Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Cell Death Dis ; 9(9): 901, 2018 Sep 05.
Article En | MEDLINE | ID: mdl-30185792

Since publication of this article, the authors wished to draw attention to an error in the materials section as a result of which they have been mis-cited ( https://www.nature.com/articles/s41422-018-0041-7 ). The dose of TNF given was not in fact 15 mg/kg body weight (as stated in the "mouse work" section), but 15 µg/kg body weight.

2.
Oncogene ; 36(15): 2066-2073, 2017 04.
Article En | MEDLINE | ID: mdl-27694901

Rearrangements of MYC or ABL proto-oncogenes lead to deregulated expression of key-regulators of cell cycle and cell survival, thereby constituting important drivers of blood cancer. Members of the BCL-2 family of apoptosis regulators contribute to oncogenic transformation downstream of these oncogenes, but the role of anti-apoptotic BCL2A1/A1 in transformation and drug resistance caused by deregulation of these oncogenes remains enigmatic. Here we analyzed the role of A1 in MYC as well as ABL kinase-driven blood cancer in mice, employing in vivo RNAi. We report that overexpression of either oncogene leads to a significant increase in A1 protein levels in otherwise A1-negative B cell progenitors, indicating a key role downstream of these oncogenes to secure survival during transformation. Knockdown of A1 by RNAi, however, did not impact on tumor latency in v-Abl-driven pre-B-ALL. In contrast, A1 knockdown in premalignant Eµ-MYC mice caused a significant reduction of transgenic pre-B cells without impacting on tumor latency as the emerging lymphomas escaped silencing of A1 expression. These findings identify A1 as a MYC target that can be induced prematurely during B cell development to aid expansion of otherwise cell-death-prone MYC transgenic pre-B cells. Hence, A1 should be considered as a putative drug target in MYC-driven blood cancer.


Lymphoma, B-Cell/genetics , Minor Histocompatibility Antigens/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Mice , Mice, Transgenic , Minor Histocompatibility Antigens/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis
3.
Oncogene ; 35(35): 4580-90, 2016 09 01.
Article En | MEDLINE | ID: mdl-26829051

In addition to their role in programmed cell death, caspases exert non-lethal functions in diverse developmental processes including cell differentiation or tissue remodeling. Terminal cell cycle exit and differentiation can be promoted by increased level of the CDK inhibitor p27(Kip1). Activated caspases cause proteolytic processing of p27, and we identified a novel caspase cleavage site in human p27 that removes a C-terminal fragment of 22 amino acids from the CDK inhibitor, including a phosphodegron. Thereby, caspases protect the inhibitor from SCF-Skp2-mediated degradation in S, G2 and M phases of the cell cycle. As a consequence, p27 becomes stabilized and remains an efficient nuclear inhibitor of cell cycle progression. Besides controlling cyclin/CDK kinase activity, p27 also regulates cytoskeletal dynamics, cell motility and cell invasion. Following processing by caspases, p27 fails to bind to RhoA and to inhibit its activation, and thereby abolishes the ability of p27 to stimulate cell migration and invasion. We propose that the stabilization of the CDK inhibitor and elimination of RhoA-induced cytoskeletal remodeling upon caspase processing could contribute to cell cycle exit and cytoskeletal remodeling during non-lethal caspase controlled differentiation processes.


Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Neoplasms/genetics , rhoA GTP-Binding Protein/genetics , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytoskeleton/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasms/pathology , Protein Binding , Proteolysis , S-Phase Kinase-Associated Proteins/genetics , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism
4.
Cell Death Differ ; 23(4): 628-39, 2016 Apr.
Article En | MEDLINE | ID: mdl-26450454

Bcl2 family proteins control mitochondrial apoptosis and its members exert critical cell type and differentiation stage-specific functions, acting as barriers against autoimmunity or transformation. Anti-apoptotic Bcl2a1/Bfl1/A1 is frequently deregulated in different types of blood cancers in humans but its physiological role is poorly understood as quadruplication of the Bcl2a1 gene locus in mice hampers conventional gene targeting strategies. Transgenic overexpression of A1, deletion of the A1-a paralogue or constitutive knockdown in the hematopoietic compartment of mice by RNAi suggested rate-limiting roles in lymphocyte development, granulopoiesis and mast cell activation. Here we report on the consequences of conditional knockdown of A1 protein expression using a reverse transactivator (rtTA)-driven approach that highlights a critical role for this Bcl2 family member in the maintenance of mature B-cell homeostasis. Furthermore, we define the A1/Bim (Bcl-2 interacting mediator of cell death) axis as a target of key kinases mediating B-cell receptor (BCR)-dependent survival signals, such as, spleen tyrosine kinase (Syk) and Brutons tyrosine kinase (Btk). As such, A1 represents a putative target for the treatment of B-cell-related pathologies depending on hyperactivation of BCR-emanating survival signals and loss of A1 expression accounts, in part, for the pro-apoptotic effects of Syk- or Btk inhibitors that rely on the 'BH3-only' protein Bim for cell killing.


B-Lymphocytes/metabolism , Gene Knockdown Techniques , Minor Histocompatibility Antigens/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Antigen, B-Cell/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Cell Survival/genetics , Humans , Mice , Minor Histocompatibility Antigens/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Antigen, B-Cell/genetics , Syk Kinase/genetics , Syk Kinase/metabolism
5.
Cell Death Differ ; 23(1): 41-51, 2016 Jan.
Article En | MEDLINE | ID: mdl-26045049

After cessation of lactation, involution of the mouse mammary gland proceeds in two distinct phases, a reversible and an irreversible one, which leads to the death and removal of alveolar cells. Cell death is preceded by the loss of STAT5 activity, which abrogates cell differentiation and gain of STAT3 activity. Despite early observations implicating BCL2 (B cell lymphoma 2) family proteins in this process, recent evidence suggests that STAT3-controlled cathepsin activity is most critical for cell death at the early stage of involution. Somewhat surprisingly, this cell death associates with but does not depend on the activation of pro-apoptotic effector caspases. However, transgenic overexpression of BCL2, that blocks caspase activation, delays involution while conditional deletion of BclX accelerates this process, suggesting that BCL2 family proteins are needed for the effective execution of involution. Here, we report on the transcriptional induction of multiple pro-apoptotic BCL2 family proteins of the 'BH3-only' subgroup during involution and the rate-limiting role of BIM in this process. Loss of Bim delayed epithelial cell clearance during involution after forced weaning in mice, whereas the absence of related Bmf had minor and loss of Bad or Noxa no impact on this process. Consistent with a contribution of BCL2 family proteins to the second wave of cell death during involution, loss of Bim reduced the number of apoptotic cells in this irreversible phase. Notably, the expression changes observed within the BCL2 family did not depend on STAT3 signalling, in line with its initiating role early in the process, but rather appear to result from relief of repression by STAT5. Our findings support the existence of a signalling circuitry regulating the irreversible phase of involution in mice by engaging BH3-only protein-driven mitochondrial apoptosis.


Apoptosis Regulatory Proteins/biosynthesis , Cell Death/genetics , Mammary Glands, Animal/metabolism , Membrane Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , STAT5 Transcription Factor/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspases/biosynthesis , Cell Differentiation/genetics , Female , Gene Expression Regulation, Developmental , Lactation/genetics , Lactation/metabolism , Mammary Glands, Animal/growth & development , Membrane Proteins/genetics , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , STAT5 Transcription Factor/biosynthesis
6.
Cell Death Differ ; 23(2): 242-52, 2016 Feb.
Article En | MEDLINE | ID: mdl-26138442

MicroRNAs (miRNAs) are a class of small, non-coding RNAs that posttranscriptionally regulate gene expression and thereby control most, if not all, biological processes. Aberrant miRNA expression has been linked to a variety of human diseases including cancer, but the underlying molecular mechanism often remains unclear. Here we have screened a miRNA expression library in a growth factor-dependent mouse pre-B-cell system to identify miRNAs with oncogenic activity. We show that miR-125b is sufficient to render pre-B cells growth factor independent and demonstrate that continuous expression of miR-125b is necessary to keep these cells in a transformed state. Mechanistically, we find that the expression of miR-125b protects against apoptosis induced by growth factor withdrawal, and that it blocks the differentiation of pre-B to immature B cells. In consequence, miR-125b-transformed cells maintain expression of their pre-B-cell receptor that provides signals for continuous proliferation and survival even in the absence of growth factor. Employing microarray analysis, we identified numerous targets of miR-125b, but only reconstitution of MAP3K11, a critical regulator of mitogen- and stress-activated kinase signaling, interferes with the cellular fitness of the transformed cells. Together, this indicates that MAP3K11 might function as an important tumor suppressor neutralized by oncomiR-125b in B-cell leukemia.


MAP Kinase Kinase Kinases/physiology , MicroRNAs/physiology , Precursor Cells, B-Lymphoid/physiology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Enzyme Repression , Female , Gene Expression Regulation, Leukemic , Humans , Leukemia/enzymology , Leukemia/genetics , Leukemia, B-Cell/enzymology , Leukemia, B-Cell/genetics , Mice , Mice, Knockout , Neoplasm Transplantation , RNA Interference , Tumor Suppressor Proteins/physiology , Mitogen-Activated Protein Kinase Kinase Kinase 11
7.
Analyst ; 140(18): 6363-72, 2015 Sep 21.
Article En | MEDLINE | ID: mdl-26236782

Mid-infrared (MIR) microscopy imaging is a vibrational spectroscopic technique that uses infrared radiation to image molecules of interest in thin tissue sections. A major advantage of this technology is the acquisition of local molecular expression profiles, while maintaining the topographic integrity of the tissue. Therefore, this technology has become an essential tool for the detection and characterization of the molecular components of many biological processes. Using this method, it is possible to investigate the spatial distribution of proteins and small molecules within biological systems by in situ analysis. In this study, we have evaluated the potential of mid-infrared microscopy imaging to study biochemical changes which distinguish between reactive lymphadenopathy and cancer in genetically modified mice with different phenotypes. We were able to demonstrate that MIR microscopy imaging and multivariate image analyses of different mouse genotypes correlated well with the morphological tissue features derived from HE staining. Using principal component analyses, we were also able to distinguish spectral clusters from different phenotype samples, particularly from reactive lymphadenopathy (follicular hyperplasia) and cancer (follicular lymphoma).


Infrared Rays , Lymphoma, Follicular/diagnosis , Microscopy/methods , Spleen/pathology , Animals , Diagnosis, Differential , Hyperplasia/diagnosis , Lymphoma, Follicular/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
8.
Cell Death Differ ; 22(11): 1803-11, 2015 Nov.
Article En | MEDLINE | ID: mdl-25857265

The receptor-interacting protein-associated ICH-1/CED-3 homologous protein with a death domain (RAIDD/CRADD) functions as a dual adaptor and is a constituent of different multi-protein complexes implicated in the regulation of inflammation and cell death. Within the PIDDosome complex, RAIDD connects the cell death-related protease, Caspase-2, with the p53-induced protein with a death domain 1 (PIDD1). As such, RAIDD has been implicated in DNA-damage-induced apoptosis as well as in tumorigenesis. As loss of Caspase-2 leads to an acceleration of tumor onset in the Eµ-Myc mouse lymphoma model, whereas loss of Pidd1 actually delays onset of this disease, we set out to interrogate the role of Raidd in cancer in more detail. Our data obtained analyzing Eµ-Myc/Raidd(-/-) mice indicate that Raidd is unable to protect from c-Myc-driven lymphomagenesis. Similarly, we failed to observe a modulatory effect of Raidd deficiency on DNA-damage-driven cancer. The role of Caspase-2 as a tumor suppressor and that of Pidd1 as a tumor promoter can therefore be uncoupled from their ability to interact with the Raidd scaffold, pointing toward the existence of alternative signaling modules engaging these two proteins in this context.


CRADD Signaling Adaptor Protein/metabolism , Caspase 2/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , CRADD Signaling Adaptor Protein/genetics , Caspase 2/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , DNA Damage/genetics , DNA Damage/radiation effects , Death Domain Receptor Signaling Adaptor Proteins/genetics , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Mice , Proto-Oncogene Proteins c-myc/genetics
9.
Cell Death Differ ; 22(9): 1477-88, 2015 Sep.
Article En | MEDLINE | ID: mdl-25698446

Terminal differentiation of B cells depends on two interconnected survival pathways, elicited by the B-cell receptor (BCR) and the BAFF receptor (BAFF-R), respectively. Loss of either signaling pathway arrests B-cell development. Although BCR-dependent survival depends mainly on the activation of the v-AKT murine thymoma viral oncogene homolog 1 (AKT)/PI3-kinase network, BAFF/BAFF-R-mediated survival engages non-canonical NF-κB signaling as well as MAPK/extracellular-signal regulated kinase and AKT/PI3-kinase modules to allow proper B-cell development. Plasma cell survival, however, is independent of BAFF-R and regulated by APRIL that signals NF-κB activation via alternative receptors, that is, transmembrane activator and CAML interactor (TACI) or B-cell maturation (BCMA). All these complex signaling events are believed to secure survival by increased expression of anti-apoptotic B-cell lymphoma 2 (Bcl2) family proteins in developing and mature B cells. Curiously, how lack of BAFF- or APRIL-mediated signaling triggers B-cell apoptosis remains largely unexplored. Here, we show that two pro-apoptotic members of the 'Bcl2 homology domain 3-only' subgroup of the Bcl2 family, Bcl2 interacting mediator of cell death (Bim) and Bcl2 modifying factor (Bmf), mediate apoptosis in the context of TACI-Ig overexpression that effectively neutralizes BAFF as well as APRIL. Surprisingly, although Bcl2 overexpression triggers B-cell hyperplasia exceeding the one observed in Bim(-/-)Bmf(-/-) mice, Bcl2 transgenic B cells remain susceptible to the effects of TACI-Ig expression in vivo, leading to ameliorated pathology in Vav-Bcl2 transgenic mice. Together, our findings shed new light on the molecular machinery restricting B-cell survival during development, normal homeostasis and under pathological conditions. Our data further suggest that Bcl2 antagonists might improve the potency of BAFF/APRIL-depletion strategies in B-cell-driven pathologies.


Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , B-Lymphocytes/physiology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Apoptosis/physiology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Bcl-2-Like Protein 11 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/metabolism , Recombinant Fusion Proteins/genetics , Signal Transduction
10.
Cell Death Dis ; 6: e1611, 2015 Jan 22.
Article En | MEDLINE | ID: mdl-25611386

Tumor necrosis factor (TNF) is a key signaling molecule orchestrating immune and inflammatory responses and possesses the capacity to trigger apoptotic as well as necroptotic cell death. Apoptotic cell death elicited by TNF has been demonstrated to engage pro-apoptotic Bcl-2 family proteins, most prominently the BH3-only protein Bid, a key substrate of caspase-8, the key effector protease downstream of TNF receptor I. Most recently, the BH3 domain-containing protein Bad (Bcl-2-antagonist of cell death) has been shown to be rate limiting for TNF-mediated cell death, suggesting possible synergy with Bid, but genetic analyses presented here demonstrate that it is dispensable for this process.


Tumor Necrosis Factor-alpha/pharmacology , bcl-Associated Death Protein/metabolism , Animals , Cell Death/drug effects , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Hepatitis/pathology , Hepatitis/prevention & control , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Mice, Inbred C57BL , Pyridines/pharmacology , Thymocytes/drug effects , Thymocytes/metabolism , bcl-Associated Death Protein/deficiency
11.
Cell Death Differ ; 22(7): 1144-57, 2015 Jul.
Article En | MEDLINE | ID: mdl-25501596

Erythropoiesis is a tightly regulated process in which multipotential hematopoietic stem cells produce mature red blood cells. Here we show that deletion of poly(ADP-ribose) polymerase-2 (PARP-2) in mice leads to chronic anemia at steady state, despite increased erythropoietin plasma levels, a phenomenon not observed in mice lacking PARP-1. Loss of PARP-2 causes shortened lifespan of erythrocytes and impaired differentiation of erythroid progenitors. In erythroblasts, PARP-2 deficiency triggers replicative stress, as indicated by the presence of micronuclei, the accumulation of γ-H2AX (phospho-histone H2AX) in S-phase cells and constitutive CHK1 and replication protein A phosphorylation. Transcriptome analyses revealed the activation of the p53-dependent DNA-damage response pathways in PARP-2-deficient cells, culminating in the upregulation of cell-cycle and cell death regulators, concomitant with G2/M arrest and apoptosis. Strikingly, while loss of the proapoptotic p53 target gene Puma restored hematocrit levels in the PARP-2-deficient mice, loss of the cell-cycle regulator and CDK inhibitor p21 leads to perinatal death by exacerbating impaired fetal liver erythropoiesis in PARP-2-deficient embryos. Although the anemia displayed by PARP-2-deficient mice is compatible with life, mice die rapidly when exposed to stress-induced enhanced hemolysis. Our results pinpoint an essential role for PARP-2 in erythropoiesis by limiting replicative stress that becomes essential in the absence of p21 and in the context of enhanced hemolysis, highlighting the potential effect that might arise from the design and use of PARP inhibitors that specifically inactivate PARP proteins.


DNA Replication , Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , Poly(ADP-ribose) Polymerases/genetics , Stress, Physiological/genetics , Animals , Apoptosis , Erythropoiesis/genetics , G2 Phase Cell Cycle Checkpoints , Gene Deletion , Histones/metabolism , Mice
12.
Oncogene ; 33(44): 5221-4, 2014 Oct 30.
Article En | MEDLINE | ID: mdl-24213575

The antiapoptotic BCL-2 protein MCL-1, which opposes mitochondrial outer membrane permeabilization, was shown to have a crucial role in the survival of hematopoietic cells. We have previously shown that, upon loss of phosphatidylinositol 3-kinase signaling, S159 of MCL-1 is phosphorylated by glycogen synthase kinase-3 (GSK-3), earmarking MCL-1 for enhanced ubiquitylation and degradation. In this study, we introduced MCL-1(wt) or the phosphorylation-deficient mutant MCL-1(S159A) in mouse BM cells, followed by adoptive transfer to recipient mice. Mice expressing MCL-1(S159A) exhibited significantly elevated white blood cell and lymphocyte counts, whereas no effect was observed on the distribution of T and B lymphocyte subsets or the numbers of monocytes, red blood cells or platelets. Expression of MCL-1(S159A) in Eµ-Myc transgenic bone marrow significantly accelerated the onset of disease, and these mice displayed increased spleen weights compared with Eµ-Myc/MCL-1(wt) mice. Our data demonstrate that the absence of MCL-1 S159 phosphorylation provides a survival advantage for hematopoietic cells in vivo and facilitates oncogenesis.


Leukocytes/metabolism , Lymphoma/pathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Bone Marrow Transplantation , Cell Survival , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukocytes/pathology , Lymph Nodes/cytology , Lymphoma/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Phosphorylation , Spleen/cytology
13.
Cell Death Dis ; 4: e942, 2013 Dec 05.
Article En | MEDLINE | ID: mdl-24309929

Much effort has been put in the discovery of ways to selectively kill p53-deficient tumor cells and targeting cell cycle checkpoint pathways has revealed promising candidates. Studies in zebrafish and human cell lines suggested that the DNA damage response kinase, checkpoint kinase 1 (Chk1), not only regulates onset of mitosis but also cell death in response to DNA damage in the absence of p53. This effect reportedly relies on ataxia telangiectasia mutated (ATM)-dependent and PIDDosome-mediated activation of Caspase-2. However, we show that genetic ablation of PIDDosome components in mice does not affect cell death in response to γ-irradiation. Furthermore, Chk1 inhibition largely failed to sensitize normal and malignant cells from p53(-/-) mice toward DNA damaging agents, and p53 status did not affect the death-inducing activity of DNA damage after Chk1 inhibition in human cancer cells. These observations argue against cross-species conservation of a Chk1-controlled cell survival pathway demanding further investigation of the molecular machinery responsible for cell death elicited by forced mitotic entry in the presence of DNA damage in different cell types and model organisms.


Caspase 2/metabolism , DNA Damage/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Caspase 2/genetics , Cell Cycle/genetics , Cell Cycle/physiology , DNA Damage/genetics , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Immunoblotting , Mice , Mice, Inbred C57BL , Mitosis/genetics , Mitosis/physiology , Tumor Suppressor Protein p53/genetics
17.
Oncogene ; 32(5): 621-30, 2013 Jan 31.
Article En | MEDLINE | ID: mdl-22430207

Proapoptotic Bcl-2 family members of the Bcl-2 homology (BH)3-only subgroup are critical for the establishment and maintenance of tissue homeostasis and can mediate apoptotic cell death in response to developmental cues or exogenously induced forms of cell stress. On the basis of the biochemical experiments as well as genetic studies in mice, the BH3-only proteins Bad and Bmf have been implicated in different proapoptotic events such as those triggered by glucose- or trophic factor-deprivation, glucocorticoids, or histone deacetylase inhibition, as well as suppression of B-cell lymphomagenesis upon aberrant expression of c-Myc. To address possible redundancies in cell death regulation and tumor suppression, we generated compound mutant mice lacking both genes. Our studies revealed lack of redundancy in most paradigms of lymphocyte apoptosis tested in tissue culture. Only spontaneous cell death of thymocytes kept in low glucose or that of pre-B cells deprived of cytokines was significantly delayed when both genes were lacking. Of note, despite these minor apoptosis defects we observed compromised lymphocyte homeostasis in vivo that affected mainly the B-cell lineage. Long-term follow-up revealed significantly reduced latency to spontaneous tumor formation in aged mice when both genes were lacking. Together our study suggests that Bad and Bmf co-regulate lymphocyte homeostasis and limit spontaneous transformation by mechanisms that may not exclusively be linked to the induction of lymphocyte apoptosis.


Adaptor Proteins, Signal Transducing/metabolism , Lymphocytes/metabolism , bcl-Associated Death Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Lymphocytes/metabolism , Cell Death , Cell Transformation, Neoplastic , Hematopoiesis , Homeostasis , Mice , Mice, Knockout , bcl-Associated Death Protein/genetics
18.
Cell Death Differ ; 20(4): 546-57, 2013 Apr.
Article En | MEDLINE | ID: mdl-23238565

Activation of NF-κB (nuclear factor of kappa light chain gene enhancer in B cells) in response to DNA damage is considered to contribute to repair of genetic lesions, increased cell survival and cytokine release. The molecular mechanisms orchestrating this cytoplasmic event involve core components of the nuclear DNA damage response machinery, including ATM-kinase (ataxia telangiectasia mutated kinase) and PARP-1 (poly (ADP-ribose) polymerase 1). The physiological consequences of defective NF-κB activation in this context, however, remain poorly investigated. Here we report on the role of the 'p53-induced protein with a death domain', PIDD, which appears rate limiting in this process, as is PARP-1. Despite impaired NF-κB activation, DNA damage did not increase cell death or reduce clonal survival of various cell types lacking PIDD, such as mouse embryonic fibroblasts or stem and progenitor cells of the hematopoietic system. Furthermore, lymphomagenesis induced by γ-irradiation (IR) was unaffected by deficiency for PIDD or PARP-1, indicating that loss of DNA damage-triggered NF-κB signalling does not affect IR-driven tumorigenesis. However, loss of either gene compromised cytokine release after acute IR injury. Hence, we propose that NF-κB's most notable function after DNA damage in primary cells is related to the release of cytokines, thereby contributing to sterile inflammation.


Cytokines/metabolism , DNA Damage , Death Domain Receptor Signaling Adaptor Proteins/metabolism , NF-kappa B/metabolism , Animals , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Cell Transformation, Neoplastic/radiation effects , Cells, Cultured , DNA Damage/radiation effects , DNA-Binding Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Radiation, Ionizing , Signal Transduction , Transcription Factor RelA/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
20.
Cell Death Differ ; 19(8): 1277-87, 2012 Aug.
Article En | MEDLINE | ID: mdl-22322859

Transforming growth factor beta (TGF-ß)- and Interleukin-2 (IL-2)-mediated signaling enables the generation and expansion of induced regulatory T (iTreg) cells that carry high hopes for the treatment of chronic inflammatory and autoimmune diseases. Knowledge about factors stabilizing their lineage commitment and lifespan, however, is limited. Here, we investigated the behavior of iTreg cells, derived from apoptosis-defective mouse mutants, during activated cell autonomous cell death, triggered by cytokine-deprivation, or activation-induced cell death (AICD) after restimulation of the T-cell receptor, and compared these responses with those of effector T cells. We observed that iTreg cells were much more sensitive to IL-2-deprivation but poorly susceptible to AICD. In fact, when apoptosis was compromised, T-cell receptor (TCR)-religation resulted in methylation-independent, ERK- and PI3K/mTOR-mediated loss of Foxp3 expression, impaired suppressive capacity and effector cytokine production. Although iTreg cells prevented colitis induction they rapidly lost Foxp3-GFP expression and gained ability to produce effector cytokines thereby imposing Th1 cell fate on resident effector cells. Surprisingly, iTreg cell conversion itself was limited by TGF-ß-mediated Bim/Bcl2L11-dependent apoptosis. Hence, the very same cytokine that drives the generation of iTreg cells can trigger their demise. Our results provide novel insights in iTreg cell biology that will assist optimization of iTreg-based therapy.


Interleukin-2/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Apoptosis/immunology , Cell Survival/immunology , Colitis/immunology , Fas Ligand Protein/immunology , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/biosynthesis , Peptide Fragments/immunology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , fas Receptor/deficiency , fas Receptor/immunology
...