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
País de afiliação
Intervalo de ano de publicação
1.
Am J Hum Genet ; 108(4): 709-721, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33735615

RESUMO

The fetal-to-adult hemoglobin switch is regulated in a developmental stage-specific manner and reactivation of fetal hemoglobin (HbF) has therapeutic implications for treatment of ß-thalassemia and sickle cell anemia, two major global health problems. Although significant progress has been made in our understanding of the molecular mechanism of the fetal-to-adult hemoglobin switch, the mechanism of epigenetic regulation of HbF silencing remains to be fully defined. Here, we performed whole-genome bisulfite sequencing and RNA sequencing analysis of the bone marrow-derived GYPA+ erythroid cells from ß-thalassemia-affected individuals with widely varying levels of HbF groups (HbF ≥ 95th percentile or HbF ≤ 5th percentile) to screen epigenetic modulators of HbF and phenotypic diversity of ß-thalassemia. We identified an ETS2 repressor factor encoded by ERF, whose promoter hypermethylation and mRNA downregulation are associated with high HbF levels in ß-thalassemia. We further observed that hypermethylation of the ERF promoter mediated by enrichment of DNMT3A leads to demethylation of γ-globin genes and attenuation of binding of ERF on the HBG promoter and eventually re-activation of HbF in ß-thalassemia. We demonstrated that ERF depletion markedly increased HbF production in human CD34+ erythroid progenitor cells, HUDEP-2 cell lines, and transplanted NCG-Kit-V831M mice. ERF represses γ-globin expression by directly binding to two consensus motifs regulating γ-globin gene expression. Importantly, ERF depletion did not affect maturation of erythroid cells. Identification of alterations in DNA methylation of ERF as a modulator of HbF synthesis opens up therapeutic targets for ß-hemoglobinopathies.


Assuntos
Epigênese Genética , Perfilação da Expressão Gênica , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Talassemia beta/genética , gama-Globinas/genética , Animais , Antígenos CD34/metabolismo , Sequência de Bases , Sistemas CRISPR-Cas/genética , Diferenciação Celular , Linhagem Celular , Criança , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , DNA Metiltransferase 3A , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Feminino , Hemoglobina Fetal/genética , Edição de Genes , Humanos , Masculino , Camundongos , Regiões Promotoras Genéticas/genética , Reprodutibilidade dos Testes , Sulfitos , Sequenciamento Completo do Genoma , Talassemia beta/patologia
2.
Biochem Biophys Res Commun ; 701: 149555, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38325179

RESUMO

Fetal-to-adult hemoglobin switching is controlled by programmed silencing of γ-globin while the re-activation of fetal hemoglobin (HbF) is an effective strategy for ameliorating the clinical severity of ß-thalassemia and sickle cell disease. The identification of enhancer RNAs (eRNAs) related to the fetal (α2γ2) to adult hemoglobin (α2ß2) switching remains incomplete. In this study, the transcriptomes of GYPA+ cells from six ß-thalassemia patients with extreme HbF levels were sequenced to identify differences in patterns of noncoding RNA expression. It is interesting that an enhancer upstream of CHD4, an HbF-related core subunit of the NuRD complex, was differentially transcribed. We found a significantly positive correlation of eRNA-CHD4 enhancer-gene interaction using the public database of FANTOM5. Specifically, the eRNA-CHD4 expression was found to be significantly higher in both CD34+ HSPCs and HUDEP-2 than those in K562 cells which commonly expressed high level of HbF, suggesting a correlation between eRNA and HbF expression. Furthermore, prediction of transcription binding sites of cis-eQTLs and the CHD4 genomic region revealed a putative interaction site between rs73264846 and ZNF410, a known transcription factor regulating HbF expression. Moreover, in-vitro validation showed that the inhibition of eRNA could reduce the expression of HBG expression in HUDEP-2 cells. Taken together, the findings of this study demonstrate that a distal enhancer contributes to stage-specific silencing of γ-globin genes through direct modulation of CHD4 expression and provide insights into the epigenetic mechanisms of NuRD-mediated hemoglobin switching.


Assuntos
Anemia Falciforme , Talassemia beta , Adulto , Humanos , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , gama-Globinas/genética , gama-Globinas/metabolismo , Talassemia beta/genética , Regulação da Expressão Gênica , Anemia Falciforme/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo
3.
Anal Chem ; 95(24): 9244-9251, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37285171

RESUMO

Identification of α-thalassemia silent carriers is challenging with conventional phenotype-based screening methods. A liquid chromatography tandem mass spectrometry (LC-MS/MS)-based approach may offer novel biomarkers to address this conundrum. In this study, we collected dried blood spot samples from individuals with three α-thalassemia subtypes for biomarker discovery and validation. We observed differential expression patterns of hemoglobin subunits among various α-thalassemia subtypes and normal controls through proteomic profiling of 51 samples in the discovery phase. Then, we developed and optimized a multiple reaction monitoring (MRM) assay to measure all detectable hemoglobin subunits. The validation phase was conducted in a cohort of 462 samples. Among the measured hemoglobin subunits, subunit µ was significantly upregulated in all the α-thalassemia groups with distinct fold changes. The hemoglobin subunit µ exhibits great potential as a novel biomarker for α-thalassemia, especially for silent α-thalassemia. We constructed predictive models based on the concentrations of hemoglobin subunits and their ratios to classify the various subtypes of α-thalassemia. In the binary classification problems of silent α-thalassemia vs normal, non-deletional α-thalassemia vs normal, and deletional α-thalassemia vs normal, the best performance of the models achieved average ROCAUCs of 0.9505, 0.9430, and 0.9976 in the cross-validation, respectively. In the multiclass model, the best performance achieved an average ROCAUC of 0.9290 in cross-validation. The performance of our MRM assay and models demonstrated that the hemoglobin subunit µ would play a vital role in screening silent α-thalassemia in clinical practice.


Assuntos
Subunidades de Hemoglobina , Talassemia alfa , Humanos , Cromatografia Líquida , Espectrometria de Massas em Tandem/métodos , Talassemia alfa/diagnóstico , Proteômica , Biomarcadores
4.
Blood ; 137(12): 1652-1657, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33227819

RESUMO

DNA methyltransferase 1 (DNMT1) is a major epigenetic regulator of the formation of large macromolecular complexes that repress human γ-globin expression by maintaining DNA methylation. However, very little is known about the association of DNMT1 variants with ß-thalassemia phenotypes. We systematically investigated associations between variants in DNMT1 and phenotypes in 1142 ß-thalassemia subjects and identified a novel missense mutation (c.2633G>A, S878F) in the DNMT1 bromo-adjacent homology-1 (BAH1) domain. We functionally characterized this mutation in CD34+ cells from patients and engineered HuDEP-2 mutant cells. Our results demonstrate that DNMT1 phosphorylation is abrogated by substituting serine with phenylalanine at position 878, resulting in lower stability and catalytic activity loss. S878F mutation also attenuated DNMT1 interactions with BCL11A, GATA1, and HDAC1/2, and reduced recruitment of DNMT1 to the γ-globin (HBG) promoters, leading to epigenetic derepression of γ-globin expression. By analyzing the F-cell pattern, we demonstrated that the effect of DNMT1 mutation on increased fetal hemoglobin (HbF) is heterocellular. Furthermore, introduction of S878F mutation into erythroid cells by clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) recapitulated γ-globin reactivation. Thus, the natural S878F DNMT1 mutation is a novel modulator of HbF synthesis and represents a potential new therapeutic target for ß-hemoglobinopathies.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/genética , Hemoglobina Fetal/genética , Talassemia beta/genética , gama-Globinas/genética , Linhagem Celular , Epigênese Genética , Humanos , Modelos Moleculares , Mutação , Regulação para Cima
5.
Acta Haematol ; 146(6): 458-464, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37573774

RESUMO

INTRODUCTION: Splenomegaly and hypersplenism are common complications of thalassemia patients due to the excessive clearance of defective red blood cells from the spleen. To date, splenectomy has been considered one of the most effective treatments for splenomegaly, reducing clinical severity among thalassemia patients. Thus, we aim to investigate the differences in splenectomy rates and hematological indices among thalassemia patients with different genotypes. METHOD: In this study, we analyzed the clinical data of thalassemia in 2,130 patients admitted to the 923rd Hospital of the People's Liberation Army from January 2006 to December 2020, and the statistical software SPSS 26.0 was applied to analyze the data. RESULT: Of the 2,130 patients with thalassemia, 265 patients underwent splenectomy. It was determined that significantly more patients with hemoglobin H (HbH) disease, a form of α-thalassemia, have undergone splenectomy than ß-thalassemia patients (20% vs. 7%). Further, HbH disease patients were diagnosed at a significantly older age than ß-thalassemia patients. CONCLUSION: The greater probability of HbH disease patients undergoing splenectomy is likely influenced by multiple factors, including their lower dependency on transfusion, leading to high spleen compensatory stress on the spleen, and the destruction of defective erythrocytes. In contrast, ß-thalassemia is clinically more severe and less tolerant of hemoglobin fluctuations. Based on these findings, clinicians are suggested to pay more attention to HbH disease patients as many of them are still under-transfused, which could lead to chronic hemolysis and more severe hepatosplenomegaly. These results might offer insight for improving the clinical management of patients with different types of thalassemia.


Assuntos
Talassemia alfa , Talassemia beta , Humanos , Talassemia beta/complicações , Talassemia beta/cirurgia , Talassemia alfa/cirurgia , Esplenectomia , Hemoglobina H , Esplenomegalia/cirurgia , Esplenomegalia/complicações
6.
Br J Haematol ; 195(5): 764-769, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34569056

RESUMO

Re-activation of fetal haemoglobin (HbF) has been proved to be an effective strategy for the treatment of ß-haemoglobinopathies. In this study, we identified TEA domain transcription factor 4 (TEAD4) as a new potential regulator of HbF by integrating public data sets with quantitative polymerase chain reaction analysis in ß-thalassaemia patients. Significant negative correlation was observed between the expression of TEAD4 and HbF levels in ß-thalassaemia patients. Functional validations of TEAD4 inhibition in both ß-thalassaemia CD34+ cells and HUDEP-2 cells indicated that depletion of TEAD4 led to a significant increase of HbF. Finally, we identified a binding motif of TEAD4 on γ-globin gene promoters; its disruption consistently led to de-repression of HbF. Taken together, these results demonstrate that TEAD4 could act as a transcriptional inhibitor of the γ-globin gene through direct binding on its promoter. Our findings demonstrate a novel role of TEAD4 on the regulation of HbF, which may benefit patients with ß-haemoglobinopathies.


Assuntos
Hemoglobina Fetal/genética , Regiões Promotoras Genéticas , Fatores de Transcrição de Domínio TEA/metabolismo , gama-Globinas/genética , Linhagem Celular , Células Eritroides/metabolismo , Regulação da Expressão Gênica , Humanos , Ligação Proteica , Talassemia beta/genética , Talassemia beta/metabolismo
7.
Br J Haematol ; 193(6): 1220-1227, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33997955

RESUMO

Reactivation of fetal haemoglobin (HbF) expression is an effective way to treat ß-thalassaemia and sickle cell anaemia. In the present study, we identified a novel GATA zinc finger domain-containing protein 2A (GATAD2A) mutation, which contributed to the elevation of HbF and ameliorated clinical severity in a patient with ß-thalassaemia, by targeted next-generation sequencing. Knockout of GATAD2A led to a significant induction of HbF in both human umbilical cord blood-derived erythroid progenitor-2 (HUDEP-2) and human cluster of differentiation (CD)34+ cells with a detectable impact on erythroid differentiation. Furthermore, heterozygous knockout of GATAD2A impaired recruitment of chromodomain helicase DNA-binding protein 4 (CHD4) to the methyl-binding domain protein 2 (MBD2)-containing nucleosome remodelling and deacetylation (NuRD) complex. Our present data suggest that mutations causing the haploinsufficiency of GATAD2A might contribute to amelioration of clinical severity in patients with ß-thalassaemia.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Nucleossomos/metabolismo , Proteínas Repressoras/deficiência , Talassemia beta/metabolismo , Acetilação , Adolescente , Linhagem Celular , Criança , Códon sem Sentido , Proteínas de Ligação a DNA/genética , Hemoglobina Fetal/genética , Haploinsuficiência , Humanos , Masculino , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Nucleossomos/genética , Proteínas Repressoras/metabolismo , Talassemia beta/genética
8.
Hemoglobin ; 45(5): 341-344, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35322741

RESUMO

We here describe a novel hemoglobin (Hb) variant, Hb Liaobu [α107(G14)Val→Leu, HBA2: c.322G>C], in a Chinese family. The structurally abnormal α chain variant could not be detected using capillary electrophoresis (CE) and was subsequently characterized by matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry (MS), and further confirmed by reversed phase high performance liquid chromatography (HPLC). Sanger sequencing revealed a novel base mutation on the α2-globin gene and RNA analysis by reverse transcription polymerase chain reaction (RT-PCR) showed the presence of an abnormal HBA transcript. The isopropanol stability test indicated the stable state of this structural Hb variant. In conclusion, a new Hb variant, Hb Liaobu, was discovered and characterized. It was proven to be a nonpathogenic variant. Our study resolved the confusion in the clinical diagnosis of individuals with this novel Hb variant in this family.


Assuntos
Hemoglobinas Anormais , Eletroforese Capilar , Hemoglobinas Anormais/análise , Hemoglobinas Anormais/genética , Humanos , Lasers , Espectrometria de Massas , Mutação , alfa-Globinas/análise , alfa-Globinas/genética
9.
Am J Hum Genet ; 101(1): 130-138, 2017 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-28669403

RESUMO

A delayed fetal-to-adult hemoglobin (Hb) switch ameliorates the severity of ß-thalassemia and sickle cell disease. The molecular mechanism underlying the epigenetic dysregulation of the switch is unclear. To explore the potential cis-variants responsible for the Hb switching, we systematically analyzed an 80-kb region spanning the ß-globin cluster using capture-based next-generation sequencing of 1142 Chinese ß-thalassemia persons and identified 31 fetal hemoglobin (HbF)-associated haplotypes of the selected 28 tag regulatory single-nucleotide polymorphisms (rSNPs) in seven linkage disequilibrium (LD) blocks. A Ly1 antibody reactive (LYAR)-binding motif disruptive rSNP rs368698783 (G/A) from LD block 5 in the proximal promoter of hemoglobin subunit gamma 1 (HBG1) was found to be a significant predictor for ß-thalassemia clinical severity by epigenetic-mediated variant-dependent HbF elevation. We found this rSNP accounted for 41.6% of ß-hemoglobinopathy individuals as an ameliorating factor in a total of 2,738 individuals from southern China and Thailand. We uncovered that the minor allele of the rSNP triggers the attenuation of LYAR and two repressive epigenetic regulators DNA methyltransferase 3 alpha (DNMT3A) and protein arginine methyltransferase 5 (PRMT5) from the HBG promoters, mediating allele-biased γ-globin elevation by facilitating demethylation of HBG core promoter CpG sites in erythroid progenitor cells from ß-thalassemia persons. The present study demonstrates that this common rSNP in the proximal Aγ-promoter is a major genetic modifier capable of ameliorating the severity of thalassemia major through the epigenetic-mediated regulation of the delayed fetal-to-adult Hb switch and provides potential targets for the treatment of ß-hemoglobinopathy.


Assuntos
Epigênese Genética , Hemoglobina Fetal/genética , Variação Genética , Talassemia beta/genética , Sequência de Bases , Células Cultivadas , Pré-Escolar , Estudos de Coortes , Feminino , Hemoglobina Fetal/metabolismo , Humanos , Lactente , Células K562 , Masculino , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Transcrição Gênica , Ativação Transcricional/genética
10.
Mol Genet Genomics ; 295(2): 505-514, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31897801

RESUMO

α-thalassemia is an inherited blood disorder commonly caused by deletions or point mutations involving one or both α-globin genes. Recent studies shed new light on the critical role of upstream enhancers multi-species conserved sequences (MCSs) in the ordered regulation of α-globin gene expression. Herein, we reported two unrelated probands with deletions in α-globin genes and MCSs, respectively. The proband from Family A is a compound heterozygote carrying a known α+ mutation (-α3.7) and a novel 60.2 kb deletion causing the absence of both α-globin genes. The proband from Family B, on the other hand, is a compound heterozygote with a known α0 mutation (--SEA) and a novel deletion involving only upstream regulatory elements MCS-R1, R2 and R3, while the α-globin genes remain intact. Notably, both these two patients suffered varied extent of anemia, indicating that the loss of enhancer elements could equally lead to reduced synthesis of α-globin. Upon these observations, we then confirmed the exact breakpoints of these two novel deletions using a targeted next-generation sequencing (NGS) previously established by our group, which may enable further elucidation of the rearrangement mechanisms on these deletions and functional dissection of MCSs. Taken together, our study reports a reliable NGS-based molecular screening approach for accurate identification of copy number variations (CNVs) in the α-globin cluster and the genetic diagnosis of these two probands may help to extend the spectrum of α-thalassemia mutations in Chinese population.


Assuntos
Elementos Alu/genética , Anemia/genética , alfa-Globinas/genética , Talassemia alfa/genética , Adulto , Anemia/sangue , Anemia/patologia , Variações do Número de Cópias de DNA/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Família Multigênica/genética , Linhagem , Mutação Puntual/genética , Deleção de Sequência/genética , Talassemia alfa/sangue , Talassemia alfa/patologia
11.
Hum Mutat ; 40(12): 2221-2229, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31286593

RESUMO

Hemoglobinopathies are the most common monogenic disorders worldwide. Substantial effort has been made to establish databases to record complete mutation spectra causing or modifying this group of diseases. We present a variant database which couples an online auxiliary diagnosis and at-risk assessment system for hemoglobinopathies (DASH). The database was integrated into the Leiden Open Variation Database (LOVD), in which we included all reported variants focusing on a Chinese population by literature peer review-curation and existing databases, such as HbVar and IthaGenes. In addition, comprehensive mutation data generated by high-throughput sequencing of 2,087 hemoglobinopathy patients and 20,222 general individuals from southern China were also incorporated into the database. These sequencing data enabled us to observe disease-causing and modifier variants responsible for hemoglobinopathies in bulk. Currently, 371 unique variants have been recorded; 265 of 371 were described as disease-causing variants, whereas 106 were defined as modifier variants, including 34 functional variants identified by a quantitative trait association study of this high-throughput sequencing data. Due to the availability of a comprehensive phenotype-genotype data set, DASH has been established to automatically provide accurate suggestions on diagnosis and genetic counseling of hemoglobinopathies. LOVD-DASH will inspire us to deal with clinical genotyping and molecular screening for other Mendelian disorders.


Assuntos
Bases de Dados Genéticas , Hemoglobinopatias/genética , Mutação , China , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Medição de Risco , Análise de Sequência de DNA
13.
Blood Cells Mol Dis ; 71: 16-22, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29475801

RESUMO

Erythroid Krüppel-like factor (EKLF/KLF1) is an erythroid-specific transcription factor whose activity is essential for erythropoiesis. The underlying mechanisms for EKLF specifically restricted to erythroid cells are of great interest but remain incompletely understood. To explore the epigenetic regulation of EKLF expression by promoter DNA methylation, we investigated the methylation status of the EKLF promoter and EKLF gene expression from a panel of human tissues. We observed that erythroid-specific hypomethylation of the EKLF promoter in adult erythroid cells was positively associated with EKLF expression. Demethylation of the EKLF promoter by 5-aza-2'-deoxycytidine led to elevated EKLF expression in non-erythroid cells. We further uncovered that EKLF promoter DNA methylation reduced the binding affinity for the transcription factors GATA1 and c-myb (MYB), which in turn silenced EKLF expression. These results suggest that hypomethylation of the EKLF promoter has functional significance in the establishment and maintenance of erythroid-specific gene expression.


Assuntos
Metilação de DNA , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Regiões Promotoras Genéticas , Sítios de Ligação , Linhagem Celular , Epigênese Genética , Células Eritroides/metabolismo , Eritropoese/genética , Fator de Transcrição GATA1/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Especificidade de Órgãos/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-myb/metabolismo
14.
J Med Genet ; 53(9): 624-33, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27247351

RESUMO

BACKGROUND: Dentin dysplasia I (DDI) is a genetically heterogeneous autosomal-dominant disorder characterised by rootless teeth with abnormal pulpal morphology, the aetiology of which presents as genetically heterogeneous. METHODS AND RESULTS: Using a cohort of a large Chinese family with 10 patients with DDI, we mapped to a 9.63 Mb candidate region for DDI on chromosome 18q21.2-q21.33. We then identified a mutation IVS7+46C>G which resulted in a novel donor splice site in intron 7 of the VPS4B gene with co-segregation of all 10 affected individuals in this family. The aberrant transcripts encompassing a new insert of 45 bp in size were detected in gingival cells from affected individuals. Protein structure prediction showed that a 15-amino acid insertion altered the ATP-binding cassette of VPS4B. The mutation resulted in significantly reduced expression of mRNA and protein and altered subcellular localisation of VPS4B, indicating a loss of function of VPS4B. Using human gingival fibroblasts, the VPS4B gene was found to act as an upstream transducer linked to Wnt/ß-catenin signalling and regulating odontogenesis. Furthermore, knockdown of vps4b in zebrafish recapitulated the reduction of tooth size and absence of teeth similar to the tooth phenotype exhibited in DDI index cases, and the zebrafish mutant phenotype could be partially rescued by wild-type human VPS4B mRNA. We also observed that vps4b depletion in the zebrafish negatively regulates the expression of some major genes involved in odontogenesis. CONCLUSIONS: This study identifies VPS4B as a disease-causing gene for DDI, which is one of the important contributors to tooth formation, through the Wnt/ß-catenin signalling pathway.


Assuntos
Adenosina Trifosfatases/genética , Displasia da Dentina/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Mutação/genética , Splicing de RNA/genética , ATPases Associadas a Diversas Atividades Celulares , Animais , Povo Asiático/genética , Sequência de Bases , Feminino , Fibroblastos/metabolismo , Humanos , Masculino , Odontogênese/genética , Linhagem , Sítios de Splice de RNA/genética , RNA Mensageiro/genética , Via de Sinalização Wnt/genética , Peixe-Zebra/genética , beta Catenina/genética
15.
Yi Chuan ; 39(3): 232-240, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28420619

RESUMO

ß-thalassemia is an autosomal recessive monogenic disease that is caused by defects in the production of ß-like globin chains. Activation of γ-globin gene and the increase in fetal hemoglobin expression have been demonstrated as one of the most important factors to ameliorate the clinical outcome of ß-thalassemia patients. In this study, 202 genes or miRNAs associated with human hemoglobin gene expression from 1802 ß-thalassemia patients were analyzed with target capture and next generation sequencing strategies in terms of functional variants that might affect hemoglobin gene expression. The subsequent bioinformatics analysis included assessments of sequence quality, the variants within the target regions and the 5'UTR with potential effects on upstream open reading frames (uORFs). Among the 41 variants in 5'UTR potentially affecting the uORFs identified in the study, two variants (chr19: 41859418 G > A and chr1:153606541 C > T) were experimentally validated with dual-luciferase assays to be capable of significantly down-regulating the expression of TGFB1 and CHTOP gene, respectively. The present study demonstrated a system suitable for evaluating the importance of variants in 5'UTRs affecting uORFs in 202 human genes associated with hemoglobin expression. Research with this approach could provide potential targets that may contribute to the clinical phenotypes and provide biomarkers for precise diagnosis of ß-thalassemia.


Assuntos
Fases de Leitura Aberta/genética , Regiões 5' não Traduzidas/genética , Biologia Computacional , Sequenciamento de Nucleotídeos em Larga Escala , Humanos
16.
Hum Genet ; 134(6): 605-12, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25800702

RESUMO

The upstream open reading frame (uORF) is a post-transcriptional regulatory element in the 5' untranslated region (5'UTR), which modulates the translation levels of main open reading frame (mORF). Earlier studies showed that disturbed uORF-mediated translation control can result in drastic changes in translation levels of mORF, leading to genetic disorders. To date, there has been no systematic investigation into the relationship between variations in patients and uORF status. Here, taking the advantage of several datasets, including gene ontology (GO) annotations and sequence feature analysis, we have examined uORF impacts in human transcripts. GO annotations indicate that uORF-containing genes are enriched in certain features such as oncogenes and transcription factors. Sequence feature analysis reveals that uORF is a factor for determination of the translation initiation site (TIS) in human transcripts. We show that genes with uORFs have lower protein expression levels than genes without uORFs in multiple human tissues. Moreover, by examining three disease variation databases, we identified uORF-altering mutations from a total of 3,740,225 variations, which are highly suspected to be associated with changed levels of gene expression. For an experimental validation, we found four mutations with significant effects on protein expression but with only modest changes in transcription levels. These findings will provide researchers on related diseases with new insights into the importance of known mutations.


Assuntos
Regiões 5' não Traduzidas/genética , Regulação da Expressão Gênica/genética , Mutação , Fases de Leitura Aberta/genética , Elementos de Resposta/genética , Linhagem Celular , Bases de Dados Genéticas , Humanos
17.
Clin Epigenetics ; 16(1): 12, 2024 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-38218889

RESUMO

The mechanism that drives the switch from fetal to adult hemoglobin (Hb) provides a therapeutic target for ß-thalassemia. We have previously identified that hypermethylation of transcription factor ERF promoter reactivated γ-globin expression. To uncover the mechanism underlying the hypermethylation of ERF promoter, we performed RNA sequencing in ß0/ß0-thalassemia patients and identified an upregulated long noncoding RNA (RP11-196G18.23) associated with HbF production. RP11-196G18.23 bound to the ERF promoter and recruited DNA methyltransferase 3A to promote DNA hypermethylation-mediated ERF downregulation, thereby ameliorating ERF-induced γ-globin inactivation. The identification of RP11-196G18.23 provides an epigenetic mechanism for the reactivation of fetal γ-globin expression for ß-hemoglobinopathies.


Assuntos
RNA Longo não Codificante , Talassemia beta , Adulto , Humanos , Talassemia beta/genética , Talassemia beta/terapia , gama-Globinas/genética , gama-Globinas/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Hemoglobina Fetal/genética , Metilação de DNA , Proteínas Repressoras/genética
18.
Heliyon ; 10(5): e26886, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38463809

RESUMO

Background: A hyperinflammatory response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection gravely worsens the clinical progression of coronavirus disease 2019 (COVID-19). Although the undesirable effects of inflammasome activation have been correlated to the severity of COVID-19, the mechanisms of this process in the asymptomatic infection and disease progression have not yet been clearly elucidated. Methods: We performed strand-specific RNA sequencing in 39 peripheral blood mononuclear cell (PBMC) samples from asymptomatic individuals(n = 10), symptomatic patients(n = 16) and healthy donors(n = 13). Results: Dysregulation of pyrin inflammasomes along with the proline-serine-threonine phosphatase-interacting protein 1 (PSTPIP1) gene was identified in SARS-COV-2 infection. Notably, the PSTPIP1 expression level showed a significant negative correlation with an adjacent long-noncoding RNA (lncRNA) RP11-797A18.6 in the asymptomatic individuals compared with the healthy controls. In addition, a decline in the nuclear factor kappa B subunit 1 (NFKB1) gene expression was observed in asymptomatic infection, followed by a rise in the mild and moderate disease stages, suggesting that altered NFKB1 expression and associated proinflammatory signals may trigger a disease progression. Conclusions: Overall, our results indicate that PSTPIP1-dependent pyrin inflammasomes-mediated pyroptosis and NF-κB activation might be potential preventive targets for COVID-19 disease development and progression.

20.
Int J Lab Hematol ; 45(1): 90-95, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36054783

RESUMO

INTRODUCTION: Though an increase in Hb A2 is one of the most key markers of ß-thal carriers, a few independent cases are reported to show elevated Hb A2 levels caused by mutations in other genes beyond ß-globin gene. METHODS: We reviewed the haematological indices of 47336 individuals to analyse the phenotype-genotype correlation and identified 1439 individuals (3.04%) positive in the elevation of Hb A2 . Globin and KLF1 genes analysis was performed, and further whole-exome sequencing was carried to dissect the genetic causes of those positive samples without ß-thalassemic or KLF1 mutations. RESULTS: Of these 1439 individuals with elevated Hb A2 , 1381 had a molecular defect in globin genes, and most were ß-thalassemic mutation; 10 had a molecular defect in KLF1 gene. Finally, among the 38 individuals without ß-thalassemic or KLF1 mutations, 7 were identified to carried a loss-of-function mutation in SUPT5H. CONCLUSION: This study has provided a mutation spectrum of SUPT5H in a cohort screening leading to the elevation of Hb A2 . According to the previous observations that individuals with a combination of ß-thal mutation and a SUPT5H variant might present moderate ß-thaelassemia, these findings emphasized the importance of comprehensive molecular diagnosis to prevent birth defects of ß-thaelassemia caused by rare mutations from modifier genes.


Assuntos
Hemoglobina A2 , Talassemia beta , Humanos , Sequenciamento do Exoma , Hemoglobina A2/genética , Hemoglobina A2/análise , Mutação , Heterozigoto , Globinas/genética , Talassemia beta/diagnóstico , Talassemia beta/genética , Genótipo , Proteínas Nucleares/genética , Fatores de Elongação da Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA