Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Intervalo de año de publicación
1.
Biomolecules ; 14(8)2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39199324

RESUMEN

Genomic imprinting refers to the epigenetic silencing of one of both alleles in a parent-of-origin-specific manner, particularly in genes regulating growth and development. Impaired genomic imprinting leading to the activation of the silenced allele, also called canonical loss-of-imprinting (LOI), is considered an early factor in oncogenesis. As LOI studies in clear cell renal cell carcinoma (ccRCC) are limited to IGF2, we performed a genome-wide analysis in 128 kidney normal solid tissue and 240 stage 1 ccRCC samples (TCGA RNA-seq data) to screen for canonical LOI in early oncogenesis. In ccRCC, we observed LOI (adj. p = 2.74 × 10-3) of HM13 (Histocompatibility Minor 13), a signal peptide peptidase involved in epitope generation. HM13 LOI samples featured HM13 overexpression, both compared to normal solid tissues (p = 3.00 × 10-7) and non-LOI (p = 1.27 × 10-2) samples. Upon adjustment for age and sex, HM13 expression was significantly associated with poor survival (p = 7.10 × 10-5). Moreover, HM13 overexpression consistently exacerbated with increasing tumor stage (p = 2.90 × 10-8). For IGF2, LOI was observed in normal solid tissues, but the prevalence did not increase in cancer. In conclusion, HM13 LOI is an early event in ccRCC, causing overexpression leading to poor prognosis.


Asunto(s)
Carcinoma de Células Renales , Impresión Genómica , Neoplasias Renales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/mortalidad , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Neoplasias Renales/metabolismo , Neoplasias Renales/mortalidad , Femenino , Masculino , Pronóstico , Persona de Mediana Edad , Regulación Neoplásica de la Expresión Génica , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Anciano
2.
Front Med (Lausanne) ; 10: 1172565, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37575996

RESUMEN

Prader-Willi syndrome (PWS) is a rare disease caused by a lack of expression of inherited imprinted genes in the paternally derived Prader-Willi critical region on chromosome 15q11.2-q13. It is characterized by poor feeding and hypotonia in infancy, intellectual disability, behavioral abnormalities, dysmorphic features, short stature, obesity, and hypogonadism. PWS is not a known cancer predisposition syndrome, but previous investigations regarding the prevalence of cancer in these patients suggest an increased risk of developing specific cancer types such as myeloid leukemia and testicular cancer. We present the results from a Swedish national population-based cohort study of 360 individuals with PWS and 18,000 matched comparisons. The overall frequency of cancer was not increased in our PWS cohort, but we found a high frequency of pediatric cancers. We also performed whole-genome sequencing of blood- and tumor-derived DNAs from a unilateral dysgerminoma in a 13-year-old girl with PWS who also developed bilateral ovarian sex cord tumors with annular tubules. In germline analysis, there were no additional findings apart from the 15q11.2-q13 deletion of the paternal allele, while a pathogenic activating KIT mutation was identified in the tumor. Additionally, methylation-specific multiplex ligation-dependent probe amplification revealed reduced methylation at the PWS locus in the dysgerminoma but not in the blood. In conclusion, our register-based study suggests an increased risk of cancer at a young age, especially testicular and ovarian tumors. We found no evidence of a general increase in cancer risk in patients with PWS. However, given our limited observational time, further studies with longer follow-up times are needed to clarify the lifetime cancer risk in PWS. We have also described the second case of locus-specific loss-of-imprinting in a germ cell tumor in PWS, suggesting a possible mechanism of carcinogenesis.

3.
Biomedicines ; 11(6)2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37371750

RESUMEN

Regulation of the human IGF2 gene displays multiple layers of control, which secures a genetically and epigenetically predetermined gene expression pattern throughout embryonal growth and postnatal life. These predominantly nuclear regulatory mechanisms converge on the function of the IGF2-H19 gene cluster on Chromosome 11 and ultimately affect IGF2 gene expression. Deregulation of such control checkpoints leads to the enhancement of IGF2 gene transcription and/or transcript stabilization, ultimately leading to IGF-II peptide overproduction. This type of anomaly is responsible for the effects observed in terms of both abnormal fetal growth and increased cell proliferation, typically observed in pediatric overgrowth syndromes and cancer. We performed a review of relevant experimental work on the mechanisms affecting the human IGF2 gene at the epigenetic, transcriptional and transcript regulatory levels. The result of our work, indeed, provides a wider and diversified scenario for IGF2 gene activation than previously envisioned by shedding new light on its extended regulation. Overall, we focused on the functional integration between the epigenetic and genetic machinery driving its overexpression in overgrowth syndromes and malignancy, independently of the underlying presence of loss of imprinting (LOI). The molecular landscape provided at last strengthens the role of IGF2 in cancer initiation, progression and malignant phenotype maintenance. Finally, this review suggests potential actionable targets for IGF2 gene- and regulatory protein target-degradation therapies.

4.
Clin Epigenetics ; 12(1): 43, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32143689

RESUMEN

BACKGROUND: Heritable epigenetic alterations have been proposed as an explanation for familial clustering of melanoma. Here we performed genome-wide DNA methylation analysis on affected family members not carrying pathogenic variants in established melanoma susceptibility genes, compared with healthy volunteers. RESULTS: All melanoma susceptibility genes showed the absence of epimutations in familial melanoma patients, and no loss of imprinting was detected. Unbiased genome-wide DNA methylation analysis revealed significantly different levels of methylation in single CpG sites. The methylation level differences were small and did not affect reported tumour predisposition genes. CONCLUSION: Our results provide no support for heritable epimutations as a cause of familial melanoma.


Asunto(s)
Metilación de ADN , Melanoma/genética , Neoplasias Cutáneas/genética , Adulto , Anciano , Islas de CpG , Genoma Humano , Humanos , Persona de Mediana Edad , Regiones Promotoras Genéticas
5.
Mol Biol Evol ; 37(2): 429-441, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31639821

RESUMEN

Genomic imprinting leads to mono-allelic expression of genes based on parent of origin. Therian mammals and angiosperms evolved this mechanism in nutritive tissues, the placenta, and endosperm, where maternal and paternal genomes are in conflict with respect to resource allocation. We used RNA-seq to analyze allelic bias in the expression of 91 known imprinted genes in term human placentas from a prospective cohort study in Mali. A large fraction of the imprinted exons (39%) deviated from mono-allelic expression. Loss of imprinting (LOI) occurred in genes with either maternal or paternal expression bias, albeit more frequently in the former. We characterized LOI using binomial generalized linear mixed models. Variation in LOI was predominantly at the gene as opposed to the exon level, consistent with a single promoter driving the expression of most exons in a gene. Some genes were less prone to LOI than others, particularly lncRNA genes were rarely expressed from the repressed allele. Further, some individuals had more LOI than others and, within a person, the expression bias of maternally and paternally imprinted genes was correlated. We hypothesize that trans-acting maternal effect genes mediate correlated LOI and provide the mother with an additional lever to control fetal growth by extending her influence to LOI of the paternally imprinted genes. Limited evidence exists to support associations between LOI and offspring phenotypes. We show that birth length and placental weight were associated with allelic bias, making this the first comprehensive report of an association between LOI and a birth phenotype.


Asunto(s)
Peso al Nacer/genética , Estatura/genética , Perfilación de la Expresión Génica/métodos , Impresión Genómica , Placenta/química , Adolescente , Femenino , Humanos , Recién Nacido , Modelos Lineales , Malí , Herencia Materna , Embarazo , Regiones Promotoras Genéticas , Estudios Prospectivos , Análisis de Secuencia de ARN , Adulto Joven
6.
J Hematol Oncol ; 10(1): 107, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28506242

RESUMEN

BACKGROUND: A microRNA (miRNA) collection on the imprinted 14q32 MEG3 region has been associated with outcome in osteosarcoma. We assessed the clinical utility of this miRNA set and their association with methylation status. METHODS: We integrated coding and non-coding RNA data from three independent annotated clinical osteosarcoma cohorts (n = 65, n = 27, and n = 25) and miRNA and methylation data from one in vitro (19 cell lines) and one clinical (NCI Therapeutically Applicable Research to Generate Effective Treatments (TARGET) osteosarcoma dataset, n = 80) dataset. We used time-dependent receiver operating characteristic (tdROC) analysis to evaluate the clinical value of candidate miRNA profiles and machine learning approaches to compare the coding and non-coding transcriptional programs of high- and low-risk osteosarcoma tumors and high- versus low-aggressiveness cell lines. In the cell line and TARGET datasets, we also studied the methylation patterns of the MEG3 imprinting control region on 14q32 and their association with miRNA expression and tumor aggressiveness. RESULTS: In the tdROC analysis, miRNA sets on 14q32 showed strong discriminatory power for recurrence and survival in the three clinical datasets. High- or low-risk tumor classification was robust to using different microRNA sets or classification methods. Machine learning approaches showed that genome-wide miRNA profiles and miRNA regulatory networks were quite different between the two outcome groups and mRNA profiles categorized the samples in a manner concordant with the miRNAs, suggesting potential molecular subtypes. Further, miRNA expression patterns were reproducible in comparing high-aggressiveness versus low-aggressiveness cell lines. Methylation patterns in the MEG3 differentially methylated region (DMR) also distinguished high-aggressiveness from low-aggressiveness cell lines and were associated with expression of several 14q32 miRNAs in both the cell lines and the large TARGET clinical dataset. Within the limits of available CpG array coverage, we observed a potential methylation-sensitive regulation of the non-coding RNA cluster by CTCF, a known enhancer-blocking factor. CONCLUSIONS: Loss of imprinting/methylation changes in the 14q32 non-coding region defines reproducible previously unrecognized osteosarcoma subtypes with distinct transcriptional programs and biologic and clinical behavior. Future studies will define the precise relationship between 14q32 imprinting, non-coding RNA expression, genomic enhancer binding, and tumor aggressiveness, with possible therapeutic implications for both early- and advanced-stage patients.


Asunto(s)
Neoplasias Óseas/genética , Cromosomas Humanos Par 14/genética , Impresión Genómica , MicroARNs/genética , Osteosarcoma/genética , ARN Neoplásico/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/mortalidad , Boston/epidemiología , Línea Celular Tumoral , Metilación de ADN , ADN de Neoplasias/genética , Conjuntos de Datos como Asunto , Supervivencia sin Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales , Curva ROC , Análisis de Supervivencia , Transcripción Genética , Resultado del Tratamiento , Utah/epidemiología
7.
Cell Stress ; 1(1): 37-54, 2017 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-31225433

RESUMEN

The long non-coding RNA (lncRNA) H19 represents a maternally expressed and epigenetically regulated imprinted gene product and is discussed to have either tumor-promoting or tumor-suppressive actions. Recently, H19 was shown to be regulated under inflammatory conditions. Therefore, aim of this study was to determine the function of H19 in hepatocellular carcinoma (HCC), an inflammation-associated type of tumor. In four different human HCC patient cohorts H19 was distinctly downregulated in tumor tissue compared to normal or non-tumorous adjacent tissue. We therefore determined the action of H19 in three different human hepatoma cell lines (HepG2, Plc/Prf5, and Huh7). Clonogenicity and proliferation assays showed that H19 overexpression could suppress tumor cell survival and proliferation after treatment with either sorafenib or doxorubicin, suggesting chemosensitizing actions of H19. Since HCC displays a highly chemoresistant tumor entity, cell lines resistant to doxorubicin or sorafenib were established. In all six chemoresistant cell lines H19 expression was significantly downregulated. The promoter methylation of the H19 gene was significantly different in chemoresistant cell lines compared to their sensitive counterparts. Chemoresistant cells were sensitized after H19 overexpression by either increasing the cytotoxic action of doxorubicin or decreasing cell proliferation upon sorafenib treatment. An H19 knockout mouse model (H19Δ3) showed increased tumor development and tumor cell proliferation after treatment with the carcinogen diethylnitrosamine (DEN) independent of the reciprocally imprinted insulin-like growth factor 2 (IGF2). In conclusion, H19 suppresses hepatocarcinogenesis, hepatoma cell growth, and HCC chemoresistance. Thus, mimicking H19 action might be a potential target to overcome chemoresistance in future HCC therapy.

8.
Proc Natl Acad Sci U S A ; 112(15): 4618-23, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825726

RESUMEN

Embryos generated with the use of assisted reproductive technologies (ART) can develop overgrowth syndromes. In ruminants, the condition is referred to as large offspring syndrome (LOS) and exhibits variable phenotypic abnormalities including overgrowth, enlarged tongue, and abdominal wall defects. These characteristics recapitulate those observed in the human loss-of-imprinting (LOI) overgrowth syndrome Beckwith-Wiedemann (BWS). We have recently shown LOI at the KCNQ1 locus in LOS, the most common epimutation in BWS. Although the first case of ART-induced LOS was reported in 1995, studies have not yet determined the extent of LOI in this condition. Here, we determined allele-specific expression of imprinted genes previously identified in human and/or mouse in day ∼105 Bos taurus indicus × Bos taurus taurus F1 hybrid control and LOS fetuses using RNAseq. Our analysis allowed us to determine the monoallelic expression of 20 genes in tissues of control fetuses. LOS fetuses displayed variable LOI compared with controls. Biallelic expression of imprinted genes in LOS was associated with tissue-specific hypomethylation of the normally methylated parental allele. In addition, a positive correlation was observed between body weight and the number of biallelically expressed imprinted genes in LOS fetuses. Furthermore, not only was there loss of allele-specific expression of imprinted genes in LOS, but also differential transcript amounts of these genes between control and overgrown fetuses. In summary, we characterized previously unidentified imprinted genes in bovines and identified misregulation of imprinting at multiple loci in LOS. We concluded that LOS is a multilocus LOI syndrome, as is BWS.


Asunto(s)
Bovinos/genética , Feto/anomalías , Impresión Genómica , Técnicas Reproductivas Asistidas/veterinaria , Alelos , Animales , Síndrome de Beckwith-Wiedemann/embriología , Síndrome de Beckwith-Wiedemann/etiología , Síndrome de Beckwith-Wiedemann/genética , Bovinos/embriología , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Gigantismo/embriología , Gigantismo/etiología , Gigantismo/genética , Humanos , Masculino , Ratones , Polimorfismo de Nucleótido Simple , Técnicas Reproductivas Asistidas/efectos adversos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Síndrome
9.
J Pathol ; 233(4): 392-401, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24838394

RESUMEN

The tumour suppressor gene RB1 is frequently silenced in many different types of human cancer, including hepatocellular carcinoma (HCC). However, mutations of the RB1 gene are relatively rare in HCC. A systematic screen for the identification of imprinted genes deregulated in human HCC revealed that RB1 shows imprint abnormalities in a high proportion of primary patient samples. Altogether, 40% of the HCC specimens (16/40) showed hyper- or hypomethylation at the CpG island in intron 2 of the RB1 gene. Re-analysis of publicly available genome-wide DNA methylation data confirmed these findings in two independent HCC cohorts. Loss of correct DNA methylation patterns at the RB1 locus leads to the aberrant expression of an alternative RB1-E2B transcript, as measured by quantitative real-time PCR. Demethylation at the intron 2 CpG island by DNMT1 knock-down or aza-deoxycytidine (DAC) treatment stimulated expression of the RB1-E2B transcript, accompanied by diminished RB1 main transcript expression. No aberrant DNA methylation was found at the RB1 locus in hepatocellular adenoma (HCA, n = 10), focal nodular hyperplasia (FNH, n = 5) and their corresponding adjacent liver tissue specimens. Deregulated RB1 expression due to hyper- or hypomethylation in intron 2 of the RB1 gene is found in tumours without loss of heterozygosity and is associated with a decrease in overall survival (p = 0.032) if caused by hypermethylation of CpG85. This unequivocally demonstrates that loss of imprinting represents an important additional mechanism for RB1 pathway inactivation in human HCC, complementing well-described molecular defects.


Asunto(s)
Carcinoma Hepatocelular/genética , Regulación hacia Abajo/genética , Genes Supresores de Tumor , Impresión Genómica/genética , Neoplasias Hepáticas/genética , Proteína de Retinoblastoma/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Islas de CpG/genética , Metilación de ADN/genética , ADN de Neoplasias/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Proteína de Retinoblastoma/metabolismo
10.
Development ; 140(22): 4480-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24154523

RESUMEN

Imprinted genes play important roles in placenta development and function. Parthenogenetic embryos, deficient in paternally expressed imprinted genes, lack extra-embryonic tissues of the trophoblast lineage. Parthenogenetic trophoblast stem cells (TSCs) are extremely difficult to derive, suggesting that an imprinted gene(s) is necessary for TSC establishment or maintenance. In a candidate study, we were able to narrow the list to one known paternally expressed gene, Sfmbt2. We show that mouse embryos inheriting a paternal Sfmbt2 gene trap null allele have severely reduced placentae and die before E12.5 due to reduction of all trophoblast cell types. We infected early embryos with lentivirus vectors expressing anti-Sfmbt2 shRNAs and found that TSC derivation was significantly reduced. Together, these observations support the hypothesis that loss of SFMBT2 results in defects in maintenance of trophoblast cell types necessary for development of the extra-embryonic tissues, the placenta in particular.


Asunto(s)
Impresión Genómica/genética , Placentación/genética , Proteínas del Grupo Polycomb/genética , Factores de Transcripción/genética , Trofoblastos/citología , Alelos , Animales , Blastocisto/citología , Blastocisto/metabolismo , Femenino , Fertilización/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hibridación Fluorescente in Situ , Patrón de Herencia/genética , Ratones , Partenogénesis/genética , Proteínas del Grupo Polycomb/metabolismo , Embarazo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Trofoblastos/metabolismo , Inactivación del Cromosoma X/genética
11.
Genet Epigenet ; 5: 17-22, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25512704

RESUMEN

Insulin-like growth factor-2 (IGF-2) is a mitogen, growth and differentiation modulator for many cell types. It is mainly expressed during the prenatal development, and its activity strongly depends on the genomic imprinting. Genomic imprinting in the chorionic tissues of spontaneously eliminated human embryos has been studied on the model of 820-AG (Apa1) of the IGF-2 gene locus. Molecular and genetic analysis was performed on the polymorphic 820-AG IGF2 locus in 107 samples of DNA extracted from the chorionic tissues of spontaneously eliminated human embryos within 5-10 weeks of gestation. Presence of AG genotype Apa1 single nucleotide polymorphisms of the IGF-2 was shown to cause more than a 7-fold increase in the risk of embryo elimination. Thus, the loss of genomic imprinting of the IGF-2 gene may be an important cause of the miscarriages in human.

12.
Am J Stem Cells ; 1(1): 59-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23671798

RESUMEN

Among the hypotheses discussing cancer formation, the cancer stem cell (CSC) theory is one receiving widespread support. One version of this theory states that changes in otherwise healthy cells can cause formation of tumor- initiating cells (TICs), which have the potential to create precancerous stem cells that can lead to CSC formation. These CSCs can be rare, in contrast to their differentiated progeny, which give rise to the vast majority of the tumor mass in most cancers. Loss of imprinting (LOI) of the insulin-like growth factor-2 (IGF2) gene is one change that can produce these TICs via an epigenetic progenitor model of tumorigenesis. While IGF2 usually supports normal cellular growth, LOI of IGF2 may lead to overexpression of the gene and moreover global chromatin instability. This modification has been observed in many forms of cancer, and given the effect of LOI of IGF2 and its role in cancer, detecting a loss of imprinting in this gene could serve as a valuable diagnostic tool. Preclinical data has shown some progress in identifying therapeutic approaches seeking to exploit this relationship. Thus, further research surrounding LOI of IGF2 could lead to increased understanding of several cancer types and enhance therapies against these diseases.

13.
Biol. Res ; 45(4): 351-356, 2012. ilus
Artículo en Inglés | LILACS | ID: lil-668685

RESUMEN

Loss of Imprinting (LOI) of IGF2 and over-expressed IGF2 are associated with tumorigenesis. Our previous epidemiological study found a relatively high frequency of IGF2 LOI in healthy mid-gestation pregnant women. The aim of this study is to determine whether the expression of IGF2 is associated with its imprinting status in healthy Chinese pregnant women. The IGF2 imprinting status of 300 pregnant women was analyzed. 20 cases of IGF2 LOI and 20 cases of IGF2 retention of imprinting (ROI) were selected randomly for IGF2 expression analysis. The expression pattern of IGF2 between the group with IGF2 ROI and group with IGF2 LOI in healthy Chinese pregnant women was evaluated by real time PCR and western blot. The result showed no significant differences between IGF2 ROI and LOI groups in mRNA and protein levels. These results imply that IGF2 imprinting status has no obvious impact on its expression. There may be some unknown important factors other than imprinting status driving IGF2 expression.


Asunto(s)
Adulto , Femenino , Humanos , Embarazo , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , ARN Mensajero/genética , China , Factor II del Crecimiento Similar a la Insulina/metabolismo , Reacción en Cadena de la Polimerasa
14.
Per Med ; 8(1): 35-43, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29768784

RESUMEN

The importance of epigenetics in normal development and tissue-specific gene expression, as well as in diseases such as cancer, is well established. DNA methylation is a primary epigenetic modification that is directly linked to the genome itself. Here, we review evidence supporting the promise of DNA methylation-based biomarkers in personalized medicine, discuss standard and emerging technologies for profiling DNA methylation on a genome-wide scale, and forecast how these approaches will be used in parallel to better understand the epigenetics of health and disease and apply that knowledge to advance the field of personalized medicine.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA