Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Biomedicines ; 10(4)2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35453543

RESUMEN

Human hepatocellular carcinoma (HCC) has a high mortality rate because of the dearth of effective treatments. Multiple studies have shown that overexpression of UPF1, a key nonsense-mediated mRNA decay (NMD) factor, reduces HCC growth through various cell signaling pathways. However, the mechanism by which UPF1 expression retards HCC proliferation through the regulation of RNA stability remains unclear. By employing various UPF1 variants and transcriptome analysis, we revealed that overexpression of UPF1 variants, not UPF1-mediated NMD, reduces HCC tumorigenesis. Additionally, UPF1 variant overexpression reduced tumorigenesis in xenografted mice. Transcriptome analysis indicated that the level of dual specificity phosphatase 1 (DUSP1) was increased by UPF1 variants via posttranscriptional regulation. The UPF1 overexpression-mediated increase of DUSP1 activated tumor suppressor signaling, ultimately inhibiting cell growth. In this study, we highlighted the function of UPF1 as a tumor suppressor in HCC growth.

2.
Nat Commun ; 11(1): 5414, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33110075

RESUMEN

The neoplastic stromal cells of giant cell tumor of bone (GCTB) carry a mutation in H3F3A, leading to a mutant histone variant, H3.3-G34W, as a sole recurrent genetic alteration. We show that in patient-derived stromal cells H3.3-G34W is incorporated into the chromatin and associates with massive epigenetic alterations on the DNA methylation, chromatin accessibility and histone modification level, that can be partially recapitulated in an orthogonal cell line system by the introduction of H3.3-G34W. These epigenetic alterations affect mainly heterochromatic and bivalent regions and provide possible explanations for the genomic instability, as well as the osteolytic phenotype of GCTB. The mutation occurs in differentiating mesenchymal stem cells and associates with an impaired osteogenic differentiation. We propose that the observed epigenetic alterations reflect distinct differentiation stages of H3.3 WT and H3.3 MUT stromal cells and add to H3.3-G34W-associated changes.


Asunto(s)
Neoplasias Óseas/genética , Tumor Óseo de Células Gigantes/genética , Histonas/genética , Osteogénesis , Neoplasias Óseas/metabolismo , Neoplasias Óseas/fisiopatología , Metilación de ADN , Epigénesis Genética , Epigenómica , Tumor Óseo de Células Gigantes/metabolismo , Tumor Óseo de Células Gigantes/fisiopatología , Histonas/metabolismo , Humanos , Mutación Missense
3.
Nucleic Acids Res ; 47(15): 7870-7885, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31226208

RESUMEN

Long interspersed elements-1 (LINE-1, L1) are retrotransposons that hold the capacity of self-propagation in the genome with potential mutagenic outcomes. How somatic cells restrict L1 activity and how this process becomes dysfunctional during aging and in cancer cells is poorly understood. L1s are enriched at lamin-associated domains, heterochromatic regions of the nuclear periphery. Whether this association is necessary for their repression has been elusive. Here we show that the sirtuin family member SIRT7 participates in the epigenetic transcriptional repression of L1 genome-wide in both mouse and human cells. SIRT7 depletion leads to increased L1 expression and retrotransposition. Mechanistically, we identify a novel interplay between SIRT7 and Lamin A/C in L1 repression. Our results demonstrate that SIRT7-mediated H3K18 deacetylation regulates L1 expression and promotes L1 association with elements of the nuclear lamina. The failure of such activity might contribute to the observed genome instability and compromised viability in SIRT7 knockout mice. Overall, our results reveal a novel function of SIRT7 on chromatin organization by mediating the anchoring of L1 to the nuclear envelope, and a new functional link of the nuclear lamina with transcriptional repression.


Asunto(s)
Genoma , Lamina Tipo A/genética , Elementos de Nucleótido Esparcido Largo , Sirtuinas/genética , Transcripción Genética , Animales , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/metabolismo , Heterocromatina/química , Heterocromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Células K562 , Lamina Tipo A/metabolismo , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocardio/citología , Miocardio/metabolismo , Lámina Nuclear/metabolismo , Lámina Nuclear/ultraestructura , Sirtuinas/deficiencia , Sirtuinas/metabolismo , Testículo/citología , Testículo/metabolismo
4.
Sci Rep ; 9(1): 4855, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30890718

RESUMEN

Protein tyrosine phosphatase, receptor type N2 (PTPRN2) encodes a major islet autoantigen in type-1 diabetes. Previous genetic studies have shown its significant association with obesity. PTPRN2 plays an important role in epigenetic regulation of metabolic diseases and cancers. We investigated CpG methylations (cg17429772 and cg158269415) in PTPRN2 by pyrosequencing from blood samples of childhood obesity (n = 638). cg158269415 had significant positive correlations with body mass index (BMI) and waist-hip ratio (WHR). Case-control analysis showed that cg158269415 methylation in blood sample was significantly more hypermethylated in obese cases (n = 252), an average of 2.93% more than that that in controls (n = 386). The cg158269415 methylation has a trimodal distribution pattern with strong dependency on nearby located rs1670344 G > A genotype. Correlations of cg158269415 with BMI and WHR were significant and strong in major G allele carriers (GG + GA). Our study showed that an epigenetic association of PTPRN2 gene with childhood obesity was under certain genetic background. The genetic and epigenetic interplay of PTPRN2 gene may implicate a mechanism of childhood obesity. Whether these small changes in DNA methylation from whole blood are causally or consequently related to childhood obesity outcome and their clinical relevance remains to be determined. However, this study presents a promising obesity risk marker that warrants further investigation.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Obesidad Infantil/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Adolescente , Niño , Islas de CpG/genética , Metilación de ADN/genética , Femenino , Humanos , Masculino , Obesidad Infantil/patología
5.
Exp Mol Med ; 51(2): 1-13, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30820026

RESUMEN

Rheumatoid arthritis (RA) is a complex chronic systematic disease with progressive destruction of the joints by invasive synoviocytes. To characterize the key regulators involved in the development of RA, we obtained multilayer epigenomics data including DNA methylation by whole-genome bisulfite sequencing, miRNA profiles, genetic variations by whole-exome sequencing, and mRNA profiles from synoviocytes of RA and osteoarthritis (OA) patients. The overall DNA methylation patterns were not much different between RA and OA, but 523 low-methylated regions (LMRs) were specific to RA. The LMRs were preferentially localized at the 5' introns and overlapped with transcription factor binding motifs for GLI1, RUNX2, and TFAP2A/C. Single base-scale differentially methylated CpGs were linked with several networks related to wound response, tissue development, collagen fibril organization, and the TGF-ß receptor signaling pathway. Further, the DNA methylation of 201 CpGs was significantly correlated with 27 expressed miRNA genes. Our interpretation of epigenomic data of the synoviocytes from RA and OA patients is an informative resource to further investigate regulatory elements and biomarkers responsible for the pathophysiology of RA and OA.


Asunto(s)
Artritis Reumatoide/genética , Epigénesis Genética , Sinoviocitos/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Islas de CpG , Metilación de ADN , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Variación Genética , Humanos , MicroARNs/genética , ARN Mensajero/genética
6.
Sci Rep ; 9(1): 1815, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755693

RESUMEN

Low-density lipoprotein Receptor Related Protein 1B (LRP1B) is homologous to the gigantic lipoprotein receptor-related protein 1 that belongs to the family of Low-density lipoprotein receptors. Previous genetic association studies of the LRP1B gene have shown its genetic association with obesity. Through exome sequencing of the LRP1B gene from a childhood severe obesity cohort (n = 692), we found novel single nucleotide polymorphism (rs431809) in intron 4, which has been significantly correlated with both body mass index (BMI) and waist-hip-ratio (WHR). Three methylations of CpG sites (cg141441481, cg01852095 and cg141441470) in the same intron were also significantly correlated with BMI and WHR. All CpG methylations had bimodal patterns, and were dependent on rs431809 genotypes. The genetic influences of obesity on the LRP1B gene may be linked to the interplay of CpG methylations in the same intron. Heritability of SNP interacts with epigenetic crosstalk in LRP1B. Genetic and epigenetic crosstalk of LRP1B gene may be implicated in the prevention and therapeutic approach to childhood obesity.


Asunto(s)
Epigénesis Genética/genética , Obesidad Infantil/genética , Receptores de LDL/genética , Adolescente , Índice de Masa Corporal , Metilación de ADN/genética , Metilación de ADN/fisiología , Exoma/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Frecuencia de los Genes/genética , Genotipo , Humanos , Intrones/genética , Masculino , Polimorfismo de Nucleótido Simple/genética , Relación Cintura-Cadera
7.
Oncotarget ; 8(40): 67473-67481, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978046

RESUMEN

Differential methylations of the HIF3A (hypoxia-inducible factor 3a) gene have been linked to body mass index (BMI). To explore the association of these methylations to childhood obesity, we measured 5 CpG methylation sites (cg27146050, cg46801562, cg22891070, cg16672562 and cg46801675) in intron 1 of the HIF3A gene by pyrosequencing, in the Korean population (mean age: 13.9 yrs, 305 obese cases and 387 controls). Two CpG methylations, cg46801562 and cg16672562, had statistically significant association with childhood obesity (P = 2.09E-9 and 1.66E-7, respectively). Notably, in the case of cg16672562, all correlations were significantly positive with BMI (beta = 0.285, P = 1.652E-13), waist-hip ratio (beta = 0.0028, P = 1.42E-15) and fasting plasma glucose level (beta = 0.0645, P = 2.61E-4), when analyzed by linear regression, with age and sex as covariates. We investigated any genetic effect of cg16672562 methylation by using 14 single nucleotide polymorphisms (SNP) identified by exome sequencing of the HIF3A locus cg16672562 methylation showed no statistically significant changes due to the 14 polymorphisms. In this study, we show that cg16672562 is the most significant blood DNA methylation marker for childhood obesity in the Korean population, and might be independent of any underlying HIF3A genetic background.

8.
Proc Natl Acad Sci U S A ; 114(28): E5635-E5644, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28630288

RESUMEN

The PIWI-interacting RNA (piRNA) pathway is essential for retrotransposon silencing. In piRNA-deficient mice, L1-overexpressing male germ cells exhibit excessive DNA damage and meiotic defects. It remains unknown whether L1 expression simply highlights piRNA deficiency or actually drives the germ-cell demise. Specifically, the sheer abundance of genomic L1 copies prevents reliable quantification of new insertions. Here, we developed a codon-optimized L1 transgene that is controlled by an endogenous mouse L1 promoter. Importantly, DNA methylation dynamics of a single-copy transgene were indistinguishable from those of endogenous L1s. Analysis of Mov10l1-/- testes established that de novo methylation of the L1 transgene required the intact piRNA pathway. Consistent with loss of DNA methylation and programmed reduction of H3K9me2 at meiotic onset, the transgene showed 1,400-fold increase in RNA expression and consequently 70-fold increase in retrotransposition in postnatal day 14 Mov10l1-/- germ cells compared with the wild-type. Analysis of adult Mov10l1-/- germ-cell fractions indicated a stage-specific increase of retrotransposition in the early meiotic prophase. However, extrapolation of the transgene data to endogenous L1s suggests that it is unlikely insertional mutagenesis alone accounts for the Mov10l1-/- phenotype. Indeed, pharmacological inhibition of reverse transcription did not rescue the meiotic defect. Cumulatively, these results establish the occurrence of productive L1 mobilization in the absence of an intact piRNA pathway but leave open the possibility of processes preceding L1 integration in triggering meiotic checkpoints and germ-cell death. Additionally, our data suggest that many heritable L1 insertions originate from individuals with partially compromised piRNA defense.


Asunto(s)
Meiosis , ARN Interferente Pequeño/metabolismo , Retroelementos , Transgenes , Regiones no Traducidas 5' , Animales , Codón , Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Masculino , Metilación , Ratones , Ratones Transgénicos , Sistemas de Lectura Abierta , Fenotipo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Espermatocitos/metabolismo , Espermatogénesis , Testículo/metabolismo
9.
Sci Rep ; 7: 45368, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28508896

RESUMEN

The cg07814318 hypermethylation of Kruppel-like factor 13 (KLF13) gene has been reported for its relevancy with Body Mass Index (BMI) from European origin. We explored the cg07814318 methylation and its cis-meQTL (cis-methylation quantitative loci) of KLF13 from a childhood obesity cohort. The cg07814318 methylation in blood was significantly associated with obesity and correlated with several obesity-related physical and biochemical traits. We examined the same loci from purified three human cell types (n = 47), i.e., pre-adipocytes, adipocytes and islets. The cg07814318 methylation pattern in pre-adipocytes and islets were significant higher in cells from subjects with a higher BMI compared with control subjects. By exome sequencing of KLF13 gene in blood with the same cohort, we found nine SNPs (single nucleotide polymorphisms) within its gene body, and two SNPs (rs11537749 and rs12595641) were as cis-meQTL of cg07814318. There was the 2.01% methylation change of cg07814318 between homozygous dominant and recessive genotypes, especially, in rs12595641. The sequencing variations within KLF13 genes could drive dynamic modifications of obesity-related CpG methylation. Differential DNA methylation patterns in the KLF13 gene determined from separate blood samples showed that this criterion could be used as a surrogate for representing overall epigenetic changes in cells related to obesity.


Asunto(s)
Proteínas de Ciclo Celular/genética , Metilación de ADN , Factores de Transcripción de Tipo Kruppel/genética , Obesidad Infantil/genética , Polimorfismo de Nucleótido Simple , Proteínas Represoras/genética , Análisis de Secuencia de ADN/métodos , Adolescente , Índice de Masa Corporal , Estudios de Cohortes , Islas de CpG , Exoma , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Sitios de Carácter Cuantitativo
10.
PLoS One ; 12(5): e0177406, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28542303

RESUMEN

Differential DNA methylation with hyperglycemia is significantly associated with Type 2 Diabetes (T2D). Longtime extended exposure to high blood glucose levels can affect the epigenetic signatures in all organs. However, the relevance of the differential DNA methylation changes with hyperglycemia in blood with pancreatic islets remains unclear. We investigated differential DNA methylation in relation to glucose homeostasis based on the Oral Glucose Tolerance Test (OGTT) in a population-based cohort. We found a total of 382 differential methylation sites from blood DNA in hyperglycemia and type 2 diabetes subgroups using a longitudinal and cross-sectional approach. Among them, three CpG sites were overlapped; they were mapped to the MSI2 and CXXC4 genes. In a DNA methylation replication study done by pyrosequencing (n = 440), the CpG site of MSI2 were shown to have strong associations with the T2D group (p value = 2.20E-16). The differential methylation of MSI2 at chr17:55484635 was associated with diabetes-related traits, in particular with insulin sensitivity (QUICKI, p value = 2.20E-16) and resistance (HOMA-IR, p value = 1.177E-07). In human pancreatic islets, at the single-base resolution (using whole-genome bisulfite sequencing), the 292 CpG sites in the ±5kb at chr17:55484635 were found to be significantly hypo-methylated in donors with T2D (average decrease = 13.91%, 95% confidence interval (CI) = 4.18~ 17.06) as compared to controls, and methylation patterns differed by sex (-9.57%, CI = -16.76~ -6.89) and age (0.12%, CI = -11.17~ 3.77). Differential methylation of the MSI2 gene (chr17:55484635) in blood and islet cells is strongly related to hyperglycemia. Our findings suggest that epigenetic perturbation on the target site of MSI2 gene in circulating blood and pancreatic islets should represent or affect hyperglycemia.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/genética , Proteínas de Unión al ARN/genética , Adulto , Anciano , Estudios de Casos y Controles , Islas de CpG , Estudios Transversales , Diabetes Mellitus Tipo 2/metabolismo , Epigénesis Genética , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/sangre , Hiperglucemia/genética , Islotes Pancreáticos/metabolismo , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Estudios Prospectivos
11.
Mol Cells ; 39(10): 750-755, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27802373

RESUMEN

Although innate color preference of motile organisms may provide clues to behavioral biases, it has remained a longstanding question. In this study, we investigated innate color preference of zebrafish larvae. A cross maze with different color sleeves around each arm was used for the color preference test (R; red, G; green, B; blue, Y; yellow). The findings showed that 5 dpf zebrafish larvae preferred blue over other colors (B > R > G > Y). To study innate color recognition further, tyrosinase mutants were generated using CRISPR/Cas9 system. As a model for oculocutaneous albinism (OCA) and color vision impairment, tyrosinase mutants demonstrated diminished color sensation, indicated mainly by hypopigmentation of the retinal pigment epithelium (RPE). Due to its relative simplicity and ease, color preference screening using zebrafish larvae is suitable for high-throughput screening applications. This system may potentially be applied to the analysis of drug effects on larval behavior or the detection of sensory deficits in neurological disorder models, such as autism-related disorders, using mutant larvae generated by the CRISPR/Cas9 technique.


Asunto(s)
Análisis Aplicado de la Conducta/métodos , Conducta Animal/fisiología , Color , Pez Cebra/fisiología , Animales , Femenino , Masculino
12.
MethodsX ; 3: 1-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27366675

RESUMEN

We have developed an NGS-based deep bisulfite sequencing protocol for the DNA methylation analysis of genomes. This approach allows the rapid and efficient construction of NGS-ready libraries with a large number of PCR products that have been individually amplified from bisulfite-converted DNA. This approach also employs a bioinformatics strategy to sort the raw sequence reads generated from NGS platforms and subsequently to derive DNA methylation levels for individual loci. The results demonstrated that this NGS-based deep bisulfite sequencing approach provide not only DNA methylation levels but also informative DNA methylation patterns that have not been seen through other existing methods.•This protocol provides an efficient method generating NGS-ready libraries from individually amplified PCR products.•This protocol provides a bioinformatics strategy sorting NGS-derived raw sequence reads.•This protocol provides deep bisulfite sequencing results that can measure DNA methylation levels and patterns of individual loci.

13.
PLoS One ; 10(12): e0145531, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26692216

RESUMEN

Peg3 is an imprinted gene that is predicted to encode a DNA-binding zinc finger protein. This was previously demonstrated through Chromatin ImmunoPrecipitation-based Sequencing experiments. In the current study, we reanalyzed the previous ChIP-Seq results and further characterized the DNA-binding motif of PEG3. According to the results, PEG3 binds to the promoters and enhancers of a subset of genes that are closely associated with the known functions of Peg3. Some of these identified targets include Tufm, Mrpl45, Cry2, Per1, Slc25a29 and Slc38a2. With this set of targets, we derived a DNA-binding motif of PEG3, 5'-GTGGCAGT-3', which also provides a tabulated matrix that can be used for predicting other unknown genomic targets. Among the newly identified targets, we analyzed in detail the two loci, Slc38a2 and Slc38a4, which are known to be involved in neutral amino acid transport. The results indicated that PEG3 likely functions as a transcriptional repressor for these two loci. Overall, the current study provides a set of genomic targets and also redefines the DNA-binding motif for the imprinted transcription factor PEG3.


Asunto(s)
ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Sistema de Transporte de Aminoácidos A/genética , Sistema de Transporte de Aminoácidos A/metabolismo , Animales , Sitios de Unión , Inmunoprecipitación de Cromatina , Criptocromos/genética , Impresión Genómica , Factores de Transcripción de Tipo Kruppel/genética , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Factor Tu de Elongación Peptídica/genética , Proteínas Circadianas Period/genética , Factores de Transcripción
14.
Epigenetics ; 10(12): 1111-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26507119

RESUMEN

Although aberrant DNA methylation within imprinted domains has been reported in a variety of neoplastic diseases, it remains largely uncharacterized in the context of carcinogenesis. In this study, we induced T-cell lymphoma in mice by employing a breeding scheme involving mouse strains, LSL-Kras(G12D) and MMTV-Cre. We then systematically surveyed imprinted domains for DNA methylation changes during tumor progression using combined bisulfite restriction analysis and NGS-based bisulfite sequencing. We detected hyper- or hypo-methylation at the imprinting control regions (ICRs) of the Dlk1, Peg10, Peg3, Grb10, and Gnas domains. These DNA methylation changes at ICRs were more prevalent and consistent than those observed at the promoter regions of well-known tumor suppressors, such as Mgmt, Fhit, and Mlh1. Thus, the changes observed at these imprinted domains are the outcome of isolated incidents affecting DNA methylation settings. Within imprinted domains, DNA methylation changes tend to be restricted to ICRs as nearby somatic differentially methylated regions and promoter regions experience no change. Furthermore, detailed analyses revealed that small cis-regulatory elements within ICRs tend to be resistant to DNA methylation changes, suggesting potential protection by unknown trans-factors. Overall, this study demonstrates that DNA methylation changes at ICRs are dynamic during carcinogenesis and advocates that detection of aberrant DNA methylation at ICRs may serve as a biomarker to enhance diagnostic procedures.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Impresión Genómica , Linfoma de Células T/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Progresión de la Enfermedad , Marcadores Genéticos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos
15.
Nucleic Acids Res ; 43(22): 10689-99, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26338779

RESUMEN

Many imprinted genes are often epigenetically affected in human cancers due to their functional linkage to insulin and insulin-like growth factor signaling pathways. Thus, the current study systematically characterized the epigenetic instability of imprinted genes in multiple human cancers. First, the survey results from TCGA (The Cancer Genome Atlas) revealed that the expression levels of the majority of imprinted genes are downregulated in primary tumors compared to normal cells. These changes are also accompanied by DNA methylation level changes in several imprinted domains, such as the PEG3, MEST and GNAS domains. Second, these DNA methylation level changes were further confirmed manually using several sets of cancer DNA. According to the results, the Imprinting Control Regions of the PEG3, MEST and GNAS domains are indeed affected in breast, lung and ovarian cancers. This DNA methylation survey also revealed that evolutionarily conserved cis-regulatory elements within these imprinted domains are very variable in both normal and cancer cells. Overall, this study highlights the epigenetic instability of imprinted domains in human cancers and further suggests its potential use as cancer biomarkers.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Impresión Genómica , Neoplasias/genética , Metilación de ADN , Humanos , Elementos Reguladores de la Transcripción
16.
Genes Cells ; 20(6): 512-20, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25847286

RESUMEN

hK2, a member of the kallikrein protease family encoded by KLK2, is expressed exclusively in prostate and is a putative adjunct tumor marker for prostate cancer screening. The T allele of rs198977, a single nucleotide polymorphism in exon 5 of KLK2, codes for W-hK2 and is associated with lower serum hK2 levels and higher risk of prostate cancer than the C allele encoding R-hK2. To elucidate the mechanism that underlies this SNP's function, we transfected plasmids expressing R-hK2 or W-hK2 into PC3, HeLa and HEK293A cells and measured the hK2 level in cell lysates and conditioned media. The level of W-hK2 was lower than R-hK2 in conditioned media but was not different from R-hK2 in cell lysates. W-hK2 was hardly colocalized with Golgi-targeted fluorescent protein whereas R-hK2 colocalized. Reporter assays related to the unfolded protein response (UPR) and phospho-eIF2α immunoblot showed that W-hK2 increased UPR activity more than R-hK2. These results indicated that W-hK2 had a defect in cellular trafficking from the ER to the Golgi complex due to its misfolding and that it activated the UPR, suggesting a mechanism to explain the association of the T allele with higher prostate cancer risk.


Asunto(s)
Calicreínas/metabolismo , Respuesta de Proteína Desplegada , Transporte Biológico Activo/genética , Estudios de Asociación Genética , Aparato de Golgi/metabolismo , Células HEK293 , Células HeLa , Humanos , Calicreínas/genética , Masculino , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/genética
17.
Sci Rep ; 4: 5417, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24957209

RESUMEN

The development of gene delivery systems into embryos is challenging due to technical difficulties, delivery efficiency and toxicity. Here, we developed an organic compound (VisuFect)-mediated gene delivery system for zygotes. The VisuFect, which is hydrophilic and Cy5.5-labeled, was conjugated with poly(A) oligo (VFA). The VFA into CHO cells showed clathrin-mediated internalization and no toxicity. The VFA successfully penetrated through the zona pellucida of fertilized eggs of various species including pigs, zebrafish, drosophilas and mice. The experiment with VisuFect-mediated delivery of the miR34c inhibitor showed similar results with direct microinjection of the miR34c inhibitor by suppressing the development of zygotes up to the blastocyst stage. Noticeable features of the VisuFect will provide great benefits for further studies on gene function in sperms and embryos.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Oligonucleótidos/genética , Transfección/métodos , Cigoto/metabolismo , Animales , Células CHO , Carbocianinas/química , Células Cultivadas , Cricetinae , Cricetulus , Drosophila , Femenino , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Ratones , Microinyecciones , Microscopía Confocal , Oligonucleótidos/administración & dosificación , Oligonucleótidos/química , Compuestos Orgánicos/química , Poli A/administración & dosificación , Poli A/química , Poli A/genética , Porcinos , Imagen de Lapso de Tiempo/métodos , Pez Cebra , Cigoto/crecimiento & desarrollo
18.
Asian J Androl ; 16(5): 694-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24799634

RESUMEN

Monocarboxylic acid transporter 2 (MCT2) transports pyruvate and lactate outside and inside of sperms, mainly as energy sources and plays roles in the regulation of spermatogenesis. We investigated the association among genetic variations in the MCT2 gene, male infertility and MCT2 expression levels in sperm. The functional and genetic significance of the intron 2 (+28201A > G, rs10506398) and 3' untranslated region (UTR) single nucleotide polymorphism (SNP) (+2626G > A, rs10506399) of MCT2 variants were investigated. Two MCT2 polymorphisms were associated with male infertility (n = 471, P < 0.05). In particular, the MCT2-3' UTR SNP (+2626 G > A) had a strong association with the oligoasthenoteratozoospermia (OAT) group. The +2626GG type had an almost 2.4-fold higher sperm count than that of the +2626AA type (+2626GG; 66 × 10 6 vs +2626AA; 27 × 10 6 , P < 0.0001). The MCT2-3' UTR SNP may be important for expression, as it is located at the MCT2 3' UTR. The average MCT2 protein amount in sperm of the +2626GG type was about two times higher than that of the +2626AA type. The results suggest that genetic variation in MCT2 has functional and clinical relevance with male infertility.


Asunto(s)
Azoospermia/genética , Transportadores de Ácidos Monocarboxílicos/genética , Oligospermia/genética , Espermatozoides/metabolismo , Estudios de Casos y Controles , Predisposición Genética a la Enfermedad , Humanos , Infertilidad Masculina/genética , Masculino , Transportadores de Ácidos Monocarboxílicos/metabolismo , Polimorfismo de Nucleótido Simple
19.
Gene ; 534(1): 107-12, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24140493

RESUMEN

Cytochrome P450, family 17, subfamily A, polypeptide 1 (CYP17A1) is a key regulatory enzyme in the steroidogenic pathway. The functional and clinical relevance of novel CYP17A1 promoter single nucleotide polymorphism (-600 C>A, rs17115149) was investigated with male infertility. Case-control association study of CYP17A1 from 456 infertile men performed with 465 normal fertile men. The rs17115149 at the promoter region of CYP17A1 was significantly associated with Oligoasthenoteratozoospermia (OAT, P=0.0015, n=265). 5-aza-dC treatment to B lymphocyte cells increased the CYP17A1 expression. Direct bisulfite sequencing of five human tissues showed that the rs17115149 is located at -600bp (-600C>A) before transcription start site within the CpG islands of CYP17A1 promoter. This -600 Cytosine of CpG site was highly methylated in colon and stomach tissues, but low methylated in adrenal gland, kidney and testis with higher CYP17A1 RNA expression. Especially, this polymorphism is statistically significant associated with testosterone levels from infertile males (n=197, P<0.05). CYP17A1 promoter polymorphism (rs17115149, -600C>A) is a functional regulatory SNP which associated with its expression possibly by epigenetic pathway, which may signify a genetic risk factor for male infertility.


Asunto(s)
Islas de CpG/genética , Infertilidad Masculina/genética , Polimorfismo de Nucleótido Simple , Esteroide 17-alfa-Hidroxilasa/genética , Testosterona/genética , Pueblo Asiatico , Estudios de Casos y Controles , Metilación de ADN , Epigénesis Genética , Estudios de Asociación Genética , Humanos , Masculino , Regiones Promotoras Genéticas , Testosterona/sangre
20.
Mol Cells ; 36(2): 145-50, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23852131

RESUMEN

During skeletal development, both osteogenic and chondrogenic programs are initiated from multipotent mesenchymal cells, requiring a number of signaling molecules, transcription factors, and downstream effectors to orchestrate the sophisticated process. Col10a1, an important downstream effector gene, has been identified as a marker for maturing chondrocytes in higher vertebrates, such as mammals and birds. In zebrafish, this gene has been shown to be expressed in both osteoblasts and chondrocytes, but no study has reported its role in osteoblast development. To initially delineate the osteogenic program from chondrogenic lineage development, we used the zebrafish col10a1 promoter to establish a transgenic zebrafish expressing a GFP reporter specifically in osteoblast-specific bone structures that do not involve cartilaginous programs. A construct harboring a -2.2-kb promoter region was found to be sufficient to drive the reporter gene in osteoblast-specific bone structures within the endogenous col10a1 expression domain, confirming that separable cis-acting elements exist for distinct cell type-specific expression of col10a1 during zebrafish skeletal development. The -2.2-kb col10a1:GFP transgenic zebrafish marking only bone structures derived from osteoblasts will undoubtedly be an invaluable tool for identifying and characterizing molecular events driving osteoblast development in zebrafish, which may further provide a differential mechanism where col10a1 is involved in the development of chondrocytes undergoing maturation in other vertebrate systems.


Asunto(s)
Animales Modificados Genéticamente , Colágeno Tipo X/genética , Proteínas Fluorescentes Verdes/genética , Osteoblastos/metabolismo , Osteogénesis , Pez Cebra/genética , Animales , Condrocitos/metabolismo , Colágeno Tipo X/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Regiones Promotoras Genéticas , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...