Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Clin Lab ; 69(5)2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37145071

RESUMEN

BACKGROUND: The goal was to study the role of the morphology, immunophenotype, karyotype and fusion gene expression in a patient with diagnosis of AML1-ETO positive acute myeloid leukemia. METHODS: A case of AML1-ETO positive acute myeloid leukemia morphologically similar to chronic myelogenous leukemia was reported. The results of the morphology, immunophenotype, karyotype and fusion gene expression were analyzed by reviewing relevant literature. RESULTS: The patient was a young boy, at the age of 13, with clinical manifestations of intermittent fatigue and fever. Blood routine: White blood cell 142.6 x 109/L, Red blood cell 0.89 x 1012/L, Hemoglobin 41 g/L, Platelet 23 x 109/L, primitive cells account for 5%. Bone marrow smear: Granulocyte system hyperplasia is obvious, visible at each stage, primitive cells account for 17%, eosinophils, basophils, and phagocytic blood cells were observed. Flow cytometry showed myeloid primitive cell population was 4.14%, immature and mature granulocytes cell population was 85.22%, and eosinophil cell population was 0.61%. The results showed that the proportion of myeloid primitive cell was high, the expression of CD34 was enhanced, the expression of CD117 was partially absent, the expression of CD38 was weakened, the expression of CD19 was weak, and a few cells expressed CD56, and the phenotype was abnormal. The proportion of granulocyte series increased and the nucleus shifted to the left. The proportion of erythroid series was decreased, and the expression of CD71 was weakened. The results of fusion gene showed AML1-ETO positive. Karyotype analysis showed clonogenic abnormality t(8;21)(q22;q22). CONCLUSIONS: The peripheral blood and bone marrow images of patients with t(8;21)(q22;q22) AML1-ETO positive are the manifestations of chronic myelogenous leukemia, suggesting that cytogenetics and molecular genetics play an irreplaceable role in the diagnosis of acute myeloid leukemia, and the comprehensive diagnostic efficiency is significantly better than that of morphology.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Leucemia Mieloide Aguda , Humanos , Proteína 1 Compañera de Translocación de RUNX1/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Médula Ósea/metabolismo , Enfermedad Crónica , Proteínas de Fusión Oncogénica/genética , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo
2.
Clin Lab ; 69(2)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36787564

RESUMEN

BACKGROUND: The aim was to improve the understanding of an AML1/ETO positive child with acute myeloid leukemia with poor prognosis. METHODS: A case of AML1/ETO positive child with acute myeloid leukemia with poor prognosis was reported. The bone marrow cell morphology, multi-parameter flow cytometry, cytogenetic or molecular genetic test results were analyzed by reviewing relevant literature. RESULTS: The patient was a young girl with clinical manifestations of respiratory tract infection. Bone marrow smears showed that myeloid primordial cells accounted for 13%, some granulocyte cell bodies are enlarged, visible pathological phenomena such as cytoplasmic vacuoles, binuclear grains, ring rods, and pseudo pelgerhuet malformations were seen (Figure 1). Flow cytometry: abnormal myeloid original cells (12.33%), expression of CD34 and HLA - DR, CD38, CD56, part of the expression of CD117, weak expression of CD13, CD33, MPO, CD19, cCD79a (Figure 2). Chromosome karyotype analysis showed that the chromosome karyotype of peripheral blood was 46, XX, t(8;21)(q22;q22). The quantitative detection result of AML1/ETO fusion gene was 42.15%, and mutations of NRAS, ASXL2, TP53 and TET2 genes were detected by second-generation sequencing. Combined with the above results, AML1/ETO positive with acute myeloid leukemia was diagnosed. CONCLUSIONS: Cytogenetics or molecular genetics is the gold standard for identification of positive AML1/ETO fusion gene. Morphological heterogeneity of AML1/ETO positive AML cells is large, which limits the morphological diagnosis of bone marrow cells to a certain extent, and the comprehensive diagnostic efficiency is significantly better than that of morphology. Leukemia fusion gene AML1/ETO refers to the fusion of AML1 gene located on human chromosome 21q22 and ETO gene 8q22, which is the most common fusion gene in acute myeloid leukemia (AML). This paper reports a case of an AML1/ETO positive child with acute myeloid leukemia with poor prognosis admitted to our hospital and reviews relevant literature.


Asunto(s)
Leucemia Mieloide Aguda , Femenino , Humanos , Niño , Proteína 1 Compañera de Translocación de RUNX1/genética , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Antígenos HLA-DR , Mutación , Proteínas de Fusión Oncogénica/genética , Pronóstico , Cromosomas Humanos Par 8/metabolismo
3.
Sci Rep ; 12(1): 14158, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35986043

RESUMEN

Acute myeloid leukemia (AML) is a malignant disease of immature myeloid cells and the most prevalent acute leukemia among adults. The oncogenic homo-tetrameric fusion protein RUNX1/ETO results from the chromosomal translocation t(8;21) and is found in AML patients. The nervy homology region 2 (NHR2) domain of ETO mediates tetramerization; this oligomerization is essential for oncogenic activity. Previously, we identified the first-in-class small-molecule inhibitor of NHR2 tetramer formation, 7.44, which was shown to specifically interfere with NHR2, restore gene expression down-regulated by RUNX1/ETO, inhibit the proliferation of RUNX1/ETO-depending SKNO-1 cells, and reduce the RUNX1/ETO-related tumor growth in a mouse model. However, no biophysical and structural characterization of 7.44 binding to the NHR2 domain has been reported. Likewise, the compound has not been characterized as to physicochemical, pharmacokinetic, and toxicological properties. Here, we characterize the interaction between the NHR2 domain of RUNX1/ETO and 7.44 by biophysical assays and show that 7.44 interferes with NHR2 tetramer stability and leads to an increase in the dimer population of NHR2. The affinity of 7.44 with respect to binding to NHR2 is Klig = 3.75 ± 1.22 µM. By NMR spectroscopy combined with molecular dynamics simulations, we show that 7.44 binds with both heteroaromatic moieties to NHR2 and interacts with or leads to conformational changes in the N-termini of the NHR2 tetramer. Finally, we demonstrate that 7.44 has favorable physicochemical, pharmacokinetic, and toxicological properties. Together with biochemical, cellular, and in vivo assessments, the results reveal 7.44 as a lead for further optimization towards targeted therapy of t(8;21) AML.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Leucemia Mieloide Aguda , Animales , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteínas de Fusión Oncogénica/metabolismo , Translocación Genética
4.
Br J Cancer ; 126(6): 917-926, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34893760

RESUMEN

BACKGROUND: Colorectal cancer (CRC) develops through chromosomal instability (CIN) or microsatellite instability (MSI) due to deficient mismatch-repair (dMMR). We aimed to characterise novel cancer-associated genes that are downregulated upon malignant transformation in microsatellite stable (MSS) CRCs, which typically exhibit CIN with proficient mismatch-repair (pMMR). METHODS: Comprehensive screening was conducted on adenomas, MSI/MSS CRCs and cell lines, followed by copy number analysis, and their genetic and prognostic relevance was confirmed in microarray and RNA-seq cohorts (n = 3262, in total). Immunohistochemistry for SH2D4A was performed in 524 specimens of adenoma, carcinoma in situ and dMMR/pMMR CRC. The functional role of SH2D4A was investigated using CRC cell lines. RESULTS: A set of 11 genes, including SH2D4A, was downregulated during the adenoma-carcinoma sequence in MSS/CIN CRCs, mainly due to chromosome 8p deletions, and their negative prognostic impact was validated in independent cohorts. All adenomas were SH2D4A positive, but a subset of CRCs (5.3%) lacked SH2D4A immunohistochemical staining, correlating with poor prognosis and scarce T cell infiltration. SH2D4A depletion did not affect cell proliferation or IL-6-induced STAT3 phosphorylation. CONCLUSIONS: Our findings suggest that downregulation of multiple genes on chromosome 8p, including SH2D4A, cooperatively contribute to tumorigenesis, resulting in the immune cold tumour microenvironment and poor prognosis.


Asunto(s)
Neoplasias Colorrectales , Linfocitos Infiltrantes de Tumor , Monosomía , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/inmunología , Cromosomas Humanos Par 8/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Reparación de la Incompatibilidad de ADN , Regulación hacia Abajo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Linfocitos Infiltrantes de Tumor/inmunología , Inestabilidad de Microsatélites , Monosomía/genética , Monosomía/inmunología , Pronóstico , Linfocitos T , Microambiente Tumoral
5.
Int J Surg Pathol ; 30(4): 462-465, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34955058

RESUMEN

MYC over-expression by immunohistochemistry (IHC) is utilised in routine pathology practice as a surrogate marker for MYC amplification, which plays a key oncogenic role in post-irradiation and chronic lymphedema-associated angiosarcoma. We present the case of a 32-year old male, who presented with high-grade angiosarcoma arising in a background of metastatic testicular teratoma. IHC for MYC showed strong nuclear expression in the angiosarcoma cells prompting the consideration of post-irradiation-induced angiosarcoma but our patient did not undergo radiotherapy. Fluorescence in-situ hybridization (FISH) excluded MYC amplification and instead showed Chromosome 8 polysomy, which accounted for the strong MYC IHC expression present, not previously described in the context of germ cell tumours. The occurrence of MYC over-expression due to polysomy illustrates a novel clinical scenario (angiosarcoma arising as somatic malignancy) where strong MYC IHC expression can be found in the absence of underlying amplification or prior radiotherapy exposure.


Asunto(s)
Hemangiosarcoma , Neoplasias Primarias Secundarias , Teratoma , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Amplificación de Genes , Hemangiosarcoma/genética , Hemangiosarcoma/patología , Humanos , Masculino , Neoplasias Primarias Secundarias/genética , Proteínas Proto-Oncogénicas c-myc/genética , Teratoma/genética
6.
PLoS One ; 16(3): e0248342, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33711044

RESUMEN

Technological advances in molecular profiling have enabled the comprehensive identification of common regions of gene amplification on chromosomes (amplicons) in muscle invasive bladder cancer (MIBC). One such region is 8q22.2, which is largely unexplored in MIBC and could harbor genes with potential for outcome prediction or targeted therapy. To investigate the prognostic role of 8q22.2 and to compare different amplicon definitions, an in-silico analysis of 357 patients from The Cancer Genome Atlas, who underwent radical cystectomy for MIBC, was performed. Amplicons were generated using the GISTIC2.0 algorithm for copy number alterations (DNA_Amplicon) and z-score normalization for mRNA gene overexpression (RNA_Amplicon). Kaplan-Meier survival analysis, univariable, and multivariable Cox proportional hazard ratios were used to relate amplicons, genes, and clinical parameters to overall (OS) and disease-free survival (DFS). Analyses of the biological functions of 8q22.2 genes and genomic events in MIBC were performed to identify potential targets. Genes with prognostic significance from the in silico analysis were validated using RT-qPCR of MIBC tumor samples (n = 46). High 8q22.2 mRNA expression (RNA-AMP) was associated with lymph node metastases. Furthermore, 8q22.2 DNA and RNA amplified patients were more likely to show a luminal subtype (DNA_Amplicon_core: p = 0.029; RNA_Amplicon_core: p = 0.01). Overexpression of the 8q22.2 gene OSR2 predicted shortened DFS in univariable (HR [CI] 1.97 [1.2; 3.22]; p = 0.01) and multivariable in silico analysis (HR [CI] 1.91 [1.15; 3.16]; p = 0.01) and decreased OS (HR [CI] 6.25 [1.37; 28.38]; p = 0.0177) in RT-qPCR data analysis. Alterations in different levels of the 8q22.2 region are associated with manifestation of different clinical characteristics in MIBC. An in-depth comprehensive molecular characterization of genomic regions involved in cancer should include multiple genetic levels, such as DNA copy number alterations and mRNA gene expression, and could lead to a better molecular understanding. In this study, OSR2 is identified as a potential biomarker for survival prognosis.


Asunto(s)
Cromosomas Humanos Par 8 , Regulación Neoplásica de la Expresión Génica , Neoplasias de los Músculos , Proteínas de Neoplasias , ARN Mensajero , ARN Neoplásico , Factores de Transcripción , Neoplasias de la Vejiga Urinaria , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de los Músculos/genética , Neoplasias de los Músculos/metabolismo , Neoplasias de los Músculos/mortalidad , Invasividad Neoplásica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Pronóstico , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Estudios Retrospectivos , Tasa de Supervivencia , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/mortalidad
9.
Genes (Basel) ; 11(12)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33316910

RESUMEN

Ring chromosome 8 (r(8)) is one of the least frequent ring chromosomes. Usually, maternal chromosome 8 forms a ring, which can be lost from cells due to mitotic instability. The 8q24 region contains the imprinted KCNK9 gene, which is expressed from the maternal allele. Heterozygous KCNK9 mutations are associated with the imprinting disorder Birk-Barel syndrome. Here, we report a 2.5-year-old boy with developmental delay, microcephaly, dysmorphic features, diffuse muscle hypotonia, feeding problems, motor alalia and noncoarse neurogenic type of disturbance of muscle electrogenesis, partially overlapping with Birk-Barel syndrome phenotype. Cytogenetic analysis of lymphocytes revealed his karyotype to be 46,XY,r(8)(p23q24.3)[27]/45,XY,-8[3]. A de novo 7.9 Mb terminal 8p23.3p23.1 deletion, a 27.1 Mb 8p23.1p11.22 duplication, and a 4.4 Mb intact segment with a normal copy number located between them, as well as a 154-kb maternal LINGO2 gene deletion (9p21.2) with unknown clinical significance were identified by aCGH + SNP array. These aberrations were confirmed by real-time PCR. According to FISH analysis, the 8p23.1-p11.22 duplication was inverted. The ring chromosome originated from maternal chromosome 8. Targeted massive parallel sequencing did not reveal the KCNK9 mutations associated with Birk-Barel syndrome. Our data allow to assume that autosomal monosomy with inactive allele of imprinted gene arising from the loss of a ring chromosome in some somatic cells may be an etiological mechanism of mosaic imprinting disorders, presumably with less severe phenotype.


Asunto(s)
Anomalías Craneofaciales/genética , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Anomalías Craneofaciales/metabolismo , Impresión Genómica/genética , Humanos , Discapacidad Intelectual/metabolismo , Cariotipo , Cariotipificación/métodos , Masculino , Proteínas de la Membrana/genética , Mosaicismo , Hipotonía Muscular/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/genética , Fenotipo , Canales de Potasio de Dominio Poro en Tándem/genética , Cromosomas en Anillo
10.
Exp Hematol ; 92: 62-74, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33152396

RESUMEN

Acute myeloid leukemia development occurs in a stepwise fashion whereby an original driver mutation is followed by additional mutations. The first type of mutations tends to be in genes encoding members of the epigenetic/transcription regulatory machinery (i.e., RUNX1, DNMT3A, TET2), while the secondary mutations often involve genes encoding members of signaling pathways that cause uncontrolled growth of such cells such as the growth factor receptors c-KIT of FLT3. Patients usually present with both types of mutations, but it is currently unclear how both mutational events shape the epigenome in developing AML cells. To this end we generated an in vitro model of t(8;21) AML by expressing its driver oncoprotein RUNX1-ETO with or without a mutated (N822K) KIT protein. Expression of N822K-c-KIT strongly increases the self-renewal capacity of RUNX1-ETO-expressing cells. Global analysis of gene expression changes and alterations in the epigenome revealed that N822K-c-KIT expression profoundly influences the open chromatin landscape and transcription factor binding. However, our experiments also revealed that double mutant cells still differ from their patient-derived counterparts, highlighting the importance of studying patient cells to obtain a true picture of how gene regulatory networks have been reprogrammed during tumorigenesis.


Asunto(s)
Cromatina/metabolismo , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda , Mutación Missense , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Transcripción Genética , Translocación Genética , Sustitución de Aminoácidos , Cromatina/patología , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/metabolismo , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Femenino , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Proteínas Proto-Oncogénicas c-kit/genética , Proteína 1 Compañera de Translocación de RUNX1/genética
11.
Genet Test Mol Biomarkers ; 24(8): 527-531, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32716213

RESUMEN

Aims: Fragile-X syndrome (FXS) is the most common inherited form of intellectual disability; it is caused by an abnormal CGG-repeat expansion at the FMR1 gene. However, a few cases of girls with mutations in the FMR1 gene have been reported in the literature. In this study, we describe the clinical and genetic assessment of a family who exhibits the unusual coexistence of FXS, an 8p23.1 deletion, and balanced translocation t(7;10)(p10;q24) in multiple members, including a symptomatic girl with FXS. Materials and Methods: All of the family members underwent comprehensive clinical and neurological examinations. All members of the family were also molecularly diagnosed using a combination of fluorescent-polymerase chain reaction (PCR), Triplet Repeat Primed-PCR, capillary electrophoresis, and karyotyping. Results: We identified a male proband and a female patient that presented with the craniofacial characteristics of FXS, neuropsychomotor developmental delay, speech delay, intellectual deficit, and a positive molecular diagnosis of FXS. Interestingly, the female patient presented with a severe phenotype also associated with the presence of 8p23.1 deletion, while the proband patient presented a balanced translocation t(7;10)(p10;q24). Moreover, we detected multiple carriers of the FXS premutation in the family. Conclusions: To our knowledge, we describe for the first time the simultaneous occurrence of FXS and an 8p23.1 deletion and their possible synergistic effects on the phenotype of a female patient. Moreover, we describe the coexistence of FXS, an 8p23.1 deletion, and a balanced translocation t(7;10)(p10;q24) in the same family.


Asunto(s)
Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/genética , Adolescente , Adulto , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Familia , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/epidemiología , Pruebas Genéticas/métodos , Heterocigoto , Humanos , Discapacidad Intelectual/genética , Masculino , México , Persona de Mediana Edad , Mutación , Linaje , Fenotipo , Translocación Genética/genética
12.
J Clin Invest ; 130(8): 4213-4217, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32369452

RESUMEN

Molecular mechanisms governing the development of the mammalian cochlea, the hearing organ, remain largely unknown. Through genome sequencing in 3 subjects from 2 families with nonsyndromic cochlear aplasia, we identified homozygous 221-kb and 338-kb deletions in a noncoding region on chromosome 8 with an approximately 200-kb overlapping section. Genomic location of the overlapping deleted region started from approximately 350 kb downstream of GDF6, which codes for growth and differentiation factor 6. Otic lineage cells differentiated from induced pluripotent stem cells derived from an affected individual showed reduced expression of GDF6 compared with control cells. Knockout of Gdf6 in a mouse model resulted in cochlear aplasia, closely resembling the human phenotype. We conclude that GDF6 plays a necessary role in early cochlear development controlled by cis-regulatory elements located within an approximately 500-kb region of the genome in humans and that its disruption leads to deafness due to cochlear aplasia.


Asunto(s)
Cromosomas Humanos Par 8 , Cóclea , Enfermedades Cocleares , Factor 6 de Diferenciación de Crecimiento , Elementos de Respuesta , Animales , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Cóclea/embriología , Cóclea/patología , Enfermedades Cocleares/embriología , Enfermedades Cocleares/genética , Enfermedades Cocleares/patología , Femenino , Factor 6 de Diferenciación de Crecimiento/biosíntesis , Factor 6 de Diferenciación de Crecimiento/genética , Humanos , Masculino , Ratones , Ratones Transgénicos
13.
Int J Lab Hematol ; 42(1): 37-45, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31725954

RESUMEN

INTRODUCTION: A prototype of good prognosis, t(8;21)-positive AML, has diverse clinical and genetic features which affect its outcome. This study aimed at evaluating the clinico-pathological spectrum of t(8;21)-positive AML and ascertaining prognostic factors influencing its outcome in the Indian subcontinent. METHODS: A retrospective analysis of 75 cases of t(8;21)-positive AML diagnosed over a period of six years (2013-2018) was carried out. Detailed clinical and laboratory data of the patients were collected from the electronic medical records and reviewed. RESULTS: Median age was 19.5 years (range 5-75 years) with a M:F of 1.7. Myeloid sarcoma was observed in 9.3% cases. There were 85% FAB AML-M2, 8% AML-M1, and 7% AML-M4 subtypes. Prominent morphological characteristics included dyspoiesis in maturing myeloid cells (83%), long thin tapered Auer rods (58%), cytoplasmic vacuoles (58%), eosinophilia (50%), and mast cells (22%). Auer rods in maturing granulocytes (4% cases) were highly suggestive of the translocation. Additional cytogenetic abnormalities were present in 53% cases. Seventy-one percent (25/35) achieved CR. The overall survival (OS) was 40%, with a median follow-up of 27 months (range 4-57 months). None of the hematological or cytogenetic factors correlated with OS, except for the presence of myeloid sarcoma which had a trend toward poor survival (P = .07). CONCLUSION: Outcome of t(8;21) AML is not influenced by any of the clinico-pathological parameters, except for a myeloid sarcoma, which may herald a poor prognosis. Recognition of this distinct subtype of AML would facilitate further molecular screening for risk stratification in resource-constrained settings.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Leucemia Mieloide Aguda , Sarcoma Mieloide , Translocación Genética , Adolescente , Adulto , Anciano , Asparaginasa/administración & dosificación , Niño , Preescolar , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/metabolismo , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/sangre , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Daunorrubicina/administración & dosificación , Supervivencia sin Enfermedad , Femenino , Humanos , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Prednisona/administración & dosificación , Proteína 1 Compañera de Translocación de RUNX1/sangre , Proteína 1 Compañera de Translocación de RUNX1/genética , Estudios Retrospectivos , Sarcoma Mieloide/sangre , Sarcoma Mieloide/tratamiento farmacológico , Sarcoma Mieloide/genética , Sarcoma Mieloide/mortalidad , Tasa de Supervivencia , Vincristina/administración & dosificación
14.
Cell Rep ; 28(12): 3022-3031.e7, 2019 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-31533028

RESUMEN

Acute myeloid leukemia (AML) is associated with mutations in transcriptional and epigenetic regulator genes impairing myeloid differentiation. The t(8;21)(q22;q22) translocation generates the RUNX1-ETO fusion protein, which interferes with the hematopoietic master regulator RUNX1. We previously showed that the maintenance of t(8;21) AML is dependent on RUNX1-ETO expression. Its depletion causes extensive changes in transcription factor binding, as well as gene expression, and initiates myeloid differentiation. However, how these processes are connected within a gene regulatory network is unclear. To address this question, we performed Promoter-Capture Hi-C assays, with or without RUNX1-ETO depletion and assigned interacting cis-regulatory elements to their respective genes. To construct a RUNX1-ETO-dependent gene regulatory network maintaining AML, we integrated cis-regulatory element interactions with gene expression and transcription factor binding data. This analysis shows that RUNX1-ETO participates in cis-regulatory element interactions. However, differential interactions following RUNX1-ETO depletion are driven by alterations in the binding of RUNX1-ETO-regulated transcription factors.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Elementos de Facilitación Genéticos , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica , Regiones Promotoras Genéticas , Proteína 1 Compañera de Translocación de RUNX1 , Factor de Transcripción AP-1 , Translocación Genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/metabolismo , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Eliminación de Gen , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
16.
Acta Haematol ; 142(2): 92-97, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31085913

RESUMEN

The platelet-derived growth factor receptor ß (PDGFRB) gene translocations lead to a spectrum of chronic myeloid neoplasms, frequently associated with eosinophilia. Clinical heterogeneity is associated with a molecular one. Here, we report a novel case of a patient harboring a t(5;8)(q33;p22) translocation, resulting in the PCM1/PDGFRB fusion. Conventional cytogenetics and RNA sequencing were performed to identify the chromosomes and the genes involved in the rearrangement, respectively. This study shows that the combination of different strategies is pivotal to fine-tune the diagnosis and the clinical management of the patient. After 1 year of treatment with imatinib, the patient achieves hematological and molecular remission. We present an attractive strategy to identify novel and/or cryptic fusions, which will be relevant for clinicians dealing with the diagnosis of the patients with myelodysplastic syndrome/myeloproliferative diseases with atypical manifestations.


Asunto(s)
Autoantígenos , Proteínas de Ciclo Celular , Mesilato de Imatinib/administración & dosificación , Leucemia Mielógena Crónica BCR-ABL Positiva , Proteínas de Fusión Oncogénica , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Análisis de Secuencia de ARN , Autoantígenos/genética , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromosomas Humanos Par 5/genética , Cromosomas Humanos Par 5/metabolismo , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Translocación Genética
17.
Ann Hematol ; 98(5): 1149-1157, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30759270

RESUMEN

t(8;16)(p11.2;p13.3)/KAT6A-CREBBP is a rare recurrent cytogenetic abnormality associated with acute myeloid leukemia (AML). We report 15 cases with t(8;16)(p11.2;p13.3). All patients were adult and had AML: 13 women and 2 men, with a median age of 50 years. Ten patients had a history of malignancy and received cytotoxic therapies before therapy-related AML (t-AML), and five patients had de novo AML. All cases of AML showed monoblastic (n = 12) or myelomonocytic (n = 3) differentiation. Hemophagocytosis was observed in seven patients. All patients had t(8;16) in the stemline: seven had t(8;16) as the sole abnormality, two had one additional abnormality, and six had a complex karyotype. KAT6A/CREBBP rearrangement was confirmed by fluorescence in situ hybridization in 13 patients who had material available for analysis. All patients received induction chemotherapy, and 11 achieved complete remission after first induction. At the time of last follow-up, nine patients (eight t-AML and one de novo AML) died and six were alive, with a median overall survival of 18.2 months. The patients with de novo AML and/or patients with non-complex karyotype showed an "undefined" overall survival. We conclude that t(8;16)(p11.2;p13.3) commonly exhibits monoblastic or myelomonocytic differentiation and commonly arises in patients with a history of cancer treated with cytotoxic therapies. Patients with de novo AML with t(8;16) or t-AML with t(8;16) without adverse prognostic factors (e.g., complex karyotype) have a good outcome.


Asunto(s)
Proteína de Unión a CREB , Cromosomas Humanos Par 16 , Cromosomas Humanos Par 8 , Histona Acetiltransferasas , Leucemia Mieloide Aguda , Proteínas de Fusión Oncogénica , Translocación Genética , Adulto , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 16/metabolismo , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Femenino , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Estudios Retrospectivos
18.
PLoS One ; 14(2): e0212850, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30794691

RESUMEN

Crohn's disease (CD) is a debilitating inflammatory bowel disease (IBD) that arises from chronic inflammation in the gastrointestinal tract. Genome-wide association studies (GWAS) have identified over 200 single nucleotide polymorphisms (SNPs) that are associated with a predisposition for developing IBD. For the majority, the causal variant and target genes affected are unknown. Here, we investigated the CD-associated SNP rs6651252 that maps to a gene desert region on chromosome 8. We demonstrate that rs6651252 resides within a Wnt responsive DNA enhancer element (WRE) and that the disease associated allele augments binding of the TCF7L2 transcription factor to this region. Using CRISPR/Cas9 directed gene editing and epigenetic modulation, we find that the rs6651252 enhancer regulates expression of the c-MYC proto-oncogene (MYC). Furthermore, we found MYC transcript levels are elevated in patient-derived colonic segments harboring the disease-associated allele in comparison to those containing the ancestral allele. These results suggest that Wnt/MYC signaling contributes to CD pathogenesis and that patients harboring the disease-associated allele may benefit from therapies that target MYC or MYC-regulated genes.


Asunto(s)
Colon/metabolismo , Enfermedad de Crohn , Elementos de Facilitación Genéticos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-myc , Alelos , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Colon/patología , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Células Epiteliales/patología , Células HCT116 , Células HEK293 , Humanos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Vía de Señalización Wnt/genética
19.
Hemoglobin ; 42(2): 108-112, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29745274

RESUMEN

Globin switching is a significant factor on blood hemoglobin (Hb) level but its molecular mechanisms have not yet been identified, however, several quantitative trait loci (QTL) and polymorphisms involved regions on chromosomes 2p, 6q, 8q and X account for variation in the γ-globin expression level. We studied the effect of interaction between a region on intron six of the TOX gene, chromosome 8q (chr8q) and XmnI locus on the γ-globin promoter, chr11p on γ-globin expression in 150 ß-thalassemia intermedia (ß-TI) patients, evaluated by statistical interaction analysis. Our results showed a significant interaction between one QTL on intron six of the TOX gene (rs9693712) and XmnI locus that effect γ-globin expression. Interchromosomal interaction mediates through transcriptional machanisms to preserve true genome architectural features, chromosomes localization and DNA bending. This interaction can be a part of the unknown molecular mechanism of globin switching and regulation of gene expression.


Asunto(s)
Regulación de la Expresión Génica , Sitios de Carácter Cuantitativo , Talasemia beta/genética , gamma-Globinas/metabolismo , Cromosomas Humanos Par 8/metabolismo , ADN/ultraestructura , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Intrones , Regiones Promotoras Genéticas , gamma-Globinas/genética
20.
Exp Mol Med ; 50(4): 1-8, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29674693

RESUMEN

The AML1-ETO fusion protein (A/E), which results from the t(8;21) translocation, is considered to be a leukemia-initiating event. Identifying the mechanisms underlying the oncogenic activity of A/E remains a major challenge. In this study, we identified a specific down-regulation of brain acid-soluble protein 1 (BASP1) in t(8;21) acute myeloid leukemia (AML). A/E recognized AML1-binding sites and recruited DNA methyltransferase 3a (DNMT3a) to the BASP1 promoter sequence, which triggered DNA methylation-mediated silencing of BASP1. Ectopic expression of BASP1 inhibited proliferation and the colony-forming ability of A/E-positive AML cell lines and led to apoptosis and cell cycle arrest. The DNMT inhibitor decitabine up-regulated the expression of BASP1 in A/E-positive AML cell lines. In conclusion, our data suggest that BASP1 silencing via promoter methylation may be involved in A/E-mediated leukemogenesis and that BASP1 targeting may be an actionable therapeutic strategy in t(8;21) AML.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Cromosomas Humanos Par 21/metabolismo , Cromosomas Humanos Par 8/metabolismo , Metilación de ADN , Leucemia Mieloide Aguda/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Represoras/biosíntesis , Translocación Genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , ADN Metiltransferasa 3A , Silenciador del Gen , Células HL-60 , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Proteínas Represoras/genética , Células THP-1 , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...