scholarly journals CNS antigen-specific control of early B-lineage development in the meninges

2021 ◽  
Author(s):  
Yan Wang ◽  
Dianyu Chen ◽  
Di Xu ◽  
Chao Huang ◽  
Danyang He ◽  
...  

The random V(D)J recombination of immunoglobulins (Ig) loci often creates autoreactive B cell progenitors expressing self-recognized B-cell receptors (BCRs)1, which are eliminated or inactivated through an autoantigen-dependent central tolerance checkpoint to prevent autoimmune reactions2,3, a process thought to be restricted to the bone marrow (BM) in the adult mammals4. Here we report that early developing B cells are also present in the meninges of mice at all ages. Single cell RNA-sequencing (scRNA-seq) analysis revealed a consecutive trajectory of meningeal developing B cells in mice and non-human primates (NHPs). Parabiosis together with lineage tracing of hematopoietic stem cells (HSCs) showed that meningeal developing B cells are continuously replenished from the HSC-derived progenitors via a circulation-independent route. Importantly, autoreactive immature B cells which recognize myelin oligodendrocyte glycoprotein (MOG)5, a central nervous system (CNS)-specific antigen, are eliminated from the meninges but not BM. Furthermore, genetic deletion of MOG restored the self-reactive B cells in the meninges. Thus, we propose that meninges function as a unique reservoir for B cell development, allowing in situ negative selection of CNS-antigen-autoreactive B cells to ensure a local non-self-reactive immune repertoire.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 95-95
Author(s):  
Sonja E. Nodland ◽  
Anna A Bajer ◽  
Tucker W. LeBien

Abstract Abstract 95 The identity and developmental potential of human lymphoid progenitors (LP) restricted to differentiate into T, B and/or NK cells at the exclusion of myeloid/erythroid lineages is not completely understood. Although LP from cord blood and marrow been described, there is no consensus as to the phenotype of LP committed to the B-lineage from either tissue source, or the identity of the cytokine signals that regulate lineage commitment of these progenitors. Resolution of this question has implications for identifying potential leukemic stem cells in acute lymphoblastic leukemia, as well as for lymphocyte reconstitution in the setting of cord blood and marrow transplantation. One of the challenges in conducting a comprehensive cellular and molecular characterization of LP is their low frequency in normal marrow and cord blood. The xenogeneic human hematopoietic stem cell/murine stromal cell (MS-5) model of lymphohematopoiesis has been shown to support the development of all human lymphoid lineages from human CD34+ hematopoietic progenitor cells (HPC). Therefore, the goal of the current study was to use the MS-5 model as a cellular resource to identify and characterize candidate LP subsets, and to evaluate the expression and function of the IL-7 receptor in B-lineage cell development. We initially determined whether expression of CD127 (the IL-7 receptor alpha chain) and CD34 were sufficient to define CD19- LP with enhanced B lymphopoietic potential. Xenogeneic cultures were initiated by plating cord blood CD34+ HPC onto MS-5, and CD19- lymphohematopoietic cells were characterized by polychromatic flow cytometry. Gating on CD19-/CD14-/CD15- cells revealed four populations: CD19-/CD34+/CD127-, CD19-/CD34+/CD127+, CD19-/CD34-/CD127+, and CD19-/CD34-/CD127-. CD19-/CD34+/CD127- cells were the predominant population during the first week of culture but sharply declined thereafter. A low frequency CD19-/CD34+/CD127+ population was first detected at 5-7 days and largely disappeared by 2 wks. The CD19-/CD34-/CD127+ population was initially detected at 1 wk, and underwent a bi-phasic increase and decrease over the following 3 wks. Evaluation of sorted fractions by quantitative PCR showed that the onset of expression of RAG1, RAG2, TDT, CD79A, VPREB, IGλ LIKE, and EBF-1 were coincident with onset of CD127 expression. Differentiation of CD34+/CD127+ to CD34-/CD127+ LP was accompanied by an ∼ 50-fold increase in expression of PAX5 and CD19, suggesting that the latter population was the immediate precursor of CD19+ B-lineage cells. When the three populations were FACS-purified on day 14 and re-plated on MS-5, a progression from CD34+/CD127- > CD34+/CD127+ > CD34-/CD127+ was suggested by the kinetics of CD19+ cell development. We have previously shown that neutralization of MS-5 produced murine IL-7 reduces the development of CD19+ cells from cord blood CD34+ HPC by ∼ 80%. When fetal liver, pediatric marrow or adult marrow CD34+ cells were used to initiate xenogeneic cultures, we observed a similar dependency on IL-7 for CD19+ cell development. However, neutralization of murine IL-7 had no effect on the development of CD34-/CD127+/CD19- LP. Surprisingly, independent of the source of CD34+ HPC used to initiate xenogeneic cultures, B-lineage cells expressing cell surface IgM developed in the absence of the canonical CD34+/CD19+ pro-B cell population present in human marrow. The absence of CD19+/CD34+ pro-B cells was not due to MS-5 xenogeneic culture conditions being non-permissive, since CD19+/CD34+ pro-B cells FACS-purified from fresh pediatric marrow and plated on MS-5 survived and differentiated over 3-4 wks of culture into surface IgM+/IgD+ naive B cells. Additionally, CD34 was stably expressed on CD19- cells in xenogeneic cultures for ∼ 2 wks. We conclude that expression of CD127 on CD19- LP and CD19+ B-lineage cells may define an early continuum in human B cell development, which at least partially encompasses a pathway that either bypasses or is distinct from development of canonical CD34+/CD19+ pro-B cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (5) ◽  
pp. 1281-1290 ◽  
Author(s):  
Phyllis W. Yu ◽  
Ruby S. Tabuchi ◽  
Roberta M. Kato ◽  
Alexander Astrakhan ◽  
Stephanie Humblet-Baron ◽  
...  

Abstract X-linked agammaglobulinemia (XLA) is a human immunodeficiency caused by mutations in Bruton tyrosine kinase (Btk) and characterized by an arrest in early B-cell development, near absence of serum immunoglobulin, and recurrent bacterial infections. Using Btk- and Tec-deficient mice (BtkTec–/–) as a model for XLA, we determined if Btk gene therapy could correct this disorder. Bone marrow (BM) from 5-fluorouracil (5FU)–treated BtkTec–/– mice was transduced with a retroviral vector expressing human Btk and transplanted into BtkTec–/– recipients. Mice engrafted with transduced hematopoietic cells exhibited rescue of both primary and peripheral B-lineage development, recovery of peritoneal B1 B cells, and correction of serum immunoglobulin M (IgM) and IgG3 levels. Gene transfer also restored T-independent type II immune responses, and B-cell antigen receptor (BCR) proliferative responses. B-cell progenitors derived from Btk-transduced stem cells exhibited higher levels of Btk expression than non-B cells; and marking studies demonstrated a selective advantage for Btk-transduced B-lineage cells. BM derived from primary recipients also rescued Btk-dependent function in secondary hosts that had received a transplant. Together, these data demonstrate that gene transfer into hematopoietic stem cells can reconstitute Btk-dependent B-cell development and function in vivo, and strongly support the feasibility of pursuing Btk gene transfer for XLA.


Blood ◽  
1993 ◽  
Vol 81 (4) ◽  
pp. 1025-1031 ◽  
Author(s):  
D Campana ◽  
E Coustan-Smith ◽  
A Manabe ◽  
M Buschle ◽  
SC Raimondi ◽  
...  

Overexpression of bcl-2 delays the onset of apoptosis in lymphohematopoietic cells. We measured levels of bcl-2 protein in normal and leukemic human B-cell progenitors with a specific monoclonal antibody and flow cytometry. Normal immature B cells had low levels of bcl-2 protein; the intensity of fluorescence, expressed as molecules of soluble fluorochrome per cell, within CD10+ cells was 3,460 +/- 1,050 (mean +/- SD; 5 samples). In 16 cases of B-lineage acute lymphoblastic leukemia (ALL), cells had levels of bcl-2 that were strikingly higher than those of their normal counterparts (33,560 +/- 14,570; P < .001 by t-test analysis). We next investigated whether the intensity of bcl-2 expression correlated with the resistance of immature B cells to in vitro culture. In 12 cases of B-lineage ALL, the cells recovered after 7 days of culture on allogeneic bone marrow stromal layers were 69% to 178% (median, 95.5%) of those originally seeded. Prolonged survival of leukemic cells in vitro was observed even in the absence of stromal layers in 6 of these 12 cases; the intensity of bcl-2 protein expression in these cases was 45,000 +/- 13,270, compared with 21,500 +/- 7,260 in the 6 cases in which greater than 99.5% of cells rapidly died by apoptosis under the same culture conditions (P = .003). Five immature B-cell lines, continuously growing in the absence of stroma, had the highest bcl-2 expression (79,400 +/- 20,330). By contrast, most normal CD19+, sIg-immature B cells died despite the presence of bone marrow stromal layers; 9.7% to 28.2% were recovered after 7 days of culture in three experiments. We conclude that abnormal bcl-2 gene expression influences the survival ability of B-cell progenitors. This may contribute to leukemogenesis and explain the aptitude of leukemic lymphoblasts to expand outside the bone marrow microenvironment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3314-3314
Author(s):  
Hongsheng Wang ◽  
Chang Hoon Lee ◽  
Chen-Feng Qi ◽  
Nishant Kerrikatte ◽  
Prafullakumar Tailor ◽  
...  

Abstract The generation of lymphoid and myeloid lineage cells from hematopoietic stem cells is controlled by multiple transcription factors regulating distinct developmental and functional aspects. Interferon consensus sequence binding protein (ICSBP)/interferon regulatory factor 8 (IRF8) is a transcription factor known to regulate the differentiation of macrophages, granulocytes, and dendritic cells. Our recent findings that IRF8 transcripts and protein are highly expressed in germinal center (GC) B cells suggest that IFR8 may also play a role in normal B cell development. In IRF8 deficient mice, the number of early B lineage cells (pre-pro-B) was reduced by 5-fold, indicating a defect in early B lineage commitment. While the numbers of late pre-B and immature B cells were moderately reduced (~2-fold), recirculating mature B cells were almost undetectable in the bone marrow of mutant mice. This deficiency in early stage B cells is correlated with increased expression of PU.1, a crucial transcription factor for myeloid and lymphoid lineage specification. Interestingly, the number of splenic transitional 1 (T1) cells was slightly increased but the numbers of T2 and follicular (FO) B-2 cells were moderately decreased in mutant mice. This indicates that positive selection of T2 cells into the mature B-2 pool is regulated by IRF8. The marginal zone (MZ) B cell and peritoneal CD11b+ B-1b cell compartments were also slightly expanded in IRF8 knockout mice. Overall, these results provide compelling evidence that IRF8 regulates B cell differentiation and function at multiple stages.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3666-3666
Author(s):  
Kathrin Händschke ◽  
Stefanie Weber ◽  
Mandy Necke ◽  
Anita Hollenbeck ◽  
Bertram Opalka ◽  
...  

Abstract The development of hematopoietic cells occurs in highly specialized microenvironments within bone marrow, thymus and spleen. Hematopoietic stem and progenitor cells are positioned at the lowest end of a bone marrow oxygen gradient, which implies a role for the hypoxic-response pathway in regulating hematopoiesis. In this pathway the von Hippel-Lindau protein (pVhl) is the central negative regulator and continuously mediates the proteasomal destruction of the hypoxia-inducible factor-1α (Hif-1α). Under hypoxic conditions Hif-1α destruction is inhibited and results in the expression of hypoxia-inducible genes. In order to study the role of pVhl in hematopoiesis we crossed Vhl conditional mice with vav-iCre mice to induce a constitutive hematopoiesis-specific Vhl deletion (VhlKOvav). As expected, we observed a dramatic expansion of spleen erythropoiesis, however bone marrow lin-Sca-1+c-kit+CD48-CD150+ hematopoietic stem cell numbers of VhlKOvav mice did not significantly differ from control mice. The most striking observation in VhlKOvav mice was that B-cell numbers in bone marrow and spleen were decreased by 53% and 78%, respectively. In order to exclude that the loss of B-cells in VhlKOvav mice was due to a B-cell extrinsic effect we crossed Vhl conditional mice to the B-cell specific deleter lines CD19-Cre and Mb1-Cre (VhlKOCD19 and VhlKOMb1 mice). Flow cytometric analysis also revealed decreased peripheral B-cell numbers in VhlKOCD19 mice and an even more pronounced B-cell loss in VhlKOMb1 mice (B-cells spleen, mean±SEM; control (n=9), 77±2.4x106; VhlKOMb1 (n=12), 1.3±0.2x106; p<0.001). This demonstrated that pVhl is cell-autonomously required for the normal development of the B-cell compartment. To more accurately define the Vhl-null B-cell developmental defect we analyzed bone marrow B-cell subsets of VhlKOMb1 and control mice. Early B-cell progenitor numbers defined by the surface markers B220, CD43, CD24, BP-1 (Hardy’s classification, fractions A-D) of VhlKOMb1 mice were not altered. In contrast, we observed a significant 41% reduction of the immature B220+CD43-IgM+IgD- and a 46% reduction of the mature B220+CD43-IgM+IgD+ bone marrow VhlKOMb1 B-cell numbers compared to controls. In peripheral blood VhlKOMb1 immature and mature B-cell numbers were even more decreased (by 81% and 86%, respectively). We hypothesized that increased CXCR4 expression, which is negatively regulated by pVhl, could be responsible for the decreased bone marrow egress of immature bone marrow B-cells. Indeed, we observed a more than twofold increase of CXCR4 expression of VhlKOMb1 compared to control bone marrow immature B-cells (MFI, mean±SEM; control (n=3), 72±15; VhlKOMb1 (n=3), 162±23; p<0.05). Strikingly, VhlKOMb1 spleens were almost devoid of follicular B220+CD21/35intmCD23+IgD+IgM+ and marginal zone B220+CD21/35highCD23-IgMhigh B-cells and accordingly follicular structures could not be observed in histological sections. In VhlKOMb1 lymph nodes the follicular B-cell numbers were also dramatically decreased. Next, we flow sorted residual splenic VhlKOMb1 B-cells and were able to confirm deletion of the Vhl-gene by PCR. Target genes of the hypoxic-response pathway such as Pgk1, Vegf and Bnip3 were 10- to more than 100-fold higher expressed in sorted VhlKOMb1 compared to control B-cells. As a possible reason for the low VhlKOMb1 peripheral B-cell numbers we identified a more than twofold reduction in CD62L expression by immature blood B-cells (MFI, mean±SEM; control (n=3), 3127±250; VhlKOMb1 (n=3), 1528±66; p<0.05) which presumably impaired their homing ability to peripheral lymphoid organs. Additionally, we detected an increased B-cell apoptosis rate of VhlKOMb1 B-cells in the spleen. Finally, we were able to show that decreased follicular splenic B-cell numbers of VhlKOCD19 mice could be completely rescued by additionally breeding Hif-1α conditional alleles into the system (Hif-1αVhlKOCD19mice). In summary, we identified pVhl as a key regulator of peripheral B-cell maturation. We show that pVhl-mediated negative regulation of the hypoxic-response pathway is required for normal peripheral B-cell differentiation. Our data suggest that B-cell pVHL loss-of-function leads to decreased bone marrow egress and decreased lymphoid organ homing of immature B-cells mediated by the dysregulation of CXCR4 and CD62L. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3699-3699
Author(s):  
Zev J. Greenberg ◽  
Darlene A. Monlish ◽  
Rachel L. Bartnett ◽  
Laura G. Schuettpelz

Abstract Acute lymphoblastic leukemia (ALL) is the most frequent pediatric malignancy, most commonly originating from the transformation of progenitor cells of the B cell lineage (B cell precursor-ALL; BCP-ALL). Treatment of patients with high-risk or relapsed disease is difficult and prognosis remains poor in pediatric patients, with an even worse survival rate for adult BCP-ALL. Previous studies have shown an association of enhanced CD53 expression with many B cell malignancies, suggesting upregulation of CD53 may be implicated in carcinogenesis or maintenance of malignant cells. CD53 is a member of the tetraspanin family of transmembrane proteins, classically involved in cell adhesion, proliferation, and survival, and expressed exclusively on hematopoietic cells. While several studies have implicated a role for CD53 in regulating mature B cell proliferation, its role in early B cell development is not yet known. To elucidate the contribution of CD53 to normal and malignant B cell development, we have generated a CD53 knockout mouse. In our CD53-/- mouse, we observe no differences in total white blood cell counts, yet the fraction of peripheral blood B cells is significantly reduced by 31% compared to wild-type (WT) controls (28.3% vs. 19.5%; p<0.005). During homeostatic B lymphopoiesis, CD53 increases through development, beginning at the pre-pro-B cell stage and reaching highest expression on mature B cells. Further investigation into the loss of B cells revealed that immature pre-B cells in the bone marrow and mature B cells in the spleen and lymph nodes are significantly diminished upon loss of CD53, resulting from increased apoptosis in CD53-/- mice. B cell differentiation of CD53-/- hematopoietic stem cells (HSCs) in vitro corroborates the dependence on CD53 for normal differentiation, as CD53-/- cultures have 26% fewer B cells than controls (p=0.033). Investigation into the signaling differences between WT and CD53-/- B cell progenitors by mass cytometry (CyTOF) suggests that decreased PI3K/Akt and MAPK signaling could be driving this loss. With the observed loss of both B cell progenitors and mature B cells in CD53-deficient mice, CD53-/- mice were recently crossed to Eμ-Myc transgenic mice, a model of B-lineage leukemia/lymphoma, to generate WT, CD53-/-, Eμ-Myc+;CD53+/+, and Eμ-Myc+;CD53-/- groups to assess whether loss of CD53 alters the pathology or survival of these mice. As observed in human patients, moribund Eμ-Myc+ mice significantly upregulate CD53 on malignant cells, suggesting a potential role for CD53 during pathogenesis. Ongoing experiments are aimed at elucidating the mechanism by which CD53 promotes homeostatic B cell development and determining the potential of CD53 as a therapeutic target for B lineage malignancies. Disclosures No relevant conflicts of interest to declare.


1997 ◽  
Vol 186 (12) ◽  
pp. 2013-2021 ◽  
Author(s):  
Martin Turner ◽  
Adam Gulbranson-Judge ◽  
Marian E. Quinn ◽  
Alice E. Walters ◽  
Ian C.M. MacLennan ◽  
...  

The tyrosine kinase Syk has been implicated as a key signal transducer from the B cell antigen receptor (BCR). We show here that mutation of the Syk gene completely blocks the maturation of immature B cells into recirculating cells and stops their entry into B cell follicles. Furthermore, using radiation chimeras we demonstrate that this developmental block is due to the absence of Syk in the B cells themselves. Syk-deficient B cells are shown to have the life span of normal immature B cells. If this is extended by over-expression of Bcl-2, they accumulate in the T zone and red pulp of the spleen in increased numbers, but still fail to mature to become recirculating follicular B cells. Despite this defect in maturation, Syk-deficient B cells were seen to give rise to switched as well as nonswitched splenic plasma cells. Normally only a proportion of immature B cells is recruited into the recirculating pool. Our results suggest that Syk transduces a BCR signal that is absolutely required for the positive selection of immature B cells into the recirculating B cell pool.


Blood ◽  
2020 ◽  
Vol 135 (17) ◽  
pp. 1452-1457 ◽  
Author(s):  
Arianna Troilo ◽  
Claudia Wehr ◽  
Iga Janowska ◽  
Nils Venhoff ◽  
Jens Thiel ◽  
...  

Abstract Common variable immunodeficiency (CVID) is a disease characterized by increased susceptibility to infections, hypogammaglobulinemia, and immune dysregulation. Although CVID is thought to be a disorder of the peripheral B-cell compartment, in 25% of patients, early B-cell development in the bone marrow is impaired. Because poor B-cell reconstitution after hematopoietic stem cell transplantation has been observed, we hypothesized that in some patients the bone marrow environment is not permissive to B-cell development. Studying the differentiation dynamics of bone marrow-derived CD34+ cells into immature B cells in vitro allowed us to distinguish patients with B-cell intrinsic defects and patients with a nonpermissive bone marrow environment. In the former, immature B cells did not develop and in the latter CD34+ cells differentiated into immature cells in vitro, but less efficiently in vivo. In a further group of patients, the uncommitted precursors were unable to support the constant development of B cells in vitro, indicating a possible low frequency or exhaustion of the precursor population. Hematopoietic stem cell transplantation would result in normal B-cell repopulation in case of intrinsic B-cell defect, but in defective B-cell repopulation in a nonpermissive environment. Our study points to the importance of the bone marrow niche in the pathogenesis of CVID.


Blood ◽  
1993 ◽  
Vol 81 (4) ◽  
pp. 1025-1031 ◽  
Author(s):  
D Campana ◽  
E Coustan-Smith ◽  
A Manabe ◽  
M Buschle ◽  
SC Raimondi ◽  
...  

Abstract Overexpression of bcl-2 delays the onset of apoptosis in lymphohematopoietic cells. We measured levels of bcl-2 protein in normal and leukemic human B-cell progenitors with a specific monoclonal antibody and flow cytometry. Normal immature B cells had low levels of bcl-2 protein; the intensity of fluorescence, expressed as molecules of soluble fluorochrome per cell, within CD10+ cells was 3,460 +/- 1,050 (mean +/- SD; 5 samples). In 16 cases of B-lineage acute lymphoblastic leukemia (ALL), cells had levels of bcl-2 that were strikingly higher than those of their normal counterparts (33,560 +/- 14,570; P < .001 by t-test analysis). We next investigated whether the intensity of bcl-2 expression correlated with the resistance of immature B cells to in vitro culture. In 12 cases of B-lineage ALL, the cells recovered after 7 days of culture on allogeneic bone marrow stromal layers were 69% to 178% (median, 95.5%) of those originally seeded. Prolonged survival of leukemic cells in vitro was observed even in the absence of stromal layers in 6 of these 12 cases; the intensity of bcl-2 protein expression in these cases was 45,000 +/- 13,270, compared with 21,500 +/- 7,260 in the 6 cases in which greater than 99.5% of cells rapidly died by apoptosis under the same culture conditions (P = .003). Five immature B-cell lines, continuously growing in the absence of stroma, had the highest bcl-2 expression (79,400 +/- 20,330). By contrast, most normal CD19+, sIg-immature B cells died despite the presence of bone marrow stromal layers; 9.7% to 28.2% were recovered after 7 days of culture in three experiments. We conclude that abnormal bcl-2 gene expression influences the survival ability of B-cell progenitors. This may contribute to leukemogenesis and explain the aptitude of leukemic lymphoblasts to expand outside the bone marrow microenvironment.


1997 ◽  
Vol 186 (8) ◽  
pp. 1299-1306 ◽  
Author(s):  
James R. Drake ◽  
Paul Webster ◽  
John C. Cambier ◽  
Ira Mellman

B cell receptor (BCR)-mediated antigen processing is a mechanism that allows class II–restricted presentation of specific antigen by B cells at relatively low antigen concentrations. Although BCR-mediated antigen processing and class II peptide loading may occur within one or more endocytic compartments, the functions of these compartments and their relationships to endosomes and lysosomes remain uncertain. In murine B cells, at least one population of class II– containing endocytic vesicles (i.e., CIIV) has been identified and demonstrated to be distinct both physically and functionally from endosomes and lysosomes. We now demonstrate the delivery of BCR-internalized antigen to CIIV within the time frame during which BCR-mediated antigen processing and formation of peptide–class II complexes occurs. Only a fraction of the BCR-internalized antigen was delivered to CIIV, with the majority of internalized antigen being delivered to lysosomes that are largely class II negative. The extensive colocalization of BCR-internalized antigen and newly synthesized class II molecules in CIIV suggests that CIIV may represent a specialized subcellular compartment for BCR-mediated antigen processing. Additionally, we have identified a putative CIIV-marker protein, immunologically related to the Igα subunit of the BCR, which further illustrates the unique nature of these endocytic vesicles.


Sign in / Sign up

Export Citation Format

Share Document