Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Chem Biol ; 15(11): 1057-1066, 2019 11.
Article in English | MEDLINE | ID: mdl-31591564

ABSTRACT

Activating the intrinsic apoptosis pathway with small molecules is now a clinically validated approach to cancer therapy. In contrast, blocking apoptosis to prevent the death of healthy cells in disease settings has not been achieved. Caspases have been favored, but they act too late in apoptosis to provide long-term protection. The critical step in committing a cell to death is activation of BAK or BAX, pro-death BCL-2 proteins mediating mitochondrial damage. Apoptosis cannot proceed in their absence. Here we show that WEHI-9625, a novel tricyclic sulfone small molecule, binds to VDAC2 and promotes its ability to inhibit apoptosis driven by mouse BAK. In contrast to caspase inhibitors, WEHI-9625 blocks apoptosis before mitochondrial damage, preserving cellular function and long-term clonogenic potential. Our findings expand on the key role of VDAC2 in regulating apoptosis and demonstrate that blocking apoptosis at an early stage is both advantageous and pharmacologically tractable.


Subject(s)
Apoptosis/physiology , Small Molecule Libraries/metabolism , Voltage-Dependent Anion Channel 2/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , Animals , Mice , Protein Binding , Voltage-Dependent Anion Channel 2/metabolism
2.
Genes Dev ; 30(10): 1240-50, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27198225

ABSTRACT

Due to the myriad interactions between prosurvival and proapoptotic members of the Bcl-2 family of proteins, establishing the mechanisms that regulate the intrinsic apoptotic pathway has proven challenging. Mechanistic insights have primarily been gleaned from in vitro studies because genetic approaches in mammals that produce unambiguous data are difficult to design. Here we describe a mutation in mouse and human Bak that specifically disrupts its interaction with the prosurvival protein Bcl-xL Substitution of Glu75 in mBak (hBAK Q77) for leucine does not affect the three-dimensional structure of Bak or killing activity but reduces its affinity for Bcl-xL via loss of a single hydrogen bond. Using this mutant, we investigated the requirement for physical restraint of Bak by Bcl-xL in apoptotic regulation. In vitro, Bak(Q75L) cells were significantly more sensitive to various apoptotic stimuli. In vivo, loss of Bcl-xL binding to Bak led to significant defects in T-cell and blood platelet survival. Thus, we provide the first definitive in vivo evidence that prosurvival proteins maintain cellular viability by interacting with and inhibiting Bak.


Subject(s)
Apoptosis/genetics , Blood Platelets/cytology , T-Lymphocytes/cytology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Line , Cell Survival/genetics , Humans , Mice , Mice, Inbred C57BL , Mutation , Protein Binding , Protein Conformation , Protein Domains/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics
3.
Am J Pathol ; 185(7): 1867-76, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26093983

ABSTRACT

E26 transformation-specific 1 (ETS1) and friend leukemia integration 1 (FLI1) are members of the ETS family of transcription factors, of which there are 28 in humans. Both genes are hemizygous in Jacobsen syndrome, an 11q contiguous gene deletion disorder involving thrombocytopenia, facial dysmorphism, growth and mental retardation, malformation of the heart and other organs, and hearing impairment associated with recurrent ear infections. To determine whether any of these defects are because of hemizygosity for ETS1 and FLI1, we characterized the phenotype of mice heterozygous for mutant alleles of Ets1 and Fli1. Fli1(+/-) mice displayed mild thrombocytopenia, as did Ets1(+/-)Fli1(+/-) animals. Fli1(+/-) and Ets1(+/-)Fli1(+/-) mice also displayed craniofacial abnormalities, including a small middle ear cavity, short nasal bone, and malformed interface between the nasal bone process and cartilaginous nasal septum. They exhibited hearing impairment, otitis media, fusions of ossicles to the middle ear wall, and deformed stapes. Hearing impairment was more penetrant and stapes malformations were more severe in Ets1(+/-)Fli1(+/-) mice than in Fli1(+/-) mice, indicating partial functional redundancy of these transcription factors during auditory development. Our findings indicate that the short nose, otitis media, and hearing impairment in Jacobsen syndrome are likely because of hemizygosity for ETS1 and FLI1.


Subject(s)
Disease Models, Animal , Haploinsufficiency , Jacobsen Distal 11q Deletion Syndrome/genetics , Mice , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-fli-1/genetics , Animals , Ear, Middle/abnormalities , Female , Genotype , Hearing Loss/genetics , Humans , Male , Mice/abnormalities , Mice/genetics , Nasal Bone/abnormalities , Otitis Media/genetics , Phenotype
4.
Blood ; 123(22): 3381-9, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24599546

ABSTRACT

The fetal/neonatal hematopoietic system must generate enough blood cells to meet the demands of rapid growth. This unique challenge might underlie the high incidence of thrombocytopenia among preterm neonates. In this study, neonatal platelet production and turnover were investigated in newborn mice. Based on a combination of blood volume expansion and increasing platelet counts, the platelet mass increased sevenfold during the first 2 weeks of murine life, a time during which thrombopoiesis shifted from liver to bone marrow. Studies applying in vivo biotinylation and mathematical modeling showed that newborn and adult mice had similar platelet production rates, but neonatal platelets survived 1 day longer in circulation. This prolonged lifespan fully accounted for the rise in platelet counts observed during the second week of murine postnatal life. A study of pro-apoptotic and anti-apoptotic Bcl-2 family proteins showed that neonatal platelets had higher levels of the anti-apoptotic protein Bcl-2 and were more resistant to apoptosis induced by the Bcl-2/Bcl-xL inhibitor ABT-737 than adult platelets. However, genetic ablation or pharmacologic inhibition of Bcl-2 alone did not shorten neonatal platelet survival or reduce platelet counts in newborn mice, indicating the existence of redundant or alternative mechanisms mediating the prolonged lifespan of neonatal platelets.


Subject(s)
Blood Platelets/physiology , Platelet Count , Thrombopoiesis/physiology , Animals , Animals, Newborn , Apoptosis/physiology , Biphenyl Compounds/pharmacology , Blood Platelets/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Humans , Infant, Newborn , Liver/cytology , Mean Platelet Volume , Megakaryocytes/physiology , Megakaryocytes/ultrastructure , Mice , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Spleen/cytology , Sulfonamides/pharmacology , Thrombopoiesis/drug effects
5.
Nat Commun ; 5: 3455, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24632563

ABSTRACT

BH3 mimetic drugs that target BCL-2 family pro-survival proteins to induce tumour cell apoptosis represent a new era in cancer therapy. Clinical trials of navitoclax (ABT-263, which targets BCL-2, BCL-XL and BCL-W) have shown great promise, but encountered dose-limiting thrombocytopenia. Recent work has demonstrated that this is due to the inhibition of BCL-XL, which is essential for platelet survival. These findings raise new questions about the established model of platelet shedding by megakaryocytes, which is thought to be an apoptotic process. Here we generate mice with megakaryocyte-specific deletions of the essential mediators of extrinsic (Caspase-8) and intrinsic (BAK/BAX) apoptosis. We show that megakaryocytes possess a Fas ligand-inducible extrinsic apoptosis pathway. However, Fas activation does not stimulate platelet production, rather, it triggers Caspase-8-mediated killing. Combined loss of Caspase-8/BAK/BAX does not impair thrombopoiesis, but can protect megakaryocytes from death in mice infected with lymphocytic choriomeningitis virus. Thus, apoptosis is dispensable for platelet biogenesis.


Subject(s)
Blood Platelets/metabolism , Aniline Compounds/pharmacology , Animals , Apoptosis/physiology , Blood Platelets/drug effects , Blotting, Western , Caspase 8/metabolism , Electrophoresis, Polyacrylamide Gel , Female , Male , Mice , Rats , Signal Transduction , Sulfonamides/pharmacology , Thrombocytopenia/metabolism
6.
Blood ; 119(24): 5850-8, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22374700

ABSTRACT

Mature megakaryocytes depend on the function of Bcl-x(L), a member of the Bcl-2 family of prosurvival proteins, to proceed safely through the process of platelet shedding. Despite this, loss of Bcl-x(L) does not prevent the growth and maturation of megakaryocytes, suggesting redundancy with other prosurvival proteins. We therefore generated mice with a megakaryocyte-specific deletion of Mcl-1, which is known to be expressed in megakaryocytes. Megakaryopoiesis, platelet production, and platelet lifespan were unperturbed in Mcl-1(Pf4Δ/Pf4Δ) animals. However, treatment with ABT-737, a BH3 mimetic compound that inhibits the prosurvival proteins Bcl-2, Bcl-x(L), and Bcl-w resulted in the complete ablation of megakaryocytes and platelets. Genetic deletion of both Mcl-1 and Bcl-x(L) in megakaryocytes resulted in preweaning lethality. Megakaryopoiesis in Bcl-x(Pf4Δ/Pf4Δ) Mcl-1(Pf4Δ/Pf4Δ) embryos was severely compromised, and these animals exhibited ectopic bleeding. Our studies indicate that the combination of Bcl-x(L) and Mcl-1 is essential for the viability of the megakaryocyte lineage.


Subject(s)
Megakaryocytes/metabolism , Megakaryocytes/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism , Alleles , Animals , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacology , Blood Cell Count , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Cell Count , Cell Death/drug effects , Cell Size , Cell Survival/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian/drug effects , Embryo, Mammalian/pathology , Fetus/drug effects , Fetus/metabolism , Fetus/pathology , Gene Deletion , Hemorrhage/pathology , Liver/drug effects , Liver/embryology , Liver/metabolism , Liver/pathology , Lymphatic Vessels/drug effects , Lymphatic Vessels/pathology , Megakaryocytes/drug effects , Megakaryocytes/ultrastructure , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/administration & dosage , Nitrophenols/pharmacology , Organ Specificity/drug effects , Piperazines/administration & dosage , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/deficiency , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Thrombopoiesis/drug effects
7.
Blood ; 119(18): 4283-90, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22294729

ABSTRACT

Apoptotic caspases, including caspase-9, are thought to facilitate platelet shedding by megakaryocytes. They are known to be activated during platelet apoptosis, and have also been implicated in platelet hemostatic responses. However, the precise requirement for, and the regulation of, apoptotic caspases have never been defined in either megakaryocytes or platelets. To establish the role of caspases in platelet production and function, we generated mice lacking caspase-9 in their hematopoietic system. We demonstrate that both megakaryocytes and platelets possess a functional apoptotic caspase cascade downstream of Bcl-2 family-mediated mitochondrial damage. Caspase-9 is the initiator caspase, and its loss blocks effector caspase activation. Surprisingly, steady-state thrombopoiesis is unperturbed in the absence of caspase-9, indicating that the apoptotic caspase cascade is not required for platelet production. In platelets, loss of caspase-9 confers resistance to the BH3 mimetic ABT-737, blocking phosphatidylserine (PS) exposure and delaying ABT-737-induced thrombocytopenia in vivo. Despite this, steady-state platelet lifespan is normal. Casp9(-/-) platelets are fully capable of physiologic hemostatic responses and functional regulation of adhesive integrins in response to agonist. These studies demonstrate that the apoptotic caspase cascade is required for the efficient death of megakaryocytes and platelets, but is dispensable for their generation and function.


Subject(s)
Apoptosis/physiology , Blood Platelets/cytology , Caspase 9/physiology , Megakaryocytes/cytology , Thrombopoiesis/physiology , Animals , Biphenyl Compounds/pharmacology , Biphenyl Compounds/toxicity , Blood Platelets/enzymology , Caspase 9/deficiency , Caspase 9/genetics , Cell Lineage , Hemostasis/drug effects , Hemostasis/physiology , Hirudins/pharmacology , Liver/embryology , Liver Transplantation , Megakaryocytes/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrophenols/pharmacology , Nitrophenols/toxicity , Piperazines/pharmacology , Piperazines/toxicity , Platelet Activation/drug effects , Platelet Activation/physiology , Radiation Chimera , Sulfonamides/pharmacology , Sulfonamides/toxicity , Thrombocytopenia/chemically induced , bcl-2-Associated X Protein/deficiency
8.
J Exp Med ; 208(10): 2017-31, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21911424

ABSTRACT

It is believed that megakaryocytes undergo a specialized form of apoptosis to shed platelets. Conversely, a range of pathophysiological insults, including chemotherapy, are thought to cause thrombocytopenia by inducing the apoptotic death of megakaryocytes and their progenitors. To resolve this paradox, we generated mice with hematopoietic- or megakaryocyte-specific deletions of the essential mediators of apoptosis, Bak and Bax. We found that platelet production was unperturbed. In stark contrast, deletion of the prosurvival protein Bcl-x(L) resulted in megakaryocyte apoptosis and a failure of platelet shedding. This could be rescued by deletion of Bak and Bax. We examined the effect on megakaryocytes of three agents that activate the intrinsic apoptosis pathway in other cell types: etoposide, staurosporine, and the BH3 mimetic ABT-737. All three triggered mitochondrial damage, caspase activation, and cell death. Deletion of Bak and Bax rendered megakaryocytes resistant to etoposide and ABT-737. In vivo, mice with a Bak(-/-) Bax(-/-) hematopoietic system were protected against thrombocytopenia induced by the chemotherapeutic agent carboplatin. Thus, megakaryocytes do not activate the intrinsic pathway to generate platelets; rather, the opposite is true: they must restrain it to survive and progress safely through proplatelet formation and platelet shedding.


Subject(s)
Apoptosis/physiology , Blood Platelets/metabolism , Megakaryocytes/cytology , Megakaryocytes/physiology , Animals , Cell Survival , Mice , Mice, Inbred C57BL , Mice, Knockout , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
9.
J Biol Chem ; 282(7): 4728-4737, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17148442

ABSTRACT

The suppressors of cytokine signaling (SOCS) proteins inhibit cytokine action by direct interaction with Janus kinases or activated cytokine receptors. In addition to the N-terminal and Src homology 2 domains that mediate these interactions, SOCS proteins contain a C-terminal SOCS box. DNA data base searches have identified a number of other protein families that possess a SOCS box, of which the ankyrin repeat and SOCS box-containing (Asb) proteins constitute the largest. Although it is known that the SOCS proteins are involved in the negative regulation of cytokine signaling, the biological and biochemical functions of the Asbs are largely undefined. Using a proteomics approach, we demonstrate that creatine kinase B (CKB) interacts with Asb-9 in a specific, SOCS box-independent manner. This interaction increases the polyubiquitylation of CKB and decreases total CKB levels within the cell. The targeting of CKB for degradation by Asb-9 was primarily SOCS box-dependent and suggests that Asb-9 acts as a specific ubiquitin ligase regulating levels of this evolutionarily conserved enzyme.


Subject(s)
Creatine Kinase/metabolism , Protein Processing, Post-Translational/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Ankyrins/metabolism , Cytokines/metabolism , Databases, Protein , Evolution, Molecular , Humans , Mice , Proteomics , Signal Transduction/physiology , src Homology Domains/physiology
SELECTION OF CITATIONS
SEARCH DETAIL