Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Genome Res ; 33(9): 1439-1454, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37798116

RESUMO

Fascioscapulohumeral muscular dystrophy (FSHD) is caused by a unique genetic mechanism that relies on contraction and hypomethylation of the D4Z4 macrosatellite array on the Chromosome 4q telomere allowing ectopic expression of the DUX4 gene in skeletal muscle. Genetic analysis is difficult because of the large size and repetitive nature of the array, a nearly identical array on the 10q telomere, and the presence of divergent D4Z4 arrays scattered throughout the genome. Here, we combine nanopore long-read sequencing with Cas9-targeted enrichment of 4q and 10q D4Z4 arrays for comprehensive genetic analysis including determination of the length of the 4q and 10q D4Z4 arrays with base-pair resolution. In the same assay, we differentiate 4q from 10q telomeric sequences, determine A/B haplotype, identify paralogous D4Z4 sequences elsewhere in the genome, and estimate methylation for all CpGs in the array. Asymmetric, length-dependent methylation gradients were observed in the 4q and 10q D4Z4 arrays that reach a hypermethylation point at approximately 10 D4Z4 repeat units, consistent with the known threshold of pathogenic D4Z4 contractions. High resolution analysis of individual D4Z4 repeat methylation revealed areas of low methylation near the CTCF/insulator region and areas of high methylation immediately preceding the DUX4 transcriptional start site. Within the DUX4 exons, we observed a waxing/waning methylation pattern with a 180-nucleotide periodicity, consistent with phased nucleosomes. Targeted nanopore sequencing complements recently developed molecular combing and optical mapping approaches to genetic analysis for FSHD by adding precision of the length measurement, base-pair resolution sequencing, and quantitative methylation analysis.


Assuntos
Distrofia Muscular Facioescapuloumeral , Sequenciamento por Nanoporos , Humanos , Distrofia Muscular Facioescapuloumeral/genética , Metilação de DNA , Processamento de Proteína Pós-Traducional , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 4/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo
2.
Cancer Sci ; 115(4): 1224-1240, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38403332

RESUMO

The transcription factor forkhead box protein O1 (FoxO1) is closely related to the occurrence and development of ovarian cancer (OC), however its role and molecular mechanisms remain unclear. Herein, we found that FoxO1 was highly expressed in clinical samples of OC patients and was significantly correlated with poor prognosis. FoxO1 knockdown inhibited the proliferation of OC cells in vitro and in vivo. ChIP-seq combined with GEPIA2 and Kaplan-Meier database analysis showed that structural maintenance of chromosome 4 (SMC4) is a downstream target of FoxO1, and FoxO1 promotes SMC4 transcription by binding to its -1400/-1390 bp promoter. The high expression of SMC4 significantly blocked the tumor inhibition effect of FoxO1 knockdown. Furtherly, FoxO1 increased SMC4 mRNA abundance by transcriptionally activating methyltransferase-like 14 (METTL14) and increasing SMC4 m6A methylation on its coding sequence region. The Cancer Genome Atlas dataset analysis confirmed a significant positive correlation between FoxO1, SMC4, and METTL14 expression in OC. In summary, this study revealed the molecular mechanisms of FoxO1 regulating SMC4 and established a clinical link between the expression of FoxO1/METTL14/SMC4 in the occurrence of OC, thus providing a potential diagnostic target and therapeutic strategy.


Assuntos
Cromossomos Humanos Par 4 , Neoplasias Ovarianas , Feminino , Humanos , Adenosina Trifosfatases/genética , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/genética , Cromossomos Humanos Par 4/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Estimativa de Kaplan-Meier , Metiltransferases/genética , Neoplasias Ovarianas/patologia
3.
Dokl Biochem Biophys ; 490(1): 50-53, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32342314

RESUMO

In order to study the effects of heat shock treatment on the distribution of rDNA contacts at the region possessing DUX genes inside chromosome 4 we used 4C approach. Our data indicate that the treatment removes the frequent rDNA contacts in this region. The recent data on involvement of superenhancers that are decorated by broad H3K27ac marks in the phase separation mechanisms and the previous data demonstrating that these broad marks are the favorite sites of rDNA contacts taken together with our data on sensitivity of the contacts to the heat shock treatment suggest that the phase separation mechanisms are involved in the reversible rDNA-mediated regulation of gene expression via the contacts.


Assuntos
Cromossomos Humanos Par 4/metabolismo , DNA Ribossômico/genética , Proteínas de Choque Térmico , Histonas/química , Proteínas de Homeodomínio/química , Núcleo Celular/metabolismo , Biblioteca Gênica , Células HEK293 , Resposta ao Choque Térmico/genética , Temperatura Alta , Humanos , Família Multigênica , Cromossomos Politênicos
4.
J Cutan Pathol ; 46(8): 599-602, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30989672

RESUMO

Capicua transcriptional repressor (CIC)-rearranged sarcomas are part of the group of Ewing-like sarcomas or atypical Ewing sarcomas which, thanks to the progress in molecular diagnosis, are being defined by particular genetic abnormalities separating this group into distinct entities with their own particular histological and immunohistochemical features, as well as different survival outcomes. We report the case of a healthy 28-year-old female presenting with a tender lesion on her forearm which after ultrasound examination was clinically favored to represent an infected sebaceous cyst. Hematoxylin-eosin staining showed a lobulated neoplasm within the subcutis composed of poorly differentiated epithelioid to round cells with a small amount of amphophilic cytoplasm. Frequent mitotic figures and tumor necrosis were present. Immunohistochemical studies showed patchy focal CD99 membranous positivity, negative WT1 and TLE1 staining and diffuse nuclear positivity for ETV4 (performed at outside laboratory). FISH analysis showed significant CIC rearrangement enabling a final diagnosis of an undifferentiated small round cell sarcoma harboring the t(4;19)(q35;q13.1) and CIC-DUX4 fusion. This case shows the importance of awareness of this entity as, unlike Ewing sarcoma, these lesions present in the soft tissues rather than bone and may, as in this case, arise in the superficial soft tissues and be submitted to a dermatopathology practice.


Assuntos
Cisto Epidérmico , Antebraço , Rearranjo Gênico , Proteínas de Homeodomínio , Proteínas de Fusão Oncogênica , Proteínas Repressoras , Sarcoma de Células Pequenas , Neoplasias Cutâneas , Adulto , Cromossomos Humanos Par 19/genética , Cromossomos Humanos Par 19/metabolismo , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 4/metabolismo , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Cisto Epidérmico/genética , Cisto Epidérmico/metabolismo , Cisto Epidérmico/patologia , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hibridização in Situ Fluorescente , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Sarcoma de Células Pequenas/genética , Sarcoma de Células Pequenas/metabolismo , Sarcoma de Células Pequenas/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Translocação Genética , Proteínas WT1/genética , Proteínas WT1/metabolismo
5.
Am J Hum Genet ; 90(5): 871-8, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22521416

RESUMO

Autosomal-recessive primary microcephaly (MCPH) is a rare congenital disorder characterized by intellectual disability, reduced brain and head size, but usually without defects in cerebral cortical architecture, and other syndromic abnormalities. MCPH is heterogeneous. The underlying genes of the seven known loci code for centrosomal proteins. We studied a family from northern Pakistan with two microcephalic children using homozygosity mapping and found suggestive linkage for regions on chromosomes 2, 4, and 9. We sequenced two positional candidate genes and identified a homozygous frameshift mutation in the gene encoding the 135 kDa centrosomal protein (CEP135), located in the linkage interval on chromosome 4, in both affected children. Post hoc whole-exome sequencing corroborated this mutation's identification as the causal variant. Fibroblasts obtained from one of the patients showed multiple and fragmented centrosomes, disorganized microtubules, and reduced growth rate. Similar effects were reported after knockdown of CEP135 through RNA interference; we could provoke them also by ectopic overexpression of the mutant protein. Our findings suggest an additional locus for MCPH at HSA 4q12 (MCPH8), further strengthen the role of centrosomes in the development of MCPH, and place CEP135 among the essential components of this important organelle in particular for a normal neurogenesis.


Assuntos
Proteínas de Transporte/genética , Deficiência Intelectual/genética , Microcefalia/genética , Mutação , Proteínas de Transporte/metabolismo , Centrossomo , Criança , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 4/metabolismo , Exoma , Éxons , Feminino , Técnicas de Silenciamento de Genes , Ligação Genética , Loci Gênicos , Homozigoto , Humanos , Deficiência Intelectual/fisiopatologia , Masculino , Microcefalia/fisiopatologia , Paquistão/epidemiologia , Linhagem , Polimorfismo de Nucleotídeo Único , Interferência de RNA , Análise de Sequência de DNA
6.
Alcohol Clin Exp Res ; 39(9): 1654-64, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26250693

RESUMO

BACKGROUND: Genetic variation in a region of chromosome 4p12 that includes the GABAA subunit gene GABRA2 has been reproducibly associated with alcohol dependence (AD). However, the molecular mechanisms underlying the association are unknown. This study examined correlates of in vitro gene expression of the AD-associated GABRA2 rs279858*C-allele in human neural cells using an induced pluripotent stem cell (iPSC) model system. METHODS: We examined mRNA expression of chromosome 4p12 GABAA subunit genes (GABRG1, GABRA2, GABRA4, and GABRB1) in 36 human neural cell lines differentiated from iPSCs using quantitative polymerase chain reaction and next-generation RNA sequencing. mRNA expression in adult human brain was examined using the BrainCloud and BRAINEAC data sets. RESULTS: We found significantly lower levels of GABRA2 mRNA in neural cell cultures derived from rs279858*C-allele carriers. Levels of GABRA2 RNA were correlated with those of the other 3 chromosome 4p12 GABAA genes, but not other neural genes. Cluster analysis based on the relative RNA levels of the 4 chromosome 4p12 GABAA genes identified 2 distinct clusters of cell lines, a low-expression cluster associated with rs279858*C-allele carriers and a high-expression cluster enriched for the rs279858*T/T genotype. In contrast, there was no association of genotype with chromosome 4p12 GABAA gene expression in postmortem adult cortex in either the BrainCloud or BRAINEAC data sets. CONCLUSIONS: AD-associated variation in GABRA2 is associated with differential expression of the entire cluster of GABAA subunit genes on chromosome 4p12 in human iPSC-derived neural cell cultures. The absence of a parallel effect in postmortem human adult brain samples suggests that AD-associated genotype effects on GABAA expression, although not present in mature cortex, could have effects on regulation of the chromosome 4p12 GABAA cluster during neural development.


Assuntos
Alcoolismo/genética , Alelos , Cromossomos Humanos Par 4/genética , Predisposição Genética para Doença/genética , Células-Tronco Neurais/fisiologia , Receptores de GABA-A/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alcoolismo/diagnóstico , Linhagem Celular , Células Cultivadas , Cromossomos Humanos Par 4/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/fisiologia , Subunidades Proteicas/biossíntese , Subunidades Proteicas/genética , Adulto Jovem
7.
Immunohematology ; 29(2): 63-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24094238

RESUMO

The JR blood group system (ISBT 032) consists of one antigen,Jra, which is of high prevalence in all populations. The rare Jr(a-) phenotype has been found mostly in Japanese and other Asian populations, but also in people of northern European ancestry, in Bedouin Arabs, and in one Mexican. Anti-Jra has caused transfusion reactions and is involved in hemolytic disease of the fetus and newborn. The Jra antigen is located on ABCG2 transporter, a multipass membrane glycoprotein (also known as the breast cancer resistance protein, BCRP), which is encoded by the ABCG2 gene on chromosome 4q22.1. The Jr(a-) phenotype mostly results from recessive inheritance of ABCG2 null alleles caused by frameshift or nonsense changes.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Alelos , Antígenos de Grupos Sanguíneos , Cromossomos Humanos Par 4 , Isoanticorpos , Proteínas de Neoplasias , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/imunologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antígenos de Grupos Sanguíneos/sangue , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/imunologia , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 4/metabolismo , Eritroblastose Fetal/sangue , Eritroblastose Fetal/genética , Eritroblastose Fetal/imunologia , Humanos , Isoanticorpos/sangue , Isoanticorpos/genética , Isoanticorpos/imunologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/metabolismo
8.
J Biol Chem ; 286(52): 44620-31, 2011 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-21937448

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD), a dominant hereditary disease with a prevalence of 7 per 100,000 individuals, is associated with a partial deletion in the subtelomeric D4Z4 repeat array on chromosome 4q. The D4Z4 repeat contains a strong transcriptional enhancer that activates promoters of several FSHD-related genes. We report here that the enhancer within the D4Z4 repeat binds the Krüppel-like factor KLF15. KLF15 was found to be up-regulated during myogenic differentiation induced by serum starvation or by overexpression of the myogenic differentiation factor MYOD. When overexpressed, KLF15 activated the D4Z4 enhancer and led to overexpression of DUX4c (Double homeobox 4, centromeric) and FRG2 (FSHD region gene 2) genes, whereas its silencing caused inactivation of the D4Z4 enhancer. In immortalized human myoblasts, the D4Z4 enhancer was activated by the myogenic factor MYOD, an effect that was abolished upon KLF15 silencing or when the KLF15-binding sites within the D4Z4 enhancer were mutated, indicating that the myogenesis-related activation of the D4Z4 enhancer was mediated by KLF15. KLF15 and several myogenesis-related factors were found to be expressed at higher levels in myoblasts, myotubes, and muscle biopsies from FSHD patients than in healthy controls. We propose that KLF15 serves as a molecular link between myogenic factors and the activity of the D4Z4 enhancer, and it thus contributes to the overexpression of the DUX4c and FRG2 genes during normal myogenic differentiation and in FSHD.


Assuntos
Cromossomos Humanos Par 4/metabolismo , Elementos Facilitadores Genéticos , Fatores de Transcrição Kruppel-Like/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/metabolismo , Proteínas Nucleares/metabolismo , Animais , Cromossomos Humanos Par 4/genética , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/genética , Células HeLa , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Desenvolvimento Muscular/genética , Músculo Esquelético/patologia , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/patologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
J Biol Chem ; 285(51): 40303-11, 2010 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20956530

RESUMO

The recent discovery of induced pluripotent stem cell (iPSC) technology provides an invaluable tool for creating in vitro representations of human genetic conditions. This is particularly relevant for those diseases that lack adequate animal models or where the species comparison is difficult, e.g. imprinting diseases such as the neurogenetic disorder Prader-Willi syndrome (PWS). However, recent reports have unveiled transcriptional and functional differences between iPSCs and embryonic stem cells that in cases are attributable to imprinting errors. This has suggested that human iPSCs may not be useful to model genetic imprinting diseases. Here, we describe the generation of iPSCs from a patient with PWS bearing a partial translocation of the paternally expressed chromosome 15q11-q13 region to chromosome 4. The resulting iPSCs match all standard criteria of bona fide reprogramming and could be readily differentiated into tissues derived from the three germ layers, including neurons. Moreover, these iPSCs retain a high level of DNA methylation in the imprinting center of the maternal allele and show concomitant reduced expression of the disease-associated small nucleolar RNA HBII-85/SNORD116. These results indicate that iPSCs may be a useful tool to study PWS and perhaps other genetic imprinting diseases as well.


Assuntos
Metilação de DNA , Impressão Genômica , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Síndrome de Prader-Willi/metabolismo , Desdiferenciação Celular/genética , Células Cultivadas , Cromossomos Humanos Par 15/genética , Cromossomos Humanos Par 15/metabolismo , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 4/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/patologia , RNA Nuclear Pequeno/biossíntese , RNA Nuclear Pequeno/genética , Translocação Genética/genética
10.
J Mol Biol ; 433(6): 166700, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33160979

RESUMO

Significant efforts have been recently made to obtain the three-dimensional (3D) structure of the genome with the goal of understanding how structures may affect gene regulation and expression. Chromosome conformational capture techniques such as Hi-C, have been key in uncovering the quantitative information needed to determine chromatin organization. Complementing these experimental tools, co-polymers theoretical methods are necessary to determine the ensemble of three-dimensional structures associated to the experimental data provided by Hi-C maps. Going beyond just structural information, these theoretical advances also start to provide an understanding of the underlying mechanisms governing genome assembly and function. Recent theoretical work, however, has been focused on single chromosome structures, missing the fact that, in the full nucleus, interactions between chromosomes play a central role in their organization. To overcome this limitation, MiChroM (Minimal Chromatin Model) has been modified to become capable of performing these multi-chromosome simulations. It has been upgraded into a fast and scalable software version, which is able to perform chromosome simulations using GPUs via OpenMM Python API, called Open-MiChroM. To validate the efficiency of this new version, analyses for GM12878 individual autosomes were performed and compared to earlier studies. This validation was followed by multi-chain simulations including the four largest human chromosomes (C1-C4). These simulations demonstrated the full power of this new approach. Comparison to Hi-C data shows that these multiple chromosome interactions are essential for a more accurate agreement with experimental results. Without any changes to the original MiChroM potential, it is now possible to predict experimentally observed inter-chromosome contacts. This scalability of Open-MiChroM allow for more audacious investigations, looking at interactions of multiple chains as well as moving towards higher resolution chromosomes models.


Assuntos
Cromatina/química , Cromossomos Humanos Par 1/química , Cromossomos Humanos Par 2/química , Cromossomos Humanos Par 3/química , Cromossomos Humanos Par 4/química , Simulação de Dinâmica Molecular , Software , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Cromatina/ultraestrutura , Cromossomos Humanos Par 1/metabolismo , Cromossomos Humanos Par 1/ultraestrutura , Cromossomos Humanos Par 2/metabolismo , Cromossomos Humanos Par 2/ultraestrutura , Cromossomos Humanos Par 3/metabolismo , Cromossomos Humanos Par 3/ultraestrutura , Cromossomos Humanos Par 4/metabolismo , Cromossomos Humanos Par 4/ultraestrutura , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Saccharum/genética , Saccharum/metabolismo , Termodinâmica , Triticum/genética , Triticum/metabolismo
11.
Ann Hematol ; 89(7): 643-52, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20195608

RESUMO

Despite recent progress in molecular research in myeloid malignancies, in subsets of patients with myelodysplastic syndrome (MDS) so far no underlying mutation was identified. In the myeloproliferative neoplasms (MPNs), the JAK2V617F alone cannot explain the phenotypic heterogeneity. In acute myeloid leukemia (AML), clinical variability exists within distinct subgroups. Thus, the search for novel molecular markers continues. Recently, mutations of the tet oncogene family member 2 (TET2) and Casitas B-cell lymphoma (CBL) genes became the focus of interest. With diverse genetic methods, TET2 on chromosome 4q24 was identified as candidate tumor suppressor gene. Sequencing studies revealed heterogeneous mutations in 10-25% of patients with acute myeloid leukemia (AML), MDS, and MPNs, while the frequency might be higher in chronic myelomonocytic leukemia (CMML). The prognostic impact is being explored. The CBL gene is involved in the degradation of tyrosine kinases. In rare cases of human AML (<2%), CBL mutants were identified, with a higher frequency in core binding factor leukemias. Presence of these mutations was suggested to be involved in aberrant FLT3 expression. In the MPNs, a 2-8% frequency of CBL mutations was reported. These novel mutations deepened insights in the mechanisms of leukemogenesis, might contribute to the identification of new therapeutic targets, and improve diagnostics in the myeloid malignancies.


Assuntos
Biomarcadores Tumorais/metabolismo , Cromossomos Humanos Par 4/metabolismo , Proteínas de Ligação a DNA/metabolismo , Leucemia Mieloide Aguda/metabolismo , Mutação , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Biomarcadores Tumorais/genética , Cromossomos Humanos Par 4/genética , Proteínas de Ligação a DNA/genética , Dioxigenases , Humanos , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-cbl/genética
12.
Eur J Haematol ; 83(5): 406-19, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19558506

RESUMO

OBJECTIVE: Gene expression profiles become increasingly more important for diagnostic procedures, allowing clinical predictions including treatment response and outcome. However, the establishment of specific and robust gene signatures from microarray data sets requires the analysis of large numbers of patients and the application of complex biostatistical algorithms. Especially in case of rare diseases and due to these constrains, diagnostic centers with limited access to patients or bioinformatic resources are excluded from implementing these new technologies. METHOD: In our study we sought to overcome these limitations and for proof of principle, we analyzed the rare t(4;11) leukemia disease entity. First, gene expression data of each t(4;11) leukemia patient were normalized by pairwise subtraction against normal bone marrow (n = 3) to identify significantly deregulated gene sets for each patient. RESULT: A 'core signature' of 186 commonly deregulated genes present in each investigated t(4;11) leukemia patient was defined. Linking the obtained gene sets to four biological discriminators (HOXA gene expression, age at diagnosis, fusion gene transcripts and chromosomal breakpoints) divided patients into two distinct subgroups: the first one comprised infant patients with low HOXA genes expression and the MLL breakpoints within introns 11/12. The second one comprised non-infant patients with high HOXA expression and MLL breakpoints within introns 9/10. CONCLUSION: A yet homogeneous leukemia entity was further subdivided, based on distinct genetic properties. This approach provided a simplified way to obtain robust and disease-specific gene signatures even in smaller cohorts.


Assuntos
Cromossomos Humanos Par 11/metabolismo , Cromossomos Humanos Par 4/metabolismo , Regulação Leucêmica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Translocação Genética , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 4/genética , Feminino , Perfilação da Expressão Gênica/métodos , Histona-Lisina N-Metiltransferase , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Masculino , Proteína de Leucina Linfoide-Mieloide/biossíntese , Proteína de Leucina Linfoide-Mieloide/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Proteínas de Fusão Oncogênica/biossíntese , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/classificação , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
13.
Nat Commun ; 10(1): 368, 2019 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-30664630

RESUMO

The molecular pathogenesis of salivary gland acinic cell carcinoma (AciCC) is poorly understood. The secretory Ca-binding phosphoprotein (SCPP) gene cluster at 4q13 encodes structurally related phosphoproteins of which some are specifically expressed at high levels in the salivary glands and constitute major components of saliva. Here we report on recurrent rearrangements [t(4;9)(q13;q31)] in AciCC that translocate active enhancer regions from the SCPP gene cluster to the region upstream of Nuclear Receptor Subfamily 4 Group A Member 3 (NR4A3) at 9q31. We show that NR4A3 is specifically upregulated in AciCCs, and that active chromatin regions and gene expression signatures in AciCCs are highly correlated with the NR4A3 transcription factor binding motif. Overexpression of NR4A3 in mouse salivary gland cells increases expression of known NR4A3 target genes and has a stimulatory functional effect on cell proliferation. We conclude that NR4A3 is upregulated through enhancer hijacking and has important oncogenic functions in AciCC.


Assuntos
Carcinoma de Células Acinares/genética , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica , Receptores de Esteroides/genética , Receptores dos Hormônios Tireóideos/genética , Neoplasias das Glândulas Salivares/genética , Proteínas e Peptídeos Salivares/genética , Translocação Genética , Células Acinares/metabolismo , Células Acinares/patologia , Animais , Carcinoma de Células Acinares/metabolismo , Carcinoma de Células Acinares/patologia , Proliferação de Células , Cromatina/química , Cromatina/metabolismo , Cromossomos Humanos Par 4/química , Cromossomos Humanos Par 4/metabolismo , Cromossomos Humanos Par 9/química , Cromossomos Humanos Par 9/metabolismo , Estudos de Coortes , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Feminino , Loci Gênicos , Humanos , Masculino , Camundongos , Família Multigênica , Cultura Primária de Células , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Neoplasias das Glândulas Salivares/metabolismo , Neoplasias das Glândulas Salivares/patologia , Glândulas Salivares/metabolismo , Glândulas Salivares/patologia , Proteínas e Peptídeos Salivares/metabolismo
14.
J Mol Cell Cardiol ; 45(6): 724-34, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18790697

RESUMO

Recent findings illustrate a critical role for ankyrin-B function in normal cardiovascular physiology. Specifically, decreased expression of ankyrin-B in mice or human mutations in the ankyrin-B gene (ANK2) results in potentially fatal cardiac arrhythmias. Despite the clear role of ankyrin-B in heart, the mechanisms underlying transcriptional regulation of ANK2 are unknown. In fact, to date there is no description of ANK2 genomic organization. The aims of this study were to provide a comprehensive description of the ANK2 gene and to evaluate the relative expression of alternative splicing events associated with ANK2 transcription in heart. Using reverse-transcriptase PCR on mRNA isolated from human hearts, we identify seven new exons associated with the ANK2 gene including an alternative first exon located approximately 145 kb upstream of the previously-identified first exon. In addition, we identify over thirty alternative splicing events associated with ANK2 mRNA transcripts. Using real-time PCR and exon boundary-spanning primers to selectively amplify these splice variants, we demonstrate that these variants are expressed at varying levels in human heart. Finally, ankyrin-B immunoblot analysis demonstrates the expression of a heterogeneous population of ankyrin-B polypeptides in heart. ANK2 consists of 53 exons that span approximately 560 kb on human chromosome 4. Additionally, our data demonstrates that ANK2 is subject to complex transcriptional regulation that likely results in differential ankyrin-B polypeptide function.


Assuntos
Processamento Alternativo , Anquirinas/biossíntese , Arritmias Cardíacas/metabolismo , Proteínas Musculares/biossíntese , Miocárdio/metabolismo , Processamento Alternativo/genética , Animais , Anquirinas/genética , Arritmias Cardíacas/genética , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 4/metabolismo , Éxons/genética , Feminino , Humanos , Masculino , Camundongos , Proteínas Musculares/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transcrição Gênica/genética
15.
PLoS One ; 13(5): e0197194, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29746601

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is a hematopoietic malignancy originating from T-cell progenitors in which differentiation is blocked at early stages. Physiological expression of specific NKL homeobox genes obeys a hematopoietic NKL-code implicated in the process of lymphopoiesis while in differentiated T-cells these genes are silenced. We propose that this developmental expression pattern underlies the observation that NKL homeobox genes are the most ubiquitous group of transcription factors deregulated in T-ALL, including TLX1, TLX3, NKX2-5 and NKX3-1. Here, we describe a novel member of the NKL homeobox gene subclass, NKX3-2 (BAPX1), which is aberrantly activated in 18% of pediatric T-ALL patients analyzed while being normally expressed in developing spleen. Identification of NKX3-2 expression in T-ALL cell line CCRF-CEM qualified these cells to model its deregulation and function in a leukemic context. Genomic and chromosomal analyses demonstrated normal configuration of the NKX3-2 locus at chromosome 4p15, thus excluding cytogenetic dysregulation. Comparative expression profiling analysis of NKX3-2 patient data revealed deregulated activity of BMP- and MAPK-signalling. These candidate pathways were experimentally confirmed to mediate aberrant NKX3-2 expression. We also show that homeobox gene SIX6, plus MIR17HG and GATA3 are downstream targets of NKX3-2 and plausibly contribute to the pathogenesis of this malignancy by suppressing T-cell differentiation. Finally, NKL homeobox gene NKX2-5 was activated by NKX3-2 in CCRF-CEM and by FOXG1 in PEER, representing mutually inhibitory activators of this translocated oncogene. Together, our findings reveal a novel oncogenic NKL homeobox gene subclass member which is aberrantly expressed in a large subset of T-ALL patients and participates in a deregulated gene network likely to arise in developing spleen.


Assuntos
Cromossomos Humanos Par 4/metabolismo , Regulação Leucêmica da Expressão Gênica , Loci Gênicos , Proteínas de Homeodomínio/biossíntese , Proteínas de Neoplasias/biossíntese , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Fatores de Transcrição/biossíntese , Linhagem Celular , Cromossomos Humanos Par 4/genética , Proteínas de Homeodomínio/genética , Humanos , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Fatores de Transcrição/genética
16.
Radiat Res ; 163(3): 324-31, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15733039

RESUMO

Delayed reproductive cell death or lethal mutations in the survivors of irradiated cells is a well-characterized end point associated with radiation-induced genomic instability. Although the mechanism for this delayed lethality has not been identified, it is thought to be a means of eliminating cells that have sustained extensive damage, thus preventing tissue disruption after radiation exposure. In this study we have tested the hypothesis that delayed reproductive cell death in chromosomally unstable GM10115 clones is due to persistently increased levels of apoptosis. Evidence for differences in apoptosis in two representative genomically unstable clones after irradiation is presented. In addition, one of the unstable clones was found to have abnormal levels of apoptosis after radiation exposure. An understanding of apoptosis in genomically unstable clones may provide insight into the maintenance of genomic instability and the mechanism by which genomically unstable cells evade cell death, potentially contributing to carcinogenesis.


Assuntos
Apoptose , Animais , Anexina A5/química , Western Blotting , Células CHO , Linhagem Celular Tumoral , Cromossomos Humanos Par 4/metabolismo , Cricetinae , Citocromos c/metabolismo , Dano ao DNA , Fragmentação do DNA , Densitometria , Relação Dose-Resposta à Radiação , Instabilidade Genômica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Hibridização in Situ Fluorescente , Marcação In Situ das Extremidades Cortadas , Metáfase , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fatores de Tempo
17.
Neoplasia ; 4(6): 544-50, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12407449

RESUMO

Human chromosome 4 was previously shown to elicit features of senescence when introduced into cell lines that map to complementation group B for senescence, including HeLa cells. Subsequently, a DNA segment encoding the pseudogene Mortality Factor 4 (MORF4) was shown to reproduce some of the effects of the intact chromosome 4 and was suggested to be a candidate mortality gene. We have identified multiple MORF4 alleles in several cell lines and tissues by sequencing and have failed to detect any cancer-specific mutations in three of the complementation group B lines (HeLa, T98G, and J82). Furthermore, MORF4 was heterozygous in these lines. These results question whether MORF4 is the chromosome 4 mortality gene. To map other candidate mortality gene(s) on this chromosome, we employed microcell-mediated monochromosome transfer to introduce either a complete copy, or defined fragments of the chromosome into HeLa cells. The introduced chromosome 4 fragments mapped the mortality gene to a region between the centromere and the marker D4S2975 (4q27), thus excluding MORF4, which maps to 4q33-q34.1. Analysis of microsatellite markers on the introduced chromosome in 59 immortal segregants identified a frequently deleted region, spanning the markers BIR0110 and D4S1557. This defines a new candidate interval of 130 kb at 4q22-q23.


Assuntos
Mapeamento Cromossômico , Cromossomos Humanos Par 4/genética , Fatores de Transcrição/genética , Alelos , Animais , Senescência Celular/genética , Coloração Cromossômica , Cromossomos Humanos Par 4/metabolismo , Células Clonais , Genes Supressores de Tumor , Teste de Complementação Genética , Genótipo , Células HeLa/metabolismo , Humanos , Perda de Heterozigosidade , Camundongos , Repetições de Microssatélites , Fenótipo , Polimorfismo Genético , Fatores de Transcrição/metabolismo
19.
PLoS One ; 8(8): e71777, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23990986

RESUMO

Gene amplification at chromosome 4q12 is a common alteration in human high grade gliomas including glioblastoma, a CNS tumour with consistently poor prognosis. This locus harbours the known oncogenes encoding the receptor tyrosine kinases PDGFRA, KIT, and VEGFR2. These receptors are potential targets for novel therapeutic intervention in these diseases, with expression noted in tumour cells and/or associated vasculature. Despite this, a detailed assessment of their relative contributions to different high grade glioma histologies and the underlying heterogeneity within glioblastoma has been lacking. We studied 342 primary high grade gliomas for individual gene amplification using specific FISH probes, as well as receptor expression in the tumour and endothelial cells by immunohistochemistry, and correlated our findings with specific tumour cell morphological types and patterns of vasculature. We identified amplicons which encompassed PDGFRA only, PDGFRA/KIT, and PDGFRA/KIT/VEGFR2, with distinct phenotypic correlates. Within glioblastoma specimens, PDGFRA amplification alone was linked to oligodendroglial, small cell and sarcomatous tumour cell morphologies, and rare MGMT promoter methylation. A younger age at diagnosis and better clinical outcome in glioblastoma patients is only seen when PDGFRA and KIT are co-amplified. IDH1 mutation was only found when all three genes are amplified; this is a subgroup which also harbours extensive MGMT promoter methylation. Whilst PDGFRA amplification was tightly linked to tumour expression of the receptor, this was not the case for KIT or VEGFR2. Thus we have identified differential patterns of gene amplification and expression of RTKs at the 4q12 locus to be associated with specific phenotypes which may reflect their distinct underlying mechanisms.


Assuntos
Cromossomos Humanos Par 4/genética , Amplificação de Genes , Glioblastoma/genética , Proteínas Proto-Oncogênicas c-kit/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Cromossomos Humanos Par 4/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/parasitologia , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Fenótipo , Prognóstico , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
20.
Eur J Med Genet ; 55(1): 37-42, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21968327

RESUMO

Wolfram syndrome, also named "DIDMOAD" (diabetes insipidus, diabetes mellitus, optic atrophy, and deafness), is an inherited association of juvenile-onset diabetes mellitus and optic atrophy as key diagnostic criteria. Renal tract abnormalities and neurodegenerative disorder may occur in the third and fourth decade. The wolframin gene, WFS1, associated with this syndrome, is located on chromosome 4p16.1. Many mutations have been described since the identification of WFS1 as the cause of Wolfram syndrome. We identified a new homozygous WFS1 mutation (c.1532T>C; p.Leu511Pro) causing Wolfram syndrome in a large inbred Turkish family. The patients showed early onset of IDDM, diabetes insipidus, optic atrophy, sensorineural hearing impairment and very rapid progression to renal failure before age 12 in three females. Ectopic expression of the wolframin mutant in HEK cells results in greatly reduced levels of protein expression compared to wild-type wolframin, strongly supporting that this mutation is disease-causing. The mutation showed perfect segregation with disease in the family, characterized by early and severe clinical manifestations.


Assuntos
Nefropatias/genética , Proteínas de Membrana/genética , Mutação , Síndrome de Wolfram/genética , Adolescente , Sequência de Aminoácidos , Criança , Pré-Escolar , Cromossomos Humanos Par 4/genética , Cromossomos Humanos Par 4/metabolismo , Consanguinidade , Feminino , Células HEK293 , Homozigoto , Humanos , Nefropatias/diagnóstico , Nefropatias/patologia , Masculino , Dados de Sequência Molecular , Linhagem , Turquia/epidemiologia , Síndrome de Wolfram/diagnóstico , Síndrome de Wolfram/patologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA