Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Blood ; 132(13): 1386-1398, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30093402

RESUMEN

Anaplastic large cell lymphomas (ALCLs) are CD30-positive T-cell non-Hodgkin lymphomas broadly segregated into ALK-positive and ALK-negative types. Although ALK-positive ALCLs consistently bear rearrangements of the ALK tyrosine kinase gene, ALK-negative ALCLs are clinically and genetically heterogeneous. About 30% of ALK-negative ALCLs have rearrangements of DUSP22 and have excellent long-term outcomes with standard therapy. To better understand this group of tumors, we evaluated their molecular signature using gene expression profiling. DUSP22-rearranged ALCLs belonged to a distinct subset of ALCLs that lacked expression of genes associated with JAK-STAT3 signaling, a pathway contributing to growth in the majority of ALCLs. Reverse-phase protein array and immunohistochemical studies confirmed the lack of activated STAT3 in DUSP22-rearranged ALCLs. DUSP22-rearranged ALCLs also overexpressed immunogenic cancer-testis antigen (CTA) genes and showed marked DNA hypomethylation by reduced representation bisulfate sequencing and DNA methylation arrays. Pharmacologic DNA demethylation in ALCL cells recapitulated the overexpression of CTAs and other DUSP22 signature genes. In addition, DUSP22-rearranged ALCLs minimally expressed PD-L1 compared with other ALCLs, but showed high expression of the costimulatory gene CD58 and HLA class II. Taken together, these findings indicate that DUSP22 rearrangements define a molecularly distinct subgroup of ALCLs, and that immunogenic cues related to antigenicity, costimulatory molecule expression, and inactivity of the PD-1/PD-L1 immune checkpoint likely contribute to their favorable prognosis. More aggressive ALCLs might be pharmacologically reprogrammed to a DUSP22-like immunogenic molecular signature through the use of demethylating agents and/or immune checkpoint inhibitors.


Asunto(s)
Metilación de ADN , Fosfatasas de Especificidad Dual/genética , Regulación Neoplásica de la Expresión Génica , Reordenamiento Génico , Linfoma Anaplásico de Células Grandes/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Antígenos de Neoplasias/genética , Fosfatasas de Especificidad Dual/inmunología , Femenino , Humanos , Linfoma Anaplásico de Células Grandes/diagnóstico , Linfoma Anaplásico de Células Grandes/inmunología , Linfoma Anaplásico de Células Grandes/patología , Masculino , Persona de Mediana Edad , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/inmunología , Fosforilación , Pronóstico , Factor de Transcripción STAT3/análisis , Transcriptoma , Escape del Tumor
2.
Blood ; 128(9): 1234-45, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27297792

RESUMEN

Peripheral T-cell lymphomas (PTCLs) represent a heterogeneous group of T-cell malignancies that generally demonstrate aggressive clinical behavior, often are refractory to standard therapy, and remain significantly understudied. The most common World Health Organization subtype is PTCL, not otherwise specified (NOS), essentially a "wastebasket" category because of inadequate understanding to assign cases to a more specific diagnostic entity. Identification of novel fusion genes has contributed significantly to improving the classification, biologic understanding, and therapeutic targeting of PTCLs. Here, we integrated mate-pair DNA and RNA next-generation sequencing to identify chromosomal rearrangements encoding expressed fusion transcripts in PTCL, NOS. Two of 11 cases had novel fusions involving VAV1, encoding a truncated form of the VAV1 guanine nucleotide exchange factor important in T-cell receptor signaling. Fluorescence in situ hybridization studies identified VAV1 rearrangements in 10 of 148 PTCLs (7%). These were observed exclusively in PTCL, NOS (11%) and anaplastic large cell lymphoma (11%). In vitro, ectopic expression of a VAV1 fusion promoted cell growth and migration in a RAC1-dependent manner. This growth was inhibited by azathioprine, a clinically available RAC1 inhibitor. We also identified novel kinase gene fusions, ITK-FER and IKZF2-ERBB4, as candidate therapeutic targets that show similarities to known recurrent oncogenic ITK-SYK fusions and ERBB4 transcript variants in PTCLs, respectively. Additional novel and potentially clinically relevant fusions also were discovered. Together, these findings identify VAV1 fusions as recurrent and targetable events in PTCLs and highlight the potential for clinical sequencing to guide individualized therapy approaches for this group of aggressive malignancies.


Asunto(s)
Linfoma de Células T Periférico/genética , Proteínas de Fusión Oncogénica/genética , Anciano , Animales , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células Jurkat , Linfoma de Células T Periférico/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Proteínas de Fusión Oncogénica/metabolismo
3.
Blood ; 125(20): 3118-27, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25833963

RESUMEN

Peripheral T-cell lymphomas (PTCLs) are generally aggressive non-Hodgkin lymphomas with poor overall survival rates following standard therapy. One-third of PTCLs express interferon regulatory factor-4 (IRF4), a tightly regulated transcription factor involved in lymphocyte growth and differentiation. IRF4 drives tumor growth in several lymphoid malignancies and has been proposed as a candidate therapeutic target. Because direct IRF4 inhibitors are not clinically available, we sought to characterize the mechanism by which IRF4 expression is regulated in PTCLs. We demonstrated that IRF4 is constitutively expressed in PTCL cells and drives Myc expression and proliferation. Using an inhibitor screen, we identified nuclear factor κB (NF-κB) as a candidate regulator of IRF4 expression and cell proliferation. We then demonstrated that the NF-κB subunits p52 and RelB were transcriptional activators of IRF4. Further analysis showed that activation of CD30 promotes p52 and RelB activity and subsequent IRF4 expression. Finally, we showed that IRF4 transcriptionally regulates CD30 expression. Taken together, these data demonstrate a novel positive feedback loop involving CD30, NF-κB, and IRF4; further evidence for this mechanism was demonstrated in human PTCL tissue samples. Accordingly, NF-κB inhibitors may represent a clinical means to disrupt this feedback loop in IRF4-positive PTCLs.


Asunto(s)
Factores Reguladores del Interferón/genética , Antígeno Ki-1/metabolismo , Linfoma de Células T Periférico/genética , Linfoma de Células T Periférico/metabolismo , FN-kappa B/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Proliferación Celular , Variaciones en el Número de Copia de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Genes myc , Células Germinativas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Polimorfismo Genético , Transcripción Genética
4.
Appl Immunohistochem Mol Morphol ; 23(8): 580-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25390351

RESUMEN

Anaplastic large cell lymphoma (ALCL) is one of the most common T-cell non-Hodgkin lymphomas and has 2 main subtypes: an anaplastic lymphoma kinase (ALK)-positive subtype characterized by ALK gene rearrangements and an ALK-negative subtype that is poorly understood. We recently identified recurrent rearrangements of the DUSP22 locus on 6p25.3 in both primary cutaneous and systemic ALK-negative ALCLs. This study aimed to determine the relationship between these rearrangements and expression of the chemokine receptor gene, CCR8. CCR8 has skin-homing properties and has been suggested to play a role in limiting extracutaneous spread of primary cutaneous ALCLs. However, overexpression of CCR8 has also been reported in systemic ALK-negative ALCLs. As available antibodies for CCR8 have shown lack of specificity, we examined CCR8 expression using quantitative real-time PCR in frozen tissue and RNA in situ hybridization (ISH) in paraffin tissue. Both approaches showed higher CCR8 expression in ALCLs with DUSP22 rearrangements than in nonrearranged cases (PCR: 19.5-fold increase, P=0.01; ISH: 3.3-fold increase, P=0.0008). CCR8 expression was not associated with cutaneous presentation, cutaneous biopsy site, or cutaneous involvement during the disease course. These findings suggest that CCR8 expression in ALCL is more closely related to the presence of DUSP22 rearrangements than to cutaneous involvement and that the function of CCR8 may extend beyond its skin-homing properties in this disease. This study also underscores the utility of RNA-ISH as a paraffin-based method for investigating gene expression when reliable antibodies for immunohistochemical analysis are not available.


Asunto(s)
Fosfatasas de Especificidad Dual/genética , Linfoma Anaplásico de Células Grandes/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Receptores CCR8/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Adulto Joven
5.
Genes Chromosomes Cancer ; 52(11): 1097-102, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23999969

RESUMEN

Chromosomal translocations leading to expression of abnormal fusion proteins play a major role in the pathogenesis of various hematologic malignancies. The recent development of high-throughput, "deep" sequencing has allowed discovery of novel translocations leading to a rapid increase in understanding these diseases. Translocations involving the anaplastic lymphoma kinase (ALK) gene leading to ALK fusion proteins originally were discovered in anaplastic large cell lymphomas (ALCLs). Among ALCLs, NPM1-ALK fusions are most common and lead to nuclear localization of the fusion protein. Here, we present a 50-year-old male with ALCL demonstrating cytoplasmic ALK immunoreactivity only, suggesting the presence of a non-NPM1 fusion partner. We performed deep RNA sequencing of tumor tissue from this patient and identified a novel transcript fusing Exon 6 of TRAF1 to Exon 20 of ALK. The TRAF1-ALK fusion transcript was confirmed at the mRNA level by Sanger sequencing and the fusion protein was visualized by Western blot. The discovery of this TRAF1-ALK fusion expands the diversity of known ALK fusion partners and highlights the power of deep sequencing for fusion transcript discovery. © 2013 Wiley Periodicals, Inc.


Asunto(s)
Linfoma Anaplásico de Células Grandes/genética , Proteínas Tirosina Quinasas Receptoras/genética , Análisis de Secuencia de ARN , Factor 1 Asociado a Receptor de TNF/genética , Quinasa de Linfoma Anaplásico , Fusión Génica , Humanos , Masculino , Persona de Mediana Edad , Nucleofosmina , Translocación Genética
6.
Am J Surg Pathol ; 37(7): 978-86, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23759932

RESUMEN

The traditional model of hematopoiesis is based on unidirectional maturation of hematopoietic precursors into lineage-committed cells. However, recent studies indicate that mature B lymphocytes may demonstrate significant lineage plasticity. We and others have reported transdifferentiation of follicular lymphomas (FLs) into clonally related histiocytic/dendritic cell neoplasms. Here, we describe 2 patients with FL who developed clonally related Langerhans cell neoplasms. The first was a 52-year-old man diagnosed with FL, grade 1. He received immunochemotherapy and had stable disease for 8 years. He then developed increasing lymphadenopathy, and lymph node biopsy showed Langerhans cell sarcoma with no evidence of FL. The second patient was a 77-year-old woman who presented with lymphadenopathy, an abdominal mass, and pulmonary nodules. Lymph node biopsy showed both Langerhans cell histiocytosis and minimal involvement by FL, grade 1. In each case, a combination of immunoglobulin gene rearrangement and fluorescence in situ hybridization studies provided evidence to support a clonal relationship between the FL and Langerhans cell neoplasm. These cases provide striking examples of neoplastic transdifferentiation and expand the spectrum of lesions clonally identical to otherwise typical FL. Awareness of this phenomenon may aid in diagnosis when histologically dissimilar tumors arise synchronously or metachronously in patients with lymphoma.


Asunto(s)
Transdiferenciación Celular , Sarcoma de Células de Langerhans/patología , Células de Langerhans/patología , Linfoma Folicular/patología , Neoplasias Primarias Secundarias/patología , Anciano , Células Clonales , Femenino , Citometría de Flujo , Reordenamiento Génico , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Inmunofenotipificación , Hibridación Fluorescente in Situ , Sarcoma de Células de Langerhans/genética , Sarcoma de Células de Langerhans/metabolismo , Células de Langerhans/metabolismo , Linfoma Folicular/genética , Linfoma Folicular/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo
7.
Blood ; 120(11): 2280-9, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22855598

RESUMEN

Peripheral T-cell lymphomas (PTCLs) are aggressive malignancies of mature T lymphocytes with 5-year overall survival rates of only ∼ 35%. Improvement in outcomes has been stymied by poor understanding of the genetics and molecular pathogenesis of PTCL, with a resulting paucity of molecular targets for therapy. We developed bioinformatic tools to identify chromosomal rearrangements using genome-wide, next-generation sequencing analysis of mate-pair DNA libraries and applied these tools to 16 PTCL patient tissue samples and 6 PTCL cell lines. Thirteen recurrent abnormalities were identified, of which 5 involved p53-related genes (TP53, TP63, CDKN2A, WWOX, and ANKRD11). Among these abnormalities were novel TP63 rearrangements encoding fusion proteins homologous to ΔNp63, a dominant-negative p63 isoform that inhibits the p53 pathway. TP63 rearrangements were seen in 11 (5.8%) of 190 PTCLs and were associated with inferior overall survival; they also were detected in 2 (1.2%) of 164 diffuse large B-cell lymphomas. As TP53 mutations are rare in PTCL compared with other malignancies, our findings suggest that a constellation of alternate genetic abnormalities may contribute to disruption of p53-associated tumor suppressor function in PTCL.


Asunto(s)
Reordenamiento Génico , Linfoma de Células T Periférico/genética , Mutación , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/química , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Análisis Mutacional de ADN , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/mortalidad , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células T Periférico/metabolismo , Linfoma de Células T Periférico/mortalidad , Linfoma de Células T Periférico/patología , Masculino , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Oxidorreductasas/química , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Estados Unidos , Oxidorreductasa que Contiene Dominios WW
8.
Blood ; 117(3): 915-9, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21030553

RESUMEN

The genetics of peripheral T-cell lymphomas are poorly understood. The most well-characterized abnormalities are translocations involving ALK, occurring in approximately half of anaplastic large cell lymphomas (ALCLs). To gain insight into the genetics of ALCLs lacking ALK translocations, we combined mate-pair DNA library construction, massively parallel ("Next Generation") sequencing, and a novel bioinformatic algorithm. We identified a balanced translocation disrupting the DUSP22 phosphatase gene on 6p25.3 and adjoining the FRA7H fragile site on 7q32.3 in a systemic ALK-negative ALCL. Using fluorescence in situ hybridization, we demonstrated that the t(6;7)(p25.3;q32.3) was recurrent in ALK-negative ALCLs. Furthermore, t(6;7)(p25.3;q32.3) was associated with down-regulation of DUSP22 and up-regulation of MIR29 microRNAs on 7q32.3. These findings represent the first recurrent translocation reported in ALK-negative ALCL and highlight the utility of massively parallel genomic sequencing to discover novel translocations in lymphoma and other cancers.


Asunto(s)
Cromosomas Humanos Par 6/genética , Cromosomas Humanos Par 7/genética , Linfoma Anaplásico de Células Grandes/genética , Translocación Genética , Adulto , Anciano , Anciano de 80 o más Años , Quinasa de Linfoma Anaplásico , Secuencia de Bases , Rotura Cromosómica , Puntos de Rotura del Cromosoma , Fosfatasas de Especificidad Dual/genética , Femenino , Humanos , Hibridación Fluorescente in Situ , Linfoma Anaplásico de Células Grandes/patología , Masculino , MicroARNs/genética , Persona de Mediana Edad , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Datos de Secuencia Molecular , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas Receptoras , Análisis de Secuencia de ADN/métodos
9.
Cancer Res ; 69(19): 7518-23, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19773435

RESUMEN

IER3 (formerly IEX-1) encodes a 27-kDa glycoprotein that regulates death receptor-induced apoptosis, interacts with NF-kappaB pathways, and increases expression rapidly in response to cellular stresses such as irradiation. Animal models, gene expression microarray experiments, and functional studies in cell lines have suggested a potential role for IER3 in oncogenesis, but, to date, no abnormalities of IER3 at the DNA level have been reported in patients with neoplasia. Here, we describe breakpoint cloning of a t(6;9)(p21;q34) translocation from a patient with a myelodysplastic syndrome (MDS), facilitated by conversion technology and array-based comparative genomic hybridization, which revealed a rearrangement translocating the IER3 coding region away from critical flanking/regulatory elements and to a transcript-poor chromosomal region, markedly decreasing expression. Using split-signal and locus-specific fluorescence in situ hybridization (FISH) probes, we analyzed 204 patients with diverse hematological malignancies accompanied by clonal chromosome 6p21 abnormalities, and found 8 additional patients with MDS with IER3 rearrangements (translocations or amplification). Although FISH studies on 157 additional samples from patients with MDS and a normal-karyotype were unrevealing, and sequencing the IER3 coding and proximal promoter regions of 74 MDS patients disclosed no point mutations, reverse transcription-PCR results suggested that dysregulated expression of IER3 is common in MDS (61% >4-fold increase or decrease in expression with decreased expression primarily in early MDS and increased expression primarily in later MDS progressing toward leukemia), consistent with findings in previous microarray experiments. These data support involvement of IER3 in the pathobiology of MDS.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas de la Membrana/genética , Síndromes Mielodisplásicos/genética , Anciano , Animales , Secuencia de Bases , Cromosomas Humanos Par 6 , Cromosomas Humanos Par 9 , Hibridación Genómica Comparativa , Amplificación de Genes , Reordenamiento Génico , Células Madre Hematopoyéticas/citología , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Datos de Secuencia Molecular , Mutación Puntual , Polimorfismo Genético , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Leuk Lymphoma ; 49(5): 989-93, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18464119

RESUMEN

Alternative pre-mRNA splicing alters gene expression and protein function, and aberrant splicing patterns can be associated with neoplasia. The potential role of disordered RNA splicing in myelodysplastic syndrome (MDS) is unexplored. We analysed the splicing repertoire of CDC25C- a gene localised to chromosome 5q31 and encoding a cyclin/cyclin-dependent-kinase regulatory phosphatase critical for cell cycle checkpoint control - in MDS, acute myeloid leukemia, chronic lymphocytic leukemia and healthy tissues. Five novel splicing isoforms were detected, and the splicing patterns were generally distinct in neoplastic samples compared with healthy controls. One of the novel isoforms, which we have termed CDC25C-6, occurred in 58% of the samples in our cohort. The results of this study suggest the possibility of aberrant splicing contributing to the phenotype in MDS and other haematologic malignancies.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Síndromes Mielodisplásicos/genética , Empalme del ARN , Fosfatasas cdc25/genética , Estudios de Casos y Controles , Ciclo Celular , Cromosomas Humanos Par 5 , Humanos , Isoformas de Proteínas , Precursores del ARN
14.
Br J Haematol ; 139(3): 439-42, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17910635

RESUMEN

Patients with clonal myeloid disorders, especially myelodysplastic syndromes (MDS), may acquire alpha-thalassaemia. To estimate the prevalence of this erythrocyte phenotype, we examined brilliant cresyl blue-stained blood smears from 201 patients with neoplastic myeloid disorders and 282 controls (195 non-clonal anaemia, 62 with medical illnesses without anaemia and 25 healthy persons). Haemoglobin H inclusions were detected in 8/100 patients with MDS (8%) and 2/81 (2.5%) patients with myeloproliferative disorders, but in none of the acute leukaemia patients or controls. We conclude that the emergence of thalassaemic clones may be relatively common in the disordered marrow milieu of MDS.


Asunto(s)
Síndromes Mielodisplásicos/complicaciones , Trastornos Mieloproliferativos/complicaciones , Talasemia alfa/etiología , Enfermedad Aguda , Anciano , Índices de Eritrocitos , Eritrocitos/química , Femenino , Hemoglobina H/análisis , Humanos , Leucemia Mieloide/sangre , Leucemia Mieloide/complicaciones , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/sangre , Trastornos Mieloproliferativos/sangre , Estudios Prospectivos
15.
Leuk Res ; 31(5): 623-8, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-16870250

RESUMEN

BACKGROUND: For most cases of idiopathic acquired sideroblastic anemia (IASA), the molecular pathogenesis is unknown, despite the consistent morphological signature of abundant pathological ringed sideroblasts with their characteristic iron-engorged mitochondria. Moderately elevated free erythrocyte protoporphyrin (FEP) levels have been described in IASA, suggesting that the activity of ferrochelatase, the enzyme that catalyzes the final step in heme biosynthesis (incorporation of ferrous iron into protoporphyrin), might be diminished in erythroid progenitor cells from IASA patients. METHODS: We confirmed FEP elevation in IASA, then pursued a candidate gene approach that included screening the gene encoding ferrochelatase, FECH, for promoter and coding region mutations and mRNA expression changes in bone marrow from 37 patients with IASA. RESULTS: The analytical techniques employed detected mutations in a test cohort of previously undiagnosed patients with biochemical evidence for erythropoietic protoporphyria, a condition resulting from germline mutations in FECH, but somatic missense mutations of FECH and its promoter were not observed in IASA patients. FECH was modestly overexpressed in progenitor cells from patients with IASA, compared with MDS patients without sideroblasts and healthy controls. In addition, we analyzed ABCB7 and PUS1, genes implicated in congenital sideroblastic anemia syndromes, but again found no coding mutations in acquired cases. CONCLUSION: We conclude that acquired mutations in the factors currently known to cause inherited sideroblastic anemias are uncommon in IASA.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Anemia Refractaria/genética , Anemia Sideroblástica/genética , Eritroblastos/patología , Ferroquelatasa/genética , Hidroliasas/genética , Mutación Puntual/genética , Protoporfirinas/genética , Médula Ósea/metabolismo , Médula Ósea/patología , Células Cultivadas , Eritrocitos/metabolismo , Eritrocitos/patología , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...