22 research outputs found

    Role of PKC during B cell development and transformation

    Get PDF
    The objective of this thesis is to determine the role of specific PKC isotypes during B cell development and transformation. B cell generation systems were validated both in vitro and in vivo, by coculturing haematopoietic progenitor cells (HPCs) on the calvanial cell line, 0P9, or by adoptively transferring HPCs into recombinase-activating gene 1-deficient (RAG-1-/-) mice, respectively. In both cases, mature B cells were generated as determined by analysing surface B cell marker expression. Coupling of these in vitro and in vivo B cell generation systems with a retroviral gene transfer technique, plasmids-encoding PKC mutants in the retroviral vector MIEV were stably expressed in foetal liver (FL)-derived HPCs from wild type mice and cultured to assess the ability of individual PKC isoforms to modulate the development or transformation of B cells. Of note, expression of a plasmid-encoding dominant negative PKCalpha (PKCalpha-KR) in HPCs and placement in B cell generation system in vitro or in vivo resulted in the generation of a population of cells that displayed an enhanced proliferative capacity. Analysis of PKCalpha-KR-expressing cells in vitro revealed that these cells incorporated BrdU significantly more than the MIEV control, and unexpectedly upregulated cell cycle regulators, p21waf-1 and p27kip-1. Of surprise, PKCalpha-KR-expressing cells phenotypically resemble human B cell chronic lymphocytic leukaemia (CLL) cells. Expression of constitutively active PKCalpha, PKCalpha-CAT, or dominant negative PKCalpha, PKCalpha-KR in HPCs caused significant decrease in cell number. CLL is characterised by the accumulation of long-lived phenotypically mature B cells with the distinctive phenotype: CD19hi, CDS+, CD23+, IgMdim, which are deficient in apoptosis and have undergone cell cycle arrest in the G0/G1 phase. Closer analysis of PKCalpha-KR-expressing cells uncovered that these cells undergo cell cycle arrest in the absence of growth factors and stroma and consistent with their ability to escape growth factor withdrawal-induced apoptosis, exhibited elevated levels of Bcl-2 and Mcl-1 expression. Upon stimulation with IL-7, PKCalpha-KR-expressing cells showed explosive proliferation, suggesting that IL-7 is a proliferation factor for these cells. In accordance with this, IL-7R expression was upregulated in these cells, which may contribute to the increased sensitivity to IL-7. Mice injected with wildtype PKCalpha-KR-HPCs bore solid intraperitonial tumours at the injection site and the cells from both tumour and spleen showed CLL-like phenotype. Interestingly, splenocytes from these mice were cycling whereas the tumour cells were arrested at the G0/G1 stage, probably reflecting the two phases of this disease, a quiescent stage and an extensive proliferative stage, respectively. The expansion of the leukaemic cells was halted when they were cultured on 0P9-DL1, 0P9 cells with ectopic expression of Notch ligand, DL1, suggesting that Notch signalling mediates tumour suppression in CLL cells

    Generation of a poor prognostic chronic lymphocytic leukemia-like disease model: PKC subversion induces up-regulation of PKC II expression in B lymphocytes

    Get PDF
    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 PKCĪ± leads to an up-regulation of PKCĪ²II 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 PKCĪ² 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 PKCĪ²II in chronic lymphocytic leukemia-like disease progression and suggest a role for PKCĪ± subversion in creating permissive conditions for leukemogenesis

    MicroRNA-155 Protects Group 2 Innate Lymphoid Cells From Apoptosis to Promote Type-2 Immunity.

    Get PDF
    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

    MicroRNA-155 controls affinity-based selection by protecting c-MYC+ B cells from apoptosis.

    Get PDF
    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.R. Nakagawa was supported by a Marie Curie Incoming Fellowship from the European Unionā€™s Seventh Framework Programme for research, technological development, and demonstration. E. Vigorito was supported by the Medical Research Council grants G1001781 and G0700287 and by the Biotechnology and Biological Sciences Research Council. M. Meyer-Hermann was supported by the German Federal Ministry of Education and Research within the Measures for the Establishment of Systems Medicine, project SYSIMIT (BMBF eMed project SYSIMIT, FKZ: 01ZX1308B) and by the Human Frontier Science Program (RGP0033/2015)

    mTORC1-selective activation of translation elongation promotes disease progression in chronic lymphocytic leukemia

    Get PDF
    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 (PKCĪ±-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

    MicroRNA-155 Protects Group 2 Innate Lymphoid Cells From Apoptosis to Promote Type-2 Immunity

    Get PDF
    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 ā€œalarminā€ administration or Nippostrongylus brasiliensis parasitic worm infection. miR-155 is upregulated in ILC2 in response to both stimuli and miR-155āˆ’/āˆ’ 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

    The miR-155-PU.1 axis acts on Pax5 to enable efficient terminal B cell differentiation.

    Get PDF
    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

    Death-associated protein kinase (DAPK) and signal transduction: regulation in cancer

    No full text
    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
    corecore