Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
BMC Cancer ; 17(1): 874, 2017 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-29262808

RESUMO

BACKGROUND: Histones undergo extensive post-translational modifications and this epigenetic regulation plays an important role in modulating transcriptional programs capable of driving cancer progression. Acetylation of histone H3K18, associated with gene activation, is enhanced by P300 and opposed by the deacetylase Sirtuin2 (SIRT2). As these enzymes represent an important target for cancer therapy, we sought to determine whether the underlying genes are altered during prostate cancer (PCa) progression. METHODS: Tissue microarrays generated from 71 radical prostatectomy patients were initially immunostained for H3K18Ac, P300 and SIRT2. Protein levels were quantified using VECTRA automation and correlated with clinicopathologic parameters. The Cancer Genome Atlas (TGCA, n = 499) and Gene Expression Omnibus (n = 504) databases were queried for expression, genomic and clinical data. Statistics were performed using SPSSv23. RESULTS: Nuclear histone H3K18Ac staining increases in primary cancer (p = 0.05) and further in metastases (p < 0.01) compared to benign on tissue arrays. P300 protein expression increases in cancer (p = 0.04) and metastases (p < 0.001). A progressive decrease in nuclear SIRT2 staining occurs comparing benign to cancer or metastases (p = 0.04 and p = 0.03 respectively). Decreased SIRT2 correlates with higher grade cancer (p = 0.02). Time to Prostate Specific Antigen (PSA) recurrence is shorter in patients exhibiting high compared to low H3K18Ac expression (350 vs. 1542 days respectively, P = 0.03). In GEO, SIRT2 mRNA levels are lower in primary and metastatic tumors (p = 0.01 and 0.001, respectively). TGCA analysis demonstrates SIRT2 deletion in 6% and increasing clinical stage, positive margins and lower PSA recurrence-free survival in patients with SIRT2 loss/deletion (p = 0.01, 0.04 and 0.04  respectively). In this dataset, a correlation between decreasing SIRT2 and increasing P300 mRNA expression occurs in tumor samples (R = -0.46). CONCLUSIONS: In multiple datasets, decreases in SIRT2 expression portend worse clinicopathologic outcomes. Alterations in SIRT2-H3K18Ac suggest altered P300 activity and identify a subset of tumors that could benefit from histone deacetylation inhibition.


Assuntos
Biomarcadores Tumorais/genética , Proteína p300 Associada a E1A/metabolismo , Histonas/metabolismo , Recidiva Local de Neoplasia/mortalidade , Neoplasias da Próstata/mortalidade , Processamento de Proteína Pós-Traducional , Sirtuína 2/genética , Acetilação , Idoso , Biomarcadores Tumorais/metabolismo , Progressão da Doença , Proteína p300 Associada a E1A/genética , Epigênese Genética , Seguimentos , Histonas/química , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/cirurgia , Prognóstico , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Deleção de Sequência , Taxa de Sobrevida
2.
J Cell Biochem ; 116(7): 1165-70, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25581900

RESUMO

Genomic imprinting is an epigenetic mechanism that restricts gene expression to one inherited allele. Improper maintenance of imprinting has been implicated in a number of human diseases and developmental syndromes. Assays are needed that can quantify the contribution of each paternal allele to a gene expression profile. We have developed a rapid, sensitive quantitative assay for the measurement of individual allelic ratios termed Pyrosequencing for Imprinted Expression (PIE). Advantages of PIE over other approaches include shorter experimental time, decreased labor, avoiding the need for restriction endonuclease enzymes at polymorphic sites, and prevent heteroduplex formation which is problematic in quantitative PCR-based methods. We demonstrate the improved sensitivity of PIE including the ability to detect differences in allelic expression down to 1%. The assay is capable of measuring genomic heterozygosity as well as imprinting in a single run. PIE is applied to determine the status of Insulin-like Growth Factor-2 (IGF2) imprinting in human and mouse tissues.


Assuntos
Impressão Genômica , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Fator de Crescimento Insulin-Like II/genética , Análise de Sequência de DNA/métodos , Alelos , Animais , Perfilação da Expressão Gênica , Heterozigoto , Humanos , Masculino , Camundongos , Polimorfismo de Nucleotídeo Único , Próstata/metabolismo , Sensibilidade e Especificidade
3.
Curr Opin Urol ; 25(1): 83-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25405932

RESUMO

PURPOSE OF REVIEW: There is a major deficit in our ability to detect and predict the clinical behavior of prostate cancer (PCa). Epigenetic changes are associated with PCa development and progression. This review will focus on recent results in the clinical application of diagnostic and prognostic epigenetic markers. RECENT FINDINGS: The development of high throughput technology has seen an enormous increase in the discovery of new markers that encompass epigenetic changes including those in DNA methylation and histone modifications. Application of these findings to urine and other biofluids, but also cancer and noncancerous prostate tissue, has resulted in new biomarkers. There has been a recent commercial development of a DNA methylation-based assay for identifying PCa risk from normal biopsy tissue. Other biomarkers are currently in the validation phase and encompass combinations of multiple genes. SUMMARY: Epigenetic changes improve the specificity and sensitivity of PCa diagnosis and have the potential to help determine clinical prognosis. Additional studies will not only provide new and better biomarker candidates, but also have the potential to inform new therapeutic strategies given the reversibility of these processes.


Assuntos
Biomarcadores Tumorais/urina , Epigênese Genética , Neoplasias da Próstata/diagnóstico , Metilação de DNA , Histonas/metabolismo , Humanos , Masculino , Prognóstico , Próstata/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo
4.
Clin Epigenetics ; 12(1): 80, 2020 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-32503656

RESUMO

BACKGROUND: The chromatin insulator CCCTC-binding factor (CTCF) displays tissue-specific DNA binding sites that regulate transcription and chromatin organization. Despite evidence linking CTCF to the protection of epigenetic states through barrier insulation, the impact of CTCF loss on genome-wide DNA methylation sites in human cancer remains undefined. RESULTS: Here, we demonstrate that prostate and breast cancers within The Cancer Genome Atlas (TCGA) exhibit frequent copy number loss of CTCF and that this loss is associated with increased DNA methylation events that occur preferentially at CTCF binding sites. CTCF sites differ among tumor types and result in tissue-specific methylation patterns with little overlap between breast and prostate cancers. DNA methylation and transcriptome profiling in vitro establish that forced downregulation of CTCF leads to spatially distinct DNA hypermethylation surrounding CTCF binding sites, loss of CTCF binding, and decreased gene expression that is also seen in human tumors. DNA methylation inhibition reverses loss of expression at these CTCF-regulated genes. CONCLUSION: These findings establish CTCF loss as a major mediator in directing localized DNA hypermethylation events in a tissue-specific fashion and further support its role as a driver of the cancer phenotype.


Assuntos
Sítios de Ligação/genética , Fator de Ligação a CCCTC/genética , Metilação de DNA/genética , Neoplasias/genética , Neoplasias da Mama/genética , Fator de Ligação a CCCTC/metabolismo , Cromatina/metabolismo , Ilhas de CpG/genética , Regulação para Baixo/genética , Epigenômica/métodos , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Masculino , Fenótipo , Neoplasias da Próstata/genética
5.
Clin Epigenetics ; 11(1): 168, 2019 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-31779677

RESUMO

BACKGROUND: An epigenetic field of cancer susceptibility exists for prostate cancer (PC) that gives rise to multifocal disease in the peripheral prostate. In previous work, genome-wide DNA methylation profiling identified altered regions in the normal prostate tissue of men with PC. In the current multicenter study, we examined the predictive strength of a panel of loci to detect cancer presence and grade in patients with negative biopsy tissue. RESULTS: Four centers contributed benign prostate biopsy tissues blocks from 129 subjects that were either tumor associated (TA, Grade Group [GG] ≥ 2, n = 77) or non-tumor associated (NTA, n = 52). Biopsies were analyzed using pyrosequencing for DNA methylation encompassing CpG loci near CAV1, EVX1, FGF1, NCR2, PLA2G16, and SPAG4 and methylation differences were detected within all gene regions (p < 0.05). A multiplex regression model for biomarker performance incorporating a gene combination discriminated TA from NTA tissues (area under the curve [AUC] 0.747, p = 0.004). A multiplex model incorporating all the above genes and clinical information (PSA, age) identified patients with GG ≥ 2 PC (AUC 0.815, p < 0.0001). In patients with cancer, increased variation in gene methylation levels occurs between biopsies across the prostate. CONCLUSIONS: A widespread epigenetic field defect is utilized to detect GG ≥ 2 PC in patients with histologically negative biopsies. These alterations in non-tumor cells display increased heterogeneity of methylation extent and are spatially distant from tumor foci. These findings have the potential to decrease the need for repeated prostate biopsy.


Assuntos
Biomarcadores Tumorais/genética , Metilação de DNA , Neoplasias da Próstata/diagnóstico , Análise de Sequência de DNA/métodos , Idoso , Biópsia , Detecção Precoce de Câncer , Epigênese Genética , Redes Reguladoras de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
6.
Cancer Res ; 77(19): 5236-5247, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28775169

RESUMO

Loss of imprinting (LOI) is an epigenetic event that relaxes an allele-specific restriction on gene expression. One gene that experiences LOI is the paracrine insulin-like growth factor IGF2, which occurs commonly in human prostate tissues during aging and tumorigenesis. However, the relationship between IGF2 LOI and prostate tumorigenesis has not been established functionally. In this study, we created a mouse model with CTCF-binding site mutations at the Igf2-H19 imprint control region that abolishes CTCF insulator activity, resulting in biallelic Igf2 expression that mimics increased levels seen with aging-induced LOI. We found that Igf2 LOI increased the prevalence and severity of prostatic intraepithelial neoplasia (PIN), a premalignant lesion. Engineering Nkx3.1 deficiency into our model increased the frequency of PIN lesions in an additive fashion. Prostates harboring LOI displayed increased MAPK signaling and epithelial proliferation. In human prostate tissue arrays, we documented a positive correlation in benign tissues of IGF2 levels with phospho-ERK and phospho-AKT levels. Overall, our results establish that Igf2 LOI is sufficient on its own to increase rates of neoplastic development in the prostate by upregulating critical cancer-associated signaling pathways. Cancer Res; 77(19); 5236-47. ©2017 AACR.


Assuntos
Regulação Neoplásica da Expressão Gênica , Impressão Genômica/genética , Fator de Crescimento Insulin-Like II/genética , Próstata/patologia , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Animais , Apoptose , Proliferação de Células , Epigênese Genética , Feminino , Proteínas de Homeodomínio/fisiologia , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Próstata/metabolismo , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Fatores de Transcrição/fisiologia , Células Tumorais Cultivadas
7.
PLoS One ; 12(2): e0172048, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28234906

RESUMO

PURPOSE: Androgen deprivation therapy (ADT) commonly leads to incomplete cell death and the fate of persistent cells involves, in part, a senescent phenotype. Senescence is terminal growth arrest in response to cell stress that is characterized by increased lysosomal-ß-galactosidase (GLB1) the origin of senescence associated-ß-gal activity (SA-ß-gal). In the current study senescence is examined in vivo after ADT use in a neoadjuvant trial. METHODS AND MATERIALS: Tissue microarrays were generated from prostate cancer specimens (n = 126) from a multicenter neoadjuvant ADT trial. Arrays were subjected to multiplexed immunofluorescent staining for GLB1, Ki67, cleaved caspase 3 (CC3) and E-cadherin. Automated quantitative imaging was performed using Vectra™ and expression correlated with clinicopathologic features. RESULTS: Tissue was analyzed from 59 patients treated with neoadjuvant ADT and 67 receiving no therapy preoperatively. Median follow-up was 85.3 mo and median ADT treatment was 5 mo. In PC treated with neoadjuvant ADT, GLB1 expression increased in intermediate Gleason score (GS 6-7; p = 0.001), but not high grade (GS 8-10) cancer. Significantly higher levels of GLB1 were seen in tissues undergoing neoadjuvant ADT longer than 5 months compared to untreated tissues (p = 0.002). In contrast, apoptosis significantly increased earlier (1-4 mo) after ADT treatment (p<0.5). CONCLUSIONS: Increased GLB1 after neoadjuvant ADT occurs primarily among more clinically favorable intermediate grade cancers and enrichment of the phenotype occurs in a temporally prolonged fashion. Senescence may explain the persistence of PCa cells after ADT. Given concerns for the detrimental longer term presence of senescent cells, targeting these cells for removal may improve outcomes.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Senescência Celular , Terapia Neoadjuvante/métodos , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Antígenos CD , Apoptose , Caderinas/metabolismo , Caspase 3/metabolismo , Proliferação de Células , Humanos , Masculino , Gradação de Tumores , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Antígeno Prostático Específico/sangue , Análise Serial de Tecidos , beta-Galactosidase/metabolismo
8.
ACS Chem Biol ; 12(11): 2804-2814, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28949514

RESUMO

There is a tremendous need for novel strategies aimed at directly assessing activities of histone modifiers to probe epigenetic determinants associated with disease progression. Here, we developed a high-throughput peptide microarray assay to identify altered histone lysine (de)acetylation activity in prostate cancer (PCa). This microarray-based activity assay revealed up-regulated histone acetyltransferase (HAT) activity against specific histone H3 sites in a castrate-resistant (CR) PCa cell line compared to its hormone-sensitive (HS) isogenic counterpart. NAD+-dependent deacetylation assays revealed down-regulated sirtuin activity in validated CR lines. Levels of acetyltransferases GCN5, PCAF, CBP, and p300 were unchanged between matched HS and CR cell lines. However, autoacetylation of p300 at K1499, a modification known to enhance HAT activity and a target of deacetylation by SIRT2, was highly elevated in CR cells, while SIRT2 protein level was reduced in CR cells. Interrogation of HS and matched CR xenograft lines reveals that H3K18 hyperacetylation, increased p300 activity, and decreased SIRT2 expression are associated with progression to CR in 8/12 (66%). Tissue microarray analysis revealed that hyperacetylation of H3K18 is a feature of CRPC. Inhibition of p300 results in lower H3K18ac levels and increased expression of androgen receptors. Thus, a novel histone array identifies altered enzyme activities during the progression to CRPC and may be utilized in a personalized medicine approach. Reduced SIRT2 expression and increased p300 activity lead to a concerted mechanism of hyperacetylation at specific histone lysine sites (H3K9, H3K14, and H3K18) in CRPC.


Assuntos
Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Neoplasias da Próstata/enzimologia , Sirtuínas/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Histonas/genética , Humanos , Masculino , Camundongos , Orquiectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Análise Serial de Proteínas , Sirtuína 2/metabolismo , Xenopus
9.
PLoS One ; 10(4): e0124366, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25876105

RESUMO

PURPOSE: Senescence is a terminal growth arrest that functions as a tumor suppressor in aging and precancerous cells and is a response to selected anticancer compounds. Lysosomal-ß-galactosidase (GLB1) hydrolyzes ß-galactose from glycoconjugates and is the origin of senescence-associated ß-gal activity (SA-ß-gal). Using a new GLB1 antibody, senescence biology was investigated in prostate cancer (PCa) tissues. EXPERIMENTAL DESIGN: In vitro characterization of GLB1 was determined in primary prostate epithelial cell cultures passaged to replicative senescence and in therapy-induced senescence in PCa lines using chemotherapeutic agents. FFPE tissue microarrays were subjected to immunofluorescent staining for GLB1, Ki67 and HP1γ and automated quantitative imaging initially using AQUA in exploratory samples and Vectra in a validation series. RESULTS: GLB1 expression accumulates in replicative and induced senescence and correlates with senescent morphology and P16 (CDKN2) expression. In tissue arrays, quantitative imaging detects increased GLB1 expression in high-grade prostatic intraepithelial neoplasia (HGPIN), known to contain senescent cells, and cancer compared to benign prostate tissues (p<0.01) and senescent cells contain low Ki67 and elevated HP1γ. Within primary tumors, elevated GLB1 associates with lower T stage (p=0.01), localized versus metastatic disease (p=0.0003) and improved PSA-free survival (p=0.03). Increased GLB1 stratifies better PSA-free survival in intermediate grade PCa (0.01). Tissues that elaborate higher GLB1 display increased uniformity of expression. CONCLUSION: Increased GLB1 is a valuable marker in formalin-fixed paraffin-embedded (FFPE) tissues for the senescence-like phenotype and associates with improved cancer outcomes. This protein addresses a lack of senescence markers and should be applicable to study the biologic role of senescence in other cancers.


Assuntos
Regulação Neoplásica da Expressão Gênica , Calicreínas/genética , Recidiva Local de Neoplasia/genética , Antígeno Prostático Específico/genética , Neoplasias da Próstata/genética , beta-Galactosidase/genética , Idoso , Antineoplásicos/farmacologia , Aziridinas/farmacologia , Benzoquinonas/farmacologia , Biomarcadores/metabolismo , Senescência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Formaldeído , Humanos , Calicreínas/metabolismo , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Inclusão em Parafina , Cultura Primária de Células , Próstata/efeitos dos fármacos , Próstata/metabolismo , Próstata/patologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Transdução de Sinais , Análise de Sobrevida , Fixação de Tecidos , beta-Galactosidase/metabolismo
10.
Neoplasia ; 16(12): 1018-27, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25499215

RESUMO

Abnormal expression and function of chromatin regulators results in the altered chromatin structure seen in cancer. The chromatin regulator CTCF, its cofactor CHD8, and antagonistic paralogue BORIS have wide-ranging effects on gene regulation. Their concurrent expression and regulation was examined in benign, localized, and metastatic prostate cancer (PCa) arrays with extended follow-up using an automated quantitative imaging system, VECTRA. Epithelial staining was quantified and compared against a range of clinicopathologic variables. CHD8 expression was decreased in HGPIN, localized, and metastatic PCa compared to benign (P < .001). CHD8 promoter hypermethylation, assessed by Quantitative Pyrosequencing, occurred in over 45% of primary cancers in this population as well as the TGCA database. Treatment of cell lines with the demethylating agent 5-Aza-2'-deoxycytidine reinduced expression. An interesting dichotomy for CHD8 was observed within primary cancers, with higher nuclear protein expression associated with adverse clinical outcomes including extracapsular extension (P = .007), presence of metastases (P = .025) and worse PSA-recurrence free survival (P = .048). CHD8 outperformed Gleason score and predicted biochemical failure within intermediate grade prostate cancers. The BORIS/CTCF expression ratio increased in localized (P = .03) and metastatic PCa (P = .006) and was associated with higher Gleason score (P = .02), increased tumor volume (P = .02) and positive margins (P = .04). Per cell heterogeneity of expression revealed all protein expression to be more heterogeneous in cancerous tissue (both P < .001), especially high grade (P < .01). In the first detailed analysis in cancer, a marked loss of CHD8 expression and increased BORIS/CTCF ratio indicate frequent disruption of CTCF and its effector genes in PCa.


Assuntos
Carcinoma in Situ/genética , Cromatina/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias da Próstata/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Fator de Ligação a CCCTC , Carcinoma in Situ/patologia , Ilhas de CpG/genética , Metilação de DNA , Humanos , Masculino , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas
11.
PLoS One ; 9(2): e88052, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24558376

RESUMO

Genomic imprinting is the allele-specific expression of a gene based on parental origin. Loss of imprinting(LOI) of Insulin-like Growth Factor 2 (IGF2) during aging is important in tumorigenesis, yet the regulatory mechanisms driving this event are largely unknown. In this study oxidative stress, measured by increased NF-κB activity, induces LOI in both cancerous and noncancerous human prostate cells. Decreased expression of the enhancer-blocking element CCCTC-binding factor(CTCF) results in reduced binding of CTCF to the H19-ICR (imprint control region), a major factor in the allelic silencing of IGF2. This ICR then develops increased DNA methylation. Assays identify a recruitment of the canonical pathway proteins NF-κB p65 and p50 to the CTCF promoter associated with the co-repressor HDAC1 explaining gene repression. An IκBα super-repressor blocks oxidative stress-induced activation of NF-κB and IGF2 imprinting is maintained. In vivo experiments using IκBα mutant mice with continuous NF-κB activation demonstrate increased IGF2 LOI further confirming a central role for canonical NF-κB signaling. We conclude CTCF plays a central role in mediating the effects of NF-κB activation that result in altered imprinting both in vitro and in vivo. This novel finding connects inflammation found in aging prostate tissues with the altered epigenetic landscape.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator de Crescimento Insulin-Like II/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo , Alelos , Animais , Fator de Ligação a CCCTC , Linhagem Celular Tumoral , Metilação de DNA , Epigênese Genética , Epigenômica , Inativação Gênica , Impressão Genômica , Humanos , Inflamação , Masculino , Camundongos , Mutação , Neoplasias da Próstata/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais
12.
Epigenetics ; 8(10): 1039-42, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23973940

RESUMO

We have developed a rapid and sensitive quantitative assay for the measurement of individual allelic ratios. This assay minimizes time and labor, the need for special restriction endonuclease enzymes for polymorphic sites, and avoids heteroduplex formation seen with traditional quantitative PCR-based methods. It has improved sensitivity compared to other methods and is capable of distinguishing 1% differences in allelic expression. This assay, termed Pyrosequencing for Imprinted Expression (PIE), involves the use of an intron-crossing PCR primer to generate the first PCR product. We applied the assay to analyze Insulin-like Growth Factor-2 (IGF2) imprinting in both human and mouse prostate tissues.


Assuntos
Expressão Gênica , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Análise de Sequência de DNA/métodos , Alelos , Animais , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Camundongos , Próstata/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA