scholarly journals Transcriptional profiling of human intestinal plasma cells reveals effector functions beyond antibody production

2019 ◽  
Vol 7 (10) ◽  
pp. 1399-1407 ◽  
Author(s):  
Omri Snir ◽  
Chakravarthi Kanduri ◽  
Knut E A Lundin ◽  
Geir Kjetil Sandve ◽  
Ludvig M Sollid
Bone Research ◽  
2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Yibo Gan ◽  
Jian He ◽  
Jun Zhu ◽  
Zhengyang Xu ◽  
Zhong Wang ◽  
...  

AbstractA comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.


PLoS ONE ◽  
2017 ◽  
Vol 12 (8) ◽  
pp. e0183264 ◽  
Author(s):  
Luis M. Valor ◽  
Beatriz Rodríguez-Bayona ◽  
Ana B. Ramos-Amaya ◽  
José A. Brieva ◽  
Antonio Campos-Caro

2012 ◽  
Vol 12 (3) ◽  
pp. 213-221 ◽  
Author(s):  
Beatriz Leon ◽  
Andre Ballesteros-Tato ◽  
Ravi S. Misra ◽  
Wojciech Wojciechowski ◽  
Frances E. Lund

Blood ◽  
2014 ◽  
Vol 123 (22) ◽  
pp. 3440-3451 ◽  
Author(s):  
Ping Zhou ◽  
Xun Ma ◽  
Lakshmanan Iyer ◽  
Chakra Chaulagain ◽  
Raymond L. Comenzo

Key PointsImmunoglobulin light-chain and antibody production by plasma cells is significantly reduced by siRNA for the light-chain constant region. In plasma cells making intact antibodies, knockdown of light chains can cause terminal ER stress because of unpaired heavy chains.


2020 ◽  
Vol 11 (11) ◽  
Author(s):  
Baohong Luo ◽  
Yikang Zhan ◽  
Minqi Luo ◽  
Huimin Dong ◽  
Jun Liu ◽  
...  

Abstract Long-lived plasma cells (LLPCs) are robust specialized antibody-secreting cells that mainly stay in the bone marrow and can persist a lifetime. As they can be generated by inducing the differentiation of B-lymphocytes, we investigated the possibility that human LLPCs might be engineered to express α-PD-1 monoclonal antibody to substitute recombinant α-PD-1 antitumor immunotherapy. To this end, we inserted an α-PD-1 cassette into the GAPDH locus through Cas9/sgRNA-guided specific integration in B-lymphocytes, which was mediated by an integrase-defective lentiviral vector. The edited B cells were capable of differentiating into LLPCs both in vitro and in vivo. Transcriptional profiling analysis confirmed that these cells were typical LLPCs. Importantly, these cells secreted de novo antibodies persistently, which were able to inhibit human melanoma growth via an antibody-mediated checkpoint blockade in xenograft-tumor mice. Our work suggests that the engineered LLPCs may be utilized as a vehicle to constantly produce special antibodies for long-term cellular immunotherapy to eradicate tumors and cellular reservoirs for various pathogens including human immunodeficiency virus type 1 (HIV-1) and hepatitis B virus (HBV).


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e14004-e14004
Author(s):  
Katja Klausz ◽  
Amir Karimzadeh-Tabrizi ◽  
Malena Buck ◽  
Anna Otte ◽  
Steffen Krohn ◽  
...  

e14004 Background: Monoclonal antibodies are established treatment options for B cell-derived malignancies, but relapse is still the major challenge. Novel target structures may open alternative avenues to develop effective antibody therapies. Here, we characterized the novel tetravalent antibody ‘EBU-141 Tetra’ and identified the glycoantigen CD75s (α-2,6-sialylated lactosamines) as suitable target structure for antibody-based therapy. CD75s was detected on most B cell lymphomas, including Burkitt’s lymphoma, FL, DLBCL, MCL, CLL, and plasma cell tumors. Classical Hodgkin lymphomas were consistently negative while reactivity on individual cases of peripheral T cell lymphoma was seen. To evaluate CD75s as a target for antibody therapy, we generated a tetravalent, Fc-engineered chEBU-141 IgG1 antibody with enhanced avidity for CD75s and potent effector functions. Methods: ‘EBU-141 Tetra’ was produced by transient transfection and purified by affinity chromatography. Direct anti-tumor effects and Fc-mediated effector functions were investigated in cell proliferation assays, by fluorescence microscopy and in 51Cr release experiments using lymphoma and myeloma cell lines and patient-derived tumor cells. Peripheral blood mononuclear cells and monocyte-derived macrophages of healthy donors were used as human effector cells in the experiments. Results: ‘EBU-141 Tetra’ showed improved binding to CD75s on cell surface of mature B cell lymphoma as well as myeloma plasma cells compared to the conventional chimeric antibody chEBU-141 IgG1. The higher avidity for CD75s resulted in markedly improved ADCC activity of ‘EBU-141 Tetra’ against Daudi Burkitt’s lymphoma, U266 plasma cells and CLL patient-derived tumor cells with EC50 values in the low nanomolar range. In addition, ‘EBU-141 Tetra’ demonstrated efficient phagocytosis of Burkitt’s lymphoma and myeloma cell lines. Thus, the novel tetravalent, chimeric, Fc-engineered antibody ‘EBU-141 Tetra’ efficiently recruits immune effector cells for tumor cell lysis. Conclusions: Our findings further demonstrate that highly potent IgG-like antibodies against glycan-structures can be generated from mouse IgM antibodies and may open a new therapeutic window for therapy of patients with mature B cell lymphomas and multiple myeloma.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2034-2034 ◽  
Author(s):  
Heiko Bruns ◽  
Hanna Gehlen ◽  
Jens Nolting ◽  
Shirin Pasemann ◽  
Peter Brossart ◽  
...  

Abstract Introduction: The bone marrow niche plays a critical role in determining the fate of malignant plasma cells in multiple myeloma (MM). Macrophages are an abundant component of the stromal cell compartment and are believed to support proliferation, survival, and drug resistance of MM cells. Conversely, macrophages can directly kill tumor cells and participate in antitumor immune responses as effector cells. Moreover, macrophages are key immune effector cells for the therapeutic effect of monoclonal antibodies. Lenalidomide, an immunomodulatory drug (IMiD®) is used for the treatment of MM, also in the combination with therapeutic antibodies. Lenalidomide is thought to target the stromal support, but its precise influence on the phenotype or the effector functions of macrophages is still unclear. Methods: To investigate the effect of lenalidomide on the interaction between macrophages and malignant plasma cells in vitro, we coincubated lenalidomide pretreated macrophages with several MM cell lines, and analysed the viability, proliferation and phenotype. For in vivo studies we utilized 5TMM mice, a suitable animal model for MM. Animals were treated with lenalidomide (50 mg/kg 5days/week) for 3 weeks, and the effector functions and phenotype of isolated bone marrow macrophages were analyzed. In addition, macrophages in the bone marrow of MM patients treated with lenalidomide were characterized by immunohistochemistry and flow cytometry. Results: We showed, that infiltrating macrophages in the bone marrow of MM patients display an anti-inflammatory M2-like phenotype characterized by the expression of surface marker CD163, CD206, PD-L1 and cytokine/chemokine secretion (e.g. IL10, CXCL10, APRIL, BAFF and RANKL). Incubation of macrophages with lenalidomide in vitro, substantially changed their transcriptional program (e.g. downregulation of IRF4 and upregulation of IRF5) and their phenotype (e.g. downregulation of the surfaces marker CD163, CD206, and upregulation of CD16, CD64, CD40 and CD86). Furthermore, we show that lenalidomide treatment decreases the expression of RANKL, BAFF and APRIL, while tumoricidal effector molecules (e.g. TRAIL, cathelicidine, Granzyme B) were increased. When lenalidomide treated macrophages were cocultured with MM cells significant cytotoxicity was detected, for all MM cell lines tested. In contrast, untreated macrophages promote tumor growth and viability of MM cells. Conclusion: Lenalidomide in vitro influences macrophages by reverting an anti-inflammatory M2 like profile to a more immunogenic phenotype. In addition it impacts on the support function by decreasing the secretion of important growth factors for B-cells. Similar results were observed in first in vivo studies. Taken together our results imply that lenalidomide interrupts an important stromal cell function thereby influencing survival of MM cells. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4620-4620
Author(s):  
Howard S Oster ◽  
Naamit Deshet-Unger ◽  
Max Gassmann ◽  
Moshe Mittelman ◽  
Drorit Neumann

Abstract Introduction: Recombinant human erythropoietin (EPO) treats anemia, but EPO also has non-erythroid effects. We have previously shown that EPO has anti-neoplastic immunomodulating effects in both patients and mice (Mittelman PNAS 2001; Mittelman Eur J Haematol 2004). EPO effects were demonstrated in both the cellular and humoral immune systems (Katz Acta Haematol 2005; Katz Eur J Immunol 2007; Prutchi-Sagiv Br J Haematol 2006; Prutchi-Sagiv Exp Hematol 2008; Lifshitz Mol Immunol 2009, Hassan Ren Fail 2003). In a previous study we found that EPO was associated with an improved antibody response to the seasonal influenza vaccine in patients (Oster Exp Hematol 2013). B-cell maturation begins in the bone marrow (BM), and continues primarily in the spleen. The cells mature either to marginal zone (MZ) or to Follicular B-cells, both of which can progress to antibody producing plasma cells (PC). This study evaluates EPO's effects on B-cell maturation and antibody production. Methods and Results: Two murine models: 1) Mice were injected (INJ) with either recombinant human EPO (rHuEPO 180units) or saline 3 times over one week (9+8 mice respectively). 2) Transgenic mice from the Tg6 line (TG), with constitutively increased levels of EPO from birth vs wild type (9+8) mice. The total B220+ (a pan B marker) cell number in EPO mice of both murine models was significantly reduced in the BM (similar to Singbrant Blood 2011; see Table). In the spleen, the total number of B220+ cells was similar, irrespective of EPO exposure. However, some B-cell populations were different (Table): splenic MZ precursor (MZP, B220+/CD21hi/CD24mid/CD23hi) as well as MZ B-cell (B220+/CD21hi/CD24mid/CD23lo) numbers were significantly smaller in EPO mice compared with controls. Splenic PC (B220-/CD138+) were tested in TG mice and their number was greater than in the WT controls (5+6 mice, respectively; see Table). Finally, serum antibodies and light chains were studied and found to be increased in TG compared with WT mice (3+4 mice). IgA: 140±14.1 vs 47±5.0 (x104 ng/ml), p<0.005; kappa TG/WT ratio: 1.6±0.08, p=0.005; and lambda TG/WT ratio: 2.0±0.18, p=0.03. Conclusions: Our findings demonstrate a multistep process, with reduced BM B-cells, reduced splenic MZP and MP cells, followed by increased splenic PC and increased antibody production. EPO may be involved in stimulating this dynamic process and as such may have the additional clinical application of augmenting the humoral immune response in patients.Table.Injected (EPO vs Saline) miceTransgenic vs Wild Type mice(mean%±SEM)EPOSalineTGWTBM B220+, total10.9 ±0.6**28.6 ±1.717.7 ±1.8**30.2 ±1.8Spleen MZP2.1 ±0.2**4.8 ±0.24. 9 ±0.6**9.4 ±1.2Spleen MZ2.2 ±0.4**4.4 ±0.43.8 ±0.5*6.4 ±0.9Spleen PCN/A2.5 ±0.4**0.5 ±0.1*depicts p<0.05; **depicts p<0.005; EPO - erythropoietin, TG - transgenic, WT - wild type, BM - bone marrow, MZ - marginal zone, MZP - marginal zone precursors, PC - plasma cells Disclosures Mittelman: XTL Biotech company, interested in EPO: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document