Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Proc Natl Acad Sci U S A ; 112(23): 7201-6, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26015568

ABSTRACT

B-cell lymphoma 2 (BCL-2) ovarian killer (BOK) is a BCL-2 family protein with high homology to the multidomain proapoptotic proteins BAX and BAK, yet Bok(-/-) and even Bax(-/-)Bok(-/-) and Bak(-/-)Bok(-/-) mice were reported to have no overt phenotype or apoptotic defects in response to a host of classical stress stimuli. These surprising findings were interpreted to reflect functional compensation among the BAX, BAK, and BOK proteins. However, BOK cannot compensate for the severe apoptotic defects of Bax(-/-)Bak(-/-) mice despite its widespread expression. Here, we independently developed Bok(-/-) mice and found that Bok(-/-) cells are selectively defective in their response to endoplasmic reticulum (ER) stress stimuli, consistent with the predominant subcellular localization of BOK at the ER. Whereas Bok(-/-) mouse embryonic fibroblasts exposed to thapsigargin, A23187, brefeldin A, DTT, geldanamycin, or bortezomib manifested reduced activation of the mitochondrial apoptotic pathway, the death response to other stimuli such as etoposide, staurosporine, or UV remained fully intact. Multiple organs in Bok(-/-) mice exhibited resistance to thapsigargin-induced apoptosis in vivo. Although the ER stress agents activated the unfolded protein response, both ATF4 and CHOP activation were diminished in Bok(-/-) cells and mice. Importantly, BAX and BAK were unable to compensate for the defective apoptotic response to ER stress observed in SV40-transformed and primary Bok(-/-) cells, and in vivo. These findings support a selective and distinguishing role for BOK in regulating the apoptotic response to ER stress, revealing--to our knowledge--the first bona fide apoptotic defect linked to Bok deletion.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/physiology , Activating Transcription Factor 4/metabolism , Animals , Annexin A5/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Cells, Cultured , Endoplasmic Reticulum/enzymology , Enzyme Activation , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Transcription Factor CHOP/metabolism
3.
Cell Metab ; 19(2): 272-84, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24506868

ABSTRACT

The homeostatic balance of hepatic glucose utilization, storage, and production is exquisitely controlled by hormonal signals and hepatic carbon metabolism during fed and fasted states. How the liver senses extracellular glucose to cue glucose utilization versus production is not fully understood. We show that the physiologic balance of hepatic glycolysis and gluconeogenesis is regulated by Bcl-2-associated agonist of cell death (BAD), a protein with roles in apoptosis and metabolism. BAD deficiency reprograms hepatic substrate and energy metabolism toward diminished glycolysis, excess fatty acid oxidation, and exaggerated glucose production that escapes suppression by insulin. Genetic and biochemical evidence suggests that BAD's suppression of gluconeogenesis is actuated by phosphorylation of its BCL-2 homology (BH)-3 domain and subsequent activation of glucokinase. The physiologic relevance of these findings is evident from the ability of a BAD phosphomimic variant to counteract unrestrained gluconeogenesis and improve glycemia in leptin-resistant and high-fat diet models of diabetes and insulin resistance.


Subject(s)
Energy Metabolism/physiology , Gluconeogenesis/physiology , Liver/metabolism , bcl-Associated Death Protein/metabolism , Animals , Energy Metabolism/genetics , Gluconeogenesis/genetics , Mice , Mice, Mutant Strains , Phosphorylation , bcl-Associated Death Protein/genetics
4.
Blood ; 123(6): 884-93, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24352880

ABSTRACT

Mantle cell lymphoma (MCL) is a highly aggressive B-cell lymphoma resistant to conventional chemotherapy. Although defined by the characteristic t(11;14) translocation, MCL has not been recapitulated in transgenic mouse models of cyclin D1 overexpression alone. Indeed, several genetic aberrations have been identified in MCL that may contribute to its pathogenesis and chemoresistance. Of particular interest is the frequent biallelic deletion of the proapoptotic BCL-2 family protein BIM. BIM exerts its pro-death function via its α-helical BH3 death domain that has the dual capacity to inhibit antiapoptotic proteins such as BCL-2 and MCL-1 and directly trigger proapoptotic proteins such as the mitochondrial executioner protein BAX. To evaluate a functional role for Bim deletion in the pathogenesis of MCL, we generated cyclin D1-transgenic mice harboring Bim-deficient B cells. In response to immunization, Eµ(CycD1)CD19(CRE)Bim(fl/fl) mice manifested selective expansion of their splenic mantle zone compartment. Three distinct immune stimulation regimens induced lymphomas with histopathologic and molecular features of human MCL in a subset of mice. Thus, deletion of Bim in B cells, in the context of cyclin D1 overexpression, disrupts a critical control point in lymphoid maturation and predisposes to the development of MCL. This genetic proof of concept for MCL pathogenesis suggests an opportunity to reactivate the death pathway by pharmacologic mimicry of proapoptotic BIM.


Subject(s)
Apoptosis Regulatory Proteins/physiology , B-Lymphocytes/pathology , Cyclin D1/physiology , Lymphoma, Mantle-Cell/pathology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Animals , B-Lymphocytes/metabolism , Bcl-2-Like Protein 11 , Cell Cycle , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunophenotyping , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Neuron ; 74(4): 719-30, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22632729

ABSTRACT

Neuronal excitation can be substantially modulated by alterations in metabolism, as evident from the anticonvulsant effect of diets that reduce glucose utilization and promote ketone body metabolism. We provide genetic evidence that BAD, a protein with dual functions in apoptosis and glucose metabolism, imparts reciprocal effects on metabolism of glucose and ketone bodies in brain cells. These effects involve phosphoregulation of BAD and are independent of its apoptotic function. BAD modifications that reduce glucose metabolism produce a marked increase in the activity of metabolically sensitive K(ATP) channels in neurons, as well as resistance to behavioral and electrographic seizures in vivo. Seizure resistance is reversed by genetic ablation of the K(ATP) channel, implicating the BAD-K(ATP) axis in metabolic control of neuronal excitation and seizure responses.


Subject(s)
Energy Metabolism/physiology , Hippocampus/metabolism , KATP Channels/metabolism , Seizures/metabolism , bcl-Associated Death Protein/metabolism , Animals , Apoptosis/physiology , Astrocytes/metabolism , Cells, Cultured , Electroencephalography , Hippocampus/physiopathology , Mice , Mice, Transgenic , Neurons/metabolism , Oxygen Consumption/physiology , Phosphorylation , Seizures/chemically induced , Seizures/physiopathology , bcl-Associated Death Protein/genetics
6.
J Clin Invest ; 122(6): 2018-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22622039

ABSTRACT

Cancer cells subvert the natural balance between cellular life and death, achieving immortality through pathologic enforcement of survival pathways and blockade of cell death mechanisms. Pro-apoptotic BCL-2 family proteins are frequently disarmed in relapsed and refractory cancer through genetic deletion or interaction-based neutralization by overexpressed antiapoptotic proteins, resulting in resistance to chemotherapy and radiation treatments. New pharmacologic strategies are urgently needed to overcome these formidable apoptotic blockades. We harnessed the natural killing activity of BCL-2-interacting mediator of cell death (BIM), which contains one of the most potent BH3 death domains of the BCL-2 protein family, to restore BH3-dependent cell death in resistant hematologic cancers. A hydrocarbon-stapled peptide modeled after the BIM BH3 helix broadly targeted BCL-2 family proteins with high affinity, blocked inhibitory antiapoptotic interactions, directly triggered proapoptotic activity, and induced dose-responsive and BH3 sequence-specific cell death of hematologic cancer cells. The therapeutic potential of stapled BIM BH3 was highlighted by the selective activation of cell death in the aberrant lymphoid infiltrates of mice reconstituted with BIM-deficient bone marrow and in a human AML xenograft model. Thus, we found that broad and multimodal targeting of the BCL-2 family pathway can overcome pathologic barriers to cell death.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Membrane Proteins/pharmacology , Peptides/pharmacology , Proto-Oncogene Proteins/pharmacology , Animals , Apoptosis/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Neoplasm Transplantation , Protein Structure, Secondary , Transplantation, Heterologous
7.
J Clin Invest ; 120(10): 3673-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20890041

ABSTRACT

Apoptosis of motor neurons is a well-documented feature in amyotrophic lateral sclerosis (ALS) and related motor neuron diseases (MNDs). However, the role of apoptosis in the pathogenesis of these diseases remains unresolved. One possibility is that the affected motor neurons only succumb to apoptosis once they have exhausted functional capacity. If true, blocking apoptosis should confer no therapeutic benefit. To directly investigate this idea, we tested whether tissue-specific deletion in the mouse CNS of BCL2-associated X protein (BAX) and BCL2-homologous antagonist/killer (BAK), 2 proapoptotic BCL-2 family proteins that together represent an essential gateway to the mitochondrial apoptotic pathway, would protect against motor neuron degeneration. We found that neuronal deletion of Bax and Bak in a mouse model of familial ALS not only halted neuronal loss, but prevented axonal degeneration, symptom onset, weight loss, and paralysis and extended survival. These results show that motor neurons damaged in ALS activate the mitochondrial apoptotic pathway early in the disease process and that apoptotic signaling directly contributes to neuromuscular degeneration and neuronal dysfunction. Hence, inhibiting apoptosis upstream of mitochondrial permeabilization represents a possible therapeutic strategy for preserving functional motor neurons in ALS and other MNDs.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Apoptosis , Mitochondria/physiology , Motor Neurons/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , Amyotrophic Lateral Sclerosis/therapy , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Superoxide Dismutase/physiology , Superoxide Dismutase-1 , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2-Associated X Protein/antagonists & inhibitors
8.
Sci Signal ; 2(85): ra48, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706873

ABSTRACT

The proapoptotic proteins BAX and BAK constitute the mitochondrial apoptotic gateway that executes cellular demise after integrating death signals. The lethal BAK is kept in check by voltage-dependent anion channel 2 (VDAC2), a mammalian-restricted VDAC isoform. Here, we provide evidence showing a critical role for the VADC2-BAK complex in determining thymocyte survival in vivo. Genetic depletion of Vdac2 in the thymus resulted in excessive cell death and hypersensitivity to diverse death stimuli including engagement of the T cell receptor. These phenotypes were completely rescued by the concurrent deletion of Bak but not that of Bax. Thus, the VDAC2-BAK axis provides a mechanism that governs the homeostasis of thymocytes. Our study reveals a sophisticated built-in rheostat that likely fine-tunes immune competence to balance autoimmunity and immunodeficiency.


Subject(s)
Clonal Deletion/physiology , T-Lymphocytes/cytology , Voltage-Dependent Anion Channel 2/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/genetics , Apoptosis/physiology , Autoimmunity/genetics , Autoimmunity/physiology , CD3 Complex/immunology , Clonal Deletion/genetics , Dimerization , Female , Gene Knockout Techniques , Genotype , Ion Transport/genetics , Ion Transport/physiology , Male , Mice , Mice, Knockout , Mitochondrial Membranes/physiology , Thymus Gland/cytology , Voltage-Dependent Anion Channel 2/deficiency , Voltage-Dependent Anion Channel 2/genetics , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/physiology
9.
Blood ; 113(12): 2805-15, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19064728

ABSTRACT

During hematopoiesis, myeloid cell leukemia-1 (MCL-1) mediates the survival of bone marrow progenitors and lymphocytes. However, its requirement during myeloid cell differentiation, development, and effector function is less clear. Lineage-specific deletion of MCL-1 in myeloid precursors results in neutropenia due to death during differentiation. The loss of mature neutrophils induced by Mcl-1 deletion was not rescued by genetic deletion of proapoptotic Bim and Puma or by exogenous cytokine treatment. However, blockade of intrinsic apoptosis by lineage-specific deletion of both multidomain proapoptotics Bax and Bak was capable of rescuing the neutropenia associated with Mcl-1 deletion. In the monocytic lineage, despite efficient Mcl-1 deletion, monocytes and macrophages undergo normal development. During the phagocytosis of extracellular bacteria, macrophages concomitantly increase the expression of both MCL-1 and BIM. However, Mcl-1-deficient macrophages exhibit increased sensitivity to death during bacterial phagocytosis that can be abolished by codeletion of Bim. These data suggest that MCL-1 may be necessary to antagonize BIM during macrophage effector responses. Thus, MCL-1 plays selective roles in myeloid development, being required for neutrophil development and setting the threshold for apoptosis during a macrophage effector response.


Subject(s)
Granulocytes/cytology , Macrophage Activation/physiology , Macrophages, Peritoneal/cytology , Myelopoiesis/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Bcl-2-Like Protein 11 , Bone Marrow Cells/cytology , Escherichia coli , Filgrastim , Gene Deletion , Granulocyte Colony-Stimulating Factor/pharmacology , Macrophages, Peritoneal/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cell Leukemia Sequence 1 Protein , Myelopoiesis/drug effects , Organ Specificity , Phagocytosis , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Recombinant Proteins , Specific Pathogen-Free Organisms , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/physiology
10.
Nat Med ; 14(2): 144-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18223655

ABSTRACT

The proapoptotic BCL-2 family member BAD resides in a glucokinase-containing complex that regulates glucose-driven mitochondrial respiration. Here, we present genetic evidence of a physiologic role for BAD in glucose-stimulated insulin secretion by beta cells. This novel function of BAD is specifically dependent upon the phosphorylation of its BH3 sequence, previously defined as an essential death domain. We highlight the pharmacologic relevance of phosphorylated BAD BH3 by using cell-permeable, hydrocarbon-stapled BAD BH3 helices that target glucokinase, restore glucose-driven mitochondrial respiration and correct the insulin secretory response in Bad-deficient islets. Our studies uncover an alternative target and function for the BAD BH3 domain and emphasize the therapeutic potential of phosphorylated BAD BH3 mimetics in selectively restoring beta cell function. Furthermore, we show that BAD regulates the physiologic adaptation of beta cell mass during high-fat feeding. Our findings provide genetic proof of the bifunctional activities of BAD in both beta cell survival and insulin secretion.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , bcl-Associated Death Protein/metabolism , Amino Acid Sequence , Animals , Blood Glucose , Calcium/metabolism , Cell Count , Cell Survival/drug effects , Diet , Glucokinase/metabolism , Glucose/pharmacology , Humans , Hydrocarbons/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , Membrane Potential, Mitochondrial/drug effects , Mice , Models, Genetic , Molecular Sequence Data , Peptides/pharmacology , Phosphoserine/metabolism , Protein Structure, Tertiary , bcl-Associated Death Protein/chemistry , bcl-Associated Death Protein/deficiency
11.
Cell Stem Cell ; 1(3): 324-37, 2007 Sep 13.
Article in English | MEDLINE | ID: mdl-18371366

ABSTRACT

The Mixed Lineage Leukemia (MLL) gene is essential for embryonic hematopoietic stem cell (HSC) development, but its role during adult hematopoiesis is unknown. Using an inducible knockout model, we demonstrate that Mll is essential for the maintenance of adult HSCs and progenitors, with fatal bone marrow failure occurring within 3 weeks of Mll deletion. Mll-deficient cells are selectively lost from mixed bone marrow chimeras, demonstrating their failure to self-renew even in an intact bone marrow environment. Surprisingly, HSCs lacking Mll exhibit ectopic cell-cycle entry, resulting in the depletion of quiescent HSCs. In contrast, Mll deletion in myelo-erythroid progenitors results in reduced proliferation and reduced response to cytokine-induced cell-cycle entry. Committed lymphoid and myeloid cells no longer require Mll, defining the early multipotent stages of hematopoiesis as Mll dependent. These studies demonstrate that Mll plays selective and independent roles within the hematopoietic system, maintaining quiescence in HSCs and promoting proliferation in progenitors.


Subject(s)
Hematopoietic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Alleles , Animals , Bone Marrow/pathology , Cell Count , Cell Death , Cell Differentiation , Cell Line , Cell Lineage , Cell Proliferation , Cell Survival , Chimera , Erythroid Precursor Cells/cytology , Exons/genetics , Hematopoietic Stem Cells/cytology , Homeostasis , Humans , Mice , Myeloid Cells/cytology , Myeloid Progenitor Cells/cytology , Sequence Deletion , Sequence Homology, Amino Acid
12.
Genes Dev ; 20(17): 2397-409, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951254

ABSTRACT

Taspase1 was identified as the threonine endopeptidase that cleaves mixed-lineage leukemia (MLL) for proper Hox gene expression in vitro. To investigate its functions in vivo, we generated Taspase1(-/-) mice. Taspase1 deficiency results in noncleavage (nc) of MLL and MLL2 and homeotic transformations. Remarkably, our in vivo studies uncover an unexpected role of Taspase1 in the cell cycle. Taspase1(-/-) animals are smaller in size. Taspase1(-/-) mouse embryonic fibroblasts (MEFs) exhibit impaired proliferation, and acute deletion of Taspase1 leads to a marked reduction of thymocytes. Taspase1 deficiency incurs down-regulation of Cyclin Es, As, and Bs and up-regulation of p16(Ink4a) . We show that MLL and MLL2 directly target E2Fs for Cyclin expression. The uncleaved precursor MLL displays a reduced histone H3 methyl transferase activity in vitro. Accordingly, chromatin immunoprecipitation assays demonstrate a markedly decreased histone H3 K4 trimethylation at Cyclin E1 and E2 genes in Taspase1(-/-) cells. Furthermore, MLL(nc/nc;2nc/nc) MEFs are also impaired in proliferation. Our data are consistent with a model in which precursor MLLs, activated by Taspase1, target to Cyclins through E2Fs to methylate histone H3 at K4, leading to activation. Lastly, Taspase1(-/-) cells are resistant to oncogenic transformation, and Taspase1 is overexpressed in many cancer cell lines. Thus, Taspase1 may serve as a target for cancer therapeutics.


Subject(s)
Cell Cycle/physiology , Endopeptidases/physiology , Myeloid-Lymphoid Leukemia Protein/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Endopeptidases/biosynthesis , Endopeptidases/deficiency , Endopeptidases/genetics , Hydrolysis , Mice , Mice, Knockout , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/genetics
13.
Nat Genet ; 37(11): 1210-2, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16200063

ABSTRACT

Stratifin (Sfn, also called 14-3-3sigma) is highly expressed in differentiating epidermis and mediates cell cycle arrest. Sfn is repressed in cancer, but its function during development is uncharacterized. We identified an insertion mutation in the gene Sfn in repeated epilation (Er) mutant mice by positional cloning. Er/+ mice expressed a truncated Sfn protein, which probably contributes to the defects in Er/Er and Er/+ epidermis and to cancer development in Er/+ mice.


Subject(s)
Alopecia/genetics , Biomarkers, Tumor/genetics , Exonucleases/genetics , Hair Removal , Mice, Mutant Strains/anatomy & histology , Mutation/genetics , Neoplasm Proteins/genetics , Skin Neoplasms/genetics , 14-3-3 Proteins , Alopecia/pathology , Animals , Epidermal Cells , Exoribonucleases , Heterozygote , Male , Mice , Molecular Sequence Data , Phenotype
14.
Proc Natl Acad Sci U S A ; 102(34): 12005-10, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16103352

ABSTRACT

Mitochondrial permeability transition (PT) is a phenomenon induced by high levels of matrix calcium and is characterized by the opening of the PT pore (PTP). Activation of the PTP results in loss of mitochondrial membrane potential, expansion of the matrix, and rupture of the mitochondrial outer membrane. Consequently, PT has been implicated in both apoptotic and necrotic cell death. Cyclophilin D (CypD) appears to be a critical component of the PTP. To investigate the role of CypD in cell death, we created a CypD-deficient mouse. In vitro, CypD-deficient mitochondria showed an increased capacity to retain calcium and were no longer susceptible to PT induced by the addition of calcium. CypD-deficient primary mouse embryonic fibroblasts (MEFs) were as susceptible to classical apoptotic stimuli as the WT, suggesting that CypD is not a central component of cell death in response to these specific death stimuli. However, CypD-deficient MEFs were significantly less susceptible than their WT counterparts to cell death induced by hydrogen peroxide, implicating CypD in oxidative stress-induced cell death. Importantly, CypD-deficient mice displayed a dramatic reduction in brain infarct size after acute middle cerebral artery occlusion and reperfusion, strongly supporting an essential role for CypD in an ischemic injury model in which calcium overload and oxidative stress have been implicated.


Subject(s)
Apoptosis/physiology , Brain Ischemia/physiopathology , Cyclophilins/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Neurons/physiology , Voltage-Dependent Anion Channels/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/metabolism , Calcium/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Hydrogen Peroxide/toxicity , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Mitochondrial Swelling/physiology , Neurons/metabolism , Permeability , Spectrophotometry
15.
EMBO J ; 24(2): 368-81, 2005 Jan 26.
Article in English | MEDLINE | ID: mdl-15635450

ABSTRACT

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.


Subject(s)
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.
Dev Cell ; 6(3): 437-43, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15030765

ABSTRACT

The Mixed-Lineage Leukemia (MLL) gene encodes a Trithorax-related chromatin-modifying protooncogene that positively regulates Hox genes. In addition to their well-characterized roles in axial patterning, Trithorax and Polycomb family proteins perform less-understood functions in vertebrate hematopoiesis. To define the role of MLL in the development of the hematopoietic system, we examined the potential of cells lacking MLL. Mll-deficient cells could not develop into lymphocytes in adult RAG-2 chimeric animals. Similarly, in vitro differentiation of B cells required MLL. In chimeric embryos, Mll-deficient cells failed to contribute to fetal liver hematopoietic stem cell/progenitor populations. Moreover, we show that aorta-gonad-mesonephros (AGM) cells from Mll-deficient embryos lacked hematopoietic stem cell (HSC) activity despite their ability to generate hematopoietic progeny in vitro. These results demonstrate an intrinsic requirement for MLL in definitive hematopoiesis, where it is essential for the generation of HSCs in the embryo.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Proto-Oncogenes , Stem Cells/physiology , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Aorta/cytology , Aorta/embryology , Aorta/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques/methods , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Flow Cytometry/methods , Galactosides/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Histone-Lysine N-Methyltransferase , Immunohistochemistry/methods , In Situ Hybridization/methods , Indoles/metabolism , Killer Cells, Natural/physiology , Leukocyte Common Antigens/metabolism , Liver/cytology , Liver/metabolism , Lymphopoiesis/drug effects , Lymphopoiesis/physiology , Mesonephros/cytology , Mesonephros/embryology , Mesonephros/metabolism , Mice , Mice, Transgenic , Myeloid-Lymphoid Leukemia Protein , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family , Stem Cell Transplantation/methods , Stem Cells/cytology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Science ; 301(5632): 513-7, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12881569

ABSTRACT

The multidomain proapoptotic molecules BAK or BAX are required to initiate the mitochondrial pathway of apoptosis. How cells maintain the potentially lethal proapoptotic effector BAK in a monomeric inactive conformation at mitochondria is unknown. In viable cells, we found BAK complexed with mitochondrial outer-membrane protein VDAC2, a VDAC isoform present in low abundance that interacts specifically with the inactive conformer of BAK. Cells deficient in VDAC2, but not cells lacking the more abundant VDAC1, exhibited enhanced BAK oligomerization and were more susceptible to apoptotic death. Conversely, overexpression of VDAC2 selectively prevented BAK activation and inhibited the mitochondrial apoptotic pathway. Death signals activate "BH3-only" molecules such as tBID, BIM, or BAD, which displace VDAC2 from BAK, enabling homo-oligomerization of BAK and apoptosis. Thus, VDAC2, an isoform restricted to mammals, regulates the activity of BAK and provides a connection between mitochondrial physiology and the core apoptotic pathway.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Mitochondria/metabolism , Porins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Animals , BH3 Interacting Domain Death Agonist Protein , Biopolymers , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Line , Cells, Cultured , Etoposide/pharmacology , Humans , Intracellular Membranes/metabolism , Jurkat Cells , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Porins/genetics , Porins/isolation & purification , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/pharmacology , Staurosporine/pharmacology , Voltage-Dependent Anion Channel 1 , Voltage-Dependent Anion Channel 2 , Voltage-Dependent Anion Channels , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
18.
Mol Endocrinol ; 17(1): 93-106, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12511609

ABSTRACT

Somatostatin (SRIF) regulates pancreatic insulin and glucagon secretion. In the present study we describe the generation of SRIF receptor subtype 5 knockout (sst(5) KO) mice to examine the role of SRIF receptor subtypes (sst) in regulating insulin secretion and glucose homeostasis. Mice deficient in sst(5) were viable, fertile, appeared healthy, and displayed no obvious phenotypic abnormalities. Pancreatic islets isolated from sst(5) KO mice displayed increased total insulin content as compared with islets obtained from wild-type (WT) mice. Somatostatin-28 (SRIF-28) and the sst(5)/sst(1)-selective agonist compound 5/1 potently inhibited glucose-stimulated insulin secretion from WT islets. SRIF-28 inhibited insulin secretion from sst(5) KO islets with 16-fold less potency while the maximal effect of compound 5/1 was markedly diminished when compared with its effects in WT islets. sst(5) KO mice exhibited decreased blood glucose and plasma insulin levels and increased leptin and glucagon concentrations compared with WT mice. Furthermore, sst(5) KO mice displayed decreased susceptibility to high fat diet-induced insulin resistance. The results of these studies suggest sst(5) mediates SRIF inhibition of pancreatic insulin secretion and contributes to the regulation of glucose homeostasis and insulin sensitivity. Our findings suggest a potential beneficial role of sst(5) antagonists for alleviating metabolic abnormalities associated with obesity and insulin resistance.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Receptors, Somatostatin/physiology , Animals , CHO Cells , Cloning, Molecular/methods , Cricetinae , Female , Gene Targeting/methods , Homeostasis/physiology , Insulin Resistance/physiology , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout , Organ Culture Techniques , Receptors, Somatostatin/agonists , Receptors, Somatostatin/deficiency , Receptors, Somatostatin/genetics , Somatostatin/metabolism , Somatostatin-28 , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...