scholarly journals Role of TLR in B Cell Development: Signaling through TLR4 Promotes B Cell Maturation and Is Inhibited by TLR2

2005 ◽  
Vol 174 (11) ◽  
pp. 6639-6647 ◽  
Author(s):  
Elize A. Hayashi ◽  
Shizuo Akira ◽  
Alberto Nobrega
1993 ◽  
Vol 7 (4) ◽  
pp. 570-582 ◽  
Author(s):  
L M Corcoran ◽  
M Karvelas ◽  
G J Nossal ◽  
Z S Ye ◽  
T Jacks ◽  
...  

2019 ◽  
Author(s):  
Samantha A. Swenson ◽  
Tyler J. Gilbreath ◽  
Heather Vahle ◽  
R. Willow Hynes-Smith ◽  
Jared H. Graham ◽  
...  

ABSTRACTCoordination of a number of molecular mechanisms including transcription, alternative splicing, and class switch recombination are required to facilitate development, activation, and survival of B cells. Disruption of these pathways can result in malignant transformation. Recently, next generation sequencing has identified a number of novel mutations in mantle cell lymphoma (MCL) patients including the ubiquitin E3 ligase UBR5. Approximately 18% of MCL patients were found to have mutations in UBR5 with the majority of mutations within the HECT domain of the protein which can accept and transfer ubiquitin molecules to the substrate. Determining if UBR5 controls the maturation of B cells is important to fully understand malignant transformation to MCL. To elucidate the role of UBR5 in B cell maturation and activation we generated a conditional mutant disrupting UBR5’s C-terminal HECT domain. Loss of the UBR5 HECT domain leads to a block in maturation of B cells in the spleen and up-regulation of proteins associated with mRNA splicing via the spliceosome. Our studies reveal a novel role of UBR5 in B cell maturation by regulating alternative splicing of key transcripts during B cell development and suggests UBR5 mutations may promote mantle cell lymphoma initiation.KEY POINTSUtilizing a novel mouse model mimicking MCL patient mutations, the loss of UBR5’s HECT domain causes alterations in B cell development.UBR5 mutations lead to stabilization of UBR5 and aberrant splicing.


F1000Research ◽  
2018 ◽  
Vol 7 ◽  
pp. 429 ◽  
Author(s):  
Juan Carlos Yam-Puc ◽  
Lingling Zhang ◽  
Yang Zhang ◽  
Kai-Michael Toellner

B-cell development is characterized by a number of tightly regulated selection processes. Signals through the B-cell receptor (BCR) guide and are required for B-cell maturation, survival, and fate decision. Here, we review the role of the BCR during B-cell development, leading to the emergence of B1, marginal zone, and peripheral follicular B cells. Furthermore, we discuss BCR-derived signals on activated B cells that lead to germinal center and plasma cell differentiation.


2007 ◽  
Vol 179 (2) ◽  
pp. 1068-1079 ◽  
Author(s):  
Xuezhi Dai ◽  
Yuhong Chen ◽  
Lie Di ◽  
Andrew Podd ◽  
Geqiang Li ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1179-1179
Author(s):  
Zhongfa Yang ◽  
Yu Zhu ◽  
Rachel Gerstein ◽  
Alan G. Rosmarin

Abstract B lymphocytes develop in the bone marrow and later encounter antigen in lymph nodes, where they complete their development as plasma cells or B memory cells. Several key transcription factors have been identified that are required for B cell development, including Pax5, BCL6, C-MYC, and others. GABP is a tetrameric ets transcription factor that includes the DNA-binding GABP alpha protein, and the unrelated protein, GABP beta, which contains multimerization and transcriptional activation domains. GABP plays key roles in cell cycle control and mitochondrial biogenesis. It is also required for lineage specific gene expression, and it was previously shown to control gene expression of the IL-7 receptor and Pax5, both of which are required for lymphocyte development. Disruption of mouse Gabpα caused cell cycle arrest in hematopoietic stem cells (HSC), profound loss of progenitor cells, and aberrant myeloid differentiation. We created a conditional knockout model of Gabpα in B lymphocytes by breeding mice with lox-P flanked Gabpa to mice that bear Cre recombinase knocked into the B-cell specific CD19 locus; the mice also carry the Rosa 26 lox-STOP-lox YFP transgene, which permits identification and isolation of individual Gabpα null cells, based on expression of YFP. Loss of Gabpα was highly lineage specific for B lymphocytes. Gabpa null mice were healthy and vigorous through young adulthood, but some developed rectal prolapse by nine months of age, and necropsy demonstrated thinning of the intestinal wall and loss of Peyer's Patches and other lymphoid tissue. We immunologically characterized mice between 6 and 8 weeks of age, in order to minimize secondary effects of the inflammatory process associated with rectal prolapse. There was no deletion of Gabpα in T lymphocytes, and no discernable effect on T-cell subpopulations. We observed a significant reduction in Gabpα null (YFP+) B cells, in comparison with the Gabpα replete (YFP-) B cells in bone marrow and spleen. Gabpα null cells contributed to the pro-B cell population, but there was a progressively reduced contribution of Gabpα null cells to later stages of B cell maturation. We detected no Gabpα null cells among mature naive IgD+/IgM+ B cells, indicating a profound block in B cell maturation in cells that lack Gabpα. Importantly, no YFP+ CD138+ cells were detected, indicating that Gabpα null cells could not contribute to plasma cell development. We conclude that Gabp is required for full B cell maturation and plasma cell development in mice, and that its deletion is associated with loss of Peyer's Patches and rectal prolapse. GABP was previously shown to regulate expression of IL-7R and Pax5, which are expressed in lymphoid progenitor cells long before activation of CD19 expression. Thus, failure of B cell development and plasma cell formation in this CD19-Cre Gabpα null model is independent of the effect of GABP on those other B cell factors, and indicates a new, critical role for GABP in later stages of B cell and plasma cell development. Although rectal prolapse has been observed in mice with T cell defects, this represents the first demonstration that B cell defects cause such a phenotype. Disclosures Gerstein: Vertex Pharmaceuticals: Other: employer of spouse.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3588-3588
Author(s):  
Yongwei Zheng ◽  
Yuhong Chen ◽  
Xiaona You ◽  
Mei Yu ◽  
Guoping Fu ◽  
...  

Abstract Small GTPases of the Ras subfamily regulate multiple signaling pathways and control numerous biological functions. Although the three major Ras members, Kras, Hras and Nras, are highly homologous, individual Ras gene can have distinct biological functions. Embryonic lethality of Kras-deficient mice precludes study of the biological role of Kras. Here, we generated and examined mice with hematopoietic- and B cell-specific deletion of Kras. In VavCreKrasfl/fl mice with hematopoietic deletion of Kras, the populations of bone marrow (BM) pre-, immature and mature B cells were reduced. The population of peripheral follicular (FO), marginal zone (MZ) and B1 mature B cells were also reduced in VavCreKrasfl/fl relative to control mice. In addition, BM chimeric mice with B cell-specific deficiency of Kras generated by transplantation of T- and B-null Rag1-deficient mice with a mixture of BM cells from VavCreKrasfl/fl mice and B cell-deficient μMT mice displayed a marked reduction of pre-, immature and mature B cells in the BM and mature B cells in the spleen. Thus, Kras deficiency intrinsically impairs early B cell development and late B cell maturation. Further, the effect of Kras deficiency on B-cell proliferation and survival was examined. The 3H-thymidine incorporation rate of Kras-deficient, relative to control, mature B cells in response to anti-IgM or anti-IgM plus IL-4 was markedly reduced. In addition, Kras-deficient B cells displayed a marked decrease of cell cycle entry and increase of cell apoptosis upon anti-IgM or anti-IgM plus IL-4 stimulation. Thus, Kras deficiency impairs BCR-induced B cell proliferation and survival. Lastly, the role of Kras in BCR signaling was studied. The level of total BCR-activated Ras (Ras-GTP) was largely reduced in Kras-deficient B cells. BCR-induced Ca2+ flux was comparable between Kras-deficient and control B cells. BCR-induced phosphorylation of Akt and IkBa was normal in mutant relative to control B cells. However, BCR-induced activation of ERK1/2 but not JNK or p38 was impaired in Kras-deficient relative to control B cells. Consistently, BCR-induced activation of Raf-1 and MEK1/2, the upstream activators of ERK1/2, was markedly reduced in mutant B cells. In addition, pre-BCR-induced ERK1/2 activation was impaired in Kras-deficient pre-B cells. Pre-BCR-induced activation of AP-1, the transcription factors downstream of ERK1/2, was decreased in mutant pre-B cells. Thus, Kras plays an important role in BCR- and pre-BCR-mediated activation of the ERK pathway in B cells. Taken together, these findings demonstrate that Kras is the Ras family member that critically regulates early B cell development and late B cell maturation through controlling the Raf-1/MEK/ERK pathway. Disclosures No relevant conflicts of interest to declare.


2003 ◽  
Vol 198 (4) ◽  
pp. 581-589 ◽  
Author(s):  
Masaki Hikida ◽  
Sachiko Johmura ◽  
Ari Hashimoto ◽  
Mayuko Takezaki ◽  
Tomohiro Kurosaki

Two signaling pathways known to be essential for progression from immature to mature B cells are BAFF receptor (BAFF-R) and the B cell receptor (BCR). Here, we first show that phospholipase C (PLC)-γ2 is required for a BAFF-R–mediated survival signal. Then, we have examined the question of whether the reduced number of mature B cells in PLC-γ2−/− mice is caused by a defect in either BCR or BAFF-R signaling. We find that a PLC-γ2 SH2 mutant, which inhibits coupling between BCR and PLC-γ2, fails to restore B cell maturation, despite supporting BAFF-dependent survival. Therefore, our data suggest that the BAFF-R–mediated survival signal, provided by PLC-γ2, is not sufficient to promote B cell maturation, and that, in addition, activation of PLC-γ2 by BCR is required for B cell development.


Sign in / Sign up

Export Citation Format

Share Document