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











Base de datos
Intervalo de año de publicación
2.
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
3.
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
4.
Oncoimmunology ; 5(7): e1170264, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27622020

RESUMEN

HSP110 is induced by different stresses and, through its anti-apoptotic and chaperoning properties, helps the cells to survive these adverse situations. In colon cancers, HSP110 is abnormally abundant. We have recently showed that colorectal cancer (CRC) patients with microsatellite instability (MSI) had an improved response to chemotherapy because they harbor an HSP110 inactivating mutation (HSP110DE9). In this work, we have used patients' biopsies and human CRC cells grown in vitro and in vivo (xenografts) to demonstrate that (1) HSP110 is secreted by CRC cells and that the amount of this extracellular HSP110 is strongly decreased by the expression of the mutant HSP110DE9, (2) Supernatants from CRC cells overexpressing HSP110 or purified recombinant human HSP110 (LPS-free) affect macrophage differentiation/polarization by favoring a pro-tumor, anti-inflammatory profile, (3) Conversely, inhibition of HSP110 (expression of siRNA, HSP110DE9 or immunodepletion) induced the formation of macrophages with a cytotoxic, pro-inflammatory profile. (4) Finally, this effect of extracellular HSP110 on macrophages seems to implicate TLR4. These results together with the fact that colorectal tumor biopsies with HSP110 high were infiltrated with macrophages with a pro-tumoral profile while those with HSP110 low were infiltrated with macrophages with a cytotoxic profile, suggest that the effect of extracellular HSP110 function on macrophages may also contribute to the poor outcomes associated with HSP110 expression.

5.
Oncotarget ; 7(40): 64785-64797, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27579617

RESUMEN

CD45 is a pan-leukocyte protein with tyrosine phosphatase activity involved in the regulation of signal transduction in hematopoiesis. Exploiting CD45 KO mice and lentiviral shRNA, we prove the crucial role that CD45 plays in acute myeloid leukemia (AML) development and maintenance. We discovered that CD45 does not colocalize with lipid rafts on murine and human non-transformed hematopoietic cells. Using a mouse model, we proved that CD45 positioning within lipid rafts is modified during their oncogenic transformation to AML. CD45 colocalized with lipid rafts on AML cells, which contributes to elevated GM-CSF signal intensity involved in proliferation of leukemic cells. We furthermore proved that the GM-CSF/Lyn/Stat3 pathway that contributes to growth of leukemic cells could be profoundly affected, by using a new plasma membrane disrupting agent, which rapidly delocalized CD45 away from lipid rafts. We provide evidence that this mechanism is also effective on human primary AML samples and xenograft transplantation. In conclusion, this study highlights the emerging evidence of the involvement of lipid rafts in oncogenic development of AML and the targeting of CD45 positioning among lipid rafts as a new strategy in the treatment of AML.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Microdominios de Membrana/metabolismo , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Femenino , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Hematopoyesis/genética , Humanos , Lentivirus/genética , Leucemia Mieloide Aguda/patología , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
6.
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
7.
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
8.
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
9.
Cell Mol Immunol ; 13(6): 795-804, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26320740

RESUMEN

OBJECTIVE: Plasma phospholipid transfer protein (PLTP) is a key determinant of lipoprotein metabolism, and both animal and human studies converge to indicate that PLTP promotes atherogenesis and its thromboembolic complications. Moreover, it has recently been reported that PLTP modulates inflammation and immune responses. Although earlier studies from our group demonstrated that PLTP can modify macrophage activation, the implication of PLTP in the modulation of T-cell-mediated immune responses has never been investigated and was therefore addressed in the present study. Approach and results: In the present study, we demonstrated that PLTP deficiency in mice has a profound effect on CD4+ Th0 cell polarization, with a shift towards the anti-inflammatory Th2 phenotype under both normal and pathological conditions. In a model of contact hypersensitivity, a significantly impaired response to skin sensitization with the hapten-2,4-dinitrofluorobenzene (DNFB) was observed in PLTP-deficient mice compared to wild-type (WT) mice. Interestingly, PLTP deficiency in mice exerted no effect on the counts of total white blood cells, lymphocytes, granulocytes, or monocytes in the peripheral blood. Moreover, PLTP deficiency did not modify the amounts of CD4+ and CD8+ T lymphocyte subsets. However, PLTP-deficiency, associated with upregulation of the Th2 phenotype, was accompanied by a significant decrease in the production of the pro-Th1 cytokine interleukin 18 by accessory cells. CONCLUSIONS: For the first time, this work reports a physiological role for PLTP in the polarization of CD4+ T cells toward the pro-inflammatory Th1 phenotype.


Asunto(s)
Inmunidad Adaptativa , Polaridad Celular/inmunología , Proteínas de Transferencia de Fosfolípidos/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Biomarcadores/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Citometría de Flujo , Factor de Transcripción GATA3/metabolismo , Hipersensibilidad Tardía/inmunología , Hipersensibilidad Tardía/patología , Recuento de Leucocitos , Ratones Endogámicos C57BL , Proteínas de Transferencia de Fosfolípidos/deficiencia , Bazo/citología , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/metabolismo
10.
J Natl Cancer Inst ; 108(3)2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26598503

RESUMEN

BACKGROUND: Exosomes, via heat shock protein 70 (HSP70) expressed in their membrane, are able to interact with the toll-like receptor 2 (TLR2) on myeloid-derived suppressive cells (MDSCs), thereby activating them. METHODS: We analyzed exosomes from mouse (C57Bl/6) and breast, lung, and ovarian cancer patient samples and cultured cancer cells with different approaches, including nanoparticle tracking analysis, biolayer interferometry, FACS, and electron microscopy. Data were analyzed with the Student's t and Mann-Whitney tests. All statistical tests were two-sided. RESULTS: We showed that the A8 peptide aptamer binds to the extracellular domain of membrane HSP70 and used the aptamer to capture HSP70 exosomes from cancer patient samples. The number of HSP70 exosomes was higher in cancer patients than in healthy donors (mean, ng/mL ± SD = 3.5 ± 1.7 vs 0.17 ± 0.11, respectively, P = .004). Accordingly, all cancer cell lines examined abundantly released HSP70 exosomes, whereas "normal" cells did not. HSP70 had higher affinity for A8 than for TLR2; thus, A8 blocked HSP70/TLR2 association and the ability of tumor-derived exosomes to activate MDSCs. Treatment of tumor-bearing C57Bl/6 mice with A8 induced a decrease in the number of MDSCs in the spleen and inhibited tumor progression (n = 6 mice per group). Chemotherapeutic agents such as cisplatin or 5FU increase the amount of HSP70 exosomes, favoring the activation of MDSCs and hampering the development of an antitumor immune response. In contrast, this MDSC activation was not observed if cisplatin or 5FU was combined with A8. As a result, the antitumor effect of the drugs was strongly potentiated. CONCLUSIONS: A8 might be useful for quantifying tumor-derived exosomes and for cancer therapy through MDSC inhibition.


Asunto(s)
Aptámeros de Péptidos/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias del Colon/inmunología , Exosomas/inmunología , Proteínas HSP70 de Choque Térmico/metabolismo , Neoplasias Pulmonares/inmunología , Células Mieloides/inmunología , Neoplasias Ováricas/inmunología , Receptor Toll-Like 2/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/tratamiento farmacológico , Exosomas/efectos de los fármacos , Femenino , Humanos , Interferometría/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Ováricas/tratamiento farmacológico , Bazo
11.
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
12.
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
13.
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
14.
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
15.
J Clin Invest ; 121(6): 2361-70, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21537084

RESUMEN

Transcription intermediary factor 1γ (TIF1γ) was suggested to play a role in erythropoiesis. However, how TIF1γ regulates the development of different blood cell lineages and whether TIF1γ is involved in human hematological malignancies remain to be determined. Here we have shown that TIF1γ was a tumor suppressor in mouse and human chronic myelomonocytic leukemia (CMML). Loss of Tif1g in mouse HSCs favored the expansion of the granulo-monocytic progenitor compartment. Furthermore, Tif1g deletion induced the age-dependent appearance of a cell-autonomous myeloproliferative disorder in mice that recapitulated essential characteristics of human CMML. TIF1γ was almost undetectable in leukemic cells of 35% of CMML patients. This downregulation was related to the hypermethylation of CpG sequences and specific histone modifications in the gene promoter. A demethylating agent restored the normal epigenetic status of the TIF1G promoter in human cells, which correlated with a reestablishment of TIF1γ expression. Together, these results demonstrate that TIF1G is an epigenetically regulated tumor suppressor gene in hematopoietic cells and suggest that changes in TIF1γ expression may be a biomarker of response to demethylating agents in CMML.


Asunto(s)
Genes Supresores de Tumor , Leucemia Mielomonocítica Crónica/genética , Factores de Transcripción/fisiología , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Animales , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Azacitidina/farmacología , Azacitidina/uso terapéutico , Secuencia de Bases , Diferenciación Celular , Metilación de ADN , Decitabina , Femenino , Regulación Leucémica de la Expresión Génica , Hematopoyesis/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mielomonocítica Crónica/tratamiento farmacológico , Leucemia Mielomonocítica Crónica/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Regiones Promotoras Genéticas , Receptor de Factor Estimulante de Colonias de Macrófagos/biosíntesis , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Organismos Libres de Patógenos Específicos , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
16.
Cancer Res ; 71(2): 484-95, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21224349

RESUMEN

The inhibition of heat shock protein 70 (HSP70) is an emerging strategy in cancer therapy. Unfortunately, no specific inhibitors are clinically available. By yeast two-hybrid screening, we have identified multiple peptide aptamers that bind HSP70. When expressed in human tumor cells, two among these peptide aptamers-A8 and A17-which bind to the peptide-binding and the ATP-binding domains of HSP70, respectively, specifically inhibited the chaperone activity, thereby increasing the cells' sensitivity to apoptosis induced by anticancer drugs. The 13-amino acid peptide from the variable region of A17 (called P17) retained the ability to specifically inhibit HSP70 and induced the regression of subcutaneous tumors in vivo after local or systemic injection. This antitumor effect was associated with an important recruitment of macrophages and T lymphocytes into the tumor bed. Altogether, these data indicate that peptide aptamers or peptides that target HSP70 may be considered as novel lead compounds for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Aptámeros de Péptidos/farmacología , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , Péptidos/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Aptámeros de Péptidos/química , Aptámeros de Péptidos/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Humanos , Melanoma Experimental/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Péptidos/química , Péptidos/genética , Estructura Terciaria de Proteína , Ratas , Transfección
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA