Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Sci Immunol ; 9(94): eadk0092, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38579014

The transition from immunoglobulin M (IgM) to affinity-matured IgG antibodies is vital for effective humoral immunity. This is facilitated by germinal centers (GCs) through affinity maturation and preferential maintenance of IgG+ B cells over IgM+ B cells. However, it is not known whether the positive selection of the different Ig isotypes within GCs is dependent on specific transcriptional mechanisms. Here, we explored IgG1+ GC B cell transcription factor dependency using a CRISPR-Cas9 screen and conditional mouse genetics. We found that MIZ1 was specifically required for IgG1+ GC B cell survival during positive selection, whereas IgM+ GC B cells were largely independent. Mechanistically, MIZ1 induced TMBIM4, an ancestral anti-apoptotic protein that regulated inositol trisphosphate receptor (IP3R)-mediated calcium (Ca2+) mobilization downstream of B cell receptor (BCR) signaling in IgG1+ B cells. The MIZ1-TMBIM4 axis prevented mitochondrial dysfunction-induced IgG1+ GC cell death caused by excessive Ca2+ accumulation. This study uncovers a unique Ig isotype-specific dependency on a hitherto unidentified mechanism in GC-positive selection.


B-Lymphocytes , Immunoglobulin G , Membrane Proteins , Animals , Mice , Germinal Center , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Signal Transduction , Membrane Proteins/metabolism
3.
Leukemia ; 37(12): 2414-2425, 2023 12.
Article En | MEDLINE | ID: mdl-37775560

Targeted deletion of Raptor, a component of mechanistic target of rapamycin complex 1 (mTORC1), reveals an essential role for mTORC1 in initiation/maintenance of leukemia in a CLL model, resulting from a failure for haemopoietic stem/progenitor cells (HSPCs) to commit to the B cell lineage. Induction of Raptor-deficiency in NSG mice transplanted with Mx1-Raptor CLL progenitor cells (PKCα-KR-transduced HSPCs) after disease establishment revealed a reduction in CLL-like disease load and a significant increase in survival in the mice. Interestingly in an aggressive CLL-like disease model, rapamycin treatment reduced disease burden more effectively than AZD2014 (dual mTORC1/2 inhibitor), indicating a skew towards mTORC1 sensitivity with more aggressive disease. Rapamycin, but not ibrutinib, efficiently targeted the eEF2/eEF2K translation elongation regulatory axis, downstream of mTORC1, resulting in eEF2 inactivation through induction of eEF2T56 phosphorylation. mTOR inhibitor treatment of primary patient CLL cells halted proliferation, at least in part through modulation of eEF2K/eEF2 phosphorylation and expression, reduced protein synthesis and inhibited expression of MCL1, Cyclin A and Cyclin D2. Our studies highlight the importance of translation elongation as a driver of disease progression and identify inactivation of eEF2 activity as a novel therapeutic target for blocking CLL progression.


Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Animals , Mice , Mechanistic Target of Rapamycin Complex 1/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Signal Transduction , Sirolimus , Phosphorylation , Disease Progression
4.
bioRxiv ; 2023 Oct 31.
Article En | MEDLINE | ID: mdl-37609190

To increase antibody affinity against pathogens, positively selected GC-B cells initiate cell division in the light zone (LZ) of germinal centres (GCs). Among those, higher-affinity clones migrate to the dark zone (DZ) and vigorously proliferate by relying on oxidative phosphorylation (OXPHOS). However, it remains unknown how positively selected GC-B cells adapt their metabolism for cell division in the glycolysis-dominant, cell cycle arrest-inducing, hypoxic LZ microenvironment. Here, we show that microRNA (miR)-155 mediates metabolic reprogramming during positive selection to protect high-affinity clones. Transcriptome examination and mass spectrometry analysis revealed that miR-155 regulates H3K36me2 levels by directly repressing hypoxia-induced histone lysine demethylase, Kdm2a. This is indispensable for enhancing OXPHOS through optimizing the expression of vital nuclear mitochondrial genes under hypoxia. The miR-155-Kdm2a interaction is crucial to prevent excessive production of reactive oxygen species and apoptosis. Thus, miR-155-mediated epigenetic regulation promotes mitochondrial fitness in high-affinity clones, ensuring their expansion and consequently affinity maturation.

5.
PLoS Pathog ; 17(11): e1010069, 2021 11.
Article En | MEDLINE | ID: mdl-34748611

ES-62 is the major secreted protein of the parasitic filarial nematode, Acanthocheilonema viteae. The molecule exists as a large tetramer (MW, ~240kD), which possesses immunomodulatory properties by virtue of multiple phosphorylcholine (PC) moieties attached to N-type glycans. By suppressing inflammatory immune responses, ES-62 can prevent disease development in certain mouse models of allergic and autoimmune conditions, including joint pathology in collagen-induced arthritis (CIA), a model of rheumatoid arthritis (RA). Such protection is associated with functional suppression of "pathogenic" hyper-responsive synovial fibroblasts (SFs), which exhibit an aggressive inflammatory and bone-damaging phenotype induced by their epigenetic rewiring in response to the inflammatory microenvironment of the arthritic joint. Critically, exposure to ES-62 in vivo induces a stably-imprinted CIA-SF phenotype that exhibits functional responses more typical of healthy, Naïve-SFs. Consistent with this, ES-62 "rewiring" of SFs away from the hyper-responsive phenotype is associated with suppression of ERK activation, STAT3 activation and miR-155 upregulation, signals widely associated with SF pathogenesis. Surprisingly however, DNA methylome analysis of Naïve-, CIA- and ES-62-CIA-SF cohorts reveals that rather than simply preventing pathogenic rewiring of SFs, ES-62 induces further changes in DNA methylation under the inflammatory conditions pertaining in the inflamed joint, including targeting genes associated with ciliogenesis, to programme a novel "resolving" CIA-SF phenotype. In addition to introducing a previously unsuspected aspect of ES-62's mechanism of action, such unique behaviour signposts the potential for developing DNA methylation signatures predictive of pathogenesis and its resolution and hence, candidate mechanisms by which novel therapeutic interventions could prevent SFs from perpetuating joint inflammation and destruction in RA. Pertinent to these translational aspects of ES-62-behavior, small molecule analogues (SMAs) based on ES-62's active PC-moieties mimic the rewiring of SFs as well as the protection against joint disease in CIA afforded by the parasitic worm product.


Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/prevention & control , Epigenesis, Genetic , Fibroblasts/metabolism , Helminth Proteins/pharmacology , Inflammation/prevention & control , Synoviocytes/metabolism , Acanthocheilonema/metabolism , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cells, Cultured , DNA Methylation , Fibroblasts/drug effects , Fibroblasts/immunology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred DBA , Synoviocytes/drug effects , Synoviocytes/immunology
6.
Front Immunol ; 12: 661678, 2021.
Article En | MEDLINE | ID: mdl-33868314

Germinal centers (GCs) are essential sites for the production of high-affinity antibody secreting plasma cells (PCs) and memory-B cells (MBCs), which form the framework of vaccination. Affinity maturation and permissive selection in GCs are key for the production of PCs and MBCs, respectively. For these purposes, GCs positively select "fit" cells in the light zone of the GC and instructs them for one of three known B cell fates: PCs, MBCs and persistent GC-B cells as dark zone entrants. In this review, we provide an overview of the positive selection process and discuss its mechanisms and how B cell fates are instructed.


Antibodies/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Germinal Center/immunology , Immunologic Memory/immunology , Plasma Cells/immunology , Animals , Antibodies/metabolism , Antibody Affinity/immunology , B-Lymphocytes/metabolism , Germinal Center/cytology , Germinal Center/metabolism , Humans , Plasma Cells/metabolism , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism
7.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article En | MEDLINE | ID: mdl-33419925

Affinity maturation depends on how efficiently germinal centers (GCs) positively select B cells in the light zone (LZ). Positively selected GC B cells recirculate between LZs and dark zones (DZs) and ultimately differentiate into plasmablasts (PBs) and memory B cells (MBCs). Current understanding of the GC reaction presumes that cMyc-dependent positive selection of LZ B cells is a competitive affinity-dependent process; however, this cannot explain the production of GC-derived lower-affinity MBCs or retention of GC B cells with varied affinities. Here, by combining single-cell/bulk RNA sequencing and flow cytometry, we identified and characterized temporally and functionally distinct positively selected cMyc+ GC B cell subpopulations. cMyc+ LZ B cell subpopulations enriched with either higher- or lower-affinity cells diverged soon after permissive positive selection. The former subpopulation contained PB precursors, whereas the latter comprised less proliferative MBC precursors and future DZ entrants. The overall affinity of future DZ entrants was enhanced in the LZ through preferential proliferation of higher-affinity cells. Concurrently, lower-affinity cells were retained in GCs and protected from apoptosis. These findings redefine positive selection as a dynamic process generating three distinct B cell fates and elucidate how positive selection ensures clonal diversity for broad protection.


B-Lymphocytes/metabolism , Germinal Center/immunology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Clonal Selection, Antigen-Mediated , Female , Humans , Lymph Nodes , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells , Receptors, Antigen, B-Cell/genetics
8.
J Exp Med ; 217(7)2020 07 06.
Article En | MEDLINE | ID: mdl-32407433

Memory B cells (MBCs) are key for protection from reinfection. However, it is mechanistically unclear how germinal center (GC) B cells differentiate into MBCs. MYC is transiently induced in cells fated for GC expansion and plasma cell (PC) formation, so-called positively selected GC B cells. We found that these cells coexpressed MYC and MIZ1 (MYC-interacting zinc-finger protein 1 [ZBTB17]). MYC and MIZ1 are transcriptional activators; however, they form a transcriptional repressor complex that represses MIZ1 target genes. Mice lacking MYC-MIZ1 complexes displayed impaired cell cycle entry of positively selected GC B cells and reduced GC B cell expansion and PC formation. Notably, absence of MYC-MIZ1 complexes in positively selected GC B cells led to a gene expression profile alike that of MBCs and increased MBC differentiation. Thus, at the GC positive selection stage, MYC-MIZ1 complexes are required for effective GC expansion and PC formation and to restrict MBC differentiation. We propose that MYC and MIZ1 form a module that regulates GC B cell fate.


B-Lymphocytes/cytology , Cell Differentiation , Germinal Center/cytology , Immunologic Memory , Animals , B-Lymphocytes/metabolism , Cell Cycle/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytidine Deaminase/metabolism , Mice, Knockout , Protein Binding , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin-Protein Ligases/metabolism , Up-Regulation/genetics
9.
Front Immunol ; 9: 2232, 2018.
Article En | MEDLINE | ID: mdl-30356668

Group-2 innate lymphoid cells (ILC2) play critical roles in the initiation and maintenance of type-2 immune responses, predominantly through their production of the type-2 cytokines IL-5, IL-9, and IL-13. ILC2 are essential for the efficient elimination of helminth parasites, but also contribute to the detrimental type-2 immune responses that underlie diseases such as asthma and allergy. While several transcription factors have been identified that regulate the development and function of ILC2, less is known about the post-transcriptional mechanisms that regulate these processes. We identified micro-RNAs (miRNAs) that are co-ordinately regulated in ILC2 from mice exposed to two different stimuli, namely IL-33 "alarmin" administration or Nippostrongylus brasiliensis parasitic worm infection. miR-155 is upregulated in ILC2 in response to both stimuli and miR-155-/- mice had impaired IL-33-driven ILC2 responses. Using mixed bone marrow chimeras, we demonstrate that this deficit is intrinsic to ILC2 and that miR-155 protects ILC2 from apoptosis, while having little impact on ILC2 proliferation or cytokine production. These data reveal a subset of miRNAs that are regulated upon ILC2 activation and establish a specific role for miR-155 in regulating ILC2 survival following activation.


Apoptosis/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , MicroRNAs/immunology , Animals , Apoptosis/genetics , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Interleukin-33/immunology , Lymphocytes/metabolism , Lymphocytes/parasitology , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Nippostrongylus/immunology , Nippostrongylus/physiology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/parasitology
10.
Methods Mol Biol ; 1623: 147-158, 2017.
Article En | MEDLINE | ID: mdl-28589355

Retrovirus-mediated gene transfer has become a powerful tool to investigate roles of specific molecules in B cells, due to its efficiency and expeditiousness. This technology is applicable to activated B cells in order to determine effects of a gene of interest during germinal center (GC) reactions in combination with adoptive transfer. To achieve this, B cells derived from SWHEL mice expressing hen egg lysozyme (HEL)-specific B cell receptors (BCR) are stimulated with HEL antigen in vivo and then with anti-CD40 antibody ex vivo. These cells are then transduced with a retrovirus allowing bicistronic co-expression of a gene of interest and GFP, enabling differentiation of transgene positive cells. The retrovirally transduced cells are then adoptively transferred into immunized CD45.1+ congenic recipient mice, to enable differentiation between donor and host cells. This chapter describes methods for (1) activation of HEL-specific mature B cells, (2) retroviral transduction of the activated B cells, (3) adoptive transfer of the cells into recipients, and (4) analysis of the resultant mice by flow cytometry.


B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Germinal Center/cytology , Germinal Center/metabolism , Transduction, Genetic , Adaptive Immunity , Animals , Biomarkers , Cell Line , Cells, Cultured , Epitopes, B-Lymphocyte/immunology , Flow Cytometry , Genetic Vectors/genetics , Germinal Center/immunology , Humans , Mice , Muramidase/immunology , Retroviridae/genetics , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transgenes
11.
Mol Cell Biol ; 37(9)2017 05 01.
Article En | MEDLINE | ID: mdl-28167605

Thymus-derived regulatory T (tTreg) cells are key to preventing autoimmune diseases, but the mechanisms involved in their development remain unsolved. Here, we show that the C-type lectin receptor CD69 controls tTreg cell development and peripheral Treg cell homeostasis through the regulation of BIC/microRNA 155 (miR-155) and its target, suppressor of cytokine signaling 1 (SOCS-1). Using Foxp3-mRFP/cd69+/- or Foxp3-mRFP/cd69-/- reporter mice and short hairpin RNA (shRNA)-mediated silencing and miR-155 transfection approaches, we found that CD69 deficiency impaired the signal transducer and activator of transcription 5 (STAT5) pathway in Foxp3+ cells. This results in BIC/miR-155 inhibition, increased SOCS-1 expression, and severely impaired tTreg cell development in embryos, adults, and Rag2-/- γc-/- hematopoietic chimeras reconstituted with cd69-/- stem cells. Accordingly, mirn155-/- mice have an impaired development of CD69+ tTreg cells and overexpression of the miR-155-induced CD69 pathway, suggesting that both molecules might be concomitantly activated in a positive-feedback loop. Moreover, in vitro-inducible CD25+ Treg (iTreg) cell development is inhibited in Il2rγ-/-/cd69-/- mice. Our data highlight the contribution of CD69 as a nonredundant key regulator of BIC/miR-155-dependent Treg cell development and homeostasis.


Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Lectins, C-Type/genetics , MicroRNAs/genetics , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein/biosynthesis , T-Lymphocytes, Regulatory/cytology , Animals , Cell Differentiation/immunology , Cells, Cultured , Chimera/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , Organ Culture Techniques , RNA Interference , RNA, Small Interfering/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology
12.
Genes Dev ; 30(22): 2475-2485, 2016 11 15.
Article En | MEDLINE | ID: mdl-27913604

In general, cell fate is determined primarily by transcription factors, followed by epigenetic mechanisms fixing the status. While the importance of transcription factors controlling cell fate has been well characterized, epigenetic regulation of cell fate maintenance remains to be elucidated. Here we provide an obvious fate conversion case, in which the inactivation of polycomb-medicated epigenetic regulation results in conversion of T-lineage progenitors to the B-cell fate. In T-cell-specific Ring1A/B-deficient mice, T-cell development was severely blocked at an immature stage. We found that these developmentally arrested T-cell precursors gave rise to functional B cells upon transfer to immunodeficient mice. We further demonstrated that the arrest was almost completely canceled by additional deletion of Pax5 These results indicate that the maintenance of T-cell fate critically requires epigenetic suppression of the B-lineage gene program.


B-Lymphocytes/cytology , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/genetics , Gene Silencing , Polycomb-Group Proteins/metabolism , T-Lymphocytes/cytology , Animals , Cell Lineage , Gene Deletion , Gene Expression Regulation, Developmental , Immunoglobulin Heavy Chains/genetics , Mice, Inbred C57BL , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic/genetics , Ubiquitin-Protein Ligases/genetics
13.
J Clin Invest ; 126(1): 377-88, 2016 Jan.
Article En | MEDLINE | ID: mdl-26657861

The production of high-affinity antibodies by B cells is essential for pathogen clearance. Antibody affinity for antigen is increased through the affinity maturation in germinal centers (GCs). This is an iterative process in which B cells cycle between proliferation coupled with the acquisition of mutations and antigen-based positive selection, resulting in retention of the highest-affinity B cell clones. The posttranscriptional regulator microRNA-155 (miR-155) is critical for efficient affinity maturation and the maintenance of the GCs; however, the cellular and molecular mechanism by which miR-155 regulates GC responses is not well understood. Here, we utilized a miR-155 reporter mouse strain and showed that miR-155 is coexpressed with the proto-oncogene encoding c-MYC in positively selected B cells. Functionally, miR-155 protected positively selected c-MYC+ B cells from apoptosis, allowing clonal expansion of this population, providing an explanation as to why Mir155 deletion impairs affinity maturation and promotes the premature collapse of GCs. We determined that miR-155 directly inhibits the Jumonji family member JARID2, which enhances B cell apoptosis when overexpressed, and thereby promotes GC B cell survival. Our findings also suggest that there is cooperation between c-MYC and miR-155 during the normal GC response, a cooperation that may explain how c-MYC and miR-155 can collaboratively function as oncogenes.


Apoptosis , B-Lymphocytes/physiology , MicroRNAs/physiology , Proto-Oncogene Proteins c-myc/analysis , Animals , Cell Survival , Germinal Center/physiology , Mice , Mice, Inbred C57BL , MicroRNAs/analysis , Polycomb Repressive Complex 2/physiology
14.
Haematologica ; 100(4): 499-510, 2015 Apr.
Article En | MEDLINE | ID: mdl-25616575

Overwhelming evidence identifies the microenvironment as a critical factor in the development and progression of chronic lymphocytic leukemia, underlining the importance of developing suitable translational models to study the pathogenesis of the disease. We previously established that stable expression of kinase dead protein kinase C alpha in hematopoietic progenitor cells resulted in the development of a chronic lymphocytic leukemia-like disease in mice. Here we demonstrate that this chronic lymphocytic leukemia model resembles the more aggressive subset of chronic lymphocytic leukemia, expressing predominantly unmutated immunoglobulin heavy chain genes, with upregulated tyrosine kinase ZAP-70 expression and elevated ERK-MAPK-mTor signaling, resulting in enhanced proliferation and increased tumor load in lymphoid organs. Reduced function of PKCα leads to an up-regulation of PKCßII expression, which is also associated with a poor prognostic subset of human chronic lymphocytic leukemia samples. Treatment of chronic lymphocytic leukemia-like cells with the selective PKCß inhibitor enzastaurin caused cell cycle arrest and apoptosis both in vitro and in vivo, and a reduction in the leukemic burden in vivo. These results demonstrate the importance of PKCßII in chronic lymphocytic leukemia-like disease progression and suggest a role for PKCα subversion in creating permissive conditions for leukemogenesis.


B-Lymphocytes/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Protein Kinase C beta/genetics , Protein Kinase C-alpha/genetics , Animals , B-Lymphocytes/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Leukemic Infiltration/pathology , Lymphoid Tissue/pathology , Mice , Mice, Knockout , Prognosis , Protein Kinase C beta/antagonists & inhibitors , Protein Kinase C beta/metabolism , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Transduction, Genetic , Tumor Burden/drug effects , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/metabolism
15.
J Exp Med ; 211(11): 2183-98, 2014 Oct 20.
Article En | MEDLINE | ID: mdl-25288398

A single microRNA (miRNA) can regulate the expression of many genes, though the level of repression imparted on any given target is generally low. How then is the selective pressure for a single miRNA/target interaction maintained across long evolutionary distances? We addressed this problem by disrupting in vivo the interaction between miR-155 and PU.1 in mice. Remarkably, this interaction proved to be key to promoting optimal T cell-dependent B cell responses, a previously unrecognized role for PU.1. Mechanistically, miR-155 inhibits PU.1 expression, leading to Pax5 down-regulation and the initiation of the plasma cell differentiation pathway. Additional PU.1 targets include a network of genes whose products are involved in adhesion, with direct links to B-T cell interactions. We conclude that the evolutionary adaptive selection of the miR-155-PU.1 interaction is exercised through the effectiveness of terminal B cell differentiation.


B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , MicroRNAs/genetics , PAX5 Transcription Factor/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , 3' Untranslated Regions , Animals , Antibody Formation/genetics , Antibody Formation/immunology , B-Lymphocytes/immunology , Base Sequence , Binding Sites , Cell Adhesion/genetics , Cell Communication/genetics , Cell Communication/immunology , Gene Expression Regulation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphopoiesis/genetics , Mice , Mice, Knockout , MicroRNAs/chemistry , Myelopoiesis/genetics , PAX5 Transcription Factor/chemistry , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trans-Activators/chemistry , Transcription Factors/genetics
16.
Immunity ; 39(1): 136-47, 2013 Jul 25.
Article En | MEDLINE | ID: mdl-23850379

Memory B cells are essential for generating rapid and robust secondary antibody responses. It has been thought that the unique cytoplasmic domain of IgG causes the prompt activation of antigen-experienced IgG memory B cells. To assess this model, we have generated a mouse containing IgG1 B cells that have never encountered antigen. We found that, upon challenge, antigen-experienced IgG1 memory B cells rapidly differentiated into plasma cells, whereas nonexperienced IgG1 B cells did not, suggesting the importance of the stimulation history. In addition, our results suggest that repression of the Bach2 transcription factor, which results from antigen experience, contributes to predisposition of IgG1 memory B cells to differentiate into plasma cells.


B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Plasma Cells/immunology , Animals , B-Lymphocytes/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Cells, Cultured , Flow Cytometry , Gene Expression/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/immunology , PAX5 Transcription Factor/metabolism , Plasma Cells/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
17.
Eur J Immunol ; 42(4): 1005-15, 2012 Apr.
Article En | MEDLINE | ID: mdl-22531924

During hematopoietic lineage development, hematopoietic stem cells sequentially commit toward myeloid or lymphoid lineages in a tightly regulated manner, which under normal circumstances is irreversible. However, studies have established that targeted deletion of the B-lineage specific transcription factor, paired box gene 5 (Pax5), enables B cells to differentiate toward other hematopoietic lineages, in addition to generating progenitor B-cell lymphomas. Our previous studies showed that subversion of protein kinase C (PKC)-α in developing B cells transformed B-lineage cells. Here, we demonstrate that PKC-α modulation in committed CD19(+) B lymphocytes also promoted lineage conversion toward myeloid, NK-, and T-cell lineages upon Notch ligation. This occurred via a reduction in Pax5 expression resulting from a downregulation of E47, a product of the E2A gene. T-cell lineage commitment was indicated by the expression of T-cell associated genes Ptcra, Cd3e, and gene rearrangement at the Tcrb gene locus. Importantly, the lineage-converted T cells carried Igh gene rearrangements reminiscent of their B-cell origin. Our findings suggest that modulation of PKC-α induces hematopoietic-lineage plasticity in committed B-lineage cells by perturbing expression of critical B-lineage transcription factors, and deregulation of PKC-α activity/expression represents a potential mechanism for lineage trans-differentiation during malignancies.


B-Lymphocytes/immunology , Cell Dedifferentiation/immunology , Lymphoid Progenitor Cells/immunology , Myeloid Progenitor Cells/immunology , PAX5 Transcription Factor/immunology , Protein Kinase C-alpha/immunology , Animals , B-Lymphocytes/enzymology , Cell Dedifferentiation/genetics , Cell Line , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Female , Gene Rearrangement, B-Lymphocyte/genetics , Gene Rearrangement, B-Lymphocyte/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphoid Progenitor Cells/enzymology , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/immunology , Male , Mice , Mice, Inbred ICR , Myeloid Progenitor Cells/enzymology , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Receptors, Notch/genetics , Receptors, Notch/immunology , Receptors, Notch/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
18.
FEBS J ; 277(1): 74-80, 2010 Jan.
Article En | MEDLINE | ID: mdl-19878310

Death-associated protein kinase (DAPK) is a pro-apoptotic serine/threonine protein kinase that is dysregulated in a wide variety of cancers. The mechanism by which this occurs has largely been attributed to promoter hypermethylation, which results in gene silencing. However, recent studies indicate that DAPK expression can be detected in some cancers, but its function is still repressed, suggesting that DAPK activity can be subverted at a post-translational level in cancer cells. This review will focus on recent data describing potential mechanisms that may alter the expression, regulation or function of DAPK.


Apoptosis Regulatory Proteins/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Neoplasms/enzymology , Signal Transduction/physiology , Apoptosis Regulatory Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Death-Associated Protein Kinases , Epigenesis, Genetic , Germ-Line Mutation , Humans , Models, Biological , Neoplasms/drug therapy , Neoplasms/genetics , Polymorphism, Genetic , Protein Processing, Post-Translational
19.
Hematol Oncol ; 24(3): 134-8, 2006 Sep.
Article En | MEDLINE | ID: mdl-16841369

While advances have been made in the clinical treatment of chronic lymphocytic leukaemia (CLL) in recent years, it is still an incurable disease and therefore the identification of novel drug therapies is of paramount importance. Understanding the molecular mechanisms that govern the survival of CLL cells is fundamental in achieving this goal. A number of studies indicate that protein kinase C (PKC)- and phosphatidylinositol-3-kinase (PI3K)- mediated signalling pathways are central to CLL cell survival, and as such PKC has gained renewed interest as a potential drug target in CLL. This may be because it represents a closely-related family of ten protein kinases, which due to the redundancy that exists between isoforms offers an opportunity for the design of isoform specific inhibitors drugs that target leukaemic cells whilst showing reduced toxicity for normal cells. Indeed, PKC signalling pathways have already been considered as targets for specific anticancer drugs [1-3]. Therefore, this short review will focus on the effect of modulating PKC activity in CLL cells and explore whether targeting PKCs could represent a valid therapy for this leukaemia.


Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Signal Transduction , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Cancer Res ; 66(1): 527-34, 2006 Jan 01.
Article En | MEDLINE | ID: mdl-16397269

B-cell chronic lymphocytic leukemia (B-CLL) is characterized by the accumulation of long-lived mature B cells with the distinctive phenotype CD19(hi) CD5+ CD23+ IgM(lo), which are refractory to apoptosis. An increased level of apoptosis has been observed on treatment of human B-CLL cells with protein kinase C (PKC) inhibitors, suggesting that this family of protein kinases mediate survival signals within B-CLL cells. Therefore, to investigate the ability of individual PKC isoforms to transform developing B cells, we stably expressed plasmids encoding PKC mutants in fetal liver-derived hematopoietic progenitor cells (HPC) from wild-type mice and then cultured them in B-cell generation systems in vitro and in vivo. Surprisingly, we noted that expression of a plasmid-encoding dominant-negative PKC alpha (PKC alpha-KR) in HPCs and subsequent culture both in vitro and in vivo resulted in the generation of a population of cells that displayed an enhanced proliferative capacity over untransfected cells and phenotypically resemble human B-CLL cells. In the absence of growth factors and stroma, these B-CLL-like cells undergo cell cycle arrest and, consistent with their ability to escape growth factor withdrawal-induced apoptosis, exhibited elevated levels of Bcl-2 expression. These studies therefore identify a unique oncogenic trigger for the development of a B-CLL-like disease resulting from the subversion of PKC alpha signaling. Our findings uncover novel avenues not only for the study of the induction of leukemic B cells but also for the development of therapeutic drugs to combat PKC alpha-regulated transformation events.


Hematopoietic Stem Cells/enzymology , Leukemia, B-Cell/enzymology , Protein Kinase C-alpha/physiology , Animals , Apoptosis/physiology , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Cell Transformation, Neoplastic , Hematopoietic Stem Cells/pathology , Leukemia, B-Cell/pathology , Mice , Mice, Inbred ICR , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/biosynthesis , Protein Kinase C-alpha/genetics , Signal Transduction
...