Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Palliat Care ; 23(1): 8, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172871

RESUMEN

BACKGROUND: Intranasal (i.n.) drug application is a widely known and low-invasive route of administration that may be able to achieve rapid symptom control in terminally ill patients. According to the German S3 guideline "Palliative care for patients with incurable cancer", benzodiazepines, such as midazolam, are recommended for the treatment of terminal agitation. To the best of our knowledge there is no evidence for i.n. midazolam in terminally ill patients. We aim to assess the use of i.n. midazolam as an alternative to subcutaneous administration of the drug. METHODS: In this monocentric, randomised, controlled, open-label investigator initiated trial, n = 60 patients treated at the palliative care unit of a University Hospital will be treated with 5 mg midazolam i.n. versus 5 mg subcutaneous (s.c.) midazolam in the control arm when terminal agitation occurs (randomly assigned 1:1). The estimated recruitment period is 18 months. Treatment efficacy is defined as an improvement on the Richmond Agitation Sedation Scale (Palliative Version) (RASS-PAL) and a study specific numeric rating scale (NRS) before and after drug administration. Furthermore, plasma concentration determinations of midazolam will be conducted at t1 = 0 min, t2 = 5 min, and t3 = 20 min using liquid chromatography/mass spectrometry (LC-MS). The primary objective is to demonstrate non-inferiority of midazolam i.n. in comparison to midazolam s.c. for the treatment of agitation in terminally ill patients. DISCUSSION: Midazolam i.n. is expected to achieve at least equivalent reduction of terminal agitation compared to s.c. administration. In addition, plasma concentrations of midazolam i.n. are not expected to be lower than those of midazolam s.c. and the dynamics of the plasma concentration with an earlier increase could be beneficial. TRIAL REGISTRATION: German Clinical Trials Registry DRKS00026775, registered 07.07.2022, Eudra CT No.: 2021-004789-36.


Asunto(s)
Midazolam , Enfermo Terminal , Humanos , Midazolam/uso terapéutico , Cuidados Paliativos , Resultado del Tratamiento , Ansiedad , Hipnóticos y Sedantes/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
2.
Eur J Haematol ; 98(5): 485-492, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28135011

RESUMEN

OBJECTIVE: Multidrug-resistant organisms (MDRO) are a challenge in allogeneic hematopoietic cell transplantation (HCT). However, in the literature there is no comprehensive analysis on MDRO in HCT. In this retrospective, single-center analysis, we appraised prevalence and clinical impact of MDRO in 98 consecutive allogeneic HCT patients. METHOD: Prior to the conditioning (baseline) and whenever clinically indicated patients underwent a full screening for MDRO (stool and urine cultures, swabs from several body regions). RESULTS: It turned out that 26 patients were colonized by 33 MDRO, either at baseline (n=16) or at any other time until day 100 post-transplantation. Of these 26 patients, eight developed an infection with MDRO, four of them by 4MRGN Pseudomonas aeruginosa, and three of them died MDRO-related. However, there was no significant difference between MDRO-colonized and non-colonized patients regarding overall survival (OS) and non-relapse-mortality (NRM). There was only a trend toward a higher NRM in patients already colonized by MDRO at baseline. This was due to the high NRM in multidrug-resistant P. aeruginosa-colonized patients. CONCLUSION: In summary, colonization with MDRO other than P. aeruginosa had no negative impact on NRM and OS. Patients colonized by multidrug-resistant P. aeruginosa had a dismal outcome. HCT of these patients should be considered with care. Screening for MDRO in the pretransplant work-up is suggested.


Asunto(s)
Farmacorresistencia Microbiana , Resistencia a Múltiples Medicamentos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Infecciones/epidemiología , Infecciones/etiología , Adulto , Anciano , Antiinfecciosos/farmacología , Manejo de la Enfermedad , Neutropenia Febril/diagnóstico , Neutropenia Febril/etiología , Neutropenia Febril/terapia , Femenino , Humanos , Infecciones/diagnóstico , Infecciones/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Mortalidad , Prevalencia , Estudios Retrospectivos , Acondicionamiento Pretrasplante/efectos adversos , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Adulto Joven
3.
Br J Haematol ; 175(5): 917-924, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27714772

RESUMEN

The mammalian-target of rapamycin (also termed mechanistic target of rapamycin, mTOR) pathway integrates various pro-proliferative and anti-apoptotic stimuli and is involved in regulatory T-cell (TREG) development. As these processes contribute to the pathogenesis of myelodysplastic syndromes (MDS), we hypothesized that mTOR modulation with temsirolimus (TEM) might show activity in MDS. This prospective multicentre trial enrolled lower and higher risk MDS patients, provided that they were transfusion-dependent/neutropenic or relapsed/refractory to 5-azacitidine, respectively. All patients received TEM at a weekly dose of 25 mg. Of the 9 lower- and 11 higher-risk patients included, only 4 (20%) reached the response assessment after 4 months of treatment and showed stable disease without haematological improvement. The remaining patients discontinued TEM prematurely due to adverse events. Median overall survival (OS) was not reached in the lower-risk group and 296 days in the higher-risk group. We observed a significant decline of bone marrow (BM) vascularisation (P = 0·006) but were unable to demonstrate a significant impact of TEM on the balance between TREG and pro-inflammatory T-helper-cell subsets within the peripheral blood or BM. We conclude that mTOR-modulation with TEM at a dose of 25 mg per week is accompanied by considerable toxicity and has no beneficial effects in elderly MDS patients.


Asunto(s)
Síndromes Mielodisplásicos/tratamiento farmacológico , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Células Sanguíneas/patología , Médula Ósea/irrigación sanguínea , Células de la Médula Ósea/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/mortalidad , Sirolimus/farmacología , Sirolimus/uso terapéutico , Sirolimus/toxicidad , Tasa de Supervivencia , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Resultado del Tratamiento
4.
N Engl J Med ; 360(7): 692-8, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19213682

RESUMEN

Infection with the human immunodeficiency virus type 1 (HIV-1) requires the presence of a CD4 receptor and a chemokine receptor, principally chemokine receptor 5 (CCR5). Homozygosity for a 32-bp deletion in the CCR5 allele provides resistance against HIV-1 acquisition. We transplanted stem cells from a donor who was homozygous for CCR5 delta32 in a patient with acute myeloid leukemia and HIV-1 infection. The patient remained without viral rebound 20 months after transplantation and discontinuation of antiretroviral therapy. This outcome demonstrates the critical role CCR5 plays in maintaining HIV-1 infection.


Asunto(s)
Infecciones por VIH/terapia , VIH-1 , Receptores CCR5/genética , Trasplante de Células Madre , Adulto , Antirretrovirales/uso terapéutico , Antígenos CD4 , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , ADN Viral/sangre , Predisposición Genética a la Enfermedad , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , VIH-1/genética , Homocigoto , Humanos , Masculino , ARN Viral/sangre , Quimera por Trasplante , Trasplante Homólogo , Carga Viral
5.
Nat Med ; 11(2): 206-13, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15665831

RESUMEN

Infusion of endothelial progenitor cells (EPC), but not of mature endothelial cells, promotes neovascularization after ischemia. We performed gene expression profiling of EPC and endothelial cells to identify genes that might be important for the neovascularization capacity of EPC. Notably, the protease cathepsin L (CathL) was highly expressed in EPC as opposed to endothelial cells and was essential for matrix degradation and invasion by EPC in vitro. CathL-deficient mice showed impaired functional recovery following hind limb ischemia, supporting the concept of a crucial role for CathL in postnatal neovascularization. Infused CathL-deficient progenitor cells neither homed to sites of ischemia nor augmented neovascularization. Forced expression of CathL in mature endothelial cells considerably enhanced their invasive activity and sufficed to confer their capacity for neovascularization in vivo. We concluded that CathL has a critical role in the integration of circulating EPC into ischemic tissue and is required for EPC-mediated neovascularization.


Asunto(s)
Catepsinas/metabolismo , Cisteína Endopeptidasas/metabolismo , Células Endoteliales/fisiología , Neovascularización Fisiológica , Células Madre/fisiología , Animales , Biomarcadores , Catepsina L , Catepsinas/genética , Cisteína Endopeptidasas/genética , Células Endoteliales/citología , Perfilación de la Expresión Génica , Miembro Posterior/irrigación sanguínea , Miembro Posterior/fisiología , Humanos , Isquemia/metabolismo , Masculino , Ratones , Ratones Desnudos , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre/citología
6.
Future Oncol ; 6(3): 445-55, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20222800

RESUMEN

Myelodysplastic syndromes (MDS) are a heterogeneous group of diseases characterized by ineffective hematopoiesis presenting with peripheral cytopenias in combination with a hyperplastic bone marrow. MDS patients have an increased risk of disease evolution to acute leukemia. Strong efforts have been made to gain further insights into the pathobiology of MDS. Development and progression of MDS to acute myeloid leukemia is suggested to be a multistep alteration to hematopoietic stem cells consisting of class I and class II alterations: the former targeting genes that are involved in signal transduction (e.g., FLT3, RAS and KIT), whereas the latter affect transcription factors (e.g., RUNX, RARA, EVI1 and WT1). These alterations consist of not only genomic mutations but also epigenetic aberrations, which can lead to reversible gene silencing. However, whether numerical and structural alterations of chromosomes and/or single genes or epigenetic changes represent the initiating event or, more likely, secondary events remains part of the discussion. Accumulation of such defects may finally cause the leukemic transformation of MDS.


Asunto(s)
Transformación Celular Neoplásica/genética , Síndromes Mielodisplásicos/genética , Lesiones Precancerosas/genética , Animales , Progresión de la Enfermedad , Humanos
7.
J Transl Med ; 7: 7, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19146667

RESUMEN

BACKGROUND: When comparing myelogenous blasts from bone marrow and peripheral blood, immunophenotyping usually show a strong correlation of expression of surface antigens. However, it remains to be determined, whether this correlation also exists on the level of protein expression. METHOD: Therefore, we investigated both bone marrow and peripheral blood blast cells from six patients with newly diagnosed acute myeloid leukemia (AML) using conventional two-dimensional electrophoresis in the first dimension and linear polyacrylamide gels (12%) in the second dimension. Proteins were visualized using the silver staining method and image analysis was performed using the PDQuest system. RESULTS: For each patient over 80 proteins were evaluated in the sample from peripheral blood and bone marrow. We could demonstrate that the protein expression profile of bone marrow did not significantly differ from the expression patterns of peripheral blast cells. CONCLUSION: The proteome-set of leukemic blast cells from marrow and blood, does not differ substantially when drawn from AML patients with over 80 percent blast cells in both compartments. This indicates that in AML, blasts from peripheral blood samples can be considered suitable for investigations of the proteome using 2D-electrophoresis.


Asunto(s)
Células Sanguíneas/metabolismo , Células de la Médula Ósea/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteoma/análisis , Adulto , Antígenos de Superficie/metabolismo , Línea Celular , Electroforesis en Gel Bidimensional , Femenino , Humanos , Inmunofenotipificación , Focalización Isoeléctrica , Masculino , Persona de Mediana Edad , Adulto Joven
8.
J Cell Biol ; 167(1): 123-33, 2004 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-15466481

RESUMEN

Disruption of latent TGF-beta binding protein (LTBP)-4 expression in the mouse leads to abnormal lung development and colorectal cancer. Lung fibroblasts from these mice produced decreased amounts of active TGF-beta, whereas secretion of latent TGF-beta was significantly increased. Expression and secretion of TGF-beta2 and -beta3 increased considerably. These results suggested that TGF-beta activation but not secretion would be severely impaired in LTBP-4 -/- fibroblasts. Microarrays revealed increased expression of bone morphogenic protein (BMP)-4 and decreased expression of its inhibitor gremlin. This finding was accompanied by enhanced expression of BMP-4 target genes, inhibitors of differentiation 1 and 2, and increased deposition of fibronectin-rich extracellular matrix. Accordingly, increased expression of BMP-4 and decreased expression of gremlin were observed in mouse lung. Transfection of LTBP-4 rescued the -/- fibroblast phenotype, while LTBP-1 was inefficient. Treatment with active TGF-beta1 rescued BMP-4 and gremlin expression to wild-type levels. Our results indicate that the lack of LTBP-4-mediated targeting and activation of TGF-beta1 leads to enhanced BMP-4 signaling in mouse lung.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Northern Blotting , Proteína Morfogenética Ósea 4 , Diferenciación Celular , ADN Complementario/metabolismo , Electroforesis en Gel de Poliacrilamida , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Unión a TGF-beta Latente , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/química , Fenotipo , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba
9.
Int J Eat Disord ; 42(4): 293-300, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19040272

RESUMEN

OBJECTIVE: Changes of the peripheral blood cell count in patients with anorexia nervosa (AN) are frequent. Anemia and leukopenia are observed in one-third of these patients. Examination of the bone marrow reveals in almost 50% of the patients with AN signs of bone marrow atrophy and can additionally suffer from a gelatinous bone marrow transformation. METHOD: Published studies and investigations concerning hematological changes in patients with AN were reviewed. RESULTS: Anemia and mild neutropenia are detectable in almost one-third of these patients, whereas thrombocytopenia is rather uncommon. The exact mechanism for these findings is still unclear, but 50% of AN-patients with hematological changes display morphological signs of partial bone marrow atrophy. DISCUSSION: Changes of the peripheral blood cell count in patients with AN is a frequent observation but the peripheral blood cell count cannot predict the severity of bone marrow atrophy. All hematological and morphological alterations disappear completely and rapidly after sufficient refeeding.


Asunto(s)
Anemia/etiología , Anorexia Nerviosa/sangre , Anorexia Nerviosa/complicaciones , Médula Ósea/patología , Neutropenia/etiología , Anemia/sangre , Anemia/epidemiología , Anorexia Nerviosa/patología , Atrofia , Recuento de Células Sanguíneas , Eritropoyesis , Glicosaminoglicanos/análisis , Humanos , Neutropenia/sangre , Neutropenia/epidemiología , Trombocitopenia/sangre , Trombocitopenia/epidemiología , Trombocitopenia/etiología
10.
J Clin Invest ; 115(9): 2382-92, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16100571

RESUMEN

Forkhead box O (Foxo) transcription factors are emerging as critical transcriptional integrators among pathways regulating differentiation, proliferation, and survival, yet the role of the distinct Foxo family members in angiogenic activity of endothelial cells and postnatal vessel formation has not been studied. Here, we show that Foxo1 and Foxo3a are the most abundant Foxo isoforms in mature endothelial cells and that overexpression of constitutively active Foxo1 or Foxo3a, but not Foxo4, significantly inhibits endothelial cell migration and tube formation in vitro. Silencing of either Foxo1 or Foxo3a gene expression led to a profound increase in the migratory and sprout-forming capacity of endothelial cells. Gene expression profiling showed that Foxo1 and Foxo3a specifically regulate a nonredundant but overlapping set of angiogenesis- and vascular remodeling-related genes. Whereas angiopoietin 2 (Ang2) was exclusively regulated by Foxo1, eNOS, which is essential for postnatal neovascularization, was regulated by Foxo1 and Foxo3a. Consistent with these findings, constitutively active Foxo1 and Foxo3a repressed eNOS protein expression and bound to the eNOS promoter. In vivo, Foxo3a deficiency increased eNOS expression and enhanced postnatal vessel formation and maturation. Thus, our data suggest an important role for Foxo transcription factors in the regulation of vessel formation in the adult.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Neovascularización Fisiológica/fisiología , Animales , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Isquemia/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
11.
Exp Hematol ; 35(4): 572-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17379067

RESUMEN

OBJECTIVE: Retinoids are crucial in the regulation of fundamental cellular processes including terminal differentiation of both normal and malignant myeloid progenitors. The aim of this study was to identify and characterize retinoic acid (RA) target genes. METHODS AND RESULTS: RTP801 is a recently cloned stress response gene that acts as a negative regulator of the mTOR pathway. Here we identified RTP801 as a novel early RA target gene in myeloid cells. RTP801 mRNA levels are induced in acute myeloid leukemia (AML) cell lines during RA-dependent differentiation and are differentially expressed during maturation of normal CD34(+) cells. The myeloid-specific, differentiation-related transcription factor C/EBPepsilon also induces RTP801 expression. Overexpression of RTP801 in the U937 leukemic cells leads to growth inhibition and apoptosis. Conversely, silencing of endogenous RTP801 by shRNA reduces RA-induced differentiation of the U937 cells. Downregulation of RTP801 also abrogates hypoxia-induced inhibition of mTOR in those cells. CONCLUSION: Taken together, our data suggest that RTP801 is an important RA-regulated gene involved in myeloid differentiation, which could represent a therapeutic target in leukemia.


Asunto(s)
Diferenciación Celular/genética , Leucemia Mieloide/patología , Factores de Transcripción/genética , Enfermedad Aguda , Apoptosis , División Celular , Granulocitos/citología , Humanos , Leucemia Mieloide/genética , Fosforilación , Proteínas Quinasas/metabolismo , ARN Mensajero/genética , Serina-Treonina Quinasas TOR , Células U937
12.
Leuk Res ; 31(1): 27-32, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16764926

RESUMEN

Lipid kinase PIK3CA mutations have been described in several cancers. They clustered in two 'hot spots' located in helical (exon 9) and kinase (exon 20) domains associated with increased kinase activity strongly suggesting oncogenic potential. Mutational analysis of previously unexamined tumors showed an amino acid change from threonine to alanine (T1025A) in exon 20 in one of 28 endometrial cancer samples and 6 endometrial cell lines. Additionally, a silent polymorphism (T1025T) was found in two of 20 MDS samples, one of 43 NHL samples, two of 40 osteosarcoma samples and Ishikawa. The polymorphism was established by identifying two of 92 normal samples with the same change. No PIK3CA mutations were found in acute myeloid leukemia (AML), myelodysplastic syndromes (MDS) and non-Hodgkin lymphomas (NHL) as well as in osteosarcomas, prostate and ovarian cancer samples. Additionally, a previously unidentified PIK3CA pseudogene spanning exons 9-13 on chromosome 22 was discovered.


Asunto(s)
Leucemia/genética , Linfoma/genética , Mutación , Fosfatidilinositol 3-Quinasas/genética , Seudogenes , Neoplasias Óseas/genética , Línea Celular Tumoral , Cromosomas Humanos Par 22 , Fosfatidilinositol 3-Quinasa Clase I , Neoplasias Endometriales/genética , Exones , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Linfoma no Hodgkin/genética , Masculino , Osteosarcoma/genética , Neoplasias Ováricas/genética , Neoplasias de la Próstata/genética , Valores de Referencia
13.
Oncogene ; 24(9): 1589-97, 2005 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-15674331

RESUMEN

CCAAT/enhancer-binding proteins (C/EBPs) are a family of highly conserved transcription factors that have important roles in normal myelopoiesis as well as associated with myeloid disorders. The chronic myelogenous leukemia (CML) cell lines, KCL22 and K562, express exceptionally low levels of endogenous C/EBPs and provide a good model to test the effects of C/EBPs on myeloid differentiation. To explore the possibility that C/EBPdelta can promote differentiation in BCR-ABL-positive cells, we generated stable KCL22 and K562 clones that expressed an inducible C/EBPdelta gene. C/EBPdelta expression resulted in G0/G1 proliferative arrest and a moderate increase in apoptosis of the KCL22 and the K562 cells. Within 4 days of inducing expression of C/EBPdelta, myeloid differentiation of the CML blast cells occurred as shown by morphologic changes and induction of secondary granule-specific genes. We also showed that during granulocytic differentiation of KCL22 cells, the C/EBPdelta protein was detected in immunocomplexes with both Rb and E2F1. Furthermore, expression of C/EBPdelta was associated with downregulation of c-Myc and cyclin E and upregulation of the cyclin-dependent kinase inhibitor p27(Kip1) in both the KCL22 and K562 cell lines. These results show that expression of C/EBPdelta in BCR-ABL-positive leukemic cells in blast crisis is sufficient for neutrophil differentiation and point to the therapeutic potential of ectopic induction of C/EBPdelta in the acute phase of CML.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas de Fusión bcr-abl/análisis , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Factores de Transcripción/genética , Proteína delta de Unión al Potenciador CCAAT , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Sustancias de Crecimiento/farmacología , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Neutrófilos/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Leuk Res ; 30(1): 98-102, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16039715

RESUMEN

Methylation profile was analyzed in 10 childhood acute lymphoblastic leukemia (ALL) and nine adult ALL cases. Four genes (p15, p16, RARbeta, FHIT) had methylation in both diseases, four genes (p14, Rb, MLH1, DAPK) showed no methylation in both diseases, and the two genes (APC, RIZ) demonstrated methylation only in adult ALL. Methylation of the RARbeta was more frequent in adult ALL than that in childhood ALL (p=0.01). The number of patients with methylation of multiple genes was higher in adult ALL than that in childhood ALL (p=0.006). Moreover, overall frequency of methylation was higher in adult ALL than that in childhood ALL (p=0.01).


Asunto(s)
Islas de CpG , Metilación de ADN , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
15.
FASEB J ; 19(8): 974-6, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15824087

RESUMEN

Endothelial progenitor cells (EPCs) contribute to postnatal neovascularization. Risk factors for coronary artery disease reduce the number of EPCs in humans. Since EPC apoptosis might be a potential mechanism to regulate the number of EPCs, we investigated the effects of oxidative stress and HMG-CoA-reductase inhibitors (statins) on EPC apoptosis. Atorvastatin, mevastatin, or VEGF prevented EPC apoptosis induced by H2O2. The antiapoptotic effect was reversed by inhibition of the PI3K/Akt pathway. Forkhead transcription factors (FOXO1, FOXO3a, FOXO4) exert proapoptotic effects and are phosphorylated and, thereby, inactivated by Akt. Therefore, we elucidated the involvement of forkhead transcription factors. Atorvastatin induced the phosphorylation of the predominant forkhead factor FOXO4 in EPCs. In addition, atorvastatin reduced the expression of the proapoptotic forkhead-regulated protein Bim in a PI3K-dependent manner. Consistently, overexpression of FOXO4 activated the Bim promoter as determined by reporter gene expression and stimulated the expression of Bim, resulting in an increased EPC apoptosis. Statins failed to prevent EPC apoptosis induced by overexpression of Bim or nonphosphorylatable FOXO4, suggesting that the protective effects of statins depend on this pathway. In summary, our results show that FOXO-dependent expression of Bim plays a pivotal role for EPC apoptosis. Statins reduce oxidative stress-induced EPC apoptosis, inactivate FOXO4, and down-regulate Bim.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Expresión Génica , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/fisiología , Atorvastatina , Proteína 11 Similar a Bcl2 , Proteínas de Ciclo Celular , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/fisiología , Citometría de Flujo , Factores de Transcripción Forkhead , Expresión Génica/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Leucocitos Mononucleares , Proteínas de la Membrana/fisiología , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/fisiología , Pirroles/farmacología , Células Madre/fisiología , Transfección , Venas Umbilicales
16.
Circ Res ; 92(9): 1049-55, 2003 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-12676819

RESUMEN

Endothelial progenitor cells (EPCs) play an important role in postnatal neovascularization of ischemic tissue. Ex vivo expansion of EPCs might be useful for potential clinical cell therapy of myocardial ischemia. However, cultivation of primary cells leads to cellular aging (senescence), thereby severely limiting the proliferative capacity. Therefore, we investigated whether statins might be able to prevent senescence of EPCs. EPCs were isolated from peripheral blood and characterized. After ex vivo cultivation, EPCs became senescent as determined by acidic beta-galactosidase staining. Atorvastatin or mevastatin dose-dependently inhibited the onset of EPC senescence in culture. Moreover, atorvastatin increased proliferation of EPCs as assessed by BrdU incorporation and colony-forming capacity. Whereas geranylgeranylpyrophosphate or farnesylpyrophosphate reduced the senescence inhibitory effect of atorvastatin, NO synthase inhibition, antioxidants, or Rho kinase inhibitors had no effect. To get further insights into the underlying downstream effects of statins, we measured telomerase activity and determined the expression of various cell cycle regulatory genes by using a microarray assay. Whereas telomerase activity did not change, atorvastatin modulated expression of cell cycle genes including upregulation of cyclins and downregulation of the cell cycle inhibitor p27Kip1. Taken together, statins inhibited senescence of EPCs independent of NO, reactive oxygen species, and Rho kinase, but dependent on geranylgeranylpyrophosphate. Atorvastatin-mediated prevention of EPC senescence appears to be mediated by the regulation of various cell cycle proteins. The inhibition of EPC senescence and induction of EPC proliferation by statins in vitro may importantly improve the functional activity of EPCs for potential cell therapy.


Asunto(s)
Endotelio Vascular/citología , Genes cdc , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/análogos & derivados , Lovastatina/farmacología , Pirroles/farmacología , Células Madre/efectos de los fármacos , Atorvastatina , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Ciclinas/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Células Madre/metabolismo , Células Madre/fisiología , Telomerasa/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
17.
Eur J Radiol Open ; 3: 134-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27437433

RESUMEN

PURPOSE: Computed tomography (CT) scans are an important source of ionizing irradiation (IR) in medicine that can induce a variety of DNA damage in human tissues. With technological improvements CT scans at reduced absorbed doses became feasible presumably lowering genotoxic side effects. MATERIALS AND METHODS: For measuring DNA damage we performed γH2AX foci microscopy in peripheral blood mononuclear cells (PBMC) after exposure to reduced and conventional absorbed radiation doses using 3rd generation dual-source CT (DSCT) technology. RESULTS: CT scans performed at reduced absorbed doses of 3 mGy induced significant lower levels (p < 0.0001) of DNA damage (0.05 focus per cell ± 0.01 [mean ± standard error of mean]) at 5 min after IR compared to conventional absorbed doses of 15 mGy (0.30 focus per cell ± 0.03). With ongoing DNA repair background γH2AX foci levels (0.05 focus per cell) were approached at 24 h after CT with both protocols. CONCLUSION: Our results provide evidence that reduced absorbed doses mediated by adjusted tube current in 3rd generation DSCT induce lower levels of DNA damage in PBMC compared to conventional absorbed doses suggesting a lower genotoxic risk for state-of-the-art tube current reduced CT protocols.

18.
Sci Rep ; 6: 35181, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27748377

RESUMEN

The aim of this study was to systematically evaluate the cumulative radiation exposure and the associated lifetime-cancer-risk from diagnostic imaging in patients with Hodgkin-lymphoma-(HL) or diffuse-large-B-cell-lymphoma (DLBCL). 99 consecutive patients (53-males) diagnosed with HL or DLBCL were included in the study and followed. Based on the imaging reports, organ and effective-doses-(ED) were calculated individually for each patient and the excess lifetime risks were estimated. The average ED in the first year after diagnosis was significantly different for men (59 ± 33 mSv) and women (74 ± 33 mSv)-(p < 0.05). The mean cumulative ED in each of the following 5 years was 16 ± 16 mSv without significant differences between men and women-(p > 0.05). Over all years, more than 90% of the ED resulted from CT. The average cumulative radiation risk estimated for the first year was significantly lower for men (0.76 ± 0.41%) as compared to women (1.28 ± 0.54%)-(p < 0.05). The same was found for each of the subsequent 5-years (men-0.18 ± 0.17%; women-0.28 ± 0.25%)-(p < 0.05). In conclusion, for HL and DLBCL patients investigated in this study, a cumulative radiation risk of about 1 excess cancer per 100 patients is estimated for diagnostic imaging procedures performed during both the first year after diagnosis and a follow-up period of 5 years.


Asunto(s)
Diagnóstico por Imagen/efectos adversos , Enfermedad de Hodgkin/diagnóstico por imagen , Tablas de Vida , Linfoma de Células B Grandes Difuso/diagnóstico por imagen , Neoplasias Inducidas por Radiación/epidemiología , Dosis de Radiación , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Exposición a la Radiación , Riesgo , Adulto Joven
19.
Oncogene ; 23(47): 7846-53, 2004 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-15361842

RESUMEN

Myelofibrosis with myeloid metaplasia (MMM) is a clonal stem-cell disorder that leads to ineffective hematopoiesis, bone marrow fibrosis, and extramedullary hematopoiesis. The molecular mechanisms underlying the development of this myeloproliferative syndrome are currently unknown. In order to identify tumor suppressor genes that may be involved in the disease process, we performed an analysis for loss of heterozygosity (LOH) in CD34+ cells from 29 patients with MMM. We observed a frequency of allelic loss on chromosomal arm 3p in 24% of cases. Detailed mapping of 3p revealed a distinct region of deletion at 3p24. Among the genes known to map within this region is the retinoic acid receptor-beta (RARbeta2) gene. To determine whether RARbeta2 gene activity is diminished in this disease, we analysed its expression in CD34+ cells from 17 patients with MMM using quantitative PCR. Our results indicate that expression of RARbeta2 is significantly decreased in 100% of patient samples compared to that in CD34+ cells from 10 normal individuals. Since allelic loss at 3p24 occurs in <25% of patients, we investigated the contribution of epigenetic modifications to RARbeta2 inactivity. Using methylation-specific PCR, we found hypermethylation of RARbeta2 in 16 of 18 patients (89%), while the methylated form of the gene was absent in CD34+ cells from nine normal individuals. Our results suggest that RARbeta2 acts as a tumor suppressor gene in MMM and that epigenetic changes are the most significant determinants of RARbeta2 gene activity in these patients.


Asunto(s)
Genes Supresores de Tumor , Mielofibrosis Primaria/genética , Receptores de Ácido Retinoico/genética , Secuencia de Bases , Células de la Médula Ósea/patología , Mapeo Cromosómico , Cromosomas Humanos Par 3/genética , Cartilla de ADN , Marcadores Genéticos , Humanos , Cariotipificación , Pérdida de Heterocigocidad , Metaplasia/genética , Repeticiones de Microsatélite/genética , Reacción en Cadena de la Polimerasa/métodos , Mielofibrosis Primaria/patología
20.
Exp Hematol ; 30(4): 318-23, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11937266

RESUMEN

OBJECTIVE: The human insulin-like growth factor 2 (IGF2) gene was thought to be imprinted and expressed only from the paternal allele in normal tissue. MATERIALS AND METHODS: Initially, we analyzed the imprinting status of IGF2 in bone marrow cells from 49 patients with myelodysplastic syndromes (MDS) utilizing the Apa I polymorphism of IGF2. Thirteen bone marrow and 14 peripheral blood samples from normal individuals served as controls. We utilized normal peripheral blood T lymphocytes to examine the relationship between genomic imprinting and cell proliferation. Expression of IGF2 was quantified by real-time PCR and proliferation of T cells was measured by 3H-thymidine incorporation. Furthermore, methylation status of the imprinting controlling region (ICR) was analyzed by subcloning and sequencing of genomic DNA after sodium bisulfite modification. RESULTS: Among 24 patients who were heterozygous for IGF2, loss of imprinting (LOI) occurred in 22 cases (92%). Surprisingly, LOI of IGF2 occurred in the normal bone marrow cells, but the normal peripheral blood cells showed retention of imprinting (ROI). Unstimulated normal T cells showed ROI. After 24 hours of exposure to PHA, these cells changed their IGF2 imprinting status from ROI to LOI. Concomitantly, their IGF2 RNA levels increased up to sixfold and their proliferation increased 10- to 20-fold. In contrast, normal T cells not stimulated with PHA did not develop LOI of IGF2, had negligible levels of IGF2 RNA, and did not increase their proliferation. In unstimulated T cells, the CpG islands of the ICR were completely methylated on one allele and nearly completely unmethylated on the other allele. After PHA stimulation, the CpG islands at the ICR became completely methylated on both alleles. CONCLUSION: LOI of IGF2 is strongly associated with cell proliferation and is not limited to cancer cells.


Asunto(s)
Impresión Genómica , Células Madre Hematopoyéticas/metabolismo , Factor II del Crecimiento Similar a la Insulina/genética , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Estudios de Casos y Controles , División Celular , Islas de CpG/genética , Metilación de ADN , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Síndromes Mielodisplásicos/patología , Polimorfismo Genético , ARN Mensajero/biosíntesis , Linfocitos T/citología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA