Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(16): E2306-15, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27044077

RESUMEN

Current approaches in tissue engineering are geared toward generating tissue-specific stem cells. Given the complexity and heterogeneity of tissues, this approach has its limitations. An alternate approach is to induce terminally differentiated cells to dedifferentiate into multipotent proliferative cells with the capacity to regenerate all components of a damaged tissue, a phenomenon used by salamanders to regenerate limbs. 5-Azacytidine (AZA) is a nucleoside analog that is used to treat preleukemic and leukemic blood disorders. AZA is also known to induce cell plasticity. We hypothesized that AZA-induced cell plasticity occurs via a transient multipotent cell state and that concomitant exposure to a receptive growth factor might result in the expansion of a plastic and proliferative population of cells. To this end, we treated lineage-committed cells with AZA and screened a number of different growth factors with known activity in mesenchyme-derived tissues. Here, we report that transient treatment with AZA in combination with platelet-derived growth factor-AB converts primary somatic cells into tissue-regenerative multipotent stem (iMS) cells. iMS cells possess a distinct transcriptome, are immunosuppressive, and demonstrate long-term self-renewal, serial clonogenicity, and multigerm layer differentiation potential. Importantly, unlike mesenchymal stem cells, iMS cells contribute directly to in vivo tissue regeneration in a context-dependent manner and, unlike embryonic or pluripotent stem cells, do not form teratomas. Taken together, this vector-free method of generating iMS cells from primary terminally differentiated cells has significant scope for application in tissue regeneration.


Asunto(s)
Azacitidina/farmacología , Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Células Cultivadas , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Especificidad de Órganos/fisiología
2.
Carcinogenesis ; 37(1): 10-17, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26498247

RESUMEN

Lynch syndrome is an autosomal dominant disorder that predisposes carriers of DNA mismatch repair (MMR) gene mutations to early-onset cancer. Germline testing screens exons and splice sites for mutations, but does not examine introns or RNA transcripts for alterations. Pathogenic mutations have not been detected in ~30% of suspected Lynch syndrome cases with standard screening practices. We present a 38-year-old male with a clinicopathological and family history consistent with Lynch syndrome, including loss of MSH2 expression in his tumor. Germline testing revealed normal MSH2 coding sequence, splice sites and exon copy number, however, cDNA sequencing identified an aberrant MSH2 transcript lacking exons 2-6. An inversion PCR on germline DNA identified an ~18kb unbalanced, paracentric inversion within MSH2, with breakpoints in a long terminal repeat in intron 1 and an Alu repeat in intron 6. The 3' end of the inversion had a 1.2 kb deletion and an 8 bp insertion at the junction with intron 6. Screening of 55 additional Australian patients presenting with MSH2-deficient tumors who were negative in germline genetic tests for MSH2 mutations identified another inversion-positive patient. We propose an Alu-mediated recombination model to explain the origin of the inversion. Our study illustrates the potential value of cDNA screening to identify patients with cryptic MMR gene rearrangements, clarifies why standard testing may not detect some pathogenic alterations, and provides a genetic test for screening individuals with suspected Lynch syndrome that present with unexplained MSH2-deficient tumors.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Exones , Proteína 2 Homóloga a MutS/genética , Inversión de Secuencia , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Neoplasias Colorrectales Hereditarias sin Poliposis/sangre , Análisis Mutacional de ADN/métodos , ADN Complementario/sangre , ADN Complementario/genética , ADN de Neoplasias/sangre , ADN de Neoplasias/genética , Reordenamiento Génico , Mutación de Línea Germinal , Humanos , Masculino , Datos de Secuencia Molecular , Linaje
3.
PLoS Genet ; 9(7): e1003636, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935509

RESUMEN

Hypomethylating agents reactivate tumor suppressor genes that are epigenetically silenced in cancer. Inevitably these genes are resilenced, leading to drug resistance. Using the MLH1 tumor suppressor gene as a model, we showed that decitabine-induced re-expression was dependent upon demethylation and eviction of promoter nucleosomes. Following decitabine withdrawal, MLH1 was rapidly resilenced despite persistent promoter demethylation. Single molecule analysis at multiple time points showed that gene resilencing was initiated by nucleosome reassembly on demethylated DNA and only then was followed by remethylation and stable silencing. Taken together, these data establish the importance of nucleosome positioning in mediating resilencing of drug-induced gene reactivation and suggest a role for therapeutic targeting of nucleosome assembly as a mechanism to overcome drug resistance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Metilación de ADN/genética , Resistencia a Antineoplásicos/genética , Proteínas Nucleares/genética , Nucleosomas/genética , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/genética , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Decitabina , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Homólogo 1 de la Proteína MutL , Nucleosomas/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos
4.
Hum Mutat ; 36(6): 622-30, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25762362

RESUMEN

Lynch syndrome is a hereditary cancer syndrome caused by a constitutional mutation in one of the mismatch repair genes. The implementation of predictive testing and targeted preventative surveillance is hindered by the frequent finding of sequence variants of uncertain significance in these genes. We aimed to determine the pathogenicity of previously reported variants (c.-28A>G and c.-7C>T) within the MLH1 5'untranslated region (UTR) in two individuals from unrelated suspected Lynch syndrome families. We investigated whether these variants were associated with other pathogenic alterations using targeted high-throughput sequencing of the MLH1 locus. We also determined their relationship to gene expression and epigenetic alterations at the promoter. Sequencing revealed that the c.-28A>G and c.-7C>T variants were the only potentially pathogenic alterations within the MLH1 gene. In both individuals, the levels of transcription from the variant allele were reduced to 50% compared with the wild-type allele. Partial loss of expression occurred in the absence of constitutional epigenetic alterations within the MLH1 promoter. We propose that these variants may be pathogenic due to constitutional partial loss of MLH1 expression, and that this may be associated with intermediate penetrance of a Lynch syndrome phenotype. Our findings provide further evidence of the potential importance of noncoding variants in the MLH1 5'UTR in the pathogenesis of Lynch syndrome.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Desequilibrio Alélico , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Expresión Génica , Variación Genética , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Regiones no Traducidas 5' , Edad de Inicio , Islas de CpG , Metilación de ADN , Epigénesis Genética , Femenino , Estudios de Asociación Genética , Sitios Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Repeticiones de Microsatélite , Homólogo 1 de la Proteína MutL , Mutación , Linaje
5.
J Biol Chem ; 287(25): 21530-40, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22549788

RESUMEN

Although serum albumin has an established function as a transport protein, evidence is emerging that serum albumin may also have a role as a molecular chaperone. Using established techniques to characterize chaperone interactions, this study demonstrates that bovine serum albumin: 1) preferentially binds stressed over unstressed client proteins; 2) forms stable, soluble, high molecular weight complexes with stressed client proteins; 3) reduces the aggregation of client proteins when it is present at physiological levels; and 4) inhibits amyloid formation by both WT and L55P transthyretin. Although the antiaggregatory effect of serum albumin is maintained in the presence of physiological levels of Ca(2+) and Cu(2+), the presence of free fatty acids significantly alters this activity: stabilizing serum albumin at normal levels but diminishing chaperone-like activity at high concentrations. Moreover, here it is shown that depletion of albumin from human plasma leads to a significant increase in aggregation under physiologically relevant heat and shear stresses. This study demonstrates that serum albumin possesses chaperone-like properties and that this activity is maintained under a number of physiologically relevant conditions.


Asunto(s)
Amiloide/química , Chaperonas Moleculares/química , Albúmina Sérica Bovina/química , Amiloide/metabolismo , Animales , Bovinos , Humanos , Chaperonas Moleculares/metabolismo , Prealbúmina/química , Prealbúmina/metabolismo , Unión Proteica , Estabilidad Proteica , Albúmina Sérica Bovina/metabolismo
6.
J Proteome Res ; 11(7): 3561-72, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22587632

RESUMEN

Breast cancer is a molecularly heterogeneous disease, and predicting response to chemotherapy remains a major clinical challenge. To minimize adverse side-effects or cumulative toxicity in patients unlikely to benefit from treatment, biomarkers indicating treatment efficacy are critically needed. iTRAQ labeling coupled with multidimensional LC-MS/MS of the enriched mitochondria and endoplasmic reticulum fraction, key organelles regulating apoptosis, has led to the discovery of several differentially abundant proteins in breast cancer cells treated with the chemotherapeutic agent doxorubicin followed by the death receptor ligand, TRAIL, among 571 and 801 unique proteins identified in ZR-75-1 and MDA-MB-231 breast cancer cell lines, respectively. The differentially abundant proteins represent diverse biological processes associated with cellular assembly and organization, molecular transport, oxidative stress, cell motility, cell death, and cancer. Despite many differences in molecular phenotype between the two breast cancer cell lines, a comparison of their subproteomes following drug treatment revealed three proteins displaying common regulation: PPIB, AHNAK, and SLC1A5. Changes in these proteins, detected by iTRAQ, were confirmed by immunofluorescence, visualized by confocal microscopy. These novel potential biomarkers may have clinical utility for assessing response to cancer treatment and may provide insight into new therapeutic targets for breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Proteoma/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Sistema de Transporte de Aminoácidos ASC/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular , Quimioterapia Adyuvante , Ciclofilinas/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Proteínas de la Membrana/metabolismo , Redes y Vías Metabólicas , Antígenos de Histocompatibilidad Menor , Mitocondrias/metabolismo , Terapia Neoadyuvante , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo , Proteómica , Espectrometría de Masas en Tándem
7.
Sci Signal ; 15(719): eabg9782, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35104163

RESUMEN

Superresolution techniques have advanced our understanding of complex cellular structures and processes but require the attachment of fluorophores to targets through tags or antibodies, which can be bulky and result in underlabeling. To overcome these limitations, we developed a technique to visualize the nanoscale binding locations of signaling proteins by taking advantage of their native interaction domains. Here, we demonstrated that pPAINT (protein point accumulation in nanoscale topography) is a new, single-molecule localization microscopy (SMLM) technique and used it to investigate T cell signaling by visualizing the Src homology 2 (SH2) domain, which is common in signaling molecules. When SH2 domain-containing proteins relocate to the plasma membrane, the domains selectively, transiently, and reversibly bind to preferred phosphorylated tyrosine residues on receptors. This transient binding yields the stochastic blinking events necessary for SMLM when observed with total internal reflection microscopy and enables quantification of binding coefficients in intact cells. We used pPAINT to reveal the binding sites of several T cell receptor-proximal signaling molecules, including Zap70, PI3K, Grb2, Syk, Eat2, and SHP2, and showed that the probes could be multiplexed. We showed that the binding half-life of the tandem SH2 domain of PI3K correlated with binding site cluster size at the immunological synapses of T cells, but that longer binding lifetimes were associated with smaller clusters for the monovalent SH2 domain of Eat2. These results demonstrate the potential of pPAINT for investigating phosphotyrosine-mediated signaling processes at the plasma membrane.


Asunto(s)
Microscopía , Dominios Homologos src , Secuencia de Aminoácidos , Sitios de Unión , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica
8.
Clin Cancer Res ; 24(18): 4602-4611, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29898989

RESUMEN

Purpose:MLH1 is a major tumor suppressor gene involved in the pathogenesis of Lynch syndrome and various sporadic cancers. Despite their potential pathogenic importance, genomic regions capable of regulating MLH1 expression over long distances have yet to be identified.Experimental Design: Here, we use chromosome conformation capture (3C) to screen a 650-kb region flanking the MLH1 locus to identify interactions between the MLH1 promoter and distal regions in MLH1-expressing and nonexpressing cells. Putative enhancers were functionally validated using luciferase reporter assays, chromatin immunoprecipitation, and CRISPR-Cas9-mediated deletion of endogenous regions. To evaluate whether germline variants in the enhancer might contribute to impaired MLH1 expression in patients with suspected Lynch syndrome, we also screened germline DNA from a cohort of 74 patients with no known coding mutations or epimutations at the MLH1 promoter.Results: A 1.8-kb DNA fragment, 35 kb upstream of the MLH1 transcription start site enhances MLH1 gene expression in colorectal cells. The enhancer was bound by CTCF and CRISPR-Cas9-mediated deletion of a core binding region impairs endogenous MLH1 expression. A total of 5.4% of suspected Lynch syndrome patients have a rare single-nucleotide variant (G > A; rs143969848; 2.5% in gnomAD European, non-Finnish) within a highly conserved CTCF-binding motif, which disrupts enhancer activity in SW620 colorectal carcinoma cells.Conclusions: A CTCF-bound region within the MLH1-35 enhancer regulates MLH1 expression in colorectal cells and is worthy of scrutiny in future genetic screening strategies for suspected Lynch syndrome associated with loss of MLH1 expression. Clin Cancer Res; 24(18); 4602-11. ©2018 AACR.


Asunto(s)
Factor de Unión a CCCTC/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales/genética , Homólogo 1 de la Proteína MutL/genética , Adulto , Anciano , Anciano de 80 o más Años , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Metilación de ADN/genética , Reparación de la Incompatibilidad de ADN/genética , Elementos de Facilitación Genéticos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Mutación de Línea Germinal/genética , Humanos , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética
9.
Cancer Genet ; 209(11): 497-500, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27886675

RESUMEN

Lynch syndrome is a hereditary cancer syndrome caused by the autosomal dominant inheritance of loss-of-function mutations in DNA mismatch repair (MMR) genes. Approximately one quarter of clinically suspected cases have no identifiable germline mutation in any MMR gene, a condition known as Lynch-like syndrome (LLS). MCM9 was recently identified as the DNA helicase in the mammalian MMR complex and loss of helicase activity results in microsatellite instability. We hypothesized that pathogenic variants in MCM9 may account for LLS. The 5'UTR and coding region of MCM9 were sequenced in germline DNA of 109 Australian patients with LLS and variants were cross-referenced with three population-based databases (dbSNP144, 1000 Genomes, ExAC). The functional effect of variants was assessed in silico with PolyPhen-2, SIFT and CONDEL. Fifteen variants that included six common SNPs and nine variants of unknown significance (VUS) were identified. We conclude that VUS occur in MCM9 in a small proportion of LLS patients and MCM9 mutations are unlikely to explain most LLS cases.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Mutación de Línea Germinal , Proteínas de Mantenimiento de Minicromosoma/genética , Análisis de Secuencia de ADN/métodos , Regiones no Traducidas 5' , Adulto , Anciano , Anciano de 80 o más Años , Australia , Biología Computacional , Simulación por Computador , Bases de Datos Genéticas , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Sistemas de Lectura Abierta
10.
Mol Cancer Res ; 14(12): 1217-1228, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27671336

RESUMEN

Laterally spreading tumors (LST) are colorectal adenomas that develop into extremely large lesions with predominantly slow progression to cancer, depending on lesion subtype. Comparing and contrasting the molecular profiles of LSTs and colorectal cancers offers an opportunity to delineate key molecular alterations that drive malignant transformation in the colorectum. In a discovery cohort of 11 LSTs and paired normal mucosa, we performed a comprehensive and unbiased screen of the genome, epigenome, and transcriptome followed by bioinformatics integration of these data and validation in an additional 84 large, benign colorectal lesions. Mutation rates in LSTs were comparable with microsatellite-stable colorectal cancers (2.4 vs. 2.6 mutations per megabase); however, copy number alterations were infrequent (averaging only 1.5 per LST). Frequent genetic, epigenetic, and transcriptional alterations were identified in genes not previously implicated in colorectal neoplasia (ANO5, MED12L, EPB41L4A, RGMB, SLITRK1, SLITRK5, NRXN1, ANK2). Alterations to pathways commonly mutated in colorectal cancers, namely, the p53, PI3K, and TGFß pathways, were rare. Instead, LST-altered genes converged on axonal guidance, Wnt, and actin cytoskeleton signaling. These integrated omics data identify molecular features associated with noncancerous LSTs and highlight that mutation load, which is relatively high in LSTs, is a poor predictor of invasive potential. IMPLICATIONS: The novel genetic, epigenetic, and transcriptional changes associated with LST development reveal important insights into why some adenomas do not progress to cancer. The finding that LSTs exhibit a mutational load similar to colorectal carcinomas has implications for the validity of molecular biomarkers for assessing cancer risk. Mol Cancer Res; 14(12); 1217-28. ©2016 AACR.


Asunto(s)
Adenoma/genética , Neoplasias Colorrectales/genética , Redes Reguladoras de Genes , Genómica/métodos , Biología Computacional/métodos , Metilación de ADN , Epigénesis Genética , Femenino , Dosificación de Gen , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Estudio de Asociación del Genoma Completo/métodos , Humanos , Mutación , Análisis de Secuencia de ARN/métodos
11.
JAMA Oncol ; 1(7): 953-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26181641

RESUMEN

IMPORTANCE: Constitutional hypermethylation of 1 allele throughout the soma (constitutional epimutation) is an accepted mechanism of cancer predisposition. Understanding the origin and inheritance of epimutations is important for assessing cancer risk in affected families. OBSERVATIONS: We report a 29-year-old man with early-onset colorectal cancer who showed a constitutional MLH1 epimutation (approximately 50% of alleles methylated and allele-specific loss of MLH1 expression) that was stable over a 16-year period. The epimutation was inherited without a genetic alteration from his asymptomatic mother. She showed methylation on the same allele but in less than 5% of her somatic cells. CONCLUSIONS AND RELEVANCE: These findings indicate that low-level somatic mosaicism for an epimutation in an asymptomatic parent can produce a nonmosaic constitutional epimutation in a child. Asymptomatic low-level methylation in some individuals may be associated with substantial cancer risk to their offspring.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Adenocarcinoma/genética , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Epigénesis Genética , Mosaicismo , Proteínas Nucleares/genética , Adenocarcinoma/patología , Adolescente , Adulto , Anciano , Neoplasias Colorrectales/patología , Metilación de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Herencia , Humanos , Masculino , Homólogo 1 de la Proteína MutL , Linaje , Fenotipo , Medición de Riesgo , Factores de Riesgo , Adulto Joven
12.
Clin Epigenetics ; 6(1): 32, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25530820

RESUMEN

BACKGROUND: Splicing is more efficient when coupled with transcription and it has been proposed that nucleosomes enriched in exons are important for splice site recognition. Lynch syndrome is a familial cancer syndrome that can be caused by the autosomal dominant inheritance of splice site mutations in the MutL homolog 1 (MLH1) gene. To better understand the role of nucleosomes in splicing, we used MLH1 splice site mutations in Lynch syndrome cases as a model to investigate if abnormal splicing was associated with altered nucleosome positioning at exon-intron boundaries. FINDINGS: Nucleosome Occupancy and Methylome sequencing (NOMe-seq) was used to determine the allele-specific positioning of nucleosomes around heterozygous splice site mutations in lymphoblastoid cells lines (LCLs) derived from six Lynch syndrome patients. These mutations were previously shown to cause exon skipping in five of the six patients. Allele-specific high-resolution nucleosome mapping across exons and exon-intron boundaries revealed high levels of nucleosomes across all regions examined. Alleles containing donor or acceptor splice site mutations showed no consistent alteration in nucleosome positioning or occupancy. CONCLUSION: Nucleosomes were enriched at MLH1 exons in LCLs derived from Lynch syndrome patients, and in this model system the positioning of nucleosomes was unaltered at exon-intron boundaries containing splice site mutations. Thus, these splice site mutations alone do not significantly change the local organisation of nucleosomes.

13.
Epigenetics ; 9(10): 1422-30, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25437056

RESUMEN

Gene silencing in cancer frequently involves hypermethylation and dense nucleosome occupancy across promoter regions. How a promoter transitions to this silent state is unclear. Using colorectal adenomas, we investigated nucleosome positioning, DNA methylation, and gene expression in the early stages of gene silencing. Genome-wide gene expression correlated with highly positioned nucleosomes upstream and downstream of a nucleosome-depleted transcription start site (TSS). Hypermethylated promoters displayed increased nucleosome occupancy, specifically at the TSS. We investigated 2 genes, CDH1 and CDKN2B, which were silenced in adenomas but lacked promoter hypermethylation. Instead, silencing correlated with loss of nucleosomes from the -2 position upstream of the TSS relative to normal mucosa. In contrast, permanent CDH1 silencing in carcinoma cells was characterized by promoter hypermethylation and dense nucleosome occupancy. Our findings suggest that silenced genes transition through an intermediary stage involving altered promoter nucleosome positioning, before permanent silencing by hypermethylation and dense nucleosome occupancy.


Asunto(s)
Metilación de ADN , Silenciador del Gen , Nucleosomas/genética , Regiones Promotoras Genéticas , Sitio de Iniciación de la Transcripción , Adenoma/genética , Anciano , Antígenos CD , Cadherinas/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Epigénesis Genética , Femenino , Humanos , Masculino , Persona de Mediana Edad
14.
Epigenetics ; 9(8): 1092-100, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24837038

RESUMEN

In mouse models, loss of the candidate tumor suppressor gene Ubiquitin Specific Protease 44 (USP44) is associated with aneuploidy and cancer. USP44 is also transcriptionally silenced in human cancers. Here we investigated the molecular mechanism of USP44 silencing and whether this correlated with aneuploidy in colorectal adenomas. DNA methylation at the USP44 CpG island (CGI) promoter was measured using combined bisulfite restriction analysis (COBRA) in colorectal cancer (CRC) cell lines (n = 18), and with COBRA and bisulfite sequencing in colorectal adenomas (n = 89) and matched normal colonic mucosa (n = 51). The USP44 CGI was hypermethylated in all CRC cell lines, in most colorectal adenomas (79 of 89, 89%) but rarely in normal mucosa samples (3 of 51, 6%). USP44 expression was also compared between normal mucosa and paired hypermethylated adenomas in six patients using qRT-PCR. Hypermethylation of the USP44 CGI in adenomas was associated with a 1.8 to 5.5-fold reduction in expression compared with paired normal mucosa. Treatment of CRC cell lines with the DNA hypomethylating agent decitabine resulted in a 14 to 270-fold increase in USP44 expression. Whole genome SNP array data showed that gain or loss of individual chromosomes occurred in adenomas, but hypermethylation did not correlate with more aneuploidy. In summary, our data shows that USP44 is epigenetically inactivated in colorectal adenomas, but this alone is not sufficient to cause aneuploidy in colorectal neoplasia.


Asunto(s)
Adenoma/genética , Aneuploidia , Neoplasias Colorrectales/genética , Epigénesis Genética , Proteasas Ubiquitina-Específicas/genética , Adenoma/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Islas de CpG , Decitabina , Expresión Génica , Genes Supresores de Tumor , Humanos , Mucosa Intestinal/metabolismo , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Recto/metabolismo , Ubiquitina Tiolesterasa , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA