Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 33(19): 1711-1725, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39045627

RESUMEN

Human cell line models, including the neuronal precursor line LUHMES, are important for investigating developmental transcriptional dynamics within imprinted regions, particularly the 15q11-q13 Angelman (AS) and Prader-Willi (PWS) syndrome locus. AS results from loss of maternal UBE3A in neurons, where the paternal allele is silenced by a convergent antisense transcript UBE3A-ATS, a lncRNA that terminates at PWAR1 in non-neurons. qRT-PCR analysis confirmed the exclusive and progressive increase in UBE3A-ATS in differentiating LUHMES neurons, validating their use for studying UBE3A silencing. Genome-wide transcriptome analyses revealed changes to 11 834 genes during neuronal differentiation, including the upregulation of most genes within the 15q11-q13 locus. To identify dynamic changes in chromatin loops linked to transcriptional activity, we performed a HiChIP validated by 4C, which identified two neuron-specific CTCF loops between MAGEL2-SNRPN and PWAR1-UBE3A. To determine if allele-specific differentially methylated regions (DMR) may be associated with CTCF loop anchors, whole genome long-read nanopore sequencing was performed. We identified a paternally hypomethylated DMR near the SNRPN upstream loop anchor exclusive to neurons and a paternally hypermethylated DMR near the PWAR1 CTCF anchor exclusive to undifferentiated cells, consistent with increases in neuronal transcription. Additionally, DMRs near CTCF loop anchors were observed in both cell types, indicative of allele-specific differences in chromatin loops regulating imprinted transcription. These results provide an integrated view of the 15q11-q13 epigenetic landscape during LUHMES neuronal differentiation, underscoring the complex interplay of transcription, chromatin looping, and DNA methylation. They also provide insights for future therapeutic approaches for AS and PWS.


Asunto(s)
Factor de Unión a CCCTC , Diferenciación Celular , Cromosomas Humanos Par 15 , Metilación de ADN , Impresión Genómica , Neuronas , Transcriptoma , Ubiquitina-Proteína Ligasas , Humanos , Impresión Genómica/genética , Factor de Unión a CCCTC/metabolismo , Factor de Unión a CCCTC/genética , Cromosomas Humanos Par 15/genética , Neuronas/metabolismo , Metilación de ADN/genética , Transcriptoma/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Diferenciación Celular/genética , Síndrome de Angelman/genética , Síndrome de Angelman/patología , ARN Largo no Codificante/genética , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/patología , Síndrome de Prader-Willi/metabolismo , Proteínas Nucleares snRNP/genética , Proteínas Nucleares snRNP/metabolismo , Alelos , Línea Celular , Epigenoma
2.
Dev Dyn ; 252(2): 276-293, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36058892

RESUMEN

BACKGROUND: Although splicing is an integral part of the expression of many genes in our body, genetic syndromes with spliceosomal defects affect only specific tissues. To help understand the mechanism, we investigated the expression pattern of a core protein of the major spliceosome, SmB/B' (Small Nuclear Ribonucleoprotein Polypeptides B/B'), which is encoded by SNRPB. Loss-of-function mutations of SNRPB in humans cause cerebro-costo-mandibular syndrome (CCMS) characterized by rib gaps, micrognathia, cleft palate, and scoliosis. Our expression analysis focused on the affected structures as well as non-affected tissues, using chick and mouse embryos as model animals. RESULTS: Embryos at young stages (gastrula) showed ubiquitous expression of SmB/B'. However, the level and pattern of expression became tissue-specific as differentiation proceeded. The regions relating to CCMS phenotypes such as cartilages of ribs and vertebrae and palatal mesenchyme express SmB/B' in the nucleus sporadically. However, cartilages that are not affected in CCMS also showed similar expressions. Another spliceosomal gene, SNRNP200, which mutations cause retinitis pigmentosa, was also prominently expressed in cartilages in addition to the retina. CONCLUSION: The expression of SmB/B' is spatiotemporally regulated during embryogenesis despite the ubiquitous requirement of the spliceosome, however, the expression pattern is not strictly correlated with the phenotype presentation.


Asunto(s)
Discapacidad Intelectual , Empalmosomas , Humanos , Animales , Ratones , Empalmosomas/genética , Proteínas Nucleares snRNP/genética , Ribonucleoproteínas Nucleares Pequeñas , Discapacidad Intelectual/genética
3.
Am J Med Genet A ; 191(5): 1425-1429, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36814386

RESUMEN

Variants in genes encoding core components of the spliceosomes are associated with craniofacial syndromes, collectively called craniofacial spliceosomopathies. SNRPE encodes a core component of pre-mRNA processing U-rich small nuclear ribonuclear proteins (UsnRNPs). Heterozygous variants in SNRPE have been reported in six families with isolated hypotrichosis simplex in addition to one case of isolated non syndromic congenital microcephaly. Here, we report a patient with a novel blended phenotype of microcephaly and congenital atrichia with multiple congenital anomalies due to a de novo intronic SNRPE deletion, c.82-28_82-16del, which results in exon skipping. As discussed within, this phenotype, which we propose be named SNRPE-related syndromic microcephaly and hypotrichosis, overlaps other craniofacial splicesosomopathies.


Asunto(s)
Anomalías Múltiples , Hipotricosis , Microcefalia , Humanos , Microcefalia/diagnóstico , Microcefalia/genética , Microcefalia/complicaciones , Fenotipo , Alopecia/complicaciones , Hipotricosis/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Proteínas Nucleares snRNP/genética
4.
J Med Genet ; 59(7): 719-722, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099539

RESUMEN

BACKGROUND: Prader-Willi syndrome (PWS) is an imprinting disorder caused by the absence of paternal expressed genes in the Prader-Willi critical region (PWCR) on chromosome 15q11.2-q13. Three molecular mechanisms have been known to cause PWS, including a deletion in the PWCR, uniparental disomy 15 and imprinting defects. RESULTS: We report the first case of PWS associated with a single-nucleotide SNRPN variant in a 10-year-old girl presenting with clinical features consistent with PWS, including infantile hypotonia and feeding difficulty, developmental delay with cognitive impairment, excessive eating with central obesity, sleep disturbances, skin picking and related behaviour issues. Whole-exome sequencing revealed a de novo mosaic nonsense variant of the SNRPN gene (c.73C>T, p.R25X) in 10% of DNA isolated from buccal cells and 19% of DNA from patient-derived lymphoblast cells. DNA methylation study did not detect an abnormal methylation pattern in the SNRPN locus. Parental origin studies showed a paternal source of an intronic single-nucleotide polymorphism within the locus in proximity to the SNRPN variant. CONCLUSIONS: This is the first report that provides evidence of a de novo point mutation of paternal origin in SNRPN as a new disease-causing mechanism for PWS. This finding suggests that gene sequencing should be considered as part of the diagnostic workup in patients with clinical suspicion of PWS.


Asunto(s)
Síndrome de Prader-Willi , Niño , Femenino , Humanos , Cromosomas Humanos Par 15/genética , ADN , Metilación de ADN/genética , Impresión Genómica , Mucosa Bucal , Síndrome de Prader-Willi/diagnóstico , Síndrome de Prader-Willi/genética , Proteínas Nucleares snRNP/genética , Polimorfismo de Nucleótido Simple
5.
Hum Mutat ; 43(11): 1567-1575, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35842787

RESUMEN

Prader-Willi syndrome (PWS; MIM# 176270) is a neurodevelopmental disorder caused by the loss of expression of paternally imprinted genes within the PWS region located on 15q11.2. It is usually caused by either maternal uniparental disomy of chromosome 15 (UPD15) or 15q11.2 recurrent deletion(s). Here, we report a healthy carrier of a balanced X;15 translocation and her two daughters, both with the karyotype 45,X,der(X)t(X;15)(p22;q11.2),-15. Both daughters display symptoms consistent with haploinsufficiency of the SHOX gene and PWS. We explored the architecture of the derivative chromosomes and investigated effects on gene expression in patient-derived neural cells. First, a multiplex ligation-dependent probe amplification methylation assay was used to determine the methylation status of the PWS-region revealing maternal UPD15 in daughter 2, explaining her clinical symptoms. Next, short read whole genome sequencing and 10X genomics linked read sequencing was used to pinpoint the exact breakpoints of the translocation. Finally, we performed transcriptome sequencing on neuroepithelial stem cells from the mother and from daughter 1 and observed biallelic expression of genes in the PWS region (including SNRPN) in daughter 1. In summary, our multi-omics analysis highlights two different PWS mechanisms in one family and provide an example of how structural variation can affect imprinting through long-range interactions.


Asunto(s)
Metilación de ADN , Síndrome de Prader-Willi , Cromosomas Humanos Par 15/genética , Femenino , Impresión Genómica , Humanos , Síndrome de Prader-Willi/genética , Translocación Genética , Disomía Uniparental/genética , Proteínas Nucleares snRNP/genética
6.
BMC Genomics ; 23(1): 744, 2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36348279

RESUMEN

BACKGROUND: Alternative splicing (AS) is an important channel for gene expression regulation and protein diversification, in addition to a major reason for the considerable differences in the number of genes and proteins in eukaryotes. In plants, U2 small nuclear ribonucleoprotein B″ (U2B″), a component of splicing complex U2 snRNP, plays an important role in AS. Currently, few studies have investigated plant U2B″, and its mechanism remains unclear. RESULT: Phylogenetic analysis, including gene and protein structures, revealed that U2B″ is highly conserved in plants and typically contains two RNA recognition motifs. Subcellular localisation showed that OsU2B″ is located in the nucleus and cytoplasm, indicating that it has broad functions throughout the cell. Elemental analysis of the promoter region showed that it responded to numerous external stimuli, including hormones, stress, and light. Subsequent qPCR experiments examining response to stress (cold, salt, drought, and heavy metal cadmium) corroborated the findings. The prediction results of protein-protein interactions showed that its function is largely through a single pathway, mainly through interaction with snRNP proteins. CONCLUSION: U2B″ is highly conserved in the plant kingdom, functions in the nucleus and cytoplasm, and participates in a wide range of processes in plant growth and development.


Asunto(s)
Ribonucleoproteína Nuclear Pequeña U2 , Empalmosomas , Proteínas Nucleares snRNP/genética , Ribonucleoproteína Nuclear Pequeña U2/química , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Filogenia , Secuencia de Aminoácidos , ARN Nuclear Pequeño/genética , Empalme del ARN
7.
Ann Hum Genet ; 86(2): 71-79, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34779508

RESUMEN

Angelman syndrome (AS) (OMIM#105830) is an imprinting disorder caused due to alterations in the maternal chr 15q11-13 region. Majority of cases can be diagnosed by methylation-specific polymerase chain reaction (MS-PCR) of SNRPN gene and by UBE3A sequencing, however, about 10% of cases with AS phenotype remain undiagnosed. Differential diagnoses of AS can be detected by chromosomal microarray (CMA) and clinical exome sequencing (CES). In this study, 30 cases with AS features were evaluated by MS-PCR, CMA, and CES. SNRPN MS-PCR confirmed AS in eight (26%), CMA and CES diagnosed nine (30%) cases. One case was identified with a novel variant c.1125C > T in GABRG3, located at 15q12 region, which is currently not associated with any syndrome. The GABRG3 gene is also speculated to be imprinted, a MS-PCR assay was designed to confirm its differential parental methylation status. This assay identified another case with altered GABRG3 methylation. The two cases with GABRG3 alteration-sequence change and methylation indicate that GABRG3 may be associated with a subtype of AS or a new related syndrome. Performing GABRG3 MS-PCR and sequencing of a larger group of patients with AS phenotype and normal SNPRN and UBE3A status will help in establishing exact genotype-phenotype correlation.


Asunto(s)
Síndrome de Angelman , Receptores de GABA-A , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Metilación de ADN , Impresión Genómica , Humanos , Fenotipo , Receptores de GABA-A/genética , Proteínas Nucleares snRNP/genética
8.
Mol Reprod Dev ; 89(7): 290-297, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35698757

RESUMEN

Imprinted genes are inherited with different DNA methylation patterns depending on the maternal or paternal origin of the allele. In cattle (Bos taurus), abnormal methylation of these genes is linked to the large offspring syndrome, a neonatal overgrowth phenotype analogous to the human Beckwith-Wiedemann syndrome. We hypothesized that in bovine oocytes, some of the methylation patterns on maternally imprinted genes are acquired in the last phase of folliculogenesis. The pyrosequencing analysis of IGF2R, KCNQ1, PLAGL1, and SNRPN imprinted genes showed no clear progression of methylation in oocytes from follicles 1-2 mm (late pre antral/early antral) and up. Instead, these oocytes displayed complete methylation at the imprinted differentially methylated regions (>80%). Other mechanisms related to imprint maintenance should be investigated to explain the hypomethylation at IGF2R, KCNQ1, PLAGL1, and SNRPN maternally imprinted sites observed in some bovine embryos.


Asunto(s)
Metilación de ADN , Impresión Genómica , Animales , Bovinos , Proteínas de Ciclo Celular , Humanos , Canal de Potasio KCNQ1/genética , Oogénesis , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Nucleares snRNP/genética
9.
Nucleic Acids Res ; 48(2): 895-911, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31799625

RESUMEN

The assembly of snRNP cores, in which seven Sm proteins, D1/D2/F/E/G/D3/B, form a ring around the nonameric Sm site of snRNAs, is the early step of spliceosome formation and essential to eukaryotes. It is mediated by the PMRT5 and SMN complexes sequentially in vivo. SMN deficiency causes neurodegenerative disease spinal muscular atrophy (SMA). How the SMN complex assembles snRNP cores is largely unknown, especially how the SMN complex achieves high RNA assembly specificity and how it is released. Here we show, using crystallographic and biochemical approaches, that Gemin2 of the SMN complex enhances RNA specificity of SmD1/D2/F/E/G via a negative cooperativity between Gemin2 and RNA in binding SmD1/D2/F/E/G. Gemin2, independent of its N-tail, constrains the horseshoe-shaped SmD1/D2/F/E/G from outside in a physiologically relevant, narrow state, enabling high RNA specificity. Moreover, the assembly of RNAs inside widens SmD1/D2/F/E/G, causes the release of Gemin2/SMN allosterically and allows SmD3/B to join. The assembly of SmD3/B further facilitates the release of Gemin2/SMN. This is the first to show negative cooperativity in snRNP assembly, which provides insights into RNA selection and the SMN complex's release. These findings reveal a basic mechanism of snRNP core assembly and facilitate pathogenesis studies of SMA.


Asunto(s)
Proteínas del Tejido Nervioso/química , Proteínas de Unión al ARN/química , ARN/química , Empalmosomas/química , Proteínas Nucleares snRNP/química , Cristalografía por Rayos X , Humanos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Proteínas del Tejido Nervioso/genética , Conformación Proteica , ARN/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteínas Nucleares Pequeñas/química , Ribonucleoproteínas Nucleares Pequeñas/genética , Proteínas del Complejo SMN/química , Proteínas del Complejo SMN/genética , Empalmosomas/genética , Proteínas Nucleares snRNP/genética
10.
PLoS Genet ; 15(10): e1008460, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31671093

RESUMEN

Malfunction of pre-mRNA processing factors are linked to several human diseases including cancer and neurodegeneration. Here we report the identification of a de novo heterozygous missense mutation in the SNRPE gene (c.65T>C (p.Phe22Ser)) in a patient with non-syndromal primary (congenital) microcephaly and intellectual disability. SNRPE encodes SmE, a basal component of pre-mRNA processing U snRNPs. We show that the microcephaly-linked SmE variant is unable to interact with the SMN complex and as a consequence fails to assemble into U snRNPs. This results in widespread mRNA splicing alterations in fibroblast cells derived from this patient. Similar alterations were observed in HEK293 cells upon SmE depletion that could be rescued by the expression of wild type but not mutant SmE. Importantly, the depletion of SmE in zebrafish causes aberrant mRNA splicing alterations and reduced brain size, reminiscent of the patient microcephaly phenotype. We identify the EMX2 mRNA, which encodes a protein required for proper brain development, as a major mis-spliced down stream target. Together, our study links defects in the SNRPE gene to microcephaly and suggests that alterations of cellular splicing of specific mRNAs such as EMX2 results in the neurological phenotype of the disease.


Asunto(s)
Empalme Alternativo , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Mutación Missense , Factores de Transcripción/genética , Proteínas Nucleares snRNP/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Linaje , Empalme del ARN , ARN Mensajero/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Secuenciación del Exoma , Pez Cebra , Proteínas Nucleares snRNP/química , Proteínas Nucleares snRNP/metabolismo
11.
Zygote ; 30(5): 638-647, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35603594

RESUMEN

High rates of infertility in type 2 diabetic (T2DM) men have led to attempts to understand the mechanisms involved in this process. This condition can be investigated from at least two aspects, namely sperm quality indices and epigenetic alterations. Epigenetics science encompasses the phenomena that can lead to inherited changes independently of the genetics. This study has been performed to test the hypothesis of the relationship between T2DM and the epigenetic profile of the sperm, as well as sperm quality indices. This research included 42 individuals referred to the infertility clinic of Royan Institute, Iran in 2019-2021. The study subjects were assigned to three groups: normozoospermic non-diabetic (control), normozoospermic diabetic (DN) and non-normozoospermic diabetic (D.Non-N). Sperm DNA fragmentation was evaluated using the sperm chromatin structure assay technique. The global methylation level was examined using 5-methyl cytosine antibody and the methylation status in differentially methylated regions of H19, MEST, and SNRPN was assessed using the methylation-sensitive high-resolution melting technique. The results showed that the sperm global methylation in spermatozoa of D.Non-N group was significantly reduced compared with the other two groups (P < 0.05). The MEST and H19 genes were hypomethylated in the spermatozoa of D.Non-N individuals, but the difference level was not significant for MEST. The SNRPN gene was significantly hypermethylated in these individuals (P < 0.05). The results of this study suggest that T2DM alters the methylation profile and epigenetic programming in spermatozoa of humans and that these methylation changes may ultimately influence the fertility status of men with diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Impresión Genómica , Cromatina/metabolismo , Citosina/metabolismo , Metilación de ADN , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Masculino , Semen/metabolismo , Espermatozoides/metabolismo , Proteínas Nucleares snRNP/genética , Proteínas Nucleares snRNP/metabolismo
12.
Hum Mol Genet ; 28(2): 220-229, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30260400

RESUMEN

Angelman syndrome, Prader-Will syndrome and Dup15q syndrome map to a cluster of imprinted genes located at 15q11-q13. Imprinting at this domain is regulated by an imprinting control region consisting of two distinct elements, the Angelman syndrome imprinting center (AS-IC) and the Prader-Willi syndrome imprinting center (PWS-IC). Individuals inheriting deletions of the AS-IC exhibit reduced expression of the maternally expressed UBE3A gene and biallelic expression of paternal-only genes. We have previously demonstrated that AS-IC activity partly consists of providing transcription across the PWS-IC in oocytes, and that these transcripts are necessary for maternal imprinting of Snrpn. Here we report a novel mouse mutation that truncates transcripts prior to transiting the PWS-IC and results in a domain-wide imprinting defect. These results confirm a transcription-based model for imprint setting at this domain. The imprinting defect can be preempted by removal of the transcriptional block in oocytes, but not by its removal in early embryos. Imprinting defect mice exhibit several traits often found in individuals with Angelman syndrome imprinting defects.


Asunto(s)
Síndrome de Angelman/genética , Modelos Animales de Enfermedad , Impresión Genómica , Animales , Metilación de ADN , Exones , Femenino , Regulación de la Expresión Génica , Masculino , Herencia Materna , Ratones , Mutación , Oocitos/metabolismo , Proteínas Nucleares snRNP/genética
13.
J Assist Reprod Genet ; 38(8): 2041-2048, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33786731

RESUMEN

PURPOSE: To evaluate the association between the DNA methylation of specific genes and sperm DNA integrity status in human sperm samples. METHODS: A total of 166 semen samples were evaluated (86 controls and 80 cases with impaired sperm DNA integrity). We detected the methylation status of 257 CpG sites among two imprinted genes (H19 and SNRPN) and four non-imprinted genes related to male infertility (MTHFR, GSTM1, DAZL, and CREM) by using a targeted next-generation sequencing method. RESULTS: Differential methylation was found in 43 CpG sites of the promoters of the six candidate genes. H19, SNRPN, MTHFR, DAZL, GSTM1, and CREM contained 22, 12, 1, 4, 0, and 4 differentially methylated CpG sites (P<0.05), respectively. The imprinting genes were associated with relatively higher rates of differentially methylated CpG sites (28.21% in H19 and 41.38% in SNRPN) than the non-imprinting genes. One CpG site in H19 remained significant after performing strict Bonferroni correction. CONCLUSION: In this study, we found that different site-specific DNA methylation signatures were correlated with sperm DNA integrity status. Further studies are needed to investigate the specific mechanisms leading to the epigenetic modifications.


Asunto(s)
Metilación de ADN , ADN/química , Epigénesis Genética , Impresión Genómica , Infertilidad Masculina/patología , ARN Largo no Codificante/genética , Proteínas Nucleares snRNP/genética , Adulto , Estudios de Casos y Controles , Modulador del Elemento de Respuesta al AMP Cíclico/genética , ADN/genética , Glutatión Transferasa/genética , Humanos , Infertilidad Masculina/genética , Masculino , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Proteínas de Unión al ARN/genética , Espermatozoides/metabolismo
14.
J Cell Mol Med ; 24(17): 9898-9907, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32693431

RESUMEN

Congenital heart disease (CHD) with extracardiac malformations (EM) is the most common multiple malformation, resulting from the interaction between genetic abnormalities and environmental factors. Most studies have attributed the causes of CHD with EM to chromosomal abnormalities. However, multi-system dysplasia is usually caused by both genetic mutations and epigenetic dysregulation. The epigenetic mechanisms underlying the pathogenesis of CHD with EM remain unclear. In this study, we investigated the mechanisms of imprinting alterations, including those of the Small nuclear ribonucleoprotein polypeptide N (SNRPN), PLAG1 like zinc finger 1 (ZAC1) and inositol polyphosphate-5-phosphatase F (INPP5F) genes, in the pathogenesis of CHD with EM. The methylation levels of SNRPN, ZAC1, and INPP5F genes were analysed by the MassARRAY platform in 24 children with CHD with EM and 20 healthy controls. The expression levels of these genes were detected by real-time polymerase chain reaction (PCR). The correlation between methylation regulation and gene expression was confirmed using 5-azacytidine (5-Aza) treated cells. The methylation levels of SNRPN and ZAC1 genes were significantly increased in CHD with EM, while that of INPP5F was decreased. The methylation alterations of these genes were negatively correlated with expression. Risk analysis showed that abnormal hypermethylation of SNRPN and ZAC1 resulted in 5.545 and 7.438 times higher risks of CHD with EM, respectively, and the abnormal hypomethylation of INPP5F was 8.38 times higher than that of the control group. We concluded that abnormally high methylation levels of SNRPN and ZAC1 and decreased levels of INPP5F imply an increased risk of CHD with EM by altering their gene functions. This study provides evidence of imprinted regulation in the pathogenesis of multiple malformations.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cardiopatías Congénitas/genética , Inositol Polifosfato 5-Fosfatasas/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Nucleares snRNP/genética , Azacitidina/química , Niño , Preescolar , Metilación de ADN/genética , Epigénesis Genética/genética , Femenino , Expresión Génica/genética , Impresión Genómica/genética , Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/patología , Humanos , Lactante , Masculino
15.
J Anat ; 236(5): 931-945, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31884688

RESUMEN

Ribs are seldom affected by developmental disorders, however, multiple defects in rib structure are observed in the spliceosomal disease cerebro-costo-mandibular syndrome (CCMS). These defects include rib gaps, found in the posterior part of the costal shaft in multiple ribs, as well as missing ribs, shortened ribs and abnormal costotransverse articulations, which result in inadequate ventilation at birth and high perinatal mortality. The genetic mechanism of CCMS is a loss-of-function mutation in SNRPB, a component of the major spliceosome, and knockdown of this gene in vitro affects the activity of the Wnt/ß-catenin and bone morphogenic protein (BMP) pathways. The aim of the present study was to investigate whether altering these pathways in vivo can recapitulate rib gaps and other rib abnormalities in the model animal. Chick embryos were implanted with beads soaked in Wnt/ß-catenin and BMP pathway modulators during somitogenesis, and incubated until the ribs were formed. Some embryos were harvested in the preceding days for analysis of the chondrogenic marker Sox9, to determine whether pathway modulation affected somite patterning or chondrogenesis. Wnt/ß-catenin inhibition manifested characteristic rib phenotypes seen in CCMS, including rib gaps (P < 0.05) and missing ribs (P < 0.05). BMP pathway activation did not cause rib gaps but yielded missing rib (P < 0.01) and shortened rib phenotypes (P < 0.05). A strong association with vertebral phenotypes was also noted with BMP4 (P < 0.001), including scoliosis (P < 0.05), a feature associated with CCMS. Reduced expression of Sox9 was detected with Wnt/ß-catenin inhibition, indicating that inhibition of chondrogenesis precipitated the rib defects in the presence of Wnt/ß-catenin inhibitors. BMP pathway activators also reduced Sox9 expression, indicating an interruption of somite patterning in the manifestation of rib defects with BMP4. The present study demonstrates that local inhibition of the Wnt/ß-catenin and activation of the BMP pathway can recapitulate rib defects, such as those observed in CCMS. The balance of Wnt/ß-catenin and BMP in the somite is vital for correct rib morphogenesis, and alteration of the activity of these two pathways in CCMS may perturb this balance during somite patterning, leading to the observed rib defects.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Discapacidad Intelectual/genética , Micrognatismo/genética , Costillas/anomalías , Factor de Transcripción SOX9/genética , Vía de Señalización Wnt/fisiología , Proteínas Nucleares snRNP/genética , Animales , Embrión de Pollo , Condrogénesis/genética , Mutación , Transducción de Señal/fisiología
16.
Mol Cell ; 45(6): 814-25, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22387027

RESUMEN

CpG islands (CGIs) function as promoters for approximately 60% of human genes. Most of these elements remain protected from CpG methylation, a prevalent epigenetic modification associated with transcriptional silencing. Here, we report that methylation-resistant CGI promoters are characterized by significant strand asymmetry in the distribution of guanines and cytosines (GC skew) immediately downstream from their transcription start sites. Using innovative genomics methodologies, we show that transcription through regions of GC skew leads to the formation of long R loop structures. Furthermore, we show that GC skew and R loop formation potential is correlated with and predictive of the unmethylated state of CGIs. Finally, we provide evidence that R loop formation protects from DNMT3B1, the primary de novo DNA methyltransferase in early development. Altogether, these results suggest that protection from DNA methylation is a built-in characteristic of the DNA sequence of CGI promoters that is revealed by the cotranscriptional formation of R loop structures.


Asunto(s)
Islas de CpG , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Animales , Apolipoproteínas E/genética , Citosina , Metilación de ADN , Epigénesis Genética , Genoma Humano , Guanina , Humanos , Ratones , Plásmidos/genética , Sitio de Iniciación de la Transcripción , Proteínas Nucleares snRNP/genética
17.
J Assist Reprod Genet ; 37(9): 2211-2221, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32572674

RESUMEN

PURPOSE: Changes in DNA methylation modifications have been associated with male infertility. With the development of assisted reproductive technologies (ARTs), abnormal DNA methylation in sperm, especially in imprinted genes, may impact the health of offspring and requires an in-depth study. METHODS: In this study, we collected abnormal human semen samples, including asthenospermic, oligospermic, oligoasthenospermic and deformed sperm, and investigated the methylation of imprinted genes by reduced representation bisulfite sequencing (RRBS) and bisulfite amplicon sequencing on the Illumina platform. RESULTS: The differentially methylated regions (DMRs) of imprinted genes, including H19, GNAS, MEG8 and SNRPN, were different in the abnormal semen groups. MEG8 DMR methylation in the asthenospermic group was significantly increased. Furthermore, higher methylation levels of MEG8, GNAS and SNRPN DMR in the oligospermic and oligoasthenospermic groups and a decrease in the H19 DMR methylation level in the oligospermic group were observed. However, the methylation levels of these regions varied greatly among the different semen samples and among individual sperm within the same semen sample. The SNP rs2525883 genotype in the H19 DMR affected DNA methylation. Moreover, DNA methylation levels differed in the abnormal semen groups in the non-imprinted genomic regions, including repetitive sequence DNA transposons and long/short interspersed nuclear elements (LINEs and SINEs). CONCLUSION: Our study established that imprinted gene DMRs, such as H19, GNAS, SNRPN and MEG8, were differentially methylated in the abnormal semen groups with obvious inter- and intra-sample heterogeneities. These results suggest that special attention needs to be paid to possible epigenetic risks during reproduction.


Asunto(s)
Astenozoospermia/genética , Metilación de ADN/genética , Impresión Genómica/genética , Infertilidad Masculina/genética , Adulto , Astenozoospermia/patología , Cromograninas/genética , Epigenómica , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Infertilidad Masculina/patología , Masculino , Persona de Mediana Edad , ARN Largo no Codificante/genética , Semen/metabolismo , Espermatozoides/metabolismo , Espermatozoides/patología , Adulto Joven , Proteínas Nucleares snRNP/genética
18.
Int J Mol Sci ; 21(12)2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32545483

RESUMEN

The core spliceosomal Sm proteins were recently proposed as cancer-selective lethal targets in non-small cell lung cancer (NSCLC). In contrast, the loss of the commonly mutated cancer target SF3B1 appeared to be toxic to non-malignant cells as well. In the current study, the transcriptomes of A549 NSCLC cells, in which SF3B1 or SNRPD3 was silenced, were compared using RNA sequencing. The skipping of exon 4 of the proteasomal subunit beta type-3 (PSMB3) mRNA, resulting in a shorter PSMB3-S variant, occurred only after silencing SNRPD3. This observation was extended to the other six Sm genes. Remarkably, the alternative splicing of PSMB3 mRNA upon Sm gene silencing was not observed in non-malignant IMR-90 lung fibroblasts. Furthermore, PSMB3 was found to be overexpressed in NSCLC clinical samples and PSMB3 expression correlated with Sm gene expression. Moreover, a high PSMB3 expression corresponds to worse survival in patients with lung adenocarcinomas. Finally, silencing the canonical full-length PSMB3-L, but not the shorter PSMB3-S variant, was cytotoxic and was accompanied by a decrease in proteasomal activity. Together, silencing Sm genes, but not SF3B1, causes a cytotoxic alternative splicing switch in the PSMB3 mRNA in NSCLC cells only.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Perfilación de la Expresión Génica/métodos , Neoplasias Pulmonares/genética , Fosfoproteínas/genética , Complejo de la Endopetidasa Proteasomal/genética , Factores de Empalme de ARN/genética , Proteínas Nucleares snRNP/genética , Células A549 , Empalme Alternativo , Regulación hacia Abajo , Exones , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Degradación de ARNm Mediada por Codón sin Sentido , Análisis de Secuencia de ARN
19.
Zhonghua Nan Ke Xue ; 26(2): 154-159, 2020 Feb.
Artículo en Zh | MEDLINE | ID: mdl-33346420

RESUMEN

OBJECTIVE: To investigate the significance of cytogenetic and molecular genetic diagnosis of a special type of secondary sexual dysplasia and the applicability of various methods for its detection. METHODS: Using karyotype analysis, array comparative genomic hybridization (aCGH), multiplex ligation-dependent probe amplification (MLPA) and methylation-specific PCR (MS-PCR), we diagnosed and differentially diagnosed a case of secondary sexual dysplasia. RESULTS: Abnormalities were not found in the karyotype analysis or the SRY and AZF gene detection, nor chromosomal duplication and deletion in the initial SurePrint G3 Human CGH Array Kit8×60K.SurePrint G3 unrestricteda CGH ISCA v2,88×60K, however, identified a 68.9 kb deletion of chromosome 15 (hg19:25190737-25259677). MLPA revealed the deletion of exon 3 of the SNRPN gene. MS-PCR showed a significant decrease in the paternal fragment signals, but no difference in the maternal fragment signals between the sample from the patient and that from the control. CONCLUSIONS: The patient was confirmed with Prader-Willi syndrome by various methods of detection.


Asunto(s)
Síndrome de Prader-Willi , Hibridación Genómica Comparativa , Metilación de ADN , Exones , Humanos , Cariotipificación , Síndrome de Prader-Willi/diagnóstico , Síndrome de Prader-Willi/genética , Eliminación de Secuencia , Proteínas Nucleares snRNP/genética
20.
RNA Biol ; 16(8): 1001-1009, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31107149

RESUMEN

In contrast to cytoplasmic localization of spliced mRNAs, many spliced lncRNAs are localized in the nucleus. To investigate the mechanism, we used lncRNA MEG3 as a reporter and mapped a potent nuclear retention element (NRE), deletion of this element led to striking export of MEG3 from the nucleus to the cytoplasm. Insertion of the NRE resulted in nuclear retention of spliced lncRNA as well as spliced mRNA. We further purified RNP assembled on the NRE in vitro and identified the proteins by mass spectrometry. Screen using siRNA revealed depletion of U1 snRNP components SNRPA, SNRNP70 or SNRPD2 caused significant cytoplasmic localization of MEG3 reporter transcripts. Co-knockdown these factors in HFF1 cells resulted in an increased cytoplasmic distribution of endogenous lncRNAs. Together, these data support a model that U1 snRNP components restrain spliced lncRNAs in the nucleus via the interaction with nuclear retention element.


Asunto(s)
Ribonucleoproteína Nuclear Pequeña U1/genética , Proteínas Nucleares snRNP/genética , Núcleo Celular/genética , Citoplasma/genética , Citosol/metabolismo , Células HeLa , Humanos , Empalme del ARN/genética , ARN Largo no Codificante/genética , Ribonucleoproteína Nuclear Pequeña U1/química , Empalmosomas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA