Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell ; 159(7): 1549-62, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525874

RESUMEN

Activated caspases are a hallmark of apoptosis induced by the intrinsic pathway, but they are dispensable for cell death and the apoptotic clearance of cells in vivo. This has led to the suggestion that caspases are activated not just to kill but to prevent dying cells from triggering a host immune response. Here, we show that the caspase cascade suppresses type I interferon (IFN) production by cells undergoing Bak/Bax-mediated apoptosis. Bak and Bax trigger the release of mitochondrial DNA. This is recognized by the cGAS/STING-dependent DNA sensing pathway, which initiates IFN production. Activated caspases attenuate this response. Pharmacological caspase inhibition or genetic deletion of caspase-9, Apaf-1, or caspase-3/7 causes dying cells to secrete IFN-ß. In vivo, this precipitates an elevation in IFN-ß levels and consequent hematopoietic stem cell dysfunction, which is corrected by loss of Bak and Bax. Thus, the apoptotic caspase cascade functions to render mitochondrial apoptosis immunologically silent.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Interferón Tipo I/metabolismo , Transducción de Señal , Animales , Caspasa 9/genética , Caspasa 9/metabolismo , Caspasas/clasificación , Cruzamientos Genéticos , ADN Mitocondrial/metabolismo , Femenino , Células Madre Hematopoyéticas/metabolismo , Interferón Tipo I/inmunología , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL
2.
EMBO J ; 42(12): e112712, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37139896

RESUMEN

cGAS-STING signalling is induced by detection of foreign or mislocalised host double-stranded (ds)DNA within the cytosol. STING acts as the major signalling hub, where it controls production of type I interferons and inflammatory cytokines. Basally, STING resides on the ER membrane. Following activation STING traffics to the Golgi to initiate downstream signalling and subsequently to endolysosomal compartments for degradation and termination of signalling. While STING is known to be degraded within lysosomes, the mechanisms controlling its delivery remain poorly defined. Here we utilised a proteomics-based approach to assess phosphorylation changes in primary murine macrophages following STING activation. This identified numerous phosphorylation events in proteins involved in intracellular and vesicular transport. We utilised high-temporal microscopy to track STING vesicular transport in live macrophages. We subsequently identified that the endosomal complexes required for transport (ESCRT) pathway detects ubiquitinated STING on vesicles, which facilitates the degradation of STING in murine macrophages. Disruption of ESCRT functionality greatly enhanced STING signalling and cytokine production, thus characterising a mechanism controlling effective termination of STING signalling.


Asunto(s)
Inmunidad Innata , Proteínas de la Membrana , Ratones , Animales , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Transducción de Señal/fisiología , Macrófagos/metabolismo , Nucleotidiltransferasas/metabolismo , ADN , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética
3.
Genes Dev ; 30(10): 1240-50, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27198225

RESUMEN

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.


Asunto(s)
Apoptosis/genética , Plaquetas/citología , Linfocitos T/citología , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína bcl-X/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Línea Celular , Supervivencia Celular/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación , Unión Proteica , Conformación Proteica , Dominios Proteicos/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/genética
4.
Nat Chem Biol ; 15(11): 1057-1066, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31591564

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Bibliotecas de Moléculas Pequeñas/metabolismo , Canal Aniónico 2 Dependiente del Voltaje/fisiología , Proteína Destructora del Antagonista Homólogo bcl-2/fisiología , Animales , Ratones , Unión Proteica , Canal Aniónico 2 Dependiente del Voltaje/metabolismo
5.
Blood ; 132(2): 197-209, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29784641

RESUMEN

The circulating life span of blood platelets is regulated by the prosurvival protein BCL-XL It restrains the activity of BAK and BAX, the essential prodeath mediators of intrinsic apoptosis. Disabling the platelet intrinsic apoptotic pathway in mice by deleting BAK and BAX results in a doubling of platelet life span and concomitant thrombocytosis. Apoptotic platelets expose phosphatidylserine (PS) via a mechanism that is distinct from that driven by classical agonists. Whether there is any role for apoptotic PS in platelet function in vivo, however, is unclear. Apoptosis has also been associated with the platelet storage lesion (PSL), the constellation of biochemical deteriorations that occur during blood bank storage. In this study, we investigated the role of BAK/BAX-mediated apoptosis in hemostasis and thrombosis and in the development of the PSL. We show that although intrinsic apoptosis is rapidly induced during storage at 37°C, it is not detected when platelets are kept at the standard storage temperature of 22°C. Remarkably, loss of BAK and BAX did not prevent the development of the PSL at either temperature. BAK/BAX-deficient mice exhibited increased bleeding times and unstable thrombus formation. This phenotype was not caused by impaired PS exposure, but was associated with a defect in granule release from aged platelets. Strikingly, rejuvenation of BAK/BAX-deficient platelets in vivo completely rescued the observed hemostatic defects. Thus, apoptotic culling of old platelets from the bloodstream is essential to maintain a functional, hemostatically reactive platelet population. Inhibiting intrinsic apoptosis in blood banked platelets is unlikely to yield significant benefit.


Asunto(s)
Apoptosis , Plaquetas/metabolismo , Susceptibilidad a Enfermedades , Animales , Apoptosis/genética , Biomarcadores , Tiempo de Sangría , Recuento de Células Sanguíneas , Coagulación Sanguínea , Caspasas/metabolismo , Supervivencia Celular/genética , Femenino , Genotipo , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Transducción de Señal , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
6.
Blood ; 119(24): 5850-8, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22374700

RESUMEN

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.


Asunto(s)
Megacariocitos/metabolismo , Megacariocitos/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo , Alelos , Animales , Compuestos de Bifenilo/administración & dosificación , Compuestos de Bifenilo/farmacología , Recuento de Células Sanguíneas , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Plaquetas/patología , Recuento de Células , Muerte Celular/efectos de los fármacos , Tamaño de la Célula , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/patología , Feto/efectos de los fármacos , Feto/metabolismo , Feto/patología , Eliminación de Gen , Hemorragia/patología , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , Hígado/patología , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/patología , Megacariocitos/efectos de los fármacos , Megacariocitos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Nitrofenoles/administración & dosificación , Nitrofenoles/farmacología , Especificidad de Órganos/efectos de los fármacos , Piperazinas/administración & dosificación , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/deficiencia , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Trombopoyesis/efectos de los fármacos
7.
Cell Rep ; 36(3): 109430, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289356

RESUMEN

While the intrinsic apoptosis pathway is thought to play a central role in shaping the B cell lineage, its precise role in mature B cell homeostasis remains elusive. Using mice in which mature B cells are unable to undergo apoptotic cell death, we show that apoptosis constrains follicular B (FoB) cell lifespan but plays no role in marginal zone B (MZB) cell homeostasis. In these mice, FoB cells accumulate abnormally. This intensifies intercellular competition for BAFF, resulting in a contraction of the MZB cell compartment, and reducing the growth, trafficking, and fitness of FoB cells. Diminished BAFF signaling dampens the non-canonical NF-κB pathway, undermining FoB cell growth despite the concurrent triggering of a protective p53 response. Thus, MZB and FoB cells exhibit a differential requirement for the intrinsic apoptosis pathway. Homeostatic apoptosis constrains the size of the FoB cell compartment, thereby preventing competition-induced FoB cell atrophy.


Asunto(s)
Apoptosis , Linfocitos B/patología , Homeostasis , Animales , Formación de Anticuerpos/inmunología , Atrofia , Factor Activador de Células B/metabolismo , Recuento de Células , Diferenciación Celular/genética , Proliferación Celular/genética , Tamaño de la Célula , Supervivencia Celular/genética , Senescencia Celular/genética , Eliminación de Gen , Regulación de la Expresión Génica , Ratones Noqueados , Análisis de Secuencia de ARN , Timo/inmunología , Factores de Transcripción/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
8.
Blood Adv ; 4(7): 1270-1283, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32236527

RESUMEN

In eukaryotic cells, messenger RNA (mRNA) molecules are exported from the nucleus to the cytoplasm, where they are translated. The highly conserved protein nuclear RNA export factor1 (Nxf1) is an important mediator of this process. Although studies in yeast and in human cell lines have shed light on the biochemical mechanisms of Nxf1 function, its contribution to mammalian physiology is less clear. Several groups have identified recurrent NXF1 mutations in chronic lymphocytic leukemia (CLL), placing it alongside several RNA-metabolism factors (including SF3B1, XPO, RPS15) whose dysregulation is thought to contribute to CLL pathogenesis. We report here an allelic series of germline point mutations in murine Nxf1. Mice heterozygous for these loss-of-function Nxf1 mutations exhibit thrombocytopenia and lymphopenia, together with milder hematological defects. This is primarily caused by cell-intrinsic defects in the survival of platelets and peripheral lymphocytes, which are sensitized to intrinsic apoptosis. In contrast, Nxf1 mutations have almost no effect on red blood cell homeostasis. Comparative transcriptome analysis of platelets, lymphocytes, and erythrocytes from Nxf1-mutant mice shows that, in response to impaired Nxf1 function, the cytoplasmic representation of transcripts encoding regulators of RNA metabolism is altered in a unique, lineage-specific way. Thus, blood cell lineages exhibit differential requirements for Nxf1-mediated global mRNA export.


Asunto(s)
Linfopenia , Trombocitopenia , Animales , Células Germinativas , Linfopenia/genética , Ratones , Mutación , Proteínas de Transporte Nucleocitoplasmático/genética , ARN Viral , Proteínas de Unión al ARN/genética , Trombocitopenia/genética
9.
Science ; 359(6378)2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29472455

RESUMEN

Mitochondrial apoptosis is mediated by BAK and BAX, two proteins that induce mitochondrial outer membrane permeabilization, leading to cytochrome c release and activation of apoptotic caspases. In the absence of active caspases, mitochondrial DNA (mtDNA) triggers the innate immune cGAS/STING pathway, causing dying cells to secrete type I interferon. How cGAS gains access to mtDNA remains unclear. We used live-cell lattice light-sheet microscopy to examine the mitochondrial network in mouse embryonic fibroblasts. We found that after BAK/BAX activation and cytochrome c loss, the mitochondrial network broke down and large BAK/BAX pores appeared in the outer membrane. These BAK/BAX macropores allowed the inner mitochondrial membrane to herniate into the cytosol, carrying with it mitochondrial matrix components, including the mitochondrial genome. Apoptotic caspases did not prevent herniation but dismantled the dying cell to suppress mtDNA-induced innate immune signaling.


Asunto(s)
Apoptosis , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Citocromos c/metabolismo , ADN Mitocondrial/metabolismo , Fibroblastos , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Membranas Mitocondriales/química , Multimerización de Proteína , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/genética
10.
J Clin Invest ; 127(3): 814-829, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28134622

RESUMEN

Platelets are anuclear cells that are essential for blood clotting. They are produced by large polyploid precursor cells called megakaryocytes. Previous genome-wide association studies in nearly 70,000 individuals indicated that single nucleotide variants (SNVs) in the gene encoding the actin cytoskeletal regulator tropomyosin 4 (TPM4) exert an effect on the count and volume of platelets. Platelet number and volume are independent risk factors for heart attack and stroke. Here, we have identified 2 unrelated families in the BRIDGE Bleeding and Platelet Disorders (BPD) collection who carry a TPM4 variant that causes truncation of the TPM4 protein and segregates with macrothrombocytopenia, a disorder characterized by low platelet count. N-Ethyl-N-nitrosourea-induced (ENU-induced) missense mutations in Tpm4 or targeted inactivation of the Tpm4 locus led to gene dosage-dependent macrothrombocytopenia in mice. All other blood cell counts in Tpm4-deficient mice were normal. Insufficient TPM4 expression in human and mouse megakaryocytes resulted in a defect in the terminal stages of platelet production and had a mild effect on platelet function. Together, our findings demonstrate a nonredundant role for TPM4 in platelet biogenesis in humans and mice and reveal that truncating variants in TPM4 cause a previously undescribed dominant Mendelian platelet disorder.


Asunto(s)
Plaquetas/metabolismo , Genes Dominantes , Enfermedades Genéticas Congénitas , Mutación Missense , Trombocitopenia , Tropomiosina , Animales , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Trombocitopenia/genética , Trombocitopenia/metabolismo , Tropomiosina/genética , Tropomiosina/metabolismo
11.
Cell Metab ; 23(1): 155-64, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26603191

RESUMEN

Interleukin-18 (IL-18) is activated by Caspase-1 in inflammasome complexes and has anti-obesity effects; however, it is not known which inflammasome regulates this process. We found that mice lacking the NLRP1 inflammasome phenocopy mice lacking IL-18, with spontaneous obesity due to intrinsic lipid accumulation. This is exacerbated when the mice are fed a high-fat diet (HFD) or a high-protein diet, but not when mice are fed a HFD with low energy density (high fiber). Furthermore, mice with an activating mutation in NLRP1, and hence increased IL-18, have decreased adiposity and are resistant to diet-induced metabolic dysfunction. Feeding these mice a HFD further increased plasma IL-18 concentrations and strikingly resulted in loss of adipose tissue mass and fatal cachexia, which could be prevented by genetic deletion of IL-18. Thus, NLRP1 is an innate immune sensor that functions in the context of metabolic stress to produce IL-18, preventing obesity and metabolic syndrome.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Inflamasomas/metabolismo , Interleucina-18/biosíntesis , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Peso Corporal , Dieta Alta en Grasa/efectos adversos , Interleucina-18/genética , Hígado/metabolismo , Hígado/patología , Masculino , Síndrome Metabólico/prevención & control , Ratones Noqueados , Obesidad/etiología , Obesidad/prevención & control
12.
Nat Commun ; 5: 3455, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24632563

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Compuestos de Anilina/farmacología , Animales , Apoptosis/fisiología , Plaquetas/efectos de los fármacos , Western Blotting , Caspasa 8/metabolismo , Electroforesis en Gel de Poliacrilamida , Femenino , Masculino , Ratones , Ratas , Transducción de Señal , Sulfonamidas/farmacología , Trombocitopenia/metabolismo
13.
Cell Metab ; 18(2): 225-38, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23931754

RESUMEN

ABCA12 is involved in the transport of ceramides in skin, but it may play a wider role in lipid metabolism. We show that, in Abca12-deficient macrophages, cholesterol efflux failed to respond to activation with LXR agonists. Abca12 deficiency caused a reduction in the abundance of Abca1, Abcg1, and Lxrß. Overexpression of Lxrß reversed the effects. Mechanistically, Abca12 deficiency did not affect expression of genes involved in cholesterol metabolism. Instead, a physical association between Abca1, Abca12, and Lxrß proteins was established. Abca12 deficiency enhanced interaction between Abca1 and Lxrß and the degradation of Abca1. Overexpression of ABCA12 in HeLa-ABCA1 cells increased the abundance and stability of ABCA1. Abca12 deficiency caused an accumulation of cholesterol in macrophages and the formation of foam cells, impaired reverse cholesterol transport in vivo, and increased the development of atherosclerosis in irradiated Apoe(-/-) mice reconstituted with Apoe(-/-)Abca12(-/-) bone marrow. Thus, ABCA12 regulates the cellular cholesterol metabolism via an LXRß-dependent posttranscriptional mechanism.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Colesterol/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/deficiencia , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Transporte Biológico/genética , Línea Celular , Células Espumosas/metabolismo , Células HeLa , Humanos , Metabolismo de los Lípidos/genética , Lipoproteínas/metabolismo , Receptores X del Hígado , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Nucleares Huérfanos/biosíntesis
14.
J Exp Med ; 208(10): 2017-31, 2011 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-21911424

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Plaquetas/metabolismo , Megacariocitos/citología , Megacariocitos/fisiología , Animales , Supervivencia Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA