Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Immunity ; 45(3): 513-526, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27523270

RESUMEN

The kinases RIPK1 and RIPK3 and the pseudo-kinase MLKL have been identified as key regulators of the necroptotic cell death pathway, although a role for MLKL within the whole animal has not yet been established. Here, we have shown that MLKL deficiency rescued the embryonic lethality caused by loss of Caspase-8 or FADD. Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice were viable and fertile but rapidly developed severe lymphadenopathy, systemic autoimmune disease, and thrombocytopenia. These morbidities occurred more rapidly and with increased severity in Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice compared to Casp8(-/-)Ripk3(-/-) or Fadd(-/-)Ripk3(-/-) mice, respectively. These results demonstrate that MLKL is an essential effector of aberrant necroptosis in embryos caused by loss of Caspase-8 or FADD. Furthermore, they suggest that RIPK3 and/or MLKL may exert functions independently of necroptosis. It appears that non-necroptotic functions of RIPK3 contribute to the lymphadenopathy, autoimmunity, and excess cytokine production that occur when FADD or Caspase-8-mediated apoptosis is abrogated.


Asunto(s)
Apoptosis/fisiología , Enfermedades Autoinmunes/metabolismo , Muerte Celular/fisiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Caspasa 8/metabolismo , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo
2.
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
3.
Blood ; 126(2): 167-75, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26036803

RESUMEN

Polycomb repressive complex 2 (PRC2) is a chromatin modifier that regulates stem cells in embryonic and adult tissues. Loss-of-function studies of PRC2 components have been complicated by early embryonic dependence on PRC2 activity and the partial functional redundancy of enhancer of zeste homolog 1 (Ezh1) and enhancer of zeste homolog 2 (Ezh2), which encode the enzymatic component of PRC2. Here, we investigated the role of PRC2 in hematopoiesis by conditional deletion of suppressor of zeste 12 protein homolog (Suz12), a core component of PRC2. Complete loss of Suz12 resulted in failure of hematopoiesis, both in the embryo and the adult, with a loss of maintenance of hematopoietic stem cells (HSCs). In contrast, partial loss of PRC2 enhanced HSC self-renewal. Although Suz12 was required for lymphoid development, deletion in individual blood cell lineages revealed that it was dispensable for the development of granulocytic, monocytic, and megakaryocytic cells. Collectively, these data reveal the multifaceted role of PRC2 in hematopoiesis, with divergent dose-dependent effects in HSC and distinct roles in maturing blood cells. Because PRC2 is a potential target for cancer therapy, the significant consequences of modest changes in PRC2 activity, as well as the cell and developmental stage-specific effects, will need to be carefully considered in any therapeutic context.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Linfopoyesis/genética , Complejo Represivo Polycomb 2/fisiología , Animales , Proliferación Celular/genética , Células Cultivadas , Feto/inmunología , Feto/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejo Represivo Polycomb 2/genética
4.
Proc Natl Acad Sci U S A ; 111(16): 5884-9, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24711413

RESUMEN

Thrombopoietin (TPO) acting via its receptor, the cellular homologue of the myeloproliferative leukemia virus oncogene (Mpl), is the major cytokine regulator of platelet number. To precisely define the role of specific hematopoietic cells in TPO-dependent hematopoiesis, we generated mice that express the Mpl receptor normally on stem/progenitor cells but lack expression on megakaryocytes and platelets (Mpl(PF4cre/PF4cre)). Mpl(PF4cre/PF4cre) mice displayed profound megakaryocytosis and thrombocytosis with a remarkable expansion of megakaryocyte-committed and multipotential progenitor cells, the latter displaying biological responses and a gene expression signature indicative of chronic TPO overstimulation as the underlying causative mechanism, despite a normal circulating TPO level. Thus, TPO signaling in megakaryocytes is dispensable for platelet production; its key role in control of platelet number is via generation and stimulation of the bipotential megakaryocyte precursors. Nevertheless, Mpl expression on megakaryocytes and platelets is essential to prevent megakaryocytosis and myeloproliferation by restricting the amount of TPO available to stimulate the production of megakaryocytes from the progenitor cell pool.


Asunto(s)
Plaquetas/metabolismo , Megacariocitos/metabolismo , Células Mieloides/citología , Receptores de Trombopoyetina/metabolismo , Trombopoyesis , Animales , Antígenos CD34/metabolismo , Plaquetas/citología , Compartimento Celular , Proliferación Celular , Células Clonales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Marcación de Gen , Sitios Genéticos/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Integrasas/metabolismo , Megacariocitos/citología , Ratones , Modelos Biológicos , Células Mieloides/metabolismo , Trombocitosis , Trombopoyetina/genética , Trombopoyetina/metabolismo , Transcripción Genética
5.
Platelets ; 27(6): 497-504, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27100842

RESUMEN

The lifespan of platelets in circulation is brief, close to 10 days in humans and 5 days in mice. Bone marrow residing megakaryocytes produce around 100 billion platelets per day. In a healthy individual, the majority of platelets are not consumed by hemostatic processes, but rather their lifespan is controlled by programmed cell death, a canonical intrinsic apoptosis program. In the last decade, insights from genetically manipulated mouse models and pharmacological developments have helped to define the components of the intrinsic, or mitochondrial, apoptosis pathway that controls platelet lifespan. This review focuses on the molecular regulation of apoptosis in platelet survival, reviews thrombocytopenic conditions linked to enhanced platelet death, examines implications of chemotherapy-induced thrombocytopenia through apoptosis-inducing drugs in cancer therapy as well as discusses ex vivo aging of platelets.


Asunto(s)
Apoptosis , Plaquetas/fisiología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Conservación de la Sangre , Caspasas/metabolismo , Regulación de la Expresión Génica , Hemostasis , Humanos , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/fisiología , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacología , Transfusión de Plaquetas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/etiología , Trombocitopenia/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.
Blood Adv ; 7(8): 1560-1571, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36075025

RESUMEN

Platelets have been shown to enhance the survival of lymphoma cell lines. However, it remains unclear whether they play a role in lymphoma. Here, we investigated the potential role of platelets and/or megakaryocytes in the progression of Eµ-myc lymphoma. Eµ-myc tumor cells were transplanted into recipient wild-type (WT) control, Mpl-/-, or TpoTg mice, which exhibited normal, low, and high platelet and megakaryocyte counts, respectively. TpoTg mice that underwent transplantation exhibited enhanced lymphoma progression with increased white blood cell (WBC) counts, spleen and lymph node weights, and enhanced liver infiltration when compared with WT mice. Conversely, tumor-bearing Mpl-/- mice had reduced WBC counts, lymph node weights, and less liver infiltration than WT mice. Using an Mpl-deficient thrombocytopenic immunocompromised mouse model, our results were confirmed using the human non-Hodgkin lymphoma GRANTA cell line. Although we found that platelets and platelet-released molecules supported Eµ-myc tumor cell survival in vitro, pharmacological inhibition of platelet function or anticoagulation in WT mice transplanted with Eµ-myc did not improve disease outcome. Furthermore, transient platelet depletion or sustained Bcl-xL-dependent thrombocytopenia did not alter lymphoma progression. Cytokine analysis of the bone marrow fluid microenvironment revealed increased levels of the proinflammatory molecule interleukin 1 in TpoTg mice, whereas these levels were lower in Mpl-/- mice. Moreover, RNA sequencing of blood-resident Eµ-myc lymphoma cells from TpoTg and WT mice after tumor transplantation revealed the upregulation of hallmark gene sets associated with an inflammatory response in TpoTg mice. We propose that the proinflammatory microenvironment in TpoTg mice promotes lymphoma progression.


Asunto(s)
Linfoma , Trombocitopenia , Ratones , Animales , Humanos , Megacariocitos/metabolismo , Receptores de Trombopoyetina , Plaquetas/metabolismo , Trombocitopenia/genética , Linfoma/genética , Microambiente Tumoral
8.
J Biol Chem ; 285(15): 11667-80, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20145240

RESUMEN

In vertebrates, stathmins form a family of proteins possessing two tubulin binding repeats (TBRs), which each binds one soluble tubulin heterodimer. The stathmins thus sequester two tubulins in a phosphorylation-dependent manner, providing a link between signal transduction and microtubule dynamics. In Drosophila, we show here that a single stathmin gene (stai) encodes a family of D-stathmin proteins. Two of the D-stathmins are maternally deposited and then restricted to germ cells, and the other two are detected in the nervous system during embryo development. Like in vertebrates, the nervous system-enriched stathmins contain an N-terminal domain involved in subcellular targeting. All the D-stathmins possess a domain containing three or four predicted TBRs, and we demonstrate here, using complementary biochemical and biophysical methods, that all four predicted TBR domains actually bind tubulin. D-stathmins can indeed bind up to four tubulins, the resulting complex being directly visualized by electron microscopy. Phylogenetic analysis shows that the presence of regulated multiple tubulin sites is a conserved characteristic of stathmins in invertebrates and allows us to predict key residues in stathmin for the binding of tubulin. Altogether, our results reveal that the single Drosophila stathmin gene codes for a stathmin family similar to the multigene vertebrate one, but with particular tubulin binding properties.


Asunto(s)
Unión Proteica , Estatmina/química , Estatmina/genética , Tubulina (Proteína)/química , Animales , Dimerización , Drosophila , Células HeLa , Humanos , Hibridación in Situ , Microtúbulos/metabolismo , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Interferencia de ARN , Proteínas Recombinantes/química , Resonancia por Plasmón de Superficie
9.
Cell Death Dis ; 12(1): 133, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33510145

RESUMEN

Necroptosis is a pro-inflammatory cell death program executed by the terminal effector, mixed lineage kinase domain-like (MLKL). Previous studies suggested a role for the necroptotic machinery in platelets, where loss of MLKL or its upstream regulator, RIPK3 kinase, impacted thrombosis and haemostasis. However, it remains unknown whether necroptosis operates within megakaryocytes, the progenitors of platelets, and whether necroptotic cell death might contribute to or diminish platelet production. Here, we demonstrate that megakaryocytes possess a functional necroptosis signalling cascade. Necroptosis activation leads to phosphorylation of MLKL, loss of viability and cell swelling. Analyses at steady state and post antibody-mediated thrombocytopenia revealed that platelet production was normal in the absence of MLKL, however, platelet activation and haemostasis were impaired with prolonged tail re-bleeding times. We conclude that MLKL plays a role in regulating platelet function and haemostasis and that necroptosis signalling in megakaryocytes is dispensable for platelet production.


Asunto(s)
Plaquetas/metabolismo , Muerte Celular/fisiología , Megacariocitos/metabolismo , Necroptosis/fisiología , Animales , Humanos , Ratones
10.
Sci Rep ; 7(1): 14953, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29097774

RESUMEN

Thrombopoietin (TPO) is the master cytokine regulator of megakaryopoiesis. In addition to regulation of megakaryocyte and platelet number, TPO is important for maintaining proper hematopoietic stem cell (HSC) function. It was previously shown that a number of lymphoid genes were upregulated in HSCs from Tpo -/- mice. We investigated if absent or enhanced TPO signaling would influence normal B-lymphopoiesis. Absent TPO signaling in Mpl -/- mice led to enrichment of a common lymphoid progenitor (CLP) signature in multipotential lineage-negative Sca-1+c-Kit+ (LSK) cells and an increase in CLP formation. Moreover, Mpl -/- mice exhibited increased numbers of PreB2 and immature B-cells in bone marrow and spleen, with an increased proportion of B-lymphoid cells in the G1 phase of the cell cycle. Conversely, elevated TPO signaling in Tpo Tg mice was associated with reduced B-lymphopoiesis. Although at steady state, peripheral blood lymphocyte counts were normal in both models, Mpl -/- Eµ-myc mice showed an enhanced preneoplastic phase with increased numbers of splenic PreB2 and immature B-cells, a reduced quiescent fraction, and augmented blood lymphocyte counts. Thus, although Mpl is not expressed on lymphoid cells, TPO signaling may indirectly influence B-lymphopoiesis and the preneoplastic state in Myc-driven B-cell lymphomagenesis by lineage priming in multipotential progenitor cells.


Asunto(s)
Linfocitos B/citología , Células Progenitoras Linfoides/citología , Linfopoyesis , Transducción de Señal , Trombopoyetina/metabolismo , Animales , Linfocitos B/metabolismo , Ciclo Celular , Femenino , Células Progenitoras Linfoides/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
11.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA