scholarly journals Differential representation of B cell subsets in mixed bone marrow chimera mice due to expression of allelic variants of CD45 (CD45.1/CD45.2)

2013 ◽  
Vol 396 (1-2) ◽  
pp. 163-167 ◽  
Author(s):  
Sreemanti Basu ◽  
Avijit Ray ◽  
Bonnie N. Dittel
Blood ◽  
2002 ◽  
Vol 100 (13) ◽  
pp. 4557-4564 ◽  
Author(s):  
Thomas J. Waldschmidt ◽  
Angela Panoskaltsis-Mortari ◽  
Ronald T. McElmurry ◽  
Lorraine T. Tygrett ◽  
Patricia A. Taylor ◽  
...  

In allogeneic hematopoietic stem cell transplant recipients, restoration of humoral immunity is delayed and can remain impaired for years. In many severe combined immune deficiency (SCID) patients given haploidentical bone marrow (BM), lesions in humoral immunity are exacerbated by poor engraftment of donor B cells. The nature of these defects is important to understand as they render patients susceptible to infection. Previous work in mice suggested that in utero transplantation (IUT) of allogeneic BM might offer several advantages for the correction of primary immune deficiencies. In SCID mice given fully allogeneic BM in utero, the lymphoid compartment was restored with minimal evidence of graft-versus-host disease (GVHD). The present report examines B-cell reconstitution and function in mice that have received allogeneic IUT. Results are compared with those of adult mice given total body irradiation (TBI) followed by transplantation with allogeneic BM. In addition to enumerating the various B-cell subsets present in BM, spleen, and peritoneal cavity (PC), B-cell competence was assessed by challenging mice with T cell–independent (TI) and T cell–dependent (TD) antigens. The results demonstrated that all B-cell subsets in the BM and periphery were restored in allogeneic IUT and TBI mice, as were antibody responses after TI challenge. Upon immunization with TD antigens, however, IUT and TBI mice exhibited suboptimal responses as measured by the capacity to isotype switch and generate germinal center (GC) B cells. Thus, although allogeneic BM transplantation results in complete recovery of the B-cell compartment, certain elements of the humoral response remain defective.


Blood ◽  
2011 ◽  
Vol 117 (24) ◽  
pp. 6552-6561 ◽  
Author(s):  
Frédéric Mourcin ◽  
Caroline Breton ◽  
Julie Tellier ◽  
Priyanka Narang ◽  
Lionel Chasson ◽  
...  

Abstract In the bone marrow (BM), stromal cells constitute a supportive tissue indispensable for the generation of pro-B/pre-BI, pre-BII, and immature B lymphocytes. IL-7–producing stromal cells constitute a cellular niche for pro-B/pre-BI cells, but no specific stromal cell microenvironment was identified for pre-BII cells expressing a functional pre-B cell receptor (pre-BCR). However expression of the pre-BCR represents a crucial checkpoint during B-cell development. We recently demonstrated that the stromal cell derived-galectin1 (GAL1) is a ligand for the pre-BCR, involved in the proliferation and differentiation of normal mouse pre-BII cells. Here we show that nonhematopoietic osteoblasts and reticular cells in the BM express GAL1. We observed that pre-BII cells, unlike the other B-cell subsets, were specifically localized in close contact with GAL1+ reticular cells. We also determined that IL-7+ and GAL1+ cells represent 2 distinct mesenchymal populations with different BM localization. These results demonstrate the existence of a pre-BII specific stromal cell niche and indicate that early B cells move from IL-7+ to GAL1+ supportive BM niches during their development.


Author(s):  
Tianshu Yu ◽  
Haoyi Wang ◽  
Yajing Zhao ◽  
Yafei Yu ◽  
Yu Hou ◽  
...  

Primary immune thrombocytopenia (ITP) is an autoantibody-mediated hemorrhagic disorder where B cells play an essential role. Previous studies have focused on peripheral blood (PB), but B cells in bone marrow (BM) have not been well characterized. We aimed to explore the profile of B cell subsets and their cytokine environments in BM of ITP patients to further clarify the pathogenesis of the disease. B cell subpopulations and their cytokine/chemokine receptors were detected by flow cytometry. Plasma concentrations of cytokines/chemokines were measured by ELISA. mRNA levels of B cell-related transcription factors were determined by qPCR. Regulatory B cell (Breg) function was assessed by quantifying their inhibitory effects on monocytes and T cells in vitro. Decreased proportions of total B cells, naïve B cells and defective Bregs were observed in ITP patients compared with healthy controls (HCs), whereas elevated frequency of long-lived plasma cells was found in BM of autoantibody-positive patients. No statistical difference was observed in plasmablasts or in short-lived plasma cells between ITP patients and HCs. The immunosuppressive capacity of BM Bregs from ITP patients was considerably weaker than that from HCs. In vivo study using an active ITP murine model revealed that Breg transfusion could significantly alleviate thrombocytopenia. Moreover, over-activation of CXCL13-CXCR5 and BAFF/APRIL systems were found in ITP patient BM. Taken together, B cell subsets in BM were skewed toward a proinflammatory profile in ITP patients, suggesting the involvement of dysregulated BM B cells in the development of the disease.


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.


2013 ◽  
Vol 174 (1) ◽  
pp. 53-59 ◽  
Author(s):  
S. Agrawal ◽  
S. A. B. C. Smith ◽  
S. G. Tangye ◽  
W. A. Sewell

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 46-47
Author(s):  
Andrea Härzschel ◽  
Peter William Krenn ◽  
Elisabeth Bayer ◽  
Simone Tangermann ◽  
Geoffroy Andrieux ◽  
...  

Introduction The development and maturation of B cells is highly dependent on signals provided by the microenvironment of the lymphatic organs. As B cells move from one developmental stage and niche to the next, the integrin family of adhesion molecules provides important cues for their correct positioning and retention. The integrin adaptor protein Kindlin-3 (encoded by the Fermt3 gene) regulates integrin activity and function in a wide range of hematopoietic cell types. In this study, we aimed to define its precise role in the development and function of the different murine B cell subsets. Methods We crossed a Fermt3flox/flox mb1-cre mouse strain (hereforth called K3ΔB mice), harboring a B cell specific Kindlin-3 deletion. B cell subsets in the different lymphoid organs of these K3ΔB mice and control littermates were defined by multicolor flow cytometry. Adoptive transfer, microscopy and real-time flow cytometry were used to analyze the different steps of integrin activation. A co-culture system with OP9 stromal cells and BAFF was used to assess the in vitro differentiation potential of immature progenitors into the different mature B cell subsets. Transcriptional differences between follicular B cells isolated from spleens of K3ΔB- and control mice were assessed by transcriptome array. Results In vitro, we found that integrin activation on B cells was induced upon activation of the chemokine receptors CXCR4 and CXCR5 or the B cell receptor. This stimulation triggered adhesion of wild type B cells to integrin ligands under shear flow. The increase of VLA-4 integrin affinity to its ligand substrates during this process could also be calculated from real-time flow cytometrical analyses. In contrast, K3ΔB-derived B cells could not reach high affinity states of integrins and thus failed to adhere on the substrates upon stimulation, despite slight upregulation of chemokine receptors CXCR4 and CXCR5. B cell migration towards the respective chemokines also required Kindlin-3, even in an integrin ligand-free setting. In vivo, Kindlin-3 was required for homing of mature B cells to the bone marrow and to lymph nodes. When further characterizing K3ΔB mice by flow cytometry and immunohistochemistry we observed increased early B cell numbers in the bone marrow. Of note, marginal zone (MZ) B cells in the spleen were completely absent (Figure 1 A+B). We consequently assessed the potential of immature B cells to develop into B cells with high expression of CD21, a marker for MZ B cells, upon their co-culture with OP9 stromal cells in the presence of the B cell survival factor BAFF. While 18% of B cells differentiating from wild type bone marrow displayed high expression of CD21, the percentage of CD21 high cells recovered from Kindlin-3 deficient progenitors was significantly lower (~12%, Figure 1C). Pathways involved in these developmental differences were analyzed by a transcriptome array, revealing increased activity of the B cell receptor pathway in the knockout situation accompanied by higher, NFkappaB and Notch signaling. Conclusion/Outlook Whereas our results highlight the importance of Kindlin-3 dependent, integrin mediated cell retention and migration during B cell development they also indicate that Kindlin-3 functions in an integrin-independent manner when regulating cell motility and transcription. The complete lack of MZ B cells in the absence of Kindlin-3 is thus most likely a combination of defective retention in the MZ area and transcriptional alterations favoring the development of transitional B cells into follicular- rather than MZ B cells. Figure 1 : B-cell specific Kindlin-3 knockout leads to loss of splenic marginal zone B cells. The percentage of MZ B-cells among total splenic B cells was determined by flow cytometry in K3ΔB mice and wild type (wt) littermates (A). Immunohistochemistry staining of CD19 showed a loss of loosely packed marginal zone B cells (yellow arrows) in the absence of Kindlin-3 (B). B cells were enriched from the bone marrow of K3ΔB mice and wt littermates and cultured on a confluent layer of OP9 cells in the presence of 200 ng/ml BAFF for 72 h. Development of CD21 high/CD23 low B cells was then determined by flow cytometry (C). Figure Disclosures Greil: Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Astra zeneca: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Daiichi Sankyo, Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; F. Hoffmann-La Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; BMS/celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; MSD Merck: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, accomodations, expenses, Research Funding.


2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 234.3-235
Author(s):  
T. Fu ◽  
Y. Yang ◽  
X. Gu ◽  
C. Dong ◽  
R. Zhao ◽  
...  

Background:B cell differentiation and dysfunction play a key role in the pathogenesis of Systemic lupus erythematosus (SLE). Bone marrow (BM) is the development organ of B cells, and also the home and residence place of plasma cells and memory B cells. However, there is a lack of studies on B cells in BM with lupus.Objectives:To map the development of BM and peripheral B cells and investigate the mechanism of abnormal early B cell development in SLE.Methods:A total of 11 SLE patients and 5 age- and sex-matched controls were recruited.BM and peripheral B cell subsets were measured by flow cytometry. sorting-purified B cell subsets were subject toSingle-cell RNA sequencing (scRNA-seq) and functional studies. Plasma cytokines and secreted immunoglobulins were detected by Luminex or ELISA. Disease activity of SLE patients was measured using the SLE Disease Activity Index (SLEDAI).Results:In the present study, we find out that the percentage of monocytes in MNC (p=0.070) and plasma cells(p=0.001)in CD19+ were significantly decreased in BM of SLE, compared to healthy controls. While, SLE patients had increased T%MNC(p=0.008) and B%CD19+(p=0.002) in BM that controls. In detail, the B cell subsets of bone marrow in patients with active lupus (SLEDAI≥8 score) were seriously disordered, showing the increasing T%MNC(p=0.049), propre-B%CD19+ (p=0.006)and immature B cell%CD19+ (p=0.010) than healthy donors. propre-B%CD19+ exhibited good relationship with SLEDAI. By integrating single B cell expression profiling and repertoire analysis, we map the development of B cells in BM and peripheral and pathogenic characteristics of early B cells, especially propre-B.Conclusion:These findings demonstrated that early B cells in BM, especially propre-B are abnormally differentiated with dysregulations. BM is an important organ targeted by SLE. This studyis not only to clarify the internal mechanism of the disorder of differentiation of B cells, but also to provide new clues for the targeted diagnosis and treatment of SLE.References:[1]Palanichamy, A., et al.,Neutrophil-mediated IFN activation in the bone marrow alters B cell development in human and murine systemic lupus erythematosus.J Immunol, 2014.192(3): p. 906-18.[2]Papadaki, H.A., J.C. Marsh, and G.D. Eliopoulos,Bone marrow stem cells and stromal cells in autoimmune cytopenias.Leuk Lymphoma, 2002.43(4): p. 753-60.[3]Karrar, S. and D.S. Cunninghame Graham,Abnormal B Cell Development in Systemic Lupus Erythematosus: What the Genetics Tell Us.Arthritis Rheumatol, 2018.70(4): p. 496-507.[4]Woods, M., Y.R. Zou, and A. Davidson,Defects in Germinal Center Selection in SLE.Front Immunol, 2015.6: p. 425.[5]Upregulation of p16INK4A promotes cellular senescence of bone marrow-derived mesenchymal stem cells from systemic lupus erythematosus patients.Cell Signal, 2012.24(12): p. 2307-14.Disclosure of Interests:None declared


Author(s):  
Irene Stachura ◽  
Milton H. Dalbow ◽  
Michael J. Niemiec ◽  
Matias Pardo ◽  
Gurmukh Singh ◽  
...  

Lymphoid cells were analyzed within pulmonary infiltrates of six patients with lymphoproliferative disorders involving lungs by immunofluorescence and immunoperoxidase techniques utilizing monoclonal antibodies to cell surface antigens T11 (total T), T4 (inducer/helper T), T8 (cytotoxic/suppressor T) and B1 (B cells) and the antisera against heavy (G,A,M) and light (kappa, lambda) immunoglobulin chains. Three patients had pseudolymphoma, two patients had lymphoma and one patient had lymphomatoid granulomatosis.A mixed population of cells was present in tissue infiltrates from the three patients with pseudolymphoma, IgM-kappa producing cells constituted the main B cell type in one patient. In two patients with lymphoma pattern the infiltrates were composed exclusively of T4+ cells and IgG-lambda B cells predominated slightly in the patient with lymphomatoid granulomatosis.


Sign in / Sign up

Export Citation Format

Share Document