Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Eur J Pediatr ; 174(10): 1399-403, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25902755

RESUMEN

UNLABELLED: The most common diagnosis for pediatric thrombocytopenia is immune thrombocytopenia. Nevertheless, in atypical cases, the hypothesis of an inherited thrombocytopenia has to be investigated. We report a series of cases of a newly described entity, genetic thrombocytopenia with mutation in the ankyrine 26 gene, diagnosed from the exploration of five pediatric cases of thrombocytopenia. This entity is characterized by a moderate thrombocytopenia with normal mean platelet volume, and poorly bleeding. Its transmission is autosomal dominant. Final diagnosis is made by sequencing of a short DNA region of ANKRD26 gene. This pathology can be considered as an hematological malignancy predisposition syndrome. CONCLUSION: We report the first cohort of pediatric patients diagnosed with thrombocytopenia with mutation in the ankyrine 26. The aim is to underline the specificities of this entity in children and bring it to the knowledge of pediatricians who may be in first place to manage these patients. WHAT IS KNOWN: • Genetic thrombocytopenia with mutation in the ankyrine 26 gene is a recently described entity, which seems to be considered as a predisposition for hematologic malignancies. • The first cohort has been reported in 2011, by Noris et al., in 78 Italian adult patients. What is New: • We describe clinical and biological features of the first pediatric cohort diagnosed with genetic thrombocytopenia with mutation in the ankyrine 26 gene. • It seemed important to consider the pediatric specificities of this entity to enable pediatricians to investigate, diagnose, and manage pediatric patients and their families.


Asunto(s)
ADN/genética , Mutación , Trombocitopenia/genética , Adolescente , Adulto , Repetición de Anquirina , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Herencia , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Persona de Mediana Edad , Proteínas Nucleares , Linaje , Trombocitopenia/diagnóstico , Trombocitopenia/metabolismo
2.
Arch Pediatr ; 30(4): 212-218, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37061360

RESUMEN

BACKGROUND: Little is known about blood lymphocyte subpopulations in children with common (CO) or syndromic (SO) obesity. We aimed to describe the blood lymphocyte profiles of obese children and to search for associations with clinical phenotypes. METHODS: Main blood lymphocyte subpopulations were analyzed in 159 children with CO and 34 with SO in a retrospective cohort. Phenotypes included obesity history, body mass index (BMI) Z score, percentage fat mass, and inflammatory parameters. Correlations were performed between phenotypes and circulating lymphocyte profiles. RESULTS: Children with SO had a higher BMI Z score (5.5 ± 1.7 SD) than children with CO (4.7 ± 0.9 SD; p = 0.01). Significant differences were found for lymphocyte counts, including a higher percentage of CD19+ B cells (SO = 20.1 ± 6.7 vs. CO = 17.1 ± 6.1%, p = 0.03), despite lower absolute numbers (SO = 0.57 ± 0.20 vs. CO = 0.63 ± 1.9 g/L, p < 0.01). However, no difference in the lymphocyte profile was found between children with SO and those with the most severe CO (BMI Z score ≥ 4.7 SD). CONCLUSION: Children with SO have altered blood lymphocyte profiles with increased prevalence of CD19+ B cells, which is closely linked to the degree of obesity severity and inflammatory markers.


Asunto(s)
Obesidad Infantil , Niño , Humanos , Obesidad Infantil/epidemiología , Estudios Retrospectivos , Índice de Masa Corporal , Linfocitos
3.
Leukemia ; 37(5): 988-1005, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37019990

RESUMEN

Chromosomal rearrangements of the human KMT2A/MLL gene are associated with de novo as well as therapy-induced infant, pediatric, and adult acute leukemias. Here, we present the data obtained from 3401 acute leukemia patients that have been analyzed between 2003 and 2022. Genomic breakpoints within the KMT2A gene and the involved translocation partner genes (TPGs) and KMT2A-partial tandem duplications (PTDs) were determined. Including the published data from the literature, a total of 107 in-frame KMT2A gene fusions have been identified so far. Further 16 rearrangements were out-of-frame fusions, 18 patients had no partner gene fused to 5'-KMT2A, two patients had a 5'-KMT2A deletion, and one ETV6::RUNX1 patient had an KMT2A insertion at the breakpoint. The seven most frequent TPGs and PTDs account for more than 90% of all recombinations of the KMT2A, 37 occur recurrently and 63 were identified so far only once. This study provides a comprehensive analysis of the KMT2A recombinome in acute leukemia patients. Besides the scientific gain of information, genomic breakpoint sequences of these patients were used to monitor minimal residual disease (MRD). Thus, this work may be directly translated from the bench to the bedside of patients and meet the clinical needs to improve patient survival.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Leucemia Mieloide Aguda , Proteína de la Leucemia Mieloide-Linfoide , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Lactante , Preescolar , Niño , Adolescente , Adulto , Persona de Mediana Edad , Anciano , Leucemia Mieloide Aguda/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , N-Metiltransferasa de Histona-Lisina/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Fusión Génica
4.
Hemasphere ; 3(6): e316, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31976488

RESUMEN

The clinical and biological characteristics of children under 2 years (infants) with acute myeloid leukemia (AML) are different from those of older children. We aimed to describe the specific characteristics of this population and the potential factors that influence the prognosis. We analyzed data concerning 438 children with newly-diagnosed AML treated in the ELAM02 protocol between March 2005 and December 2011, of which 103 were under 2 years old at diagnosis. The evaluation criteria were overall survival (OS) and event-free survival (EFS) of infants vs older children. The clinical and biological features were secondary criteria. Infants presented more frequent extra-medullary presentation than older children. They had a significantly higher proportion of skin lesions and central nervous system involvement (15% vs 3%, p < 0.0001 and 26% vs 12%, p = 0.0005, respectively). The global incidence of KMT2A rearrangements was nearly 55% for infants vs 11% for older children (p < 0.0001). Median 5-year OS was 70.4% for infants vs 71.4% for older children (p = 0.83). Five-year EFS was 67% for infants vs 58% for older children (p = 0.27). Infants with AML represent a cohort of patients with specific clinical and biological features. These remarkable differences had no significant impact on their outcome in the ELAM02 protocol.

5.
Leukemia ; 32(2): 273-284, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28701730

RESUMEN

Chromosomal rearrangements of the human MLL/KMT2A gene are associated with infant, pediatric, adult and therapy-induced acute leukemias. Here we present the data obtained from 2345 acute leukemia patients. Genomic breakpoints within the MLL gene and the involved translocation partner genes (TPGs) were determined and 11 novel TPGs were identified. Thus, a total of 135 different MLL rearrangements have been identified so far, of which 94 TPGs are now characterized at the molecular level. In all, 35 out of these 94 TPGs occur recurrently, but only 9 specific gene fusions account for more than 90% of all illegitimate recombinations of the MLL gene. We observed an age-dependent breakpoint shift with breakpoints localizing within MLL intron 11 associated with acute lymphoblastic leukemia and younger patients, while breakpoints in MLL intron 9 predominate in AML or older patients. The molecular characterization of MLL breakpoints suggests different etiologies in the different age groups and allows the correlation of functional domains of the MLL gene with clinical outcome. This study provides a comprehensive analysis of the MLL recombinome in acute leukemia and demonstrates that the establishment of patient-specific chromosomal fusion sites allows the design of specific PCR primers for minimal residual disease analyses for all patients.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide Aguda/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Adulto , Niño , Aberraciones Cromosómicas , Rotura Cromosómica , Femenino , Reordenamiento Génico/genética , Humanos , Lactante , Masculino , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Translocación Genética/genética
6.
J Clin Invest ; 108(6): 851-9, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11560954

RESUMEN

The mitogen-activated protein kinase (MAPK) pathway regulates growth and survival of many cell types, and its constitutive activation has been implicated in the pathogenesis of a variety of malignancies. In this study we demonstrate that small-molecule MEK inhibitors (PD98059 and PD184352) profoundly impair cell growth and survival of acute myeloid leukemia (AML) cell lines and primary samples with constitutive MAPK activation. These agents abrogate the clonogenicity of leukemic cells but have minimal effects on normal hematopoietic progenitors. MEK blockade also results in sensitization to spontaneous and drug-induced apoptosis. At a molecular level, these effects correlate with modulation of the expression of cyclin-dependent kinase inhibitors (p27(Kip1) and p21(Waf1/CIP1)) and antiapoptotic proteins of the inhibitor of apoptosis proteins (IAP) and Bcl-2 families. Interruption of constitutive MEK/MAPK signaling therefore represents a promising therapeutic strategy in AML.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/enzimología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Supresoras de Tumor , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Citarabina/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Leucemia Mieloide Aguda/patología , Proteínas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal , Células Tumorales Cultivadas , Proteína Inhibidora de la Apoptosis Ligada a X
7.
Oncogene ; 12(2): 309-22, 1996 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-8570208

RESUMEN

The expression of E and D-type cyclins, Cyclin-Dependent Kinase (CDK) 2 and 4, as well as CDK inhibitors p21Cip1 and p27Kip1 were examined during in vitro differentiation of mouse embryonic stem (ES) cells. ES cells cultured in presence of Differentiation Inhibitory Activity/Leukemia Inhibitory Factor (DIA/LIF) express very low levels of cyclin E/CDK2 complexes, p21Cip1 and p27Kip1 CDK inhibitors, while cyclin D/CDK4-associated kinase activity is undetectable. Withdrawal of DIA/LIF, which induces differentiation, results in the progressive up-regulation of all. Up-regulation of D cyclins occurs through an increase in the steady-state levels of mRNA, concomitantly with the activation of Brachyury and Goosecoid, two early markers of mesoderm differentiation. Similarly, cells from the epiblast of the early postimplantation mouse embryo do not express any cyclin D/CDK4 complexes. These are progressively upregulated at gastrulation and early organogenesis. DIA/LIF-stimulated ES cells are not growth-arrested by overexpression of p16Ink4a, a specific inhibitor of CDK4 and CDK6. We propose that the G1/S transition may be regulated by a minimal mechanism in mouse embryonic stem cells. Induction of differentiation triggers the establishment of a more sophisticated mechanism involving both cyclin D/CDK4- and CDK inhibitor-associated control of G1-phase progression.


Asunto(s)
Proteínas de Ciclo Celular , Ciclinas/biosíntesis , Embrión de Mamíferos/metabolismo , Inhibidores Enzimáticos/metabolismo , Inhibidores de Crecimiento/fisiología , Interleucina-6 , Linfocinas/fisiología , Proteínas Asociadas a Microtúbulos/biosíntesis , Células Madre/metabolismo , Proteínas Supresoras de Tumor , Animales , Secuencia de Bases , Proteínas Portadoras/fisiología , Diferenciación Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Fase G1 , Factor Inhibidor de Leucemia , Ratones , Datos de Secuencia Molecular , Regulación hacia Arriba
8.
Oncogene ; 34(5): 631-8, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24469042

RESUMEN

Constitutional dominant loss-of-function mutations in the SPRED1 gene cause a rare phenotype referred as neurofibromatosis type 1 (NF1)-like syndrome or Legius syndrome, consisted of multiple café-au-lait macules, axillary freckling, learning disabilities and macrocephaly. SPRED1 is a negative regulator of the RAS MAPK pathway and can interact with neurofibromin, the NF1 gene product. Individuals with NF1 have a higher risk of haematological malignancies. SPRED1 is highly expressed in haematopoietic cells and negatively regulates haematopoiesis. SPRED1 seemed to be a good candidate for leukaemia predisposition or transformation. We performed SPRED1 mutation screening and expression status in 230 paediatric lymphoblastic and acute myeloblastic leukaemias (AMLs). We found a loss-of-function frameshift SPRED1 mutation in a patient with Legius syndrome. In this patient, the leukaemia blasts karyotype showed a SPRED1 loss of heterozygosity, confirming SPRED1 as a tumour suppressor. Our observation confirmed that acute leukaemias are rare complications of the Legius syndrome. Moreover, SPRED1 was significantly decreased at RNA and protein levels in the majority of AMLs at diagnosis compared with normal or paired complete remission bone marrows. SPRED1 decreased expression correlated with genetic features of AML. Our study reveals a new mechanism which contributes to deregulate RAS MAPK pathway in the vast majority of paediatric AMLs.


Asunto(s)
Manchas Café con Leche/genética , Genes ras/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia Mieloide Aguda/genética , Proteínas de la Membrana/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas Adaptadoras Transductoras de Señales , Adolescente , Manchas Café con Leche/complicaciones , Manchas Café con Leche/patología , Niño , Preescolar , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Leucemia Mieloide Aguda/complicaciones , Leucemia Mieloide Aguda/patología , Pérdida de Heterocigocidad/genética , Masculino , Proteínas de la Membrana/biosíntesis , Mutación , Neurofibromina 1/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología
10.
Leukemia ; 26(11): 2384-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22513837

RESUMEN

Myeloproliferative neoplasms are frequently associated with aberrant constitutive tyrosine kinase (TK) activity resulting from chimaeric fusion genes or point mutations such as BCR-ABL1 or JAK2 V617F. We report here the cloning and functional characterization of two novel fusion genes BCR-RET and FGFR1OP-RET in chronic myelomonocytic leukemia (CMML) cases generated by two balanced translocations t(10;22)(q11;q11) and t(6;10)(q27;q11), respectively. The two RET fusion genes leading to the aberrant activation of RET, are able to transform hematopoietic cells and skew the hematopoietic differentiation program towards the monocytic/macrophage lineage. The RET fusion genes seem to constitutively mimic the same signaling pathway as RAS mutations frequently involved in CMML. One patient was treated with Sorafenib, a specific inhibitor of the RET TK function, and demonstrated cytological and clinical remissions.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Leucemia Mielomonocítica Crónica/patología , Monocitos/citología , Proteínas Proto-Oncogénicas c-ret/genética , Secuencia de Bases , Cartilla de ADN , Humanos , Hibridación Fluorescente in Situ , Leucemia Mielomonocítica Crónica/genética , Mutación Puntual , Reacción en Cadena de la Polimerasa/métodos , Translocación Genética
15.
Immunogenetics ; 53(6): 468-76, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11685457

RESUMEN

Proteins with transmembrane domains are classified in different families based on their structure, amino acid homology, and function. In this study, we report the identification, sequence, and expression profile of a new member of the CD20/FcepsilonRIbeta family, CD20/FcepsilonRIbeta family member 4 (CFFM4). The CFFM4 gene contains seven exons and six introns and is transcribed into an mRNA encoding a 240-amino acid protein with four hydrophobic regions. The CFFM4 protein shares a high degree of homology with the other members of the family, especially in the hydrophobic regions where several amino acids are conserved. However, the CFFM4 protein can be distinguished from the other members of the family based on the length of the second extracellular loop and the absence of an immunoreceptor tyrosine-based activation motif signal. Another distinct characteristic is that CFFM4 mRNA expression is not limited to the hematopoietic lineage. CFFM4 was detected by Northern dot blot in a variety of normal and cancerous tissues. CFFM4 expression was also compared in developmentally early hematopoietic human bone marrow CD34+ stem cells versus peripheral blood-derived CD14+ mature monocytes, in the undifferentiated versus differentiated myelomonocytic U937 cell line, and in acute myelogenous leukemia FAB1 versus FAB5. In each of these systems, cellular myelomonocytic differentiation correlated with an increase in CFFM4 mRNA expression. Such results indicate that CFFM4 is associated with mature cellular function in the monocytic lineage and like CD20 and FcepsilonRIbeta, it may be a component of a receptor complex involved in signal transduction.


Asunto(s)
Antígenos CD20/genética , Antígenos CD/biosíntesis , Antígenos CD/genética , Receptores de IgE/biosíntesis , Receptores de IgE/genética , Enfermedad Aguda , Secuencia de Aminoácidos , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Clonación Molecular , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Proteínas de la Membrana , Datos de Secuencia Molecular , Monocitos/metabolismo , ARN Mensajero/biosíntesis , Homología de Secuencia de Aminoácido , Distribución Tisular , Células Tumorales Cultivadas , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA