Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Cell Death Dis ; 15(4): 251, 2024 Apr 08.
Article En | MEDLINE | ID: mdl-38589365

Cell death mediated by genetically defined signaling pathways influences the health and dynamics of all tissues, however the tissue specificity of cell death pathways and the relationships between these pathways and human disease are not well understood. We analyzed the expression profiles of an array of 44 cell death genes involved in apoptosis, necroptosis, and pyroptosis cell death pathways across 49 human tissues from GTEx, to elucidate the landscape of cell death gene expression across human tissues, and the relationship between tissue-specific genetically determined expression and the human phenome. We uncovered unique cell death gene expression profiles across tissue types, suggesting there are physiologically distinct cell death programs in different tissues. Using summary statistics-based transcriptome wide association studies (TWAS) on human traits in the UK Biobank (n ~ 500,000), we evaluated 513 traits encompassing ICD-10 defined diagnoses and laboratory-derived traits. Our analysis revealed hundreds of significant (FDR < 0.05) associations between genetically regulated cell death gene expression and an array of human phenotypes encompassing both clinical diagnoses and hematologic parameters, which were independently validated in another large-scale DNA biobank (BioVU) at Vanderbilt University Medical Center (n = 94,474) with matching phenotypes. Cell death genes were highly enriched for significant associations with blood traits versus non-cell-death genes, with apoptosis-associated genes enriched for leukocyte and platelet traits. Our findings are also concordant with independently published studies (e.g. associations between BCL2L11/BIM expression and platelet & lymphocyte counts). Overall, these results suggest that cell death genes play distinct roles in their contribution to human phenotypes, and that cell death genes influence a diverse array of human traits.


Genome-Wide Association Study , Transcriptome , Humans , Genome-Wide Association Study/methods , Phenotype , Cell Death/genetics , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease
2.
Exp Hematol ; 103: 60-72.e5, 2021 11.
Article En | MEDLINE | ID: mdl-34563605

Myelodysplastic syndrome (MDS) is characterized by persistent cytopenias and evidence of morphologic dysplasia in the bone marrow (BM). Excessive hematopoietic programmed cell death (PCD) and inflammation have been observed in the bone marrow of patients with MDS, and are thought to play a significant role in the pathogenesis of the disease. Necroptosis is a major pathway of PCD that incites inflammation; however, the role of necroptosis in human MDS has not been extensively investigated. To assess PCD status in newly diagnosed MDS, we performed immunofluorescence staining with computational image analysis of formalin-fixed, paraffin-embedded BM core biopsies using cleaved caspase-3 (apoptosis marker) and necroptosis markers (receptor-interacting serine/threonine-protein kinase 1 [RIPK1], phospho-mixed lineage kinase domain-like protein [pMLKL]). Patients with MDS, but not controls without MDS or patients with de novo acute myeloid leukemia, had significantly increased expression of RIPK1 and pMLKL but not cleaved caspase-3, which was most evident in morphologically low-grade MDS (<5% BM blasts) and in MDS with low International Prognostic Scoring System risk score. RIPK1 expression highly correlated with the distribution of CD71+ erythroid precursors but not with CD34+ blast cells. We found that necroptosis is upregulated in early/low-grade MDS relative to control participants, warranting further study to define the role of necroptosis in the pathogenesis of MDS and as a potential biomarker for the diagnosis of low-grade MDS.


Myelodysplastic Syndromes/pathology , Necroptosis , Adult , Aged , Female , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/analysis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Up-Regulation
3.
Blood ; 133(2): 107-120, 2019 01 10.
Article En | MEDLINE | ID: mdl-30413413

Hematopoiesis is a dynamic system that requires balanced cell division, differentiation, and death. The 2 major modes of programmed cell death, apoptosis and necroptosis, share molecular machinery but diverge in outcome with important implications for the microenvironment; apoptotic cells are removed in an immune silent process, whereas necroptotic cells leak cellular contents that incite inflammation. Given the importance of cytokine-directed cues for hematopoietic cell survival and differentiation, the impact on hematopoietic homeostasis of biasing cell death fate to necroptosis is substantial and poorly understood. Here, we present a mouse model with increased bone marrow necroptosis. Deletion of the proapoptotic Bcl-2 family members Bax and Bak inhibits bone marrow apoptosis. Further deletion of the BH3-only member Bid (to generate Vav CreBaxBakBid triple-knockout [TKO] mice) leads to unrestrained bone marrow necroptosis driven by increased Rip1 kinase (Ripk1). TKO mice display loss of progenitor cells, leading to increased cytokine production and increased stem cell proliferation and exhaustion and culminating in bone marrow failure. Genetically restoring Ripk1 to wild-type levels restores peripheral red cell counts as well as normal cytokine production. TKO bone marrow is hypercellular with abnormal differentiation, resembling the human disorder myelodysplastic syndrome (MDS), and we demonstrate increased necroptosis in MDS bone marrow. Finally, we show that Bid impacts necroptotic signaling through modulation of caspase-8-mediated Ripk1 degradation. Thus, we demonstrate that dysregulated necroptosis in hematopoiesis promotes bone marrow progenitor cell death that incites inflammation, impairs hematopoietic stem cells, and recapitulates the salient features of the bone marrow failure disorder MDS.


Bone Marrow Diseases/etiology , Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Inflammation/etiology , Myelodysplastic Syndromes/etiology , Necrosis , Animals , BH3 Interacting Domain Death Agonist Protein/physiology , Bone Marrow/metabolism , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/pathology , Cells, Cultured , Cytokines/metabolism , Hematopoietic Stem Cells/metabolism , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology
4.
Elife ; 72018 10 03.
Article En | MEDLINE | ID: mdl-30281024

Bcl-2 family proteins reorganize mitochondrial membranes during apoptosis, to form pores and rearrange cristae. In vitro and in vivo analysis integrated with human genetics reveals a novel homeostatic mitochondrial function for Bcl-2 family protein Bid. Loss of full-length Bid results in apoptosis-independent, irregular cristae with decreased respiration. Bid-/- mice display stress-induced myocardial dysfunction and damage. A gene-based approach applied to a biobank, validated in two independent GWAS studies, reveals that decreased genetically determined BID expression associates with myocardial infarction (MI) susceptibility. Patients in the bottom 5% of the expression distribution exhibit >4 fold increased MI risk. Carrier status with nonsynonymous variation in Bid's membrane binding domain, BidM148T, associates with MI predisposition. Furthermore, Bid but not BidM148T associates with Mcl-1Matrix, previously implicated in cristae stability; decreased MCL-1 expression associates with MI. Our results identify a role for Bid in homeostatic mitochondrial cristae reorganization, that we link to human cardiac disease.


BH3 Interacting Domain Death Agonist Protein/metabolism , Genomics , Heart Diseases/genetics , Heart Diseases/prevention & control , Mitochondria/metabolism , Mitochondria/ultrastructure , Animals , Apoptosis , BH3 Interacting Domain Death Agonist Protein/chemistry , Beclin-1/metabolism , Cell Respiration , Fibrosis , Gene Expression Regulation , Genome-Wide Association Study , Heart Diseases/pathology , Heart Ventricles/pathology , Humans , Mice, Inbred C57BL , Mitochondrial Proton-Translocating ATPases , Mutation/genetics , Myeloid Progenitor Cells/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Polymorphism, Single Nucleotide/genetics , Protein Multimerization , Protein Structure, Secondary , Protein Subunits/metabolism , Reactive Oxygen Species/metabolism , Reproducibility of Results , Up-Regulation
5.
Mitochondrion ; 28: 88-95, 2016 05.
Article En | MEDLINE | ID: mdl-27085476

Modifications of cardiolipin (CL) levels or compositions are associated with changes in mitochondrial function in a wide range of pathologies. We have made the discovery that acetaminophen remodels CL fatty acids composition from tetralinoleoyl to linoleoyltrioleoyl-CL, a remodeling that is associated with decreased mitochondrial respiration. Our data show that CL remodeling causes a shift in electron entry from complex II to the ß-oxidation electron transfer flavoprotein quinone oxidoreductase (ETF/QOR) pathway. These data demonstrate that electron entry in the respiratory chain is regulated by CL fatty acid composition and provide proof-of-concept that pharmacological intervention can be used to modify CL composition.


Acetaminophen/metabolism , Antipyretics/metabolism , Cardiolipins/metabolism , Electron Transport , Fatty Acids/metabolism , Mitochondria/drug effects , Cell Respiration/drug effects , Cells, Cultured , Humans , Mitochondria/chemistry , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Quinones/analysis
6.
Blood ; 126(9): 1057-68, 2015 Aug 27.
Article En | MEDLINE | ID: mdl-26077396

Much-needed attention has been given of late to diseases specifically associated with an expanding elderly population. Myelodysplastic syndrome (MDS), a hematopoietic stem cell-based blood disease, is one of these. The lack of clear understanding of the molecular mechanisms underlying the pathogenesis of this disease has hampered the development of efficacious therapies, especially in the presence of comorbidities. Mouse models could potentially provide new insights into this disease, although primary human MDS cells grow poorly in xenografted mice. This makes genetically engineered murine models a more attractive proposition, although this approach is not without complications. In particular, it is unclear if or how myelodysplasia (abnormal blood cell morphology), a key MDS feature in humans, presents in murine cells. Here, we evaluate the histopathologic features of wild-type mice and 23 mouse models with verified myelodysplasia. We find that certain features indicative of myelodysplasia in humans, such as Howell-Jolly bodies and low neutrophilic granularity, are commonplace in healthy mice, whereas other features are similarly abnormal in humans and mice. Quantitative hematopoietic parameters, such as blood cell counts, are required to distinguish between MDS and related diseases. We provide data that mouse models of MDS can be genetically engineered and faithfully recapitulate human disease.


Disease Models, Animal , Genetic Engineering , Hematopoietic Stem Cells/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Animals , Hematopoiesis , Humans , Mice , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/diagnosis
7.
Aging Cell ; 14(3): 382-90, 2015 Jun.
Article En | MEDLINE | ID: mdl-25727904

In aged mice, new B-cell development is diminished and the antibody repertoire becomes more autoreactive. Our studies suggest that (i) apoptosis contributes to reduced B lymphopoiesis in old age and preferentially eliminates those B-cell precursors with higher levels of the surrogate light chain (SLC) proteins (λ5/VpreB) and (ii) λ5(low) B-cell precursors generate new B cells which show increased reactivity to the self-antigen/bacterial antigen phosphorylcholine (PC). Pro-B cells in old bone marrow as well as pro-B cells from young adult λ5-deficient mice are resistant to cytokine-induced apoptosis (TNFα; TGFß), indicating that low λ5 expression in pro-B cells is sufficient to cause increased survival. Transfer of TNFα-producing 'age-associated B cells' (ABC; CD21/35(-) CD23(-)) or follicular (FO) B cells from aged mice into RAG-2 KO recipients led to preferential loss of λ5(high) pro-B cells, but retention of λ5(low), apoptosis-resistant pro-B cells. In old mice, there is increased reactivity to PC in both immature bone marrow B cells and mature splenic FO B cells. In young mice, absence of λ5 expression led to a similar increase in PC reactivity among bone marrow and splenic B cells. We propose that in old age, increased apoptosis, mediated in part by TNFα-producing B cells, results in preferential loss of SLC(high) pro-B cells within the bone marrow. Further B-cell development then occurs via an 'SLC(low)' pathway that not only impairs B-cell generation, but promotes autoreactivity within the naïve antibody repertoires in the bone marrow and periphery.


Apoptosis/immunology , Cell Cycle Checkpoints/immunology , Immunoglobulin Light Chains, Surrogate/immunology , Lymphocyte Activation/immunology , Phosphorylcholine/immunology , Precursor Cells, B-Lymphoid/metabolism , Animals , Cell Differentiation/physiology , Mice, Inbred C57BL
8.
Gut ; 62(10): 1446-55, 2013 Oct.
Article En | MEDLINE | ID: mdl-22833394

OBJECTIVE: The myeloid translocation genes (MTGs) are transcriptional corepressors with both Mtg8(-/-) and Mtgr1(-/-) mice showing developmental and/or differentiation defects in the intestine. We sought to determine the role of MTG16 in intestinal integrity. METHODS: Baseline and stress induced colonic phenotypes were examined in Mtg16(-/-) mice. To unmask phenotypes, we treated Mtg16(-/-) mice with dextran sodium sulphate (DSS) or infected them with Citrobacter rodentium and the colons were examined for ulceration and for changes in proliferation, apoptosis and inflammation. RESULTS: Mtg16(-/-) mice have altered immune subsets, suggesting priming towards Th1 responses. Mtg16(-/-) mice developed increased weight loss, diarrhoea, mortality and histological colitis and there were increased innate (Gr1(+), F4/80(+), CD11c(+) and MHCII(+); CD11c(+)) and Th1 adaptive (CD4) immune cells in Mtg16(-/-) colons after DSS treatment. Additionally, there was increased apoptosis and a compensatory increased proliferation in Mtg16(-/-) colons. Compared with wild-type mice, Mtg16(-/-) mice exhibited increased colonic CD4;IFN-γ cells in vehicle-treated and DSS-treated mice. Adoptive transfer of wild-type marrow into Mtg16(-/-) recipients did not rescue the Mtg16(-/-) injury phenotype. Isolated colonic epithelial cells from DSS-treated Mtg16(-/-) mice exhibited increased KC (Cxcl1) mRNA expression when compared with wild-type mice. Mtg16(-/-) mice infected with C rodentium had more severe colitis and greater bacterial colonisation. Last, MTG16 mRNA levels were reduced in human ulcerative colitis versus normal colon tissues. CONCLUSIONS: These observations indicate that MTG16 is critical for colonocyte survival and regeneration in response to intestinal injury and provide evidence that this transcriptional corepressor regulates inflammatory recruitment in response to injury.


Colitis/pathology , Nuclear Proteins/physiology , Transcription Factors/physiology , Adaptive Immunity , Adoptive Transfer , Animals , Bone Transplantation , Cell Proliferation , Colitis/chemically induced , Colitis/immunology , Colitis/physiopathology , Colitis, Ulcerative/metabolism , Colon/immunology , Dextran Sulfate , Enterocytes/pathology , Female , Humans , Immunity, Innate , Immunophenotyping , Intestinal Absorption/physiology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Male , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Permeability , Repressor Proteins , Th1 Cells/immunology , Transcription Factors/deficiency , Transcription Factors/genetics
9.
Biochem Biophys Res Commun ; 423(2): 224-8, 2012 Jun 29.
Article En | MEDLINE | ID: mdl-22634010

Cytochrome (cyt) c can uncouple from the respiratory chain following mitochondrial stress and catalyze lipid peroxidation. Accumulating evidence shows that this phenomenon impairs mitochondrial respiratory function and also initiates the apoptotic cascade. Therefore, under certain conditions a pharmacological approach that can inhibit cyt c catalyzed lipid peroxidation may be beneficial. We recently showed that acetaminophen (ApAP) at normal pharmacologic concentrations can prevent hemoprotein-catalyzed lipid peroxidation in vitro and in vivo by reducing ferryl heme to its ferric state. We report here, for the first time, that ApAP inhibits cytochrome c-catalyzed oxidation of unsaturated free fatty acids and also the mitochondrial phospholipid, cardiolipin. Using isolated mitochondria, we also showed that ApAP inhibits cardiolipin oxidation induced by the pro-apoptotic protein, tBid. We found that the IC(50) of the inhibition of cardiolipin oxidation by ApAP is similar in both intact isolated mitochondria and cardiolipin liposomes, suggesting that ApAP penetrates well into the mitochondria. Together with our previous results, the findings presented herein suggest that ApAP is a pleiotropic inhibitor of peroxidase catalyzed lipid peroxidation. Our study also provides a potentially novel pharmacological approach for inhibiting the cascade of events that can result from redox cycling of cyt c.


Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Cytochromes c/metabolism , Fatty Acids, Unsaturated/metabolism , Lipid Peroxidation/drug effects , Arachidonic Acid/metabolism , Cardiolipins/metabolism , Catalysis , Humans , Mitochondria/drug effects , Mitochondria/enzymology , Oxidation-Reduction/drug effects
10.
Mol Cell Biol ; 31(21): 4298-309, 2011 Nov.
Article En | MEDLINE | ID: mdl-21859891

Proapoptotic BH3-interacting death domain agonist (BID) regulates apoptosis and the DNA damage response. Following replicative stress, BID associates with proteins of the DNA damage sensor complex, including replication protein A (RPA), ataxia telangiectasia and Rad3 related (ATR), and ATR-interacting protein (ATRIP), and facilitates an efficient DNA damage response. We have found that BID stimulates the association of RPA with components of the DNA damage sensor complex through interaction with the basic cleft of the N-terminal domain of the RPA70 subunit. Disruption of the BID-RPA interaction impairs the association of ATR-ATRIP with chromatin as well as ATR function, as measured by CHK1 activation and recovery of DNA replication following hydroxyurea (HU). We further demonstrate that the association of BID with RPA stimulates the association of ATR-ATRIP to the DNA damage sensor complex. We propose a model in which BID associates with RPA and stimulates the recruitment and/or stabilization of ATR-ATRIP to the DNA damage sensor complex.


BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/genetics , Base Sequence , Cell Cycle Proteins/chemistry , Cell Line , DNA Damage , DNA Replication , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Protein A/chemistry , Signal Transduction , Stress, Physiological
11.
PLoS Pathog ; 6: e1000980, 2010 Jul 01.
Article En | MEDLINE | ID: mdl-20617182

Reovirus infection leads to apoptosis in both cultured cells and the murine central nervous system (CNS). NF-kappaB-driven transcription of proapoptotic cellular genes is required for the effector phase of the apoptotic response. Although both extrinsic death-receptor signaling pathways and intrinsic pathways involving mitochondrial injury are implicated in reovirus-induced apoptosis, mechanisms by which either of these pathways are activated and their relationship to NF-kappaB signaling following reovirus infection are unknown. The proapoptotic Bcl-2 family member, Bid, is activated by proteolytic cleavage following reovirus infection. To understand how reovirus integrates host signaling circuits to induce apoptosis, we examined proapoptotic signaling following infection of Bid-deficient cells. Although reovirus growth was not affected by the absence of Bid, cells lacking Bid failed to undergo apoptosis. Furthermore, we found that NF-kappaB activation is required for Bid cleavage and subsequent proapoptotic signaling. To examine the functional significance of Bid-dependent apoptosis in reovirus disease, we monitored fatal encephalitis caused by reovirus in the presence and absence of Bid. Survival of Bid-deficient mice was significantly enhanced in comparison to wild-type mice following either peroral or intracranial inoculation of reovirus. Decreased reovirus virulence in Bid-null mice was accompanied by a reduction in viral yield. These findings define a role for NF-kappaB-dependent cleavage of Bid in the cell death program initiated by viral infection and link Bid to viral virulence.


BH3 Interacting Domain Death Agonist Protein/physiology , Encephalitis, Viral/etiology , Reoviridae Infections/virology , Reoviridae/pathogenicity , Animals , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/deficiency , Fas-Associated Death Domain Protein/physiology , Fibroblasts/virology , Humans , L Cells , Mice , NF-kappa B/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , Signal Transduction/drug effects , Virus Replication/drug effects
12.
Methods Enzymol ; 442: 231-50, 2008.
Article En | MEDLINE | ID: mdl-18662573

The BCL-2 family of apoptotic proteins encompasses key regulators proximal to irreversible cell damage. BID, a "BH3-only" proapoptotic family member, plays a critical role in connecting death signals through surface death receptors such as Fas and tumor necrosis factor-alpha to the core apoptotic pathway at the mitochondria. BID is activated downstream of death receptors by caspase-8 cleavage and N-myristoylation to target mitochondria where it activates BAX, BAK, and the downstream apoptotic pathway. In addition to its role in apoptosis, a role has been uncovered for BID in regulating the DNA damage-induced intra-S phase checkpoint that does not require its death-promoting BH3 domain. Following DNA damage, BID is found in the nucleus where it is phosphorylated by ATM and plays a role in the intra-S phase checkpoint. This checkpoint role is dependent on ATM-mediated phosphorylation at position 78. Thus, BID has two distinct and separable functions: an apoptotic function mediated by caspase cleavage and its BH3 domain and a cell cycle/DNA repair function mediated by phosphorylation by the DNA damage kinase ATM. Studies indicate that the pro-death activity of BID is inhibited by phosphorylation. Taken together, these findings suggest interaction between the two functions of BID. An area of intense research pursuit is determining what dictates how cells respond to DNA damage. Some cells arrest the cell cycle, whereas others undergo apoptosis. We hypothesize that BID acts at the interface between the DNA damage response and apoptosis, in position to signal a cell either to undergo cell cycle arrest and initiate DNA repair or to undergo apoptosis. This chapter describes the techniques used to characterize the role of BID in apoptosis and the DNA damage response.


Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/physiology , DNA Damage , Animals , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Cells, Cultured , DNA Repair/genetics , DNA Repair/physiology , Fluorescent Antibody Technique , Mice
14.
Cell ; 122(4): 579-91, 2005 Aug 26.
Article En | MEDLINE | ID: mdl-16122425

The BCL-2 family of apoptotic proteins encompasses key regulators proximal to irreversible cell damage. The BH3-only members of this family act as sentinels, interconnecting specific death signals to the core apoptotic pathway. Our previous data demonstrated a role for BH3-only BID in maintaining myeloid homeostasis and suppressing leukemogenesis. In the absence of Bid, mice accumulate chromosomal aberrations and develop a fatal myeloproliferative disorder resembling chronic myelomonocytic leukemia. Here, we describe a role for BID in preserving genomic integrity that places BID at an early point in the path to determine the fate of a cell. We show that BID plays an unexpected role in the intra-S phase checkpoint downstream of DNA damage distinct from its proapoptotic function. We further demonstrate that this role is mediated through BID phosphorylation by the DNA-damage kinase ATM. These results establish a link between proapoptotic Bid and the DNA-damage response.


Apoptosis/genetics , Carrier Proteins/metabolism , DNA Damage/genetics , Myeloid Progenitor Cells/metabolism , Animals , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Damage/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genes, cdc/drug effects , Genes, cdc/physiology , Genomic Instability/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/metabolism , Male , Mice , Mice, Knockout , Mutagens/pharmacology , NIH 3T3 Cells , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary/genetics , S Phase/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
15.
EMBO J ; 24(2): 368-81, 2005 Jan 26.
Article En | MEDLINE | ID: mdl-15635450

Chromosomal translocations that fuse the mixed lineage leukemia (MLL) gene with multiple partners typify acute leukemias of infancy as well as therapy-related leukemias. We utilized a conditional knockin strategy to bypass the embryonic lethality caused by MLL-CBP expression and to assess the immediate effects of induced MLL-CBP expression on hematopoiesis. Within days of activating MLL-CBP, the fusion protein selectively expanded granulocyte/macrophage progenitors (GMP) and enhanced their self-renewal/proliferation. MLL-CBP altered the gene expression program of GMP, upregulating a subset of genes including Hox a9. Inhibition of Hox a9 expression by RNA interference demonstrated that MLL-CBP required Hox a9 for its enhanced cell expansion. Following exposure to sublethal gamma-irradiation or N-ethyl-N-nitrosourea (ENU), MLL-CBP mice developed myelomonocytic hyperplasia and progressed to fatal myeloproliferative disorders. These represented the spectrum of therapy-induced acute myelomonocytic leukemia/chronic myelomonocytic leukemia/myelodysplastic/myeloproliferative disorder similar to that seen in humans possessing the t(11;16). This model of MLL-CBP therapy-related myeloproliferative disease demonstrates the selectivity of this MLL fusion for GMP cells and its ability to initiate leukemogenesis in conjunction with cooperating mutations.


DNA-Binding Proteins/physiology , Myeloproliferative Disorders/chemically induced , Nuclear Proteins/physiology , Proto-Oncogenes/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Animals , CREB-Binding Protein , Histone-Lysine N-Methyltransferase , Mice , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Reverse Transcriptase Polymerase Chain Reaction
16.
Genes Dev ; 17(2): 229-39, 2003 Jan 15.
Article En | MEDLINE | ID: mdl-12533511

The proper expansion and contraction of hematopoietic cells requires tight regulation of cell death. BID, a "BH3-only" molecule, amplifies death receptor signals connecting the extrinsic to intrinsic pathways by triggering the mitochondrial pathway of apoptosis. Bid-deficient mice, as they age, spontaneously develop a myeloproliferative disorder, which progresses from myeloid hyperplasia to a fatal, clonal malignancy closely resembling chronic myelomonocytic leukemia (CMML). Thus, an apoptotic defect can result in myeloid leukemogenesis. Premalignant Bid-/- myeloid precursor cells are resistant to death receptor-induced apoptosis. Furthermore, a competitive reconstitution assay demonstrates that Bid-deficient long-term repopulating cells give rise to expanded myelomonocytic cells in vivo. Surprisingly, a single BH3-only molecule operating in the extrinsic death receptor pathway proved essential in vivo for physiologic cell death required to maintain myeloid homeostasis. Moreover, progression to CMML indicates that an upstream BH3-only molecule, BID, is required to suppress tumorigenesis.


Apoptosis/physiology , Carrier Proteins/physiology , Leukemia, Myelomonocytic, Chronic/prevention & control , Myelopoiesis/physiology , Animals , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/genetics , Chromosome Aberrations , Female , Homeostasis , Leukemia, Myelomonocytic, Chronic/etiology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis/genetics , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/prevention & control , Signal Transduction
...