Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Rep ; 14(1): 16083, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38992199

RESUMEN

Extracellular vesicles (EVs) are a new mechanism of cellular communication, by delivering their cargo into target cells to modulate molecular pathways. EV-mediated crosstalk contributes to tumor survival and resistance to cellular stress. However, the role of EVs in B-cell Acute Lymphoblastic Leukaemia (B-ALL) awaits to be thoroughly investigated. We recently published that ActivinA increases intracellular calcium levels and promotes actin polymerization in B-ALL cells. These biological processes guide cytoskeleton reorganization, which is a crucial event for EV secretion and internalization. Hence, we investigated the role of EVs in the context of B-ALL and the impact of ActivinA on this phenomenon. We demonstrated that leukemic cells release a higher number of EVs in response to ActivinA treatment, and they can actively uptake EVs released by other B-ALL cells. Under culture-induced stress conditions, EVs coculture promoted cell survival in B-ALL cells in a dose-dependent manner. Direct stimulation of B-ALL cells with ActivinA or with EVs isolated from ActivinA-stimulated cells was even more effective in preventing cell death. This effect can be possibly ascribed to the increase of vesiculation and modifications of EV-associated microRNAs induced by ActivinA. These data demonstrate that ActivinA boosts EV-mediated B-ALL crosstalk, improving leukemia survival in stress conditions.


Asunto(s)
Comunicación Celular , Supervivencia Celular , Vesículas Extracelulares , Vesículas Extracelulares/metabolismo , Humanos , Línea Celular Tumoral , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , MicroARNs/metabolismo , MicroARNs/genética
2.
Bone Marrow Transplant ; 59(2): 171-177, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37935782

RESUMEN

Analysis of donor-recipient chimerism after hematopoietic stem cell transplantation (HSCT) is of pivotal importance for patient's clinical management, especially in the context of mixed chimerism. Patients are routinely monitored for chimerism in sorted subsets of peripheral blood cells. However, measurement of chimerism in sorted immune cell subsets is technically challenging and time consuming. We here propose a novel, flow cytometry-based approach to detect donor cell chimerism in sex-mismatched HSCT. We exploit RNA PrimeFlow™ system, based on RNA hybridization, to detect mRNA from a lysine demethylase encoded by Y chromosome, KDM5D. This approach allows to distinguish male and female derived cells with around 1% sensitivity. The procedure can be coupled with multiparametric immunophenotyping to assess chimerism in specific immune cell subsets without the need for prior FACS-sorting. We apply this method to a cohort of HSCT patients (n = 10) and we show that it is consistent with standard PCR-based method. We also show that different T lymphocyte subsets display variable degrees of donor chimerism, especially in CD8+ T cell compartment where we observe an enrichment for recipient chimerism in central memory T cells. This method can be exploited to advance current knowledge on immune reconstitution focusing on specific subsets avoiding prior FACS-sorting.


Asunto(s)
Quimerismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Masculino , Femenino , Citometría de Flujo , ARN , Trasplante de Células Madre Hematopoyéticas/métodos , Subgrupos de Linfocitos T , Quimera por Trasplante , Antígenos de Histocompatibilidad Menor , Histona Demetilasas/genética
3.
Leukemia ; 37(10): 1994-2005, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37640845

RESUMEN

Complete elimination of B-cell acute lymphoblastic leukemia (B-ALL) by a risk-adapted primary treatment approach remains a clinical key objective, which fails in up to a third of patients. Recent evidence has implicated subpopulations of B-ALL cells with stem-like features in disease persistence. We hypothesized that microRNA-126, a core regulator of hematopoietic and leukemic stem cells, may resolve intratumor heterogeneity in B-ALL and uncover therapy-resistant subpopulations. We exploited patient-derived xenograft (PDX) models with B-ALL cells transduced with a miR-126 reporter allowing the prospective isolation of miR-126(high) cells for their functional and transcriptional characterization. Discrete miR-126(high) populations, often characterized by MIR126 locus demethylation, were identified in 8/9 PDX models and showed increased repopulation potential, in vivo chemotherapy resistance and hallmarks of quiescence, inflammation and stress-response pathway activation. Cells with a miR-126(high) transcriptional profile were identified as distinct disease subpopulations by single-cell RNA sequencing in diagnosis samples from adult and pediatric B-ALL. Expression of miR-126 and locus methylation were tested in several pediatric and adult B-ALL cohorts, which received standardized treatment. High microRNA-126 levels and locus demethylation at diagnosis associate with suboptimal response to induction chemotherapy (MRD > 0.05% at day +33 or MRD+ at day +78).


Asunto(s)
Linfoma de Burkitt , MicroARNs , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Adulto , Humanos , Niño , Neoplasia Residual/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , MicroARNs/genética , MicroARNs/metabolismo
5.
Cancer Cell ; 29(6): 905-921, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27300437

RESUMEN

MicroRNA (miRNA)-126 is a known regulator of hematopoietic stem cell quiescence. We engineered murine hematopoiesis to express miRNA-126 across all differentiation stages. Thirty percent of mice developed monoclonal B cell leukemia, which was prevented or regressed when a tetracycline-repressible miRNA-126 cassette was switched off. Regression was accompanied by upregulation of cell-cycle regulators and B cell differentiation genes, and downregulation of oncogenic signaling pathways. Expression of dominant-negative p53 delayed blast clearance upon miRNA-126 switch-off, highlighting the relevance of p53 inhibition in miRNA-126 addiction. Forced miRNA-126 expression in mouse and human progenitors reduced p53 transcriptional activity through regulation of multiple p53-related targets. miRNA-126 is highly expressed in a subset of human B-ALL, and antagonizing miRNA-126 in ALL xenograft models triggered apoptosis and reduced disease burden.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , MicroARNs/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis , Ciclo Celular , Diferenciación Celular , Regulación Neoplásica de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , MicroARNs/metabolismo , Neoplasias Experimentales , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Transducción de Señal , Regulación hacia Arriba
7.
Cancer Cell ; 29(2): 214-28, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26832662

RESUMEN

To investigate miRNA function in human acute myeloid leukemia (AML) stem cells (LSC), we generated a prognostic LSC-associated miRNA signature derived from functionally validated subpopulations of AML samples. For one signature miRNA, miR-126, high bioactivity aggregated all in vivo patient sample LSC activity into a single sorted population, tightly coupling miR-126 expression to LSC function. Through functional studies, miR-126 was found to restrain cell cycle progression, prevent differentiation, and increase self-renewal of primary LSC in vivo. Compared with prior results showing miR-126 regulation of normal hematopoietic stem cell (HSC) cycling, these functional stem effects are opposite between LSC and HSC. Combined transcriptome and proteome analysis demonstrates that miR-126 targets the PI3K/AKT/MTOR signaling pathway, preserving LSC quiescence and promoting chemotherapy resistance.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , MicroARNs/fisiología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Ratones SCID , MicroARNs/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
8.
EMBO Mol Med ; 5(6): 858-69, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23653322

RESUMEN

A third of patients with critical limb ischemia (CLI) will eventually require limb amputation. Therapeutic neovascularization using unselected mononuclear cells to salvage ischemic limbs has produced modest results. The TIE2-expressing monocytes/macrophages (TEMs) are a myeloid cell subset known to be highly angiogenic in tumours. This study aimed to examine the kinetics of TEMs in patients with CLI and whether these cells promote neovascularization of the ischemic limb. Here we show that there are 10-fold more circulating TEMs in CLI patients, and removal of ischemia reduces their numbers to normal levels. TEM numbers in ischemic muscle are two-fold greater than normoxic muscle from the same patient. TEMs from patients with CLI display greater proangiogenic activity than TIE2-negative monocytes in vitro. Using a mouse model of hindlimb ischemia, lentiviral-based Tie2 knockdown in TEMs impaired recovery from ischemia, whereas delivery of mouse macrophages overexpressing TIE2, or human TEMs isolated from CLI patients, rescued limb ischemia. These data suggest that enhancing TEM recruitment to the ischemic muscle may have the potential to improve limb neovascularization in CLI patients.


Asunto(s)
Isquemia/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Receptor TIE-2/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Angiopoyetina 2/metabolismo , Animales , Femenino , Humanos , Isquemia/patología , Macrófagos/inmunología , Masculino , Ratones , MicroARNs/metabolismo , Persona de Mediana Edad , Monocitos/inmunología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Neovascularización Fisiológica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Cancer Discov ; 2(5): 395-7, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22588875

RESUMEN

Acute mobilization of circulating endothelial progenitors has been implicated in tumor resistance to vascular-disrupting agents. In the current issue of Cancer Discovery, Taylor and colleagues provide novel insight into the kinetics of endothelial progenitor mobilization by vascular-disrupting agents in both mouse tumor models and cancer patients.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Resistencia a Antineoplásicos , Células Endoteliales/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Humanos , Masculino
10.
Int J Dev Biol ; 55(4-5): 495-503, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21732273

RESUMEN

During organ development and remodeling, macrophages support angiogenesis, not only by secreting proangiogenic growth factors and matrix-remodeling proteases, but also by physically interacting with the sprouting vasculature to assist the formation of complex vascular networks. Recent data further indicate that embryonic and tumor-associated macrophages express similar genetic programs, possibly suggesting convergent functions in organogenesis and tumorigenesis. In this article, we review the role of macrophages in development, tissue injury and regeneration, by focusing on the mechanisms used by subsets of these cells, such as the TIE2-expressing macrophages, to regulate angiogenesis and lymphangiogenesis in both fetal and post-natal life.


Asunto(s)
Macrófagos/fisiología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Animales , Humanos , Linfangiogénesis/fisiología , Macrófagos/citología , Neoplasias/patología , Neoplasias/fisiopatología , Neovascularización Patológica/patología , Organogénesis/fisiología , Receptor TIE-2/fisiología , Regeneración/fisiología , Heridas y Lesiones/patología , Heridas y Lesiones/fisiopatología
11.
J Clin Invest ; 121(5): 1969-73, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490397

RESUMEN

Vascular-disrupting agents (VDAs) such as combretastatin A4 phosphate (CA4P) selectively disrupt blood vessels in tumors and induce tumor necrosis. However, tumors rapidly repopulate after treatment with such compounds. Here, we show that CA4P-induced vessel narrowing, hypoxia, and hemorrhagic necrosis in murine mammary tumors were accompanied by elevated tumor levels of the chemokine CXCL12 and infiltration by proangiogenic TIE2-expressing macrophages (TEMs). Inhibiting TEM recruitment to CA4P-treated tumors either by interfering pharmacologically with the CXCL12/CXCR4 axis or by genetically depleting TEMs in tumor-bearing mice markedly increased the efficacy of CA4P treatment. These data suggest that TEMs limit VDA-induced tumor injury and represent a potential target for improving the clinical efficacy of VDA-based therapies.


Asunto(s)
Macrófagos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Estilbenos/farmacología , Animales , Antineoplásicos Fitogénicos/farmacología , Separación Celular , Quimiocina CXCL12/metabolismo , Femenino , Citometría de Flujo , Neoplasias Mamarias Animales , Ratones , Ratones Transgénicos , Necrosis/patología , Trasplante de Neoplasias , Receptor TIE-2 , Receptores CXCR4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA