Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Hematol ; 91(5): 481-5, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26852175

RESUMEN

Mantle cell lymphoma (MCL) is an aggressive lymphoid neoplasm with poor prognosis. Acquired telomerase reverse transcriptase gene promoter (TERTp) mutations are among the most frequent somatic non-coding mutations in cancers. In this study, the prevalence of TERTp mutations in 24 MCL and 21 other lymphoid neoplasias (oLN) was investigated. Eight MCL samples (33%) carried TERTp mutations, two homozygous and six heterozygous (seven C228T and one C250T), which directly correlated with higher TERT transcription, mitochondrial DNA copy number, and IGHV mutational status in MCL neoplastic cells. TERTp mutations were not found in oLN. TERTp mutations correlated with more lymphoma proliferation and tumor burden, as suggested by the higher number of lymphoma cells circulating in peripheral blood, and tended to associate with longer MCL telomeres, especially in homozygous mutants, although not statistically significant. Telomere-biology genes were overexpressed in MCL cells in comparison to healthy lymphocytes, but were not influenced by mutation status. The findings described for the first time that acquired TERTp mutations are common in MCL but not in other lymphoid neoplasms. It was also demonstrated that TERTp mutations are associated with higher TERT mRNA expression in MCL cells in vivo and higher tumor burden, suggesting these mutations as a driver event in MCL development and progression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Linfoma de Células del Manto/genética , Mutación Puntual , Regiones Promotoras Genéticas/genética , Telomerasa/genética , Transcripción Genética , Anciano , Anciano de 80 o más Años , División Celular , ADN Mitocondrial/análisis , ADN de Neoplasias/genética , Progresión de la Enfermedad , Femenino , Dosificación de Gen , Genotipo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Leucemia/genética , Linfoma/genética , Masculino , Persona de Mediana Edad , Telómero/ultraestructura , Carga Tumoral , Macroglobulinemia de Waldenström/genética
2.
Reprod Sci ; 23(5): 644-54, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26586671

RESUMEN

BACKGROUND: Physical activity is known to relieve the metabolic complications of polycystic ovary syndrome (PCOS), and exercise is also associated with telomere biology. We investigated the changes induced by progressive resistance training (PRT) in telomere content and metabolic disorder in women with PCOS and controls. PARTICIPANTS AND METHODS: Forty-five women with PCOS and 52 healthy women aged 18 to 37 years were submitted to PRT. A linear periodization of PRT was prepared based on a trend of decreasing volume and intensity throughout the training period. The volunteers performed PRT 3 times a week for 4 months. The participants' physical characteristics and hormonal concentrations were measured before and after PRT, as telomere content that was measured using quantitative real-time polymerase chain reaction. RESULTS: Briefly, Progressive resistance training reduced waist circumference, body fat percentage, plasma testosterone and sex hormone-binding globulin concentrations, glycemia, and free androgen index. Fasting insulin and insulin resistance index were greater in women with PCOS. Androstenedione and homocysteine increased after PRT. There were no differences in telomere content between controls (0.96 ± 0.3 before vs 0.85 ± 0.21 after) and women with PCOS (0.94 ± 0.33 before vs 0.88 ± 0.39 after). Adjusted analysis showed telomere shortening after PRT in all women (0.95 ± 0.31 before vs 0.86 ± 0.31 after; P = .03). In women with PCOS, increased homocysteine levels were related to telomere reduction and increased androstenedione was positively correlated with telomere content after PRT. CONCLUSIONS: Progressive resistance training had positive effects on the hormonal and physical characteristics of women with PCOS and controls, but telomere content was reduced and homocysteine level increased in all participants.


Asunto(s)
Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/terapia , Entrenamiento de Fuerza/métodos , Telómero/fisiología , Tejido Adiposo/fisiología , Adolescente , Adulto , Glucemia/metabolismo , Femenino , Humanos , Resistencia a la Insulina/fisiología , Obesidad/sangre , Obesidad/diagnóstico , Obesidad/terapia , Síndrome del Ovario Poliquístico/diagnóstico , Circunferencia de la Cintura/fisiología , Adulto Joven
4.
PLoS One ; 9(11): e113747, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25409313

RESUMEN

Telomere length measurement is an essential test for the diagnosis of telomeropathies, which are caused by excessive telomere erosion. Commonly used methods are terminal restriction fragment (TRF) analysis by Southern blot, fluorescence in situ hybridization coupled with flow cytometry (flow-FISH), and quantitative PCR (qPCR). Although these methods have been used in the clinic, they have not been comprehensively compared. Here, we directly compared the performance of flow-FISH and qPCR to measure leukocytes' telomere length of healthy individuals and patients evaluated for telomeropathies, using TRF as standard. TRF and flow-FISH showed good agreement and correlation in the analysis of healthy subjects (R(2) = 0.60; p<0.0001) and patients (R(2) = 0.51; p<0.0001). In contrast, the comparison between TRF and qPCR yielded modest correlation for the analysis of samples of healthy individuals (R(2) = 0.35; p<0.0001) and low correlation for patients (R(2) = 0.20; p = 0.001); Bland-Altman analysis showed poor agreement between the two methods for both patients and controls. Quantitative PCR and flow-FISH modestly correlated in the analysis of healthy individuals (R(2) = 0.33; p<0.0001) and did not correlate in the comparison of patients' samples (R(2) = 0.1, p = 0.08). Intra-assay coefficient of variation (CV) was similar for flow-FISH (10.8 ± 7.1%) and qPCR (9.5 ± 7.4%; p = 0.35), but the inter-assay CV was lower for flow-FISH (9.6 ± 7.6% vs. 16 ± 19.5%; p = 0.02). Bland-Altman analysis indicated that flow-FISH was more precise and reproducible than qPCR. Flow-FISH and qPCR were sensitive (both 100%) and specific (93% and 89%, respectively) to distinguish very short telomeres. However, qPCR sensitivity (40%) and specificity (63%) to detect telomeres below the tenth percentile were lower compared to flow-FISH (80% sensitivity and 85% specificity). In the clinical setting, flow-FISH was more accurate, reproducible, sensitive, and specific in the measurement of human leukocyte's telomere length in comparison to qPCR. In conclusion, flow-FISH appears to be a more appropriate method for diagnostic purposes.


Asunto(s)
Hibridación Fluorescente in Situ/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Acortamiento del Telómero/fisiología , Telómero/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anemia Aplásica/metabolismo , Anemia Aplásica/patología , Niño , Preescolar , Disqueratosis Congénita/metabolismo , Disqueratosis Congénita/patología , Femenino , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Lactante , Recién Nacido , Leucocitos/citología , Leucocitos/metabolismo , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Adulto Joven
5.
Blood ; 120(1): 207-13, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22517898

RESUMEN

Increased fibrinolysis is an important component of acute promyelocytic leukemia (APL) bleeding diathesis. APL blasts overexpress annexin II (ANXII), a receptor for tissue plasminogen activator (tPA), and plasminogen, thereby increasing plasmin generation. Previous studies suggested that ANXII plays a pivotal role in APL coagulopathy. ANXII binding to tPA can be inhibited by homocysteine and hyperhomocysteinemia can be induced by L-methionine supplementation. In the present study, we used an APL mouse model to study ANXII function and the effects of hyperhomocysteinemia in vivo. Leukemic cells expressed higher ANXII and tPA plasma levels (11.95 ng/mL in leukemic vs 10.74 ng/mL in wild-type; P = .004). In leukemic mice, administration of L-methionine significantly increased homocysteine levels (49.0 µmol/mL and < 6.0 µmol/mL in the treated and nontreated groups, respectively) and reduced tPA levels to baseline concentrations. The latter were also decreased after infusion of the LCKLSL peptide, a competitor for the ANXII tPA-binding site (11.07 ng/mL; P = .001). We also expressed and purified the p36 component of ANXII in Pichia methanolica. The infusion of p36 in wild-type mice increased tPA and thrombin-antithrombin levels, and the latter was reversed by L-methionine administration. The results of the present study demonstrate the relevance of ANXII in vivo and suggest that methionine-induced hyperhomocysteinemia may reverse hyperfibrinolysis in APL.


Asunto(s)
Anexina A2/metabolismo , Fibrinólisis/fisiología , Hiperhomocisteinemia/inducido químicamente , Leucemia Promielocítica Aguda , Metionina/farmacología , Animales , Anexina A2/farmacología , Coagulación Sanguínea/fisiología , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Fibrinolisina/metabolismo , Homocisteína/sangre , Leucemia Promielocítica Aguda/complicaciones , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Activador de Tejido Plasminógeno/sangre
6.
PLoS One ; 6(10): e26713, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22053203

RESUMEN

Promyelocytic leukemia-retinoic acid receptor alpha (PML-RARα) expression in acute promyelocytic leukemia (APL) impairs transforming growth factor beta (TGFß) signaling, leading to cell growth advantage. Halofuginone (HF), a low-molecular-weight alkaloid that modulates TGFß signaling, was used to treat APL cell lines and non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice subjected to transplantation with leukemic cells from human chorionic gonadotrophin-PML-RARα transgenic mice (TG). Cell cycle analysis using incorporated bromodeoxyuridine and 7-amino-actinomycin D showed that, in NB4 and NB4-R2 APL cell lines, HF inhibited cellular proliferation (P<0.001) and induced apoptosis (P = 0.002) after a 24-hour incubation. Addition of TGFß revealed that NB4 cells were resistant to its growth-suppressive effects and that HF induced these effects in the presence or absence of the cytokine. Cell growth inhibition was associated with up-regulation of TGFß target genes involved in cell cycle regulation (TGFB, TGFBRI, SMAD3, p15, and p21) and down-regulation of MYC. Additionally, TGFß protein levels were decreased in leukemic TG animals and HF in vivo could restore TGFß values to normal. To test the in vivo anti-leukemic activity of HF, we transplanted NOD/SCID mice with TG leukemic cells and treated them with HF for 21 days. HF induced partial hematological remission in the peripheral blood, bone marrow, and spleen. Together, these results suggest that HF has anti-proliferative and anti-leukemic effects by reversing the TGFß blockade in APL. Since loss of the TGFß response in leukemic cells may be an important second oncogenic hit, modulation of TGFß signaling may be of therapeutic interest.


Asunto(s)
Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Piperidinas/farmacología , Quinazolinonas/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Animales , Recuento de Células Sanguíneas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda/sangre , Leucemia Promielocítica Aguda/genética , Ratones , Ratones SCID , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba/efectos de los fármacos
7.
Leuk Res ; 35(5): 657-62, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21168913

RESUMEN

Indole-3-acetic acid (IAA), when oxidized by horseradish peroxidase (HRP), is transformed into cytotoxic molecules capable of inducing cell injury. The aim of this study was to test if, by targeting hematopoietic tumors with HRP-conjugated antibodies in association with IAA treatment, there is induction of apoptosis. We used two lineages of hematologic tumors: NB4, derived from acute promyelocytic leukemia (APL) and Granta-519 from mantle cell lymphoma (MCL). We also tested cells from 12 patients with acute myeloid leukemia (AML) and from 10 patients with chronic lymphocytic leukemia (CLL). HRP targeting was performed with anti-CD33 or anti-CD19 antibodies (depending on the origin of the cell), followed by incubation with goat anti-mouse antibody conjugated with HRP. Eight experimental groups were analyzed: control, HRP targeted, HRP targeted and incubated with 1, 5 and 10mM IAA, and cells not HRP targeted but incubated with 1, 5 and 10mM IAA. Apoptosis was analyzed by flow cytometry using annexin V-FITC and propidium iodide labeling. Results showed that apoptosis was dependent on the dose of IAA utilized, the duration of exposure to the prodrug and the origin of the neoplasia. Targeting HRP with antibodies was efficient in activating IAA and inducing apoptosis.


Asunto(s)
Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Hematológicas/patología , Ácidos Indolacéticos/farmacología , Adolescente , Adulto , Anticuerpos/química , Apoptosis/fisiología , Técnicas de Cultivo de Célula , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Estudios de Cohortes , Sistemas de Liberación de Medicamentos/métodos , Femenino , Peroxidasa de Rábano Silvestre/metabolismo , Peroxidasa de Rábano Silvestre/farmacología , Humanos , Inmunotoxinas/química , Inmunotoxinas/farmacología , Ácidos Indolacéticos/química , Masculino , Persona de Mediana Edad , Células Tumorales Cultivadas
9.
Blood ; 114(27): 5415-25, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-19797526

RESUMEN

Acute promyelocytic leukemia (APL) is characterized by a block in differentiation and accumulation of promyelocytes in the bone marrow and blood. The majority of APL patients harbor the t(15:17) translocation leading to expression of the fusion protein promyelocytic-retinoic acid receptor alpha. Treatment with retinoic acid leads to degradation of promyelocytic-retinoic acid receptor alpha protein and disappearance of leukemic cells; however, 30% of APL patients relapse after treatment. One potential mechanism for relapse is the persistence of cancer "stem" cells in hematopoietic organs after treatment. Using a novel sorting strategy we developed to isolate murine myeloid cells at distinct stages of differentiation, we identified a population of committed myeloid cells (CD34(+), c-kit(+), FcgammaRIII/II(+), Gr1(int)) that accumulates in the spleen and bone marrow in a murine model of APL. We observed that these cells are capable of efficiently generating leukemia in recipient mice, demonstrating that this population represents the APL cancer-initiating cell. These cells down-regulate the transcription factor CCAAT/enhancer binding protein alpha (C/EBPalpha) possibly through a methylation-dependent mechanism, indicating that C/EBPalpha deregulation contributes to transformation of APL cancer-initiating cells. Our findings provide further understanding of the biology of APL by demonstrating that a committed transformed progenitor can initiate and propagate the disease.


Asunto(s)
Células de la Médula Ósea/patología , Leucemia Promielocítica Aguda/patología , Células Mieloides/patología , Células Madre Neoplásicas/patología , Animales , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Calgranulina A/genética , Calgranulina A/metabolismo , Línea Celular Tumoral , Metilación de ADN , Femenino , Citometría de Flujo , Humanos , Estimación de Kaplan-Meier , Leucemia Promielocítica Aguda/sangre , Leucemia Promielocítica Aguda/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células U937
10.
Rev. bras. hematol. hemoter ; 30(supl.2): 33-36, jun. 2008. ilus
Artículo en Inglés | LILACS | ID: lil-496442

RESUMEN

Differentiation syndrome is a treatment complication which can occur in acute promyelocytic leukemia (APL) patients treated with all-trans retinoic acid (ATRA) or arsenic trioxide (ATO), which is characterized by enhanced leukocyte transmigration. Several cellular and molecular mechanisms participate in differentiation syndrome development. This review discusses the changes in expression of adhesion molecules induced during ATRA and ATO treatments and their possible implications in the pathogenesis of this potentially fatal complication.


A síndrome da diferenciação (DS) é um efeito colateral que pode ocorrer em pacientes com leucemia promielocítica aguda (APL) tratados com ácido all-trans-retinóico (ATRA) ou trióxido de arsênico (ATO), sendo caracterizada pelo aumento da transmigração de leucócitos. Vários mecanismos celulares e moleculares participam no desenvolvimento da DS. Esta revisão discute as mudanças na expressão de moléculas de adesão induzidas durante o tratamento com ATRA e ATO e possíveis implicações na patogênese desta complicação potencialmente fatal.


Asunto(s)
Humanos , Arsénico , Moléculas de Adhesión Celular , Leucemia Promielocítica Aguda , Retinoides
11.
Br J Haematol ; 142(1): 74-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18422993

RESUMEN

TP73 encodes for two proteins: full-length TAp73 and DeltaNp73, which have little transcriptional activity and exert dominant-negative function towards TP53 and TAp73. We compared TATP73 and DeltaNTP73 expression in acute myeloid leukaemia (AML) samples and normal CD34(+) progenitors. Both forms were more highly expressed in leukaemic cells. Amongst AML blasts, TATP73 was more expressed in AML harbouring the recurrent genetic abnormalities (RGA): PML-RARA, RUNX1-RUNX1T1 and CBFB-MYH11, whereas higher DeltaNTP73 expression was detected in non-RGA cases. TP53 expression did not vary according to DeltaNTP73/TATP73 expression ratio. Leukaemic cells with higher DeltaNTP73/TATP73 ratios were significantly more resistant to cytarabine-induced apoptosis.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Citarabina/uso terapéutico , Proteínas de Unión al ADN/genética , Genes p53/genética , Leucemia Mieloide Aguda/genética , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Apoptosis/efectos de los fármacos , Aberraciones Cromosómicas , Resistencia a Antineoplásicos , Expresión Génica , Reordenamiento Génico , Humanos , Reacción en Cadena de la Polimerasa , Recurrencia
12.
J Leukoc Biol ; 82(5): 1340-3, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17704297

RESUMEN

The use of all trans-retinoic acid (ATRA) is the basis of treatment of acute promyelocytic leukemia (APL) and represents the paradigm of differentiation therapy. In general, ATRA is well-tolerated but may be associated with a potentially lethal side-effect, referred to as retinoic acid or differentiation syndrome (DS). The cellular and molecular mechanisms of DS are poorly understood and involve changes in the adhesive qualities and cytokine secretion of leukemic cells during ATRA-induced differentiation. As leukocyte extravasation is a key event in DS pathogenesis, we analyzed the association between the polymorphisms at Exon 4 (G241R) and Exon 6 (E469K) of ICAM-1 and Exon 3 (L125V) of PECAM-1 genes with DS development in APL patients treated with ATRA and anthracyclines. DS was diagnosed in 23/127 (18.1%) APL patients at an average of 11.5 days after the start of ATRA. All patients presented respiratory distress associated with increased ground-glass opacity in chest radiographies. Other accompanying symptoms were: fever not attributable to infection (65.2%), generalized edema (37.5%), weight gain (37.5%), and impairment of renal function (8.6%). We detected an association between development of DS and the AA genotype at Codon 469 of ICAM-1 (odds ratio of 3.5; 95% confidence interval: 1.2-10.2). Conversely, no significant association was detected between G241R or L125V polymorphisms at Exon 4 of ICAM-1 and Exon 3 of PECAM-1, respectively. Our results suggest that susceptibility to DS in APL patients may be influenced by genetic variation in adhesion molecule loci.


Asunto(s)
Exones/genética , Molécula 1 de Adhesión Intercelular/genética , Leucemia Promielocítica Aguda/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Polimorfismo Genético/genética , Adulto , Antineoplásicos/efectos adversos , Diferenciación Celular , Diagnóstico Diferencial , Femenino , Humanos , Leucemia Promielocítica Aguda/complicaciones , Leucemia Promielocítica Aguda/diagnóstico , Masculino , Síndrome , Tretinoina/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...