Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 111(29): 10592-7, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002492

RESUMEN

The hematopoietic system declines with age. Myeloid-biased differentiation and increased incidence of myeloid malignancies feature aging of hematopoietic stem cells (HSCs), but the mechanisms involved remain uncertain. Here, we report that 4-mo-old mice deleted for transcription intermediary factor 1γ (Tif1γ) in HSCs developed an accelerated aging phenotype. To reinforce this result, we also show that Tif1γ is down-regulated in HSCs during aging in 20-mo-old wild-type mice. We established that Tif1γ controls TGF-ß1 receptor (Tgfbr1) turnover. Compared with young HSCs, Tif1γ(-/-) and old HSCs are more sensitive to TGF-ß signaling. Importantly, we identified two populations of HSCs specifically discriminated by Tgfbr1 expression level and provided evidence of the capture of myeloid-biased (Tgfbr1(hi)) and myeloid-lymphoid-balanced (Tgfbr1(lo)) HSCs. In conclusion, our data provide a new paradigm for Tif1γ in regulating the balance between lymphoid- and myeloid-derived HSCs through TGF-ß signaling, leading to HSC aging.


Asunto(s)
Senescencia Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factores de Transcripción/metabolismo , Envejecimiento/metabolismo , Animales , Antígenos CD/metabolismo , Separación Celular , Senescencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Células Mieloides/metabolismo , Fenotipo , Poliubiquitina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta1/farmacología , Ubiquitinación/efectos de los fármacos
2.
Proc Natl Acad Sci U S A ; 109(2): E68-75, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22065775

RESUMEN

The inactivation of the p53 tumor suppressor pathway in many cancers often increases their resistance to anticancer therapy. Here we show that a previously proposed strategy directed to Wip1 inhibition could be ineffective in tumors lacking p53. On the contrary, Wip1 overexpression sensitized these tumors to chemotherapeutic agents. This effect was mediated through interaction between Wip1 and RUNX2 that resulted, in response to anticancer treatment, in RUNX2-dependent transcriptional induction of the proapoptotic Bax protein. The potentiating effects of Wip1 overexpression on chemotherapeutic agents were directed only to tumor cells lacking p53. The overexpression of Wip1 in normal tissues provided protection from cisplatin-induced apoptosis through decreased strength of upstream signaling to p53. Thus, Wip1 phosphatase promotes apoptosis in p53-negative tumors and protects normal tissues during treatment with anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias/tratamiento farmacológico , Fosfoproteínas Fosfatasas/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Línea Celular , Cartilla de ADN/genética , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Neoplasias/metabolismo , Plásmidos/genética , Proteína Fosfatasa 2C , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
3.
FASEB J ; 27(4): 1549-60, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23288928

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by myofibroblast proliferation. Transition of epithelial/mesothelial cells into myofibroblasts [epithelial-to-mesenchymal transition (EMT)] occurs under the influence of transforming growth factor (TGF)-ß1, with Snail being a major transcription factor. We study here the role of the heat-shock protein HSP27 in fibrogenesis and EMT. In vitro, we have up- and down-modulated HSP27 expression in mesothelial and epithelial cell lines and studied the expression of different EMT markers induced by TGF-ß1. In vivo, we inhibited HSP27 with the antisense oligonucleotide OGX-427 (in phase II clinical trials as anticancer agent) in our rat subpleural/pulmonary fibrosis models. We demonstrate that HSP27 is strongly expressed during the fibrotic process in patients with IPF and in different in vivo models. We showed that HSP27 binds to and stabilizes Snail and consequently induces EMT. Conversely, HSP27 knockdown leads to Snail proteasomal degradation, thus inhibiting TGF-ß1-induced EMT. Inhibition of HSP27 with OGX-427 efficiently blocks EMT and fibrosis development. Controls in vivo were an empty adenovirus that did not induce fibrosis and a control antisense oligonucleotide. The present work opens the possibility of a new therapeutic use for HSP27 inhibitors against IPF, for which there is no conclusively effective treatment.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas de Choque Térmico HSP27/antagonistas & inhibidores , Caracoles/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Cadherinas/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Fibrosis/metabolismo , Humanos , Oligonucleótidos Antisentido/farmacología , Ratas , Ratas Sprague-Dawley , Tionucleótidos/farmacología , Factores de Transcripción/metabolismo
4.
J Biol Chem ; 286(5): 3418-28, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21127066

RESUMEN

Extracellular heat shock protein HSP90α was reported to participate in tumor cell growth, invasion, and metastasis formation through poorly understood signaling pathways. Herein, we show that extracellular HSP90α favors cell migration of glioblastoma U87 cells. More specifically, externally applied HSP90α rapidly induced endocytosis of EGFR. This response was accompanied by a transient increase in cytosolic Ca(2+) appearing after 1-3 min of treatment. In the presence of EGF, U87 cells showed HSP90α-induced Ca(2+) oscillations, which were reduced by the ATP/ADPase, apyrase, and inhibited by the purinergic P(2) inhibitor, suramin, suggesting that ATP release is requested. Disruption of lipid rafts with methyl ß-cyclodextrin impaired the Ca(2+) rise induced by extracellular HSP90α combined with EGF. Specific inhibition of TLR4 expression by blocking antibodies suppressed extracellular HSP90α-induced Ca(2+) signaling and the associated cell migration. HSPs are known to bind lipopolysaccharides (LPSs). Preincubating cells with Polymyxin B, a potent LPS inhibitor, partially abrogated the effects of HSP90α without affecting Ca(2+) oscillations observed with EGF. Extracellular HSP90α induced EGFR phosphorylation at Tyr-1068, and this event was prevented by both the protein kinase Cδ inhibitor, rottlerin, and the c-Src inhibitor, PP2. Altogether, our results suggest that extracellular HSP90α transactivates EGFR/ErbB1 through TLR4 and a PKCδ/c-Src pathway, which induces ATP release and cytosolic Ca(2+) increase and finally favors cell migration. This mechanism could account for the deleterious effects of HSPs on high grade glioma when released into the tumor cell microenvironment.


Asunto(s)
Movimiento Celular , Receptores ErbB/genética , Glioblastoma/patología , Proteínas HSP90 de Choque Térmico/fisiología , Receptor Toll-Like 4/metabolismo , Activación Transcripcional , Adenosina Trifosfato/metabolismo , Señalización del Calcio , Línea Celular Tumoral , Humanos , Microdominios de Membrana , Proteína Quinasa C-delta/metabolismo
5.
J Biol Chem ; 286(30): 26406-17, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21653699

RESUMEN

The inhibitor of apoptosis protein cIAP1 (cellular inhibitor of apoptosis protein-1) is a potent regulator of the tumor necrosis factor (TNF) receptor family and NF-κB signaling pathways in the cytoplasm. However, in some primary cells and tumor cell lines, cIAP1 is expressed in the nucleus, and its nuclear function remains poorly understood. Here, we show that the N-terminal part of cIAP1 directly interacts with the DNA binding domain of the E2F1 transcription factor. cIAP1 dramatically increases the transcriptional activity of E2F1 on synthetic and CCNE promoters. This function is not conserved for cIAP2 and XIAP, which are cytoplasmic proteins. Chromatin immunoprecipitation experiments demonstrate that cIAP1 is recruited on E2F binding sites of the CCNE and CCNA promoters in a cell cycle- and differentiation-dependent manner. cIAP1 silencing inhibits E2F1 DNA binding and E2F1-mediated transcriptional activation of the CCNE gene. In cells that express a nuclear cIAP1 such as HeLa, THP1 cells and primary human mammary epithelial cells, down-regulation of cIAP1 inhibits cyclin E and A expression and cell proliferation. We conclude that one of the functions of cIAP1 when localized in the nucleus is to regulate E2F1 transcriptional activity.


Asunto(s)
Núcleo Celular/metabolismo , Ciclina A/biosíntesis , Ciclina E/biosíntesis , Factor de Transcripción E2F1/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Elementos de Respuesta/fisiología , Transcripción Genética/fisiología , Animales , Núcleo Celular/genética , Proliferación Celular , Ciclina A/genética , Ciclina E/genética , Factor de Transcripción E2F1/genética , Silenciador del Gen , Células HeLa , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Ratones , Estructura Terciaria de Proteína
6.
Blood ; 116(1): 85-96, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20410505

RESUMEN

Heat shock protein 27 (HSP27) is a chaperone whose cellular expression increases in response to various stresses and protects the cell either by inhibiting apoptotic cell death or by promoting the ubiquitination and proteasomal degradation of specific proteins. Here, we show that globin transcription factor 1 (GATA-1) is a client protein of HSP27. In 2 models of erythroid differentiation; that is, in the human erythroleukemia cell line, K562 induced to differentiate into erythroid cells on hemin exposure and CD34(+) human cells ex vivo driven to erythroid differentiation in liquid culture, depletion of HSP27 provokes an accumulation of GATA-1 and impairs terminal maturation. More specifically, we demonstrate that, in the late stages of the erythroid differentiation program, HSP27 is phosphorylated in a p38-dependent manner, enters the nucleus, binds to GATA-1, and induces its ubiquitination and proteasomal degradation, provided that the transcription factor is acetylated. We conclude that HSP27 plays a role in the fine-tuning of terminal erythroid differentiation through regulation of GATA-1 content and activity.


Asunto(s)
Diferenciación Celular , Células Eritroides/metabolismo , Factor de Transcripción GATA1/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Animales , Antígenos CD34/sangre , Células COS , Núcleo Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Células Eritroides/citología , Células Eritroides/efectos de los fármacos , Factor de Transcripción GATA1/genética , Proteínas de Choque Térmico HSP27/genética , Células HeLa , Proteínas de Choque Térmico , Humanos , Imidazoles/farmacología , Immunoblotting , Interleucina-6/farmacología , Células K562 , Leupeptinas/farmacología , Chaperonas Moleculares , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Unión Proteica , Piridinas/farmacología , Interferencia de ARN , Factor de Crecimiento Transformador beta/farmacología , Ubiquitinación/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
FASEB J ; 24(9): 3544-54, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20418497

RESUMEN

Hexaacyl lipopolysaccharide (LPS) aggregates in aqueous media, but its partially deacylated lipid A moiety forms monomers with weaker toxicity. Because plasma phospholipid transfer protein (PLTP) transfers hexaacyl LPS, its impact on metabolism and biological activity of triacyl lipid A in mice was addressed. Triacyl lipid A bound readily to plasma high-density lipoproteins (HDLs) when active PLTP was expressed [HDL-associated lipid A after 4.5 h: 59.1+/-16.0% of total in wild-type (WT) vs. 32.5+/-10.3% in PLTP-deficient mice, P<0.05]. In the opposite to hexaacyl LPS, plasma residence time of lipid A was extended by PLTP, and proinflammatory cytokines were produced in higher amounts in WT than PLTP(-/-) mice (remaining lipid A after 8 h: 53+/-12 vs. 35+/-7%, and IL6 concentration after 4.5 h: 45.5+/-5.9 vs. 14.6+/-7.8 ng/ml, respectively; P<0.05 in all cases). After 1 wk, onset of B16-induced melanoma was observed in only 30% of lipid A-treated WT mice, whereas >80% of the untreated WT, untreated PLTP-deficient, or lipid A-treated PLTP-deficient animals bore tumors (P<0.05 in all cases). It is concluded that PLTP is essential in mediating the association of triacyl lipid A with lipoproteins, leading to extension of its residence time and to magnification of its proinflammatory and anticancer properties.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata/fisiología , Lípido A/inmunología , Lípido A/farmacología , Proteínas de Transferencia de Fosfolípidos/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Quimiocina CCL2/sangre , Citocinas/sangre , Citometría de Flujo , Inmunidad Innata/genética , Interferón gamma/sangre , Interleucina-10/sangre , Interleucina-6/sangre , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Mutantes , Proteínas de Transferencia de Fosfolípidos/genética , Factor de Necrosis Tumoral alfa/sangre
9.
J Mol Med (Berl) ; 97(5): 633-645, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30843084

RESUMEN

Cohen syndrome (CS) is a rare genetic disorder due to mutations in VPS13B gene. Among various clinical and biological features, CS patients suffer from inconsistent neutropenia, which is associated with recurrent but minor infections. We demonstrate here that this neutropenia results from an exaggerate rate of neutrophil apoptosis. Besides this increased cell death, which occurs in the absence of any endoplasmic reticulum stress or defect in neutrophil elastase (ELANE) expression or localization, all neutrophil functions appeared to be normal. We showed a disorganization of the Golgi apparatus in CS neutrophils precursors, that correlates with an altered glycosylation of ICAM-1 in these cells, as evidenced by a migration shift of the protein. Furthermore, a striking decrease in the expression of SERPINB1 gene, which encodes a critical component of neutrophil survival, was detected in CS neutrophils. These abnormalities may account for the excessive apoptosis of neutrophils leading to neutropenia in CS. KEY MESSAGES: Cohen syndrome patients' neutrophils display normal morphology and functions. Cohen syndrome patients' neutrophils have an increased rate of spontaneous apoptosis compared to healthy donors' neutrophils. No ER stress or defective ELA2 expression or glycosylation was observed in Cohen syndrome patients' neutrophils. SerpinB1 expression is significantly decreased in Cohen syndrome neutrophils as well as in VPS13B-deficient cells.


Asunto(s)
Apoptosis , Dedos/anomalías , Discapacidad Intelectual/genética , Microcefalia/genética , Hipotonía Muscular/genética , Miopía/genética , Neutropenia/genética , Neutrófilos/patología , Obesidad/genética , Degeneración Retiniana/genética , Serpinas/genética , Adolescente , Adulto , Niño , Preescolar , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Regulación hacia Abajo , Femenino , Dedos/patología , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Masculino , Microcefalia/complicaciones , Microcefalia/patología , Persona de Mediana Edad , Hipotonía Muscular/complicaciones , Hipotonía Muscular/patología , Mutación , Miopía/complicaciones , Miopía/patología , Neutropenia/etiología , Neutropenia/patología , Neutrófilos/metabolismo , Obesidad/complicaciones , Obesidad/patología , Degeneración Retiniana/complicaciones , Degeneración Retiniana/patología , Adulto Joven
10.
Br J Haematol ; 143(3): 378-82, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18729850

RESUMEN

The inv(8)(p11q13) chromosomal abnormality, described in acute myeloid leukaemias (AML), fuses the histone acetyl-transferase (HAT) MYST3 (MOZ) gene with another HAT gene, NCOA2 (TIF2). We generated a transgenic zebrafish in which the MYST3/NCOA2 fusion gene was expressed under control of the spi1 promoter. An AML developed in 2 of 180 MYST3/NCOA2-EGFP-expressing embryos, 14 and 26 months after injection of the fusion gene in a one-cell embryo, respectively. This leukaemia was characterised by an extensive invasion of kidneys by myeloid blast cells. This model, which is the first zebrafish model of AML, demonstrates the oncogenic potency of MYST3/NCOA2 fusion gene.


Asunto(s)
Histona Acetiltransferasas/genética , Leucemia Mieloide Aguda/genética , Coactivador 2 del Receptor Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Fusión Génica , Histona Acetiltransferasas/metabolismo , Riñón/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Microinyecciones/métodos , Coactivador 2 del Receptor Nuclear/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Pez Cebra/genética
11.
Cancer Res ; 66(8): 4191-7, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16618741

RESUMEN

When overexpressed, the stress protein heat shock protein 70 (HSP70) increases the oncogenic potential of cancer cells in rodent models. HSP70 also prevents apoptosis, thereby increasing the survival of cells exposed to a wide range of otherwise lethal stimuli. These protective functions of HSP70 involve its interaction with and neutralization of the adaptor molecule apoptotic protease activation factor-1, implicated in caspase activation, and the flavoprotein apoptosis-inducing factor (AIF), involved in caspase-independent cell death. We have shown previously that a peptide containing the AIF sequence involved in its interaction with HSP70 (ADD70, amino acids 150-228) binds to and neutralizes HSP70 in the cytosol, thereby sensitizing cancer cells to apoptosis induced by a variety of death stimuli. Here, we show that expression of ADD70 in tumor cells decreases their tumorigenicity in syngeneic animals without affecting their growth in immunodeficient animals. ADD70 antitumorigenic effects are associated with an increase in tumor-infiltrating cytotoxic CD8+ T cells. In addition, ADD70 sensitizes rat colon cancer cells (PROb) and mouse melanoma cells (B16F10) to the chemotherapeutic agent cisplatin. ADD70 also shows an additive effect with HSP90 inhibition by 17-allylamino-17-demethoxygeldanamycin in vitro. Altogether, these data indicate the potential interest of targeting the HSP70 interaction with AIF for cancer therapy.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Neoplasias Colorrectales/terapia , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Melanoma Experimental/terapia , Fragmentos de Péptidos/genética , Animales , Factor Inductor de la Apoptosis/biosíntesis , Benzoquinonas , Linfocitos T CD8-positivos/inmunología , Cisplatino/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Terapia Combinada , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/biosíntesis , Ratas , Ratas Desnudas , Rifabutina/análogos & derivados , Rifabutina/farmacología , Transfección
12.
FASEB J ; 20(8): 1179-81, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16641199

RESUMEN

Stress-inducible HSP27 protects cells from death through various mechanisms. We have recently demonstrated that HSP27 can also enhance the degradation of some proteins through the proteasomal pathway. Here, we show that one of these proteins is the cyclin-dependent kinase (Cdk) inhibitor p27Kip1. The ubiquitination and degradation of this protein that favors progression through the cell cycle was previously shown to involve either a Skp2-dependent mechanism,i.e., at the S-/G2-transition, or a KPC (Kip1 ubiquitination-promoting complex)-dependent mechanism, i.e.,at the G0/G1 transition. In this work, we demonstrate that, in response to serum depletion, p27Kip1 cellular content first increases then progressively decreases as cells begin to die. In this stressful condition, HSP27favors p27Kip1 ubiquitination and degradation by the proteasome. A similar observation was made in response to stress induced by the NO donor glyceryl trinitrate (GTN). HSP27-mediated ubiquitination ofp27Kip1 does not require its phosphorylation on Thr187 or Ser-10, nor does it depend on the SCFSkp2 ubiquitin ligase E3 complex. It facilitates the G1/S transition,which suggests that, in stressful conditions, HSP27might render quiescent cells competent to re-enter the cell cycle.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Fase S , Ubiquitina/metabolismo , Animales , Línea Celular , Medio de Cultivo Libre de Suero , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Fase G1 , Proteínas de Choque Térmico HSP27 , Humanos , Chaperonas Moleculares , Fosforilación , Ratas , Fase de Descanso del Ciclo Celular , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 26(9): 2160-7, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16825594

RESUMEN

OBJECTIVE: The aim of the present study was to assess the effect of alpha-tocopherol, the main vitamin E isomer on phosphatidylserine (PS) exposure at the surface of circulating erythrocytes, and to determine consequences on erythrocyte properties. METHODS AND RESULTS: In vitro alpha-tocopherol enrichment of isolated erythrocytes significantly decreased PS externalization as assessed by lower Annexin V-fluorescein isothiocyanate labeling. Plasma phospholipid transfer protein (PLTP) transfers vitamin E, and both alpha- and gamma-tocopherol accumulated in circulating erythrocytes from PLTP-deficient homozygous (PLTP-/-) mice as compared with wild-type mice. In agreement with in vitro studies, vitamin E-enriched erythrocytes from PLTP-/- mice displayed fewer externalized PS molecules than wild-type controls (-64%, P<0.05). The perturbation of phospholipid membrane asymmetry from PLTP-/- erythrocytes was accompanied by impairment of their procoagulant properties, with a 20% increase in clotting time as compared with wild-type controls (P<0.05). Less pronounced, however still significant, changes were observed in alpha-tocopherol content, procoagulant properties, and PS externalization in erythrocytes of PLTP-deficient heterozygotes. Finally, whole blood coagulation and circulating level of D-dimer, which reflects increased thrombus formation in vivo, were significantly decreased in PLTP-/- mice compared with wild-type mice. CONCLUSIONS: Vitamin E modifies PS externalization in circulating erythrocytes, thus modulating in vivo their PS-dependent procoagulant properties.


Asunto(s)
Membrana Eritrocítica/metabolismo , Fosfatidilserinas/sangre , Proteínas de Transferencia de Fosfolípidos/deficiencia , alfa-Tocoferol/farmacología , Animales , Biomarcadores/sangre , Coagulación Sanguínea/fisiología , Separación Celular , Eritrocitos/fisiología , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Homocigoto , Ratones , Ratones Noqueados , Oxidación-Reducción , Fenotipo , Tiempo de Coagulación de la Sangre Total
14.
JCI Insight ; 2(6): e90531, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28352659

RESUMEN

Better identification of severe acute graft-versus-host disease (GvHD) may improve the outcome of this life-threatening complication of allogeneic hematopoietic stem cell transplantation. GvHD induces tissue damage and the release of damage-associated molecular pattern (DAMP) molecules. Here, we analyzed GvHD patients (n = 39) to show that serum heat shock protein glycoprotein 96 (Gp96) could be such a DAMP molecule. We demonstrate that serum Gp96 increases in gastrointestinal GvHD patients and its level correlates with disease severity. An increase in Gp96 serum level was also observed in a mouse model of acute GvHD. This model was used to identify complement C3 as a main partner of Gp96 in the serum. Our biolayer interferometry, yeast two-hybrid and in silico modeling data allowed us to determine that Gp96 binds to a complement C3 fragment encompassing amino acids 749-954, a functional complement C3 hot spot important for binding of different regulators. Accordingly, in vitro experiments with purified proteins demonstrate that Gp96 downregulates several complement C3 functions. Finally, experimental induction of GvHD in complement C3-deficient mice confirms the link between Gp96 and complement C3 in the serum and with the severity of the disease.


Asunto(s)
Complemento C3/metabolismo , Enfermedad Injerto contra Huésped/sangre , Glicoproteínas de Membrana/sangre , Chaperonas Moleculares/sangre , Adolescente , Adulto , Animales , Activación de Complemento , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Persona de Mediana Edad , Adulto Joven
15.
Cancer Res ; 64(10): 3593-8, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15150117

RESUMEN

We have shown previously that the death receptor CD95 could contribute to anticancer drug-induced apoptosis of colon cancer cells. In addition, anticancer drugs cooperate with CD95 cognate ligand or agonistic antibodies to trigger cancer cell apoptosis. In the present study, we show that the anticancer drug cisplatin induces clustering of CD95 at the surface of the human colon cancer cell line HT29, an event inhibited by the inhibitor of acid sphingomyelinase (aSMase) imipramine. The cholesterol sequestering agent nystatin also prevents cisplatin-induced CD95 clustering and decreases HT29 cell sensitivity to cisplatin-induced apoptosis and the synergy between cisplatin and anti-CD95 agonistic antibodies. CD95, together with the adaptor molecule Fas-associated death domain and procaspase-8, is redistributed into cholesterol- and sphingolipid-enriched cell fractions after cisplatin treatment, suggesting plasma membrane raft involvement. Interestingly, nystatin prevents the translocation of the aSMase to the extracellular surface of plasma membrane and the production of ceramide, suggesting that these early events require raft integrity. In addition, nystatin prevents cisplatin-induced transient increase in plasma membrane fluidity that could be required for CD95 translocation. Together, these results demonstrate that cisplatin activates aSMase and induces ceramide production, which triggers the redistribution of CD95 into the plasma membrane rafts. Such redistribution contributes to cell death and sensitizes tumor cells to CD95-mediated apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Microdominios de Membrana/metabolismo , Receptor fas/metabolismo , Antineoplásicos/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Ceramidas/biosíntesis , Cisplatino/antagonistas & inhibidores , Interacciones Farmacológicas , Células HT29 , Humanos , Fluidez de la Membrana/efectos de los fármacos , Microdominios de Membrana/efectos de los fármacos , Nistatina/farmacología , Esfingomielina Fosfodiesterasa/metabolismo
16.
Cancer Res ; 64(8): 2705-11, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15087383

RESUMEN

We and others have previously reported in an in vivo rat colon cancer cell model that cell death precedes and is necessary for the development of a specific antitumoral immune response. To sensitize colon cancer cells to death, we depleted cytochrome c by stable transfection with an antisense construct. Cytochrome c depletion sensitizes human and rat colon cancer cells to a nonapoptotic, nonautophagic death induced by various stimuli. This increased sensitization to a necrosis-like cell death may be related to a decrease in cellular ATP levels and an increase in reactive oxygen species production caused by cytochrome c depletion. In vivo, depletion of cytochrome c decreases the tumorigenicity of colon cancer cells in syngeneic rats without influencing their growth in immune-deficient animals. Furthermore, decreased expression of cytochrome c in tumor cells facilitates in vivo "necrotic" cell death and the induction of a specific immune response. These results delineate a novel strategy to sensitize colon cancer cells to chemotherapy and to increase their immunogenicity in immuno-competent hosts.


Asunto(s)
Neoplasias del Colon/inmunología , Citocromos c/deficiencia , Citocromos c/inmunología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Cisplatino/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Citocromos c/biosíntesis , Citocromos c/genética , ADN sin Sentido/genética , Regulación hacia Abajo , Doxorrubicina/farmacología , Etopósido/farmacología , Células HCT116 , Células HT29 , Humanos , Macrófagos/inmunología , Ratones , Ratas , Estaurosporina/farmacología , Transfección
17.
Cancer Res ; 63(23): 8233-40, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678980

RESUMEN

Heat shock protein 70 (HSP70) inhibits apoptosis and thereby increases the survival of cells exposed to a wide range of lethal stimuli. HSP70 has also been shown to increase the tumorigenicity of cancer cells in rodent models. The protective function of this chaperone involves interaction and neutralization of the caspase activator apoptotic protease activation factor-1 and the mitochondrial flavoprotein apoptosis-inducing factor (AIF). In this work, we determined by deletional mutagenesis that a domain of AIF comprised between amino acids 150 and 228 is engaged in a molecular interaction with the substrate-binding domain of HSP70. Computer calculations favored this conclusion. On the basis of this information, we constructed an AIF-derived protein, which is cytosolic, noncytotoxic, yet maintains its capacity to interact with HSP70. This protein, designated ADD70, sensitized different human cancer cells to apoptosis induced by a variety of death stimuli by its capacity to interact with HSP70 and therefore to sequester HSP70. Thus, its chemosensitizing effect was lost in cells in which inducible HSP70 genes had been deleted. These data delineate a novel strategy for the selective neutralization of HSP70.


Asunto(s)
Flavoproteínas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de la Membrana/metabolismo , Apoptosis/fisiología , Factor Inductor de la Apoptosis , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Simulación por Computador , Flavoproteínas/genética , Proteínas Fluorescentes Verdes , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Proteínas de la Membrana/genética , Modelos Moleculares , Mutagénesis , Mapeo Peptídico , Conformación Proteica , Estructura Terciaria de Proteína , Transfección
18.
Oncotarget ; 7(19): 28160-8, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27058413

RESUMEN

Gap junctional communication between cancer cells and blood capillary cells is crucial to tumor growth and invasion. Gap junctions may transfer microRNAs (miRs) among cells. Here, we explore the impact of such a transfer in co-culture assays, using the antitumor miR-145 as an example. The SW480 colon carcinoma cells form functional gap junction composed of connexin-43 (Cx43) with human microvascular endothelial cells (HMEC). When HMEC are loaded with miR-145-5p mimics, the miR-145 level drastically increases in SW480. The functional inhibition of gap junctions, using either a gap channel blocker or siRNA targeting Cx43, prevents this increase. The transfer of miR-145 also occurs from SW480 to HMEC but not in non-contact co-cultures, excluding the involvement of soluble exosomes. The miR-145 transfer to SW480 up-regulates their Cx43 expression and inhibits their ability to promote angiogenesis. Our results indicate that the gap junctional communication can inhibit tumor growth by transferring miRs from one endothelial cell to neighboring tumor cells. This "bystander" effect could find application in cancer therapy.


Asunto(s)
Comunicación Celular/fisiología , Neoplasias del Colon/metabolismo , Células Endoteliales/metabolismo , Uniones Comunicantes/metabolismo , MicroARNs/metabolismo , Neovascularización Patológica/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Neoplasias del Colon/patología , Humanos , Neovascularización Patológica/patología
19.
Oncotarget ; 7(45): 73925-73934, 2016 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-27661112

RESUMEN

Extensive invasion and angiogenesis are hallmark features of malignant glioblastomas. Here, we co-cultured U87 human glioblastoma cells and human microvascular endothelial cells (HMEC) to demonstrate the exchange of microRNAs that initially involve the formation of gap junction communications between the two cell types. The functional inhibition of gap junctions by carbenoxolone blocks the transfer of the anti-tumor miR-145-5p from HMEC to U87, and the transfer of the pro-invasive miR-5096 from U87 to HMEC. These two microRNAs exert opposite effects on angiogenesis in vitro. MiR-5096 was observed to promote HMEC tubulogenesis, initially by increasing Cx43 expression and the formation of heterocellular gap junctions, and secondarily through a gap-junction independent pathway. Our results highlight the importance of microRNA exchanges between tumor and endothelial cells that in part involves the formation of functional gap junctions between the two cell types.


Asunto(s)
Células Endoteliales/metabolismo , Uniones Comunicantes/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , MicroARNs/genética , Comunicación Celular/genética , Línea Celular Tumoral , Expresión Génica , Glioblastoma/patología , Humanos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Transporte de ARN
20.
Exp Hematol ; 44(8): 727-739.e6, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27130375

RESUMEN

Trim33/Tif1γ (Trim33) is a member of the tripartite motif family. Using a conditional hematopoietic-specific Trim33 knock-out (Trim33(Δ/Δ)) mouse, we showed previously that Trim33 deficiency in hematopoietic stem cells leads to severe defects in hematopoiesis, resembling the main features of human chronic myelomonocytic leukemia. We also demonstrated that Trim33 is involved in hematopoietic aging through TGFß signaling. Nevertheless, how Trim33 contributes to the terminal stages of myeloid differentiation remains to be clarified. We reveal here the crucial role of Trim33 expression in the control of mature granulomonocytic differentiation. An important component of Trim33-deficient mice is the alteration of myeloid differentiation, as characterized by dysplastic features, abnormal granulocyte and monocyte maturation, and the expansion of CD11b(+)Ly6G(high)Ly6C(low) myeloid cells, which share some features with polymorphonuclear-myeloid-derived suppressor cells. Moreover, in Trim33(Δ/Δ) mice, we observed the alteration of CSF-1-mediated macrophage differentiation in association with the lack of Csf-1 receptor. Altogether, these results indicate that Trim33 deficiency leads to the expansion of a subset of myeloid cells characterizing the myelodysplastic/myeloproliferative neoplasm.


Asunto(s)
Diferenciación Celular/genética , Células Progenitoras de Granulocitos y Macrófagos/citología , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Mielopoyesis/genética , Factores de Transcripción/genética , Animales , Apoptosis/genética , Biomarcadores , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Linaje de la Célula , Movimiento Celular/genética , Modelos Animales de Enfermedad , Inmunofenotipificación , Ratones , Ratones Noqueados , Células Mieloides , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA