These selective events occur primarily during late stages of differentiation in the marrow and periphery, when newly formed B cells bear surface IgM (sIgM), but differ from mature B cells in their expression of heat-stable Ag (CD24), B220 (CD45), and sIgD. Because genes of the Bcl-2 family influence longevity, we compared the expression of Bcl-2, Bax, and A1 among immature vs mature peripheral B cells using semiquantitative reverse-transcriptase PCR. While the levels of both Bcl-2 and Bax mRNA remain constant in these two populations, A1 expression is strikingly up-regulated among mature B cells. In addition, A1 expression is low among pro- and pre-B cells, as well as in immature (sIgM+) marrow B cells. Together, these data indicate that A1 mRNA expression is low at all stages of B cell development before final maturation in the periphery and, unlike other Bcl-2 family members whose expression changes little after marrow egress, A1 is up-regulated 10-fold as cells are recruited into the long-lived peripheral B cell pool.


Subject(s)
B-Lymphocyte Subsets/cytology , DNA-Binding Proteins/metabolism , Homeodomain Proteins , Repressor Proteins , Saccharomyces cerevisiae Proteins , Animals , B-Lymphocyte Subsets/immunology , Biomarkers , Bone Marrow Cells/metabolism , Cell Survival , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Inbred BALB C , Minor Histocompatibility Antigens , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Replication Protein C , bcl-2-Associated X Protein
12.
J Immunol ; 160(8): 3743-7, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9558076

ABSTRACT

Peripheral B cells are divided into two subpopulations, B-1 and B-2, the relationship of which remains obscure. We recently showed that the Bcmd mutation in A/WySnJ mice reduces average B cell life span, yielding 90% fewer peripheral B cells. Despite this defect, A/WySnJ mice have an elevated proportion of peritoneal CD5+ B cells, suggesting that Bcmd may be the first B-cell-intrinsic gene to differentially affect the B-1 and B-2 subpopulations. To test this hypothesis in detail, we have used in vivo BrdU labeling and four-color cytofluorometry to examine the numbers and turnover rates of sIgM+CD23-CD43+ (B-1) and sIgM+CD23+CD43- (B-2) splenocytes in A/WySnJ and A/J mice. The results show the expected 90% reduction of splenic B-2 cells among A/WySnJ mice, but a normal splenic B-1 cell pool. Increased B-1 cell renewal cannot explain this undiminished pool, because BrdU labeling kinetics reveals an identical splenic B-1 subset turnover rate of approximately 4%/day in both A/WySnJ and A/J strains. Thus, B-1 cells are Bcmd-independent but B-2 cells are Bcmd-dependent, suggesting Bcmd functions in a positive signaling pathway that imparts longevity to quiescent B cells, but that is not required for cycling B cells. Moreover these results show that the requisites for maturation and longevity differ between the B-1 and B-2 subsets.


Subject(s)
Antigens, CD , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Cellular Senescence/genetics , Mutation , Animals , B-Lymphocyte Subsets/metabolism , Bromodeoxyuridine/metabolism , Cell Differentiation , Cellular Senescence/immunology , Female , Flow Cytometry , Immunoglobulin M/metabolism , Kinetics , Leukosialin , Lymphocyte Count , Male , Mice , Mice, Inbred A , Mice, Mutant Strains , Receptors, Antigen, B-Cell/metabolism , Receptors, IgE/metabolism , Sialoglycoproteins/metabolism , Signal Transduction , Spleen/cytology , Spleen/immunology
13.
Immunol Res ; 17(1-2): 3-11, 1998.
Article in English | MEDLINE | ID: mdl-9479562

ABSTRACT

More than 95% of newly formed B cells die in the short interval spanning sIgM acquisition in the bone marrow and entry into the long-lived pool, suggesting that selective events dictating B cell longevity occur at this stage. These likely include both ligand-induced deletion as well as discrete events that mediate recruitment to the long-lived recirculating pool. We are probing these events through the examination of normal B cell differentiation during this critical period: the characterization of a natural mutation that blocks late maturation, an irradiation/autoreconstitution model of marrow-derived B cell differentiation, and the identification of life span regulatory genes whose expression changes within this window.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Bone Marrow/immunology , Animals , B-Lymphocytes/cytology , Bone Marrow Cells/cytology , Cell Differentiation/immunology , Humans , Immunoglobulin M/analysis , Mutation
14.
J Immunol ; 157(2): 598-606, 1996 Jul 15.
Article in English | MEDLINE | ID: mdl-8752907

ABSTRACT

The A/WySnJ mouse provides a genetic model for studying new B cell selection into the stable peripheral B cell pool. Unlike the related A/J strain, the A/WySnJ has a single, autosomal codominant gene defect, Bcmd, resulting in a profound peripheral B cell deficiency. Here, continuous in vivo bromodeoxyuridine labeling and immunofluorescence analysis showed normal bone marrow B cell genesis but excessive B cell loss from the marrow and each peripheral pool in A/WySnJ. The A/WySnJ immature B220low/HSAhigh splenic B cell pool was 79% smaller, had a 69% slower renewal rate, and its cells had a 29% shorter average half-life than A/J. The A/WySnJ mature B220high/HSAlow splenic B cell pool was 92% smaller, had an 83% slower renewal rate, and its cells had a 56% shorter average half-life. In reciprocal chimeras, the A/WySnJ marrow failed to repopulate the peripheral B cell pool in A/J mice, whereas the A/J marrow fully reconstituted the A/WySnJ mice. Histochemistry revealed disordered splenic architecture in A/WySnJ, with few primary lymphoid follicles and a second abnormal phenotype, mastocytosis. There was no common genetic basis for B cell deficiency and mastocytosis in the F2 progeny of an (A/WySnJ x CAST/Ei)F1 intercross. We conclude that Bcmd is expressed in bone marrow cells, most likely B cells, where it hinders short-lived B cell maturation to a long-lived phenotype with the potential to form memory B cells.


Subject(s)
B-Lymphocytes/immunology , Immunologic Deficiency Syndromes/genetics , Lymphocyte Activation/genetics , Animals , B-Lymphocytes/pathology , Bone Marrow/immunology , Bone Marrow Cells , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Gene Deletion , Immunologic Deficiency Syndromes/pathology , Male , Mastocytosis/genetics , Mastocytosis/immunology , Mastocytosis/pathology , Mice , Mice, Inbred A , Mice, Mutant Strains , Radiation Chimera/immunology , Spleen/immunology , Spleen/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
15.
J Immunol ; 151(9): 4431-44, 1993 Nov 01.
Article in English | MEDLINE | ID: mdl-8409411

ABSTRACT

HSAhi B cells comprise 5 to 10% of adult mouse splenic B cells and are phenotypically and functionally immature. To assess their origin and relationship to mature, heat-stable Ag (HSA)lo B cells, we determined HSA and surface IgD phenotype among splenic B cells throughout development, as well as during reconstitution of lethally irradiated adults given adult B-depleted bone marrow. In each case, HSAhi splenic B cells predominate during the earliest stages of B cell genesis. Furthermore, 5-bromo-2'-deoxyuridine labeling experiments indicate rapid turnover within both the marrow and peripheral HSAhi pools, and adoptive transfer studies show that peripheral HSAhi splenic B cells differentiate to HSAlo within 4 days. Finally, splenic HSAhi B cells reconstitute both primary and memory humoral responses. Together, these data indicate that splenic B cells in the HSAhi subset are an intermediate maturational stage in the adult periphery.


Subject(s)
Antigens, Differentiation/analysis , B-Lymphocytes/physiology , Bone Marrow Cells , Hematopoietic Stem Cells/physiology , Spleen/cytology , Animals , B-Lymphocytes/immunology , Hematopoietic Stem Cells/immunology , Hot Temperature , Immunoglobulin D/analysis , Immunotherapy, Adoptive , Mice , Mice, Inbred BALB C , Receptors, Antigen, B-Cell/analysis , Receptors, IgE/analysis
16.
J Immunol ; 149(8): 2533-40, 1992 Oct 15.
Article in English | MEDLINE | ID: mdl-1383316

ABSTRACT

Whether recently generated peripheral B cells in adults are functionally equivalent to immature B cells in the neonatal spleen is unknown. We have identified a splenic B cell subpopulation in adults whose phenotypic and in vitro characteristics closely resemble those of neonatal B cells. These cells are defined by the cell surface phenotype heat-stable Aghi (HSAhi), and make up 10 to 15% of the adult splenic B cell pool. HSAhi B cells in adults bear the immature phenotype B220lo sIgMhi, and are 50% sIgD+. Furthermore, after sublethal irradiation, the initial wave of newly generated splenic B cells in self-reconstituting adults express a similar phenotype. In keeping with previous data on neonatal B cells, HSAhi cells from either normal or self-reconstituting mice are refractory to stimulation with anti-IgM antibodies, yet proliferate upon LPS stimulation, and generate primary antibody responses if given appropriate T cell help. In contrast to neonatal cells, HSAhi adult B cells are refractory to stimulation with PMA plus ionomycin. Together, these data suggest that peripheral HSAhi B cells in adults correspond to recently generated B cells, whose signaling characteristics are distinct from previously described B cell subsets.


Subject(s)
Antigens, Surface/analysis , B-Lymphocytes/physiology , Animals , Animals, Newborn/immunology , Antibody Formation , Cells, Cultured , Hot Temperature , Immunoglobulin M/physiology , Ionomycin/pharmacology , Leukocyte Common Antigens , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/physiology , Tetradecanoylphorbol Acetate/pharmacology
18.
J Exp Med ; 170(6): 1811-23, 1989 Dec 01.
Article in English | MEDLINE | ID: mdl-2584925

ABSTRACT

Gene conversion has been suggested as the basis for many VH allelic differences, particularly in the murine VHS107 family. Whether conversion among IgVH genes is likely to have occurred in outbred populations has not been directly addressed. The CLA-2/Cn and WSA strains, which were recently and independently derived from a feral population exhibiting low responsiveness to PC, provide the opportunity to approach this question. In previous studies, the heavy chain cDNA sequence of a PC-specific hybridoma derived from CLA-2/Cn suggested gene conversion events within the VHS107 family. Accordingly, we have examined the germline VHS107 genes of CLA-2/Cn and WSA. The results indicate that: (a) The CLA-2 and WSA strains bear an identical but novel VHS107 family haplotype, which lacks a V3 element and contains a V1, a V13, and two V11 genes; (b) low PC responsiveness in these populations is unlikely due to an inability to express the V1 member of the VHS107 gene family; and (c) when compared with the other known VHS107 haplotypes, the proportion of differences consistent with gene conversion greatly exceeds that expected by random base substitution. Thus, gene conversion events appear to have occurred with considerable frequency in the evolution of the murine VHS107 family, especially among the V3, V13, and V11 members.


Subject(s)
Gene Conversion , Genes, Immunoglobulin , Haplotypes , Alleles , Animals , Base Sequence , Mice , Mice, Inbred Strains , Molecular Sequence Data , Species Specificity
19.
J Immunol ; 143(12): 4019-24, 1989 Dec 15.
Article in English | MEDLINE | ID: mdl-2480384

ABSTRACT

IL-6 (formerly PCTGF, HP-1, BSF-2, HGF, IFN-beta 2, 26 kDa) is a recently defined lymphokine demonstrating activity on multiple cell types, including hepatocytes, thymocytes, T cells, plasmacytomas, and B cells. The biologic effects of IL-6 on lymphocytes, particularly B cells, suggest this factor may be involved in the regulation of normal immune responses. Accordingly, we have investigated the role of IL-6 in Ag-specific responses of B cells from both naive and Ag-primed mice. When Ag-primed splenic T cells were used as a source of help, naive (primary) B cell responses specific for the hemagglutinin molecule of the influenza A virus (PR8) were fully inhibited by the addition of an anti-IL-6 antiserum, and are thus IL-6 dependent. In contrast, secondary B cell responses were essentially IL-6 independent, being unaffected by this antiserum even at concentrations 10-fold higher than required to completely inhibit primary responses. This differential IL-6 requirement was further investigated by using a panel of hemagglutinin molecule-specific Th clones. Consistent with the above findings, a Th1 clone secreting biologically active IL-6 enables antibody secretion by both primary and secondary B cells, whereas Th1 clones that do not produce IL-6 support secondary responses, but fail to help primary B cell responses unless exogenous IL-6 is added. These results provide the first instance of differential lymphokine requirements among primary vs secondary B cell responses, and suggest T cell-derived IL-6 plays a critical role during the regulation of humoral immune responses. Moreover, functionally distinct Th1 clones were identified that differed in IL-6 secretion and their corresponding ability to induce Ig secretion by primary and secondary B cells.


Subject(s)
Antibodies, Viral/biosynthesis , B-Lymphocytes/metabolism , Epitopes/immunology , Interleukin-6/physiology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , Clone Cells/analysis , Immunologic Memory , Influenza A virus/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocytes, Helper-Inducer/analysis
20.
Semin Immunol ; 1(1): 79-86, 1989 Sep.
Article in English | MEDLINE | ID: mdl-15630961

ABSTRACT

The physiologic distinctions between secondary and primary lymphoid populations remain largely conceptual. Altered activation, differentiation, and compartmentalization properties likely underlie these differences, but little precise knowledge of how these parameters are affected by antigen priming exists. Because lymphoid populations are dynamic entities and priming is a temporal process, lineage and life span analyses of definable subpopulations are required for the design and interpretation of experiments to probe functional distinctions between primed versus unprimed B-cell populations. Subsequent studies, which address differences in activation requirements and collaborative potential of primed versus unprimed B-cells are further required to evaluate the cellular basis of immunologic memory. Recent advances in the ability to dissect lymphoid differentiation subsets and lineages, coupled with a rapidly expanding knowledge of molecules which mediate B-lymphocyte growth and differentiation, render these possibilities amenable to experimental analysis.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Animals , Antigens, Surface , B-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Proliferation , T-Lymphocytes, Helper-Inducer/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...