Epigenetic regulation of B cell fate and function during an immune response

2019 ◽  
Vol 288 (1) ◽  
pp. 75-84 ◽  
Author(s):  
Yan Zhang ◽  
Kim L. Good‐Jacobson
Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 130-130
Author(s):  
Hiroshi Arima ◽  
Momoko Nishikori ◽  
Yasuyuki Otsuka ◽  
Kiyotaka Izumi ◽  
Wataru Kishimoto ◽  
...  

Abstract Notch1 signaling pathway is involved in T-cell fate decision and development, but it is also known to be activated in B cells upon anti-IgM or LPS stimulation. In addition to its physiological upregulation in B cells, Notch1 signaling is often aberrantly activated in several lymphoid malignancies of B-cell origin, such as classical Hodgkin lymphoma, mantle cell lymphoma and chronic lymphocytic leukemia. However, functional roles of Notch1 in B cells have not been well elucidated to date. Here we report a novel immunomodulatory role of Notch1-activated B cells that alters T-cell immune response in an IL-33-dependent manner. Functional analysis of Notch1 in mature B cells had been hampered by its substitutability for Notch2, which is involved in early B-cell fate decision towards marginal zone B cells (Zhang et. al. J Immunol 2013). To eliminate such irrelevant effect of Notch1 on early B-cell differentiation, we generated a mouse model in which Notch1 intracellular domain (NICD), a constitutively active form of Notch1, began to be expressed in mature B cells after AICDA promoter-dependent Cre expression in germinal centers (StopFloxed-NICD Tg mice×Aicda-Cre mice, hereby designated as NICD Tg mice). In this mouse model, NICD transgene was expressed in about 5% of total splenic B cells, with normal B cell maturation and differentiation. Alternatively, subsets of splenic CD4+ T cells were significantly altered, with increase in Th2 and Treg cells and decrease in Th1 and Th17 cells. IFN-γ production by CD8+ T cells was also significantly reduced. Consequently, NICD Tg mice were susceptible to fungal infections, and more importantly, they began to die of spontaneous malignant neoplasms such as sarcoma and lymphoma at 9 months of age. The tumor development was further increased when TP53 gene was heterozygously deleted in NICD Tg mice. None of the tumors having developed in NICD Tg mice expressed the NICD transgene, suggesting that these tumors did not develop as a result of direct oncogenic effect of NICD. As serum levels of IFN-γ and TNF-α were significantly lower in NICD Tg mice than in control mice, it was rather suggested that these tumors had developed under a condition of suppressed anti-tumor immunity. To elucidate the mechanism of immunomodulatory activity of Notch1-activated B cells, we performed a comparative gene expression analysis using B cells from NICD Tg and control mice. Among several candidate genes whose expression levels were increased in Notch1-activated B cells, we focused on elevated IL-33 as a potential cause for the immunomodulation. Upregulation of IL-33 protein in Notch1-activated B cells was validated by intracellular cytokine flow cytometry. IL-33 is a cytokine that is expressed in nuclei of broad types of cells in their resting state. However, we found that it was also present in the cytoplasm of Notch1-activated B cells, suggesting that IL-33 is actively produced in these cells. To confirm whether extracellular release of IL-33 from B cells was enhanced through Notch1, we cultured splenic B cells from wild-type mice with LPS stimulation in the presence of L cells with or without Notch1 ligand Delta-like 1 (Dll1) expression. We found that IL-33 secretion from B cells was increased twofold in the presence of Dll1-positive compared to Dll1-negative L cells. As expected, the Dll1-mediated increase in IL-33 levels was successfully blocked by DAPT, a Notch signaling inhibitor. To determine whether the IL-33 secreted from Notch1-activated B cells was responsible for the functional modulation of T cells, we cultured wild-type CD4+ T cells with B cells from NICD Tg or control mice, and measured cytokine levels produced by T cells. As a result, IL-4, IL-13 and IL-10 secretion was markedly increased when T cells were cocultured with Notch1-activated B cells. Strikingly, the increase in these Th2- and Treg-associated cytokine levels was completely canceled by addition of a blocking antibody against the IL-33 receptor ST2. In summary, we have shown that Notch1-activated B cells have a novel immunomodulatory function to alter T-cell immunity towards Th2 and Treg immune response via IL-33 secretion, thereby suppressing cellular immunity. This immunomodulatory mechanism may potentially be utilized by Notch1-activated B-cell neoplasms to escape anti-tumor immunity, and we propose that the Notch1-IL-33-ST2 axis can be a promising target for immunotherapy of lymphoid malignancies. Disclosures Nishikori: Kyowa Kirin: Honoraria; Eisai: Honoraria, Research Funding; Janssen Pharmaceutical: Honoraria. Takaori-Kondo:Alexion Pharmaceuticals: Research Funding; Mochida Pharmaceutical: Research Funding; Shionogi: Research Funding; Eisai: Research Funding; Takeda Pharmaceutical: Research Funding; Astellas Pharma: Research Funding; Kyowa Kirin: Research Funding; Chugai Pharmaceutical: Research Funding; Pfizer: Research Funding; Janssen Pharmaceuticals: Speakers Bureau; Merck Sharp and Dohme: Speakers Bureau; Bristol-Myers Squibb: Speakers Bureau; Toyama Chemical: Research Funding; Cognano: Research Funding.


2014 ◽  
Vol 193 (3) ◽  
pp. 1171-1183 ◽  
Author(s):  
Marie Marquet ◽  
Armand Garot ◽  
Sébastien Bender ◽  
Claire Carrion ◽  
Pauline Rouaud ◽  
...  

2015 ◽  
Vol 194 (6) ◽  
pp. 2624-2634 ◽  
Author(s):  
Julia Ludwig ◽  
Giuseppina Federico ◽  
Sandra Prokosch ◽  
Günter Küblbeck ◽  
Sabine Schmitt ◽  
...  
Keyword(s):  
B Cell ◽  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1138-1138
Author(s):  
Rainer Hubmann ◽  
Martin Hilgarth ◽  
Susanne Schnabl ◽  
Dita Demirtas ◽  
Josef D. Schwarzmeier ◽  
...  

Abstract B-cell chronic lymphocytic leukemia (B-CLL) represents a clonal expansion of self-reactive CD5+ B-lymphocytes which seems to be resistant to apoptosis in vivo. One of the characteristics of B-CLL lymphocytes is the high expression of the B-cell differentiation/activation marker CD23 which we recently identified as a target gene for NOTCH2 signaling. NOTCH2 is implicated in the development/homeostasis of murine CD5+ (B-1a) B-cells, suggesting a function for human NOTCH2 in B-CLL leukemogenesis. Here we show that peripheral B-CLL lymphocytes overexpress a transcriptionally active form of NOTCH2 (N2IC) irrespective of their prognostic marker profile (ie. IgVH mutational status, CD38 expression, and cytogenetics). Although the majority of unstimulated B-CLL samples downregulate their N2IC activity in vitro, DNA-bound N2IC complexes could be maintained by the protein kinase C (PKC) activator TPA (12-O-tetradecanoylphorbol 13-acetate) accompanied by an upregulation of the NOTCH2 target gene CD23 and increased B-CLL cell viability. These effects are sensitive to the PKC-δ selective inhibitor Rottlerin. In 80% of B-CLL cases, NOTCH2 signaling was found to be resistant to the γ-secretase inhibitors (GSI’s) Dapt and compound E, indicative for the expression of truncated forms of NOTCH2 which do not require γ-secretase for processing and function. Inhibition of NOTCH2 either by Dapt in GSI sensitive B-CLL cases or, more specifically, by RNA interference downregulates CD23 expression on the mRNA and protein level and sensitizes B-CLL cells for apoptosis. Since self-reactive B-cells are normally eliminated either by chronic (anergy) or apoptotic (negative selection) B-cell receptor (BCR) signaling, we asked whether NOTCH2 modulates B-cell fate decisions triggered by the BCR. For this reason, we stably transduced the human B-cell line BL41 with constitutive active forms of NOTCH2 and found that NOTCH2 inhibits BCR mediated apoptosis induced by surface-IgM cross-linking. In summary, the data demonstrate that NOTCH2 signaling is deregulated in B-CLL cells and might be critically involved in the PKC-dependent maintenance of their malignant phenotype.


Mitochondrion ◽  
2018 ◽  
Vol 41 ◽  
pp. 58-65 ◽  
Author(s):  
Hector Sandoval ◽  
Srikanth Kodali ◽  
Jin Wang
Keyword(s):  
B Cell ◽  

2016 ◽  
Vol 113 (41) ◽  
pp. E6172-E6181 ◽  
Author(s):  
Alissa C. Rothchild ◽  
James R. Sissons ◽  
Shahin Shafiani ◽  
Christopher Plaisier ◽  
Deborah Min ◽  
...  

The regulation of host–pathogen interactions during Mycobacterium tuberculosis (Mtb) infection remains unresolved. MicroRNAs (miRNAs) are important regulators of the immune system, and so we used a systems biology approach to construct an miRNA regulatory network activated in macrophages during Mtb infection. Our network comprises 77 putative miRNAs that are associated with temporal gene expression signatures in macrophages early after Mtb infection. In this study, we demonstrate a dual role for one of these regulators, miR-155. On the one hand, miR-155 maintains the survival of Mtb-infected macrophages, thereby providing a niche favoring bacterial replication; on the other hand, miR-155 promotes the survival and function of Mtb-specific T cells, enabling an effective adaptive immune response. MiR-155–induced cell survival is mediated through the SH2 domain-containing inositol 5-phosphatase 1 (SHIP1)/protein kinase B (Akt) pathway. Thus, dual regulation of the same cell survival pathway in innate and adaptive immune cells leads to vastly different outcomes with respect to bacterial containment.


2002 ◽  
Vol 185 (s1) ◽  
pp. S37-S45 ◽  
Author(s):  
Christopher B. Wilson ◽  
Karen W. Makar ◽  
Mercedes Pérez‐Melgosa

2021 ◽  
Vol 233 ◽  
pp. 87-91
Author(s):  
Wei Wang ◽  
Luman Wang ◽  
Bingbing Zha
Keyword(s):  
B Cell ◽  

2021 ◽  
Vol 12 ◽  
Author(s):  
Junxia Huang ◽  
Xinzhi Xu ◽  
Ji Yang

T helper 17 (Th17) cells are characterized by the secretion of the IL-17 cytokine and are essential for the immune response against bacterial and fungal infections. Despite the beneficial roles of Th17 cells, unrestrained IL-17 production can contribute to immunopathology and inflammatory autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease. Although these diverse outcomes are directed by the activation of Th17 cells, the regulation of Th17 cells is incompletely understood. The discovery that microRNAs (miRNAs) are involved in the regulation of Th17 cell differentiation and function has greatly improved our understanding of Th17 cells in immune response and disease. Here, we provide an overview of the biogenesis and function of miRNA and summarize the role of miRNAs in Th17 cell differentiation and function. Finally, we focus on recent advances in miRNA-mediated dysregulation of Th17 cell fate in autoimmune diseases.


2016 ◽  
Vol 310 ◽  
pp. 78-88 ◽  
Author(s):  
Joseph W. Brewer ◽  
Viktoriya Solodushko ◽  
Ileana Aragon ◽  
Robert A. Barrington
Keyword(s):  
B Cell ◽  

Sign in / Sign up

Export Citation Format

Share Document