Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Thorax ; 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38373824

RESUMEN

BACKGROUND: In patients with asthma, respiratory syncytial virus (RSV) infections can cause disease exacerbation by infecting the epithelial layer of the airways, inducing subsequent immune response. The type I interferon antiviral response of epithelial cells upon RSV infection is found to be reduced in asthma in most-but not all-studies. Moreover, the molecular mechanisms causing the differences in the asthmatic bronchial epithelium in response to viral infection are poorly understood. METHODS: Here, we investigated the transcriptional response to RSV infection of primary bronchial epithelial cells (pBECs) from patients with asthma (n=8) and healthy donors (n=8). The pBECs obtained from bronchial brushes were differentiated in air-liquid interface conditions and infected with RSV. After 3 days, cells were processed for single-cell RNA sequencing. RESULTS: A strong antiviral response to RSV was observed for all cell types, for all samples (p<1e-48). Most (1045) differentially regulated genes following RSV infection were found in cells transitioning to secretory cells. Goblet cells from patients with asthma showed lower expression of genes involved in the interferon response (false discovery rate <0.05), including OASL, ICAM1 and TNFAIP3. In multiciliated cells, an impairment of the signalling pathways involved in the response to RSV in asthma was observed. CONCLUSION: Our results highlight that the response to RSV infection of the bronchial epithelium in asthma and healthy airways was largely similar. However, in asthma, the response of goblet and multiciliated cells is impaired, highlighting the need for studying airway epithelial cells at high resolution in the context of asthma exacerbation.

2.
Respir Res ; 24(1): 308, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38062491

RESUMEN

BACKGROUND: Asthma is stratified into type 2-high and type 2-low inflammatory phenotypes. Limited success has been achieved in developing drugs that target type 2-low inflammation. Previous studies have linked IL-6 signaling to severe asthma. IL-6 cooperates with soluble-IL-6Rα to activate cell signaling in airway epithelium. OBJECTIVE: We sought to study the role of sIL-6Rα amplified IL-6 signaling in airway epithelium and to develop an IL-6+ sIL-6Rα gene signature that may be used to select asthma patients who potentially respond to anti-IL-6 therapy. METHODS: Human airway epithelial cells were stimulated with combinations of IL-6, sIL-6Rα, and inhibitors, sgp130 (Olamkicept), and anti-IL-6R (Tocilizumab), to assess effects on pathway activation, epithelial barrier integrity, and gene expression. A gene signature was generated to identify IL-6 high patients using bronchial biopsies and nasal brushes. RESULTS: Soluble-IL-6Rα amplified the activation of the IL-6 pathway, shown by the increase of STAT3 phosphorylation and stronger gene induction in airway epithelial cells compared to IL-6 alone. Olamkicept and Tocilizumab inhibited the effect of IL-6 + sIL-6Rα on gene expression. We developed an IL-6 + sIL-6Rα gene signature and observed enrichment of this signature in bronchial biopsies but not nasal brushes from asthma patients compared to healthy controls. An IL-6 + sIL-6Rα gene signature score was associated with lower levels of sputum eosinophils in asthma. CONCLUSION: sIL-6Rα amplifies IL-6 signaling in bronchial epithelial cells. Higher local airway IL-6 + sIL-6Rα signaling is observed in asthma patients with low sputum eosinophils.


Asunto(s)
Asma , Interleucina-6 , Humanos , Asma/diagnóstico , Asma/tratamiento farmacológico , Asma/genética , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Inflamación , Interleucina-6/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Transducción de Señal
3.
Antimicrob Agents Chemother ; 66(1): e0154321, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34633839

RESUMEN

Antiviral therapies are urgently needed to treat and limit the development of severe COVID-19 disease. Ivermectin, a broad-spectrum anti-parasitic agent, has been shown to have anti-SARS-CoV-2 activity in Vero cells at a concentration of 5 µM. These limited in vitro results triggered the investigation of ivermectin as a treatment option to alleviate COVID-19 disease. However, in April 2021, the World Health Organization stated the following: "The current evidence on the use of ivermectin to treat COVID-19 patients is inconclusive." It is speculated that the in vivo concentration of ivermectin is too low to exert a strong antiviral effect. Here, we performed a head-to-head comparison of the antiviral activity of ivermectin and the structurally related, but metabolically more stable moxidectin in multiple in vitro models of SARS-CoV-2 infection, including physiologically relevant human respiratory epithelial cells. Both moxidectin and ivermectin exhibited antiviral activity in Vero E6 cells. Subsequent experiments revealed that these compounds predominantly act on the steps following virus cell entry. Surprisingly, however, in human-airway-derived cell models, both moxidectin and ivermectin failed to inhibit SARS-CoV-2 infection, even at concentrations of 10 µM. These disappointing results call for a word of caution in the interpretation of anti-SARS-CoV-2 activity of drugs solely based on their activity in Vero cells. Altogether, these findings suggest that even using a high-dose regimen of ivermectin, or switching to another drug in the same class, is unlikely to be useful for treatment of SARS-CoV-2 in humans.


Asunto(s)
COVID-19 , Ivermectina , Animales , Antivirales/farmacología , Chlorocebus aethiops , Células Epiteliales , Humanos , Ivermectina/farmacología , Macrólidos , SARS-CoV-2 , Células Vero , Replicación Viral
4.
Stem Cells ; 34(3): 640-52, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26887347

RESUMEN

Adult stem cells are often touted as therapeutic agents in the regenerative medicine field, however data detailing both the engraftment and functional capabilities of solid tissue derived human adult epithelial stem cells is scarce. Here we show the isolation of adult human salivary gland (SG) stem/progenitor cells and demonstrate at the single cell level in vitro self-renewal and differentiation into multilineage organoids. We also show in vivo functionality, long-term engraftment, and functional restoration in a xenotransplantation model. Indeed, transplanted human salisphere-derived cells restored saliva production and greatly improved the regenerative potential of irradiated SGs. Further selection for c-Kit expression enriched for cells with enhanced regenerative potencies. Interestingly, interaction of transplanted cells with the recipient SG may also be involved in functional recovery. Thus, we show for the first time that salispheres cultured from human SGs contain stem/progenitor cells capable of self-renewal and differentiation and rescue of saliva production. Our study underpins the therapeutic promise of salisphere cell therapy for the treatment of xerostomia.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/biosíntesis , Glándulas Salivales/citología , Trasplante de Células Madre , Xerostomía/terapia , Animales , Diferenciación Celular/genética , Diferenciación Celular/efectos de la radiación , Regulación del Desarrollo de la Expresión Génica/efectos de la radiación , Humanos , Ratones , Proteínas Proto-Oncogénicas c-kit/genética , Radiación , Glándulas Salivales/metabolismo , Glándulas Salivales/trasplante , Análisis de la Célula Individual , Células Madre/citología , Células Madre/metabolismo , Células Madre/efectos de la radiación , Xerostomía/patología
5.
Blood ; 121(13): 2452-61, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23349393

RESUMEN

The Polycomb group (PcG) protein BMI1 is a key factor in regulating hematopoietic stem cell (HSC) and leukemic stem cell self-renewal and functions in the context of the Polycomb repressive complex 1 (PRC1). In humans, each of the 5 subunits of PRC1 has paralog family members of which many reside in PRC1 complexes, likely in a mutually exclusive manner, pointing toward a previously unanticipated complexity of Polycomb-mediated silencing. We used an RNA interference screening approach to test the functionality of these paralogs in human hematopoiesis. Our data demonstrate a lack of redundancy between various paralog family members, suggestive of functional diversification between PcG proteins. By using an in vivo biotinylation tagging approach followed by liquid chromatography-tandem mass spectrometry to identify PcG interaction partners, we confirmed the existence of multiple specific PRC1 complexes. We find that CBX2 is a nonredundant CBX paralog vital for HSC and progenitor function that directly regulates the expression of the cyclin-dependent kinase inhibitor p21, independently of BMI1 that dominantly controls expression of the INK4A/ARF locus. Taken together, our data show that different PRC1 paralog family members have nonredundant and locus-specific gene regulatory activities that are essential for human hematopoiesis.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Silenciador del Gen , Sitios Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Femenino , Sangre Fetal/citología , Sangre Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen/fisiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Humanos , Recién Nacido , Familia de Multigenes/genética , Familia de Multigenes/fisiología , Embarazo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Homología de Secuencia , Especificidad por Sustrato/genética
6.
Mol Cell Proteomics ; 12(3): 626-37, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23233446

RESUMEN

Interactions between hematopoietic stem cells and their niche are mediated by proteins within the plasma membrane (PM) and changes in these interactions might alter hematopoietic stem cell fate and ultimately result in acute myeloid leukemia (AML). Here, using nano-LC/MS/MS, we set out to analyze the PM profile of two leukemia patient samples. We identified 867 and 610 unique CD34(+) PM (-associated) proteins in these AML samples respectively, including previously described proteins such as CD47, CD44, CD135, CD96, and ITGA5, but also novel ones like CD82, CD97, CD99, PTH2R, ESAM, MET, and ITGA6. Further validation by flow cytometry and functional studies indicated that long-term self-renewing leukemic stem cells reside within the CD34(+)/ITGA6(+) fraction, at least in a subset of AML cases. Furthermore, we combined proteomics with transcriptomics approaches using a large panel of AML CD34(+) (n = 60) and normal bone marrow CD34(+) (n = 40) samples. Thus, we identified eight subgroups of AML patients based on their specific PM expression profile. GSEA analysis revealed that these eight subgroups are enriched for specific cellular processes.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Células Madre Neoplásicas/metabolismo , Proteómica/métodos , Enfermedad Aguda , Antígenos CD34/genética , Antígenos CD34/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Membrana Celular/metabolismo , Cromatografía Liquida , Citometría de Flujo , Regulación Leucémica de la Expresión Génica , Humanos , Integrina alfa6/genética , Integrina alfa6/metabolismo , Nanotecnología/métodos , Análisis de Componente Principal , Proteoma/análisis , Espectrometría de Masas en Tándem
7.
Nat Genet ; 37(5): 468-70, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15821733

RESUMEN

Roberts syndrome is an autosomal recessive disorder characterized by craniofacial anomalies, tetraphocomelia and loss of cohesion at heterochromatic regions of centromeres and the Y chromosome. We identified mutations in a new human gene, ESCO2, associated with Roberts syndrome in 15 kindreds. The ESCO2 protein product is a member of a conserved protein family that is required for the establishment of sister chromatid cohesion during S phase and has putative acetyltransferase activity.


Asunto(s)
Acetiltransferasas/genética , Cromátides/fisiología , Proteínas Cromosómicas no Histona/genética , Emparejamiento Cromosómico/fisiología , Labio Leporino/genética , Fisura del Paladar/genética , Ectromelia/genética , Proteínas Nucleares/genética , Proteínas de Saccharomyces cerevisiae/genética , Acetiltransferasas/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Ectromelia/metabolismo , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Proteínas Nucleares/fisiología , Linaje , Proteínas de Saccharomyces cerevisiae/fisiología
8.
J Biol Chem ; 286(8): 6061-70, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21169357

RESUMEN

In human hematopoietic malignancies, RAS mutations are frequently observed. Yet, little is known about signal transduction pathways that mediate KRAS-induced phenotypes in human CD34(+) stem/progenitor cells. When cultured on bone marrow stroma, we observed that KRAS(G12V)-transduced cord blood (CB) CD34(+) cells displayed a strong proliferative advantage over control cells, which coincided with increased early cobblestone (CAFC) formation and induction of myelomonocytic differentiation. However, the KRAS(G12V)-induced proliferative advantage was transient. By week three no progenitors remained in KRAS(G12V)-transduced cultures and cells were all terminally differentiated into monocytes/macrophages. In line with these results, LTC-IC frequencies were strongly reduced. Both the ERK and p38 MAPK pathways, but not JNK, were activated by KRAS(G12V) and we observed that proliferation and CAFC formation were mediated via ERK, while differentiation was predominantly mediated via p38. Interestingly, we observed that KRAS(G12V)-induced proliferation and CAFC formation, but not differentiation, were largely mediated via secreted factors, since these phenotypes could be recapitulated by treating non-transduced cells with conditioned medium harvested from KRAS(G12V)-transduced cultures. Multiplex cytokine arrays and genome-wide gene expression profiling were performed to gain further insight into the mechanisms by which oncogenic KRAS(G12V) can contribute to the process of leukemic transformation. Thus, angiopoietin-like 6 (ANGPTL6) was identified as an important factor in the KRAS(G12V) secretome that enhanced proliferation of human CB CD34(+) cells.


Asunto(s)
Antígenos CD34 , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Monocitos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas ras/metabolismo , Sustitución de Aminoácidos , Proteína 6 similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Estudio de Asociación del Genoma Completo , Células Madre Hematopoyéticas/citología , Humanos , Leucemia/genética , Leucemia/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Monocitos/citología , Mutación Missense , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Transducción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas ras/genética
9.
Viruses ; 13(7)2021 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-34372541

RESUMEN

The current COVID-19 pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has an enormous impact on human health and economy. In search for therapeutic options, researchers have proposed resveratrol, a food supplement with known antiviral, anti-inflammatory, and antioxidant properties as an advantageous antiviral therapy for SARS-CoV-2 infection. Here, we provide evidence that both resveratrol and its metabolically more stable structural analog, pterostilbene, exhibit potent antiviral properties against SARS-CoV-2 in vitro. First, we show that resveratrol and pterostilbene antiviral activity in African green monkey kidney cells. Both compounds actively inhibit virus replication within infected cells as reduced virus progeny production was observed when the compound was added at post-inoculation conditions. Without replenishment of the compound, antiviral activity was observed up to roughly five rounds of replication, demonstrating the long-lasting effect of these compounds. Second, as the upper respiratory tract represents the initial site of SARS-CoV-2 replication, we also assessed antiviral activity in air-liquid interface (ALI) cultured human primary bronchial epithelial cells, isolated from healthy volunteers. Resveratrol and pterostilbene showed a strong antiviral effect in these cells up to 48 h post-infection. Collectively, our data indicate that resveratrol and pterostilbene are promising antiviral compounds to inhibit SARS-CoV-2 infection. Because these results represent laboratory findings in cells, we advocate evaluation of these compounds in clinical trials before statements are made whether these drugs are advantageous for COVID-19 treatment.


Asunto(s)
Bronquios/virología , COVID-19/virología , Células Epiteliales/virología , Resveratrol/farmacología , SARS-CoV-2/efectos de los fármacos , Estilbenos/farmacología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , COVID-19/epidemiología , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Femenino , Humanos , Masculino , Persona de Mediana Edad , SARS-CoV-2/fisiología , Células Vero , Tratamiento Farmacológico de COVID-19
10.
Exp Hematol ; 35(5): 782-92, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17577927

RESUMEN

OBJECTIVE: To determine the involvement of Rac signaling in self-renewal and expansion on bone marrow stroma of normal CD34+ cells vs leukemic CD34+ cells from acute myeloid leukemia (AML) patients. MATERIALS AND METHODS: Rac signaling was modulated by retroviral introduction of Racl-N17, Racl-V12, or by using the Rac inhibitor NSC23766. In long-term MS5 cocultures (leukemic) expansion, migration, adhesion, and presence of stem/progenitor cells were monitored in both normal as well as leukemic CD34+ cells. RESULTS: Inhibition of Rac signaling impaired migration and adhesion of cord blood (CB) CD34+ cells on MS5 stroma. Long-term inhibition of Rac during a 5-week coculture period on stroma prevented association of hematopoietic progenitors with the bone marrow stromal cells and resulted in a dramatic decrease in the primitive stem cell frequency (long-term culture-initiating cell) in a dose-dependent manner. Many of these phenotypes were reversed in the presence of activated Racl-V12, including improved migration toward, and association with, MS5 cells. CD34+ AML cells were characterized by elevated levels of Rac activity (five of seven patients) and enhanced migration and adhesion to MS5 bone marrow stroma as compared to CB CD34+ cells. A dramatic decrease was observed in the formation of leukemic cobblestone area-forming cells as well as strongly diminished clonal expansion in the presence of the Rac inhibitor NSC23766. CONCLUSION: Our data indicate that Rac signal transduction is required for the maintenance and expansion of both normal as well as leukemic stem/progenitor cells by mediating their interaction with stromal cells.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Leucemia Mieloide/metabolismo , Células del Estroma/fisiología , Proteínas de Unión al GTP rac/fisiología , Aminoquinolinas/farmacología , Antígenos CD34/inmunología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Técnicas de Cultivo de Célula/métodos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células Madre Neoplásicas , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Proteínas de Unión al GTP rac/antagonistas & inhibidores
11.
Exp Hematol ; 35(10): 1538-49, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17889721

RESUMEN

OBJECTIVE: With the emergence of the concept of the leukemia stem cell, assays to study them remain pivotal in understanding (leukemic) stem cell biology. METHODS: We have cultured acute myeloid leukemia CD34(+) cells on bone marrow stroma. Long-term expansion was monitored and self-renewal was addressed by replating of Leukemic-cobblestone area-forming cells (L-CAs). Also, lentiviral vectors were generated that could target L-CAs. RESULTS: A strong expansion was observed in about 75% of the acute myeloid leukemia cases (n = 30) and long-term cultures could be maintained for up to 24 weeks on MS5 bone marrow stromal cells. Cells that were able to initiate leukemic cobblestone areas resided in the CD34(+) population and were absent from the CD34(-) population. Self-renewal within these L-CAs was determined by sequential passaging of these L-CAs onto new MS5 stromal layers, which resulted in the generation of second, third, and fourth L-CAs, which were able to sustain long-term expansion and generated high numbers of immature undifferentiated suspension cells. CD34(+) cells that were able to initiate long-term cultures all coexpressed MEIS1 and HOXA9, and expressed elevated BMI1 levels. CONCLUSION: We present a novel long-term leukemic stem/progenitor assay in which new drugs can be tested and in which genes can be overexpressed or downmodulated using a lentiviral approach in order to obtain more insight into the process of leukemic transformation and self-renewal.


Asunto(s)
Células de la Médula Ósea/patología , Línea Celular Tumoral/patología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Ensayo de Tumor de Célula Madre , Antígenos CD34 , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral/metabolismo , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Humanos , Leucemia Mieloide Aguda/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/biosíntesis , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/biosíntesis , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Represoras/biosíntesis , Células del Estroma/metabolismo , Células del Estroma/patología , Factores de Tiempo
12.
PLoS One ; 4(9): e6936, 2009 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-19738907

RESUMEN

Cohesion between sister chromatids is essential for faithful chromosome segregation. In budding yeast, the acetyltransferase Eco1/Ctf7 establishes cohesion during DNA replication in S phase and in response to DNA double strand breaks in G2/M phase. In humans two Eco1 orthologs exist: ESCO1 and ESCO2. Both proteins are required for proper sister chromatid cohesion, but their exact function is unclear at present. Since ESCO2 has been identified as the gene defective in the rare autosomal recessive cohesinopathy Roberts syndrome (RBS), cells from RBS patients can be used to elucidate the role of ESCO2. We investigated for the first time RBS cells in comparison to isogenic controls that stably express V5- or GFP-tagged ESCO2. We show that the sister chromatid cohesion defect in the transfected cell lines is rescued and suggest that ESCO2 is regulated by proteasomal degradation in a cell cycle-dependent manner. In comparison to the corrected cells RBS cells were hypersensitive to the DNA-damaging agents mitomycin C, camptothecin and etoposide, while no particular sensitivity to UV, ionizing radiation, hydroxyurea or aphidicolin was found. The cohesion defect of RBS cells and their hypersensitivity to DNA-damaging agents were not corrected by a patient-derived ESCO2 acetyltransferase mutant (W539G), indicating that the acetyltransferase activity of ESCO2 is essential for its function. In contrast to a previous study on cells from patients with Cornelia de Lange syndrome, another cohesinopathy, RBS cells failed to exhibit excessive chromosome aberrations after irradiation in G2 phase of the cell cycle. Our results point at an S phase-specific role for ESCO2 in the maintenance of genome stability.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Fibroblastos/metabolismo , Trastornos del Crecimiento/diagnóstico , Camptotecina/farmacología , Proteínas de Ciclo Celular/metabolismo , Aberraciones Cromosómicas , Segregación Cromosómica , Anomalías Congénitas/diagnóstico , Anomalías Congénitas/genética , Daño del ADN , Etopósido/farmacología , Trastornos del Crecimiento/genética , Humanos , Lactante , Masculino , Mitomicina/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Intercambio de Cromátides Hermanas , Síndrome , Cohesinas
13.
Scand Cardiovasc J ; 41(4): 230-4, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17680510

RESUMEN

OBJECTIVES: To study the prevalence of active cytomegalovirus (CMV) infection in patients with stable and unstable conditions of coronary artery disease (CAD). DESIGN: Forty patients with acute coronary syndrome (ACS), 50 patients with stable angina and angiographically verified CAD (SA) and 50 clinically healthy controls were included. Monocytes were isolated from peripheral blood and CMV-RNA expression was determined by a nested RT-PCR assay. CMV IgM and IgG antibodies, interleukin-(IL)-6, IL-10 and CRP were measured in serum. RESULTS: The prevalence of active CMV infection was significantly higher in patients with ACS (15%) and in patients with SA (10%) compared with controls (2%) (p<0.001). The presence of an active CMV infection was associated with increased serum concentrations of IL-6. CONCLUSIONS: Active CMV infection was found to a larger extent in CAD patients than in healthy controls. The data indicate that CAD patients are more susceptible to reactivation of CMV and put new focus on the role of CMV in atherosclerosis.


Asunto(s)
Síndrome Coronario Agudo/virología , Angina de Pecho/virología , Infecciones por Citomegalovirus/epidemiología , Citomegalovirus/fisiología , Replicación Viral , Síndrome Coronario Agudo/complicaciones , Anciano , Angina de Pecho/complicaciones , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/virología , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/diagnóstico , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Monocitos/virología , Prevalencia , ARN Viral/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Blood ; 110(8): 2880-8, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17630355

RESUMEN

The transcription factor STAT5 fulfills a distinct role in the hematopoietic system, but its precise role in primitive human hematopoietic cells remains to be elucidated. Therefore, we performed STAT5 RNAi in sorted cord blood (CB) and acute myeloid leukemia (AML) CD34+ cells by lentiviral transduction and investigated effects of STAT5 downmodulation on the normal stem/progenitor cell compartment and the leukemic counterpart. STAT5 RNAi cells displayed growth impairment, without affecting their differentiation in CB and AML cultures on MS5 stroma. In CB, limiting-dilution assays demonstrated a 3.9-fold reduction in progenitor numbers. Stem cells were enumerated in long-term culture-initiating cell (LTC-IC) assays, and the average LTC-IC frequency was 3.25-fold reduced from 0.13% to 0.04% by STAT5 down-regulation. Single-cell sorting experiments of CB CD34+/CD38(-) cells demonstrated a 2-fold reduced cytokine-driven expansion, with a subsequent 2.3-fold reduction of progenitors. In sorted CD34+ AML cells with constitutive STAT5 phosphorylation (5/8), STAT5 RNAi demonstrated a reduction in cell number (72% +/- 17%) and a decreased expansion (17 +/- 15 vs 80 +/- 58 in control cultures) at week 6 on MS5 stroma. Together, our data indicate that STAT5 expression is required for the maintenance and expansion of primitive hematopoietic stem and progenitor cells, both in normal as well as leukemic hematopoiesis.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide/metabolismo , Factor de Transcripción STAT5/metabolismo , Enfermedad Aguda , Antígenos CD34/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Sangre Fetal , Citometría de Flujo , Expresión Génica , Humanos , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/genética , Factores de Tiempo
15.
Blood ; 110(4): 1317-25, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17475913

RESUMEN

The CCAAT/enhancer binding protein (C/EBP) alpha transcription factor is indispensable for myeloid differentiation. In various myeloid leukemias, C/EBPalpha is mutated or functionally impaired due to decreased C/EBPalpha expression or phosphorylation. In order to investigate the functional consequences of decreased C/EBPalpha function in AML, we reintroduced C/EBPalpha in primary CD34(+) sorted acute myeloid leukemia (AML) cells using a lentiviral approach. Self-renewal and differentiation of primary AML stem cells were studied on long-term MS5 cocultures. Activation of C/EBPalpha immediately led to a growth arrest in all AML cultures (N = 7), resulting in severely reduced expansion compared with control cultures. This growth arrest corresponded with enhanced myeloid differentiation as assessed by fluorescence-activated cell sorter (FACS) analysis for CD14, CD15, and CD11b. Myeloid differentiation was further confirmed by the up-regulation of neutrophil elastase and granulocyte colony-stimulating factor (G-CSF) receptor in C/EBPalpha transduced cells. C/EBPalpha-expressing AML CD34(+) cells failed to generate second and third leukemic cobblestone areas (L-CAs) in serial replating experiments, while control cultures could be sequentially passaged for more than 4 times, indicating that reintroduction of C/EBPalpha impaired the self-renewal capacity of the leukemic CD34(+) compartment. Together, our data indicate that low C/EBPalpha levels are necessary to maintain self-renewal and the immature character of AML stem cells.


Asunto(s)
Antígenos CD34/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/farmacología , Mielopoyesis , Células Madre/citología , Crisis Blástica , Células de la Médula Ósea/patología , Proteína alfa Potenciadora de Unión a CCAAT/genética , Ciclo Celular/genética , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Lentivirus/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Fosforilación , Células del Estroma/patología , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA