Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nano Lett ; 21(21): 9061-9068, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34672610

RESUMEN

Cell-free DNA (cfDNA) analysis, specifically circulating tumor DNA (ctDNA) analysis, provides enormous opportunities for noninvasive early assessment of cancers. To date, PCR-based methods have led this field. However, the limited sensitivity/specificity of PCR-based methods necessitates the search for new methods. Here, we describe a direct approach to detect KRAS G12D mutated genes in clinical ctDNA samples with the utmost LOD and sensitivity/specificity. In this study, MutS protein was immobilized on the tip of an atomic force microscope (AFM), and the protein sensed the mismatched sites of the duplex formed between the capture probe on the surface and mutated DNA. A noteworthy LOD (3 copies, 0.006% allele frequency) was achieved, along with superb sensitivity/specificity (100%/100%). These observations demonstrate that force-based AFM, in combination with the protein found in nature and properly designed capture probes/blockers, represents an exciting new avenue for ctDNA analysis.


Asunto(s)
ADN Tumoral Circulante , Neoplasias , Biomarcadores de Tumor/genética , ADN Tumoral Circulante/genética , Humanos , Mutación , Mutación Puntual , Sensibilidad y Especificidad
2.
Biochem Biophys Res Commun ; 558: 209-215, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32958251

RESUMEN

Tumor heterogeneity is one of the ongoing huddles in the field of colon cancer therapy. It is evident that there are countless clones which exhibit different phenotypes and therefore, single cell analysis is inevitable. Cancer stem cells (CSCs) are rare cell population within tumor which is known to function in cancer metastasis and recurrence. Although there have been trials to prove intra-tumoral heterogeneity using single cell sequencing, that of CSCs has not been clearly elucidated. Here, we articulate the presence of heterogeneous subclones within CD133 positive cancer stem cells through single cell sequencing. As a proof of principle, we performed phenotype-based high-throughput laser isolation and single cell sequencing (PHLI-seq) of CD133 positive cells in a frozen tumor tissue obtained from a patient with colorectal cancer. The result proved that CD133 positive cells were shown to be heterogeneous both in copy number and mutational profiles. Single cancer stem cell specific mutations such as RNF144A, PAK2, PARP4, ADAM21, HYDIN, KRT38 and CELSR1 could be also detected in liver metastatic tumor of the same patient. Collectively, these data suggest that single cell analysis used to spot subclones with genetic variation within rare population, will lead to new strategies to tackle colon cancer metastasis.


Asunto(s)
Antígeno AC133/metabolismo , Células Madre Neoplásicas/clasificación , Células Madre Neoplásicas/metabolismo , Anciano , Biomarcadores de Tumor/metabolismo , Separación Celular/métodos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Dosificación de Gen , Humanos , Rayos Láser , Masculino , Mutación , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Células Madre Neoplásicas/patología , Fenotipo , Análisis de la Célula Individual , Secuenciación del Exoma
3.
Int J Cancer ; 144(2): 389-401, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29978469

RESUMEN

PIK3CA is a frequently mutated gene in cancer, including about ~15 to 20% of colorectal cancers (CRC). PIK3CA mutations lead to activation of the PI3K/AKT/mTOR signaling pathway, which plays pivotal roles in tumorigenesis. Here, we investigated the mechanism of resistance of PIK3CA-mutant CRC cell lines to gedatolisib, a dual PI3K/mTOR inhibitor. Out of a panel of 29 CRC cell lines, we identified 7 harboring one or more PIK3CA mutations; of these, 5 and 2 were found to be sensitive and resistant to gedatolisib, respectively. Both of the gedatolisib-resistant cell lines expressed high levels of active glycogen synthase kinase 3-beta (GSK3ß) and harbored the same frameshift mutation (c.465_466insC; H155fs*) in TCF7, which encodes a positive transcriptional regulator of the WNT/ß-catenin signaling pathway. Inhibition of GSK3ß activity in gedatolisib-resistant cells by siRNA-mediated knockdown or treatment with a GSK3ß-specific inhibitor effectively reduced the activity of molecules downstream of mTOR and also decreased signaling through the WNT/ß-catenin pathway. Notably, GSK3ß inhibition rendered the resistant cell lines sensitive to gedatolisib cytotoxicity, both in vitro and in a mouse xenograft model. Taken together, these data demonstrate that aberrant regulation of WNT/ß-catenin signaling and active GSK3ß induced by the TCF7 frameshift mutation cause resistance to the dual PI3K/mTOR inhibitor gedatolisib. Cotreatment with GSK3ß inhibitors may be a strategy to overcome the resistance of PIK3CA- and TCF7-mutant CRC to PI3K/mTOR-targeted therapies.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos/fisiología , Morfolinas/farmacología , Triazinas/farmacología , Vía de Señalización Wnt/fisiología , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Humanos , Ratones , Mutación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
4.
EMBO Rep ; 17(9): 1343-59, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27466323

RESUMEN

Epithelial to mesenchymal transition (EMT) and mesenchymal to epithelial transition (MET) are important interconnected events in tumorigenesis controlled by complex genetic networks. However, the cues that activate EMT-initiating factors and the mechanisms that reversibly connect EMT/MET are not well understood. Here, we show that cohesin-mediated chromatin organization coordinates EMT/MET by regulating mesenchymal genes. We report that RAD21, a subunit of the cohesin complex, is expressed in epithelial breast cancer cells, whereas its expression is decreased in mesenchymal cancer. Depletion of RAD21 in epithelial cancer cells causes transcriptional activation of TGFB1 and ITGA5, inducing EMT. Reduced binding of RAD21 changes intrachromosomal chromatin interactions within the TGFB1 and ITGA5 loci, creating an active transcriptional environment. Similarly, stem cell-like cancer cells also show an open chromatin structure at both genes, which correlates with high expression levels and mesenchymal fate characteristics. Conversely, overexpression of RAD21 in mesenchymal cancer cells induces MET-specific expression patterns. These findings indicate that dynamic cohesin-mediated chromatin structures are responsible for the initiation and regulation of essential EMT-related cell fate changes in cancer.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Transición Epitelial-Mesenquimal/genética , Neoplasias/genética , Neoplasias/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Neoplasias/patología , Proteínas Nucleares/genética , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Factor de Crecimiento Transformador beta1/genética , Cohesinas
5.
Xenotransplantation ; 25(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29135052

RESUMEN

BACKGROUND: Porcine islet xenotransplantation is considered an attractive alternative treatment for type 1 diabetes mellitus. However, it is largely limited because of initial rejection due to Instant Blood-Mediated Inflammatory Reaction (IBMIR), oxidative stress, and inflammatory responses. Recently, soluble tumor necrosis factor-ɑ receptor type I (sTNF-αR) and heme oxygenase (HO)-1 genes (HO-1/sTNF-αR) have been shown to improve the viability and functionality of porcine islets after transplantation. METHODS: In this study, genetically modified mesenchymal stem cells (MSCs) expressing the HO-1/sTNF-αR genes (HO-1/sTNF-αR-MSC) were developed using an adenoviral system, and porcine islet viability and function were confirmed by in vitro tests such as GSIS, AO/PI, and the ADP/ATP ratio after coculturing with HO-1/sTNF-αR-MSCs. Subsequently, isolated porcine islets were transplanted underneath the kidney capsule of diabetic humanized mice without MSCs, with MSCs or with HO-1/sTNF-αR-MSCs. RESULTS: According to the results, the HO-1/sTNF-αR-MSC-treated group exhibited improved survival of porcine islets and could reverse hyperglycemia more than porcine islets not treated with MSCs or islets cotransplanted with MSCs. Moreover, the HO-1/sTNF-αR-MSC group maintained its morphological characteristics and the insulin secretion pattern of transplanted porcine islets similar to endogenous islets in immunocompetent humanized mice. CONCLUSIONS: Our results suggest that HO-1/sTNF-αR-MSCs are efficient tools for porcine islet xenotransplantation, and this study may provide basic information for pre-clinical animal models and future clinical trials of porcine islet xenotransplantation.


Asunto(s)
Supervivencia de Injerto , Hemo-Oxigenasa 1/genética , Xenoinjertos/inmunología , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/citología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Técnicas de Cocultivo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/terapia , Supervivencia de Injerto/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones Transgénicos , Trasplante Heterólogo/métodos
6.
Nucleic Acids Res ; 44(2): 558-72, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26420833

RESUMEN

Gene amplification is a hallmark of cancer with chromosomal instability although the underlying mechanism by which altered copy numbers are maintained is largely unclear. Cohesin, involved in sister chromatid cohesion, DNA repair, cell cycle progression and transcriptional regulation of key developmental genes, is frequently overexpressed in human cancer. Here we show that cohesin-dependent change in DNA replication controls the copy numbers of amplified genes in cancer cells with chromosomal instability. We found that the down-regulation of elevated cohesin leads to copy number-associated gene expression changes without disturbing chromosomal segregation. Highly amplified genes form typical long-range chromatin interactions, which are stabilized by enriched cohesin. The spatial proximities among cohesin binding sites within amplified genes are decreased by RAD21-knockdown, resulting in the rapid decline of amplified gene expression. After several passages, cohesin depletion inhibits DNA replication initiation by reducing the recruitment of pre-replication complexes such as minichromosome maintenance subunits 7 (MCM7), DNA polymerase α, and CDC45 at replication origins near the amplified regions, and as a result, decreases the DNA copy numbers of highly amplified genes. Collectively, our data demonstrate that cohesin-mediated chromatin organization and DNA replication are important for stabilizing gene amplification in cancer cells with chromosomal instability.


Asunto(s)
Inestabilidad Cromosómica , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Fosfoproteínas/genética , Neoplasias Gástricas/genética , Sitios de Unión , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromátides/química , Cromátides/metabolismo , Cromatina/química , Cromatina/metabolismo , Segregación Cromosómica , Hibridación Genómica Comparativa , ADN Polimerasa I/genética , ADN Polimerasa I/metabolismo , Replicación del ADN , Proteínas de Unión al ADN , Dosificación de Gen , Células HCT116 , Células Hep G2 , Humanos , Componente 7 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
7.
Am J Pathol ; 185(3): 666-78, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25576785

RESUMEN

Recent genome-wide epigenomic and transcription profiling studies have demonstrated that epigenetic silencing can encompass multiple neighboring genes, termed as long-range epigenetic silencing (LRES). Herein, we identified a novel LRES region by comparing gene expression of human colon cancer HCT116 cells with their DNA methyltransferase 1 and DNA methyltransferase 3B double-knockout derivative double-knockout cells. Ten consecutive genes spanning 3 Mb of chromosome 15q25 were coordinately silenced, with eight genes showing promoter CpG island hypermethylation and enrichment of repressive histone marks, which were evaluated by bisulfite sequencing analysis and chromatin immunoprecipitation assay. Comparison of primary gastric tumor specimens with normal tissue confirmed that the long-range silencing of this region was tumor specific. Methylation of genes within the LRES region was evaluated in 190 gastric tumor tissues using the MethyLight assay, and their association with clinicopathological features, such as older age, high-grade differentiation, and diffuse or mixed-type histology, was determined. LRES-positive gastric cancer patients (six or more methylated genes) showed lower recurrence and better survival. Our findings emphasize the differential dynamics of DNA methylation and histone modification, indicating the importance of studying the relationship of each epigenetic modification in the context of chromatin domains. Patients with LRES showed lower recurrence and better prognosis, indicating that stratifying patients according to underlying molecular features, such as LRES regions, may better predict recurrence and survival.


Asunto(s)
Cromosomas Humanos Par 15 , Epigénesis Genética , Silenciador del Gen , Recurrencia Local de Neoplasia/genética , Neoplasias Gástricas/genética , Adulto , Anciano , Islas de CpG , Metilación de ADN , Femenino , Histonas , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Regiones Promotoras Genéticas , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Tasa de Supervivencia , Adulto Joven
8.
Clin Transplant ; 30(9): 1146-51, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27411211

RESUMEN

BACKGROUND: Portal vein thrombosis (PVT) is a relative contraindication in living donor liver transplantation (LDLT). We investigated the long-term outcome of adult patients with PVT in LDLT. METHODS: Between 2004 and 2009, 471 cases of adult LDLT were performed and 56 patients had PVT (11.8%). Thrombectomy was attempted using a modified eversion technique. We evaluated the outcome of patients with PVT according to grade and compared with no-PVT patients. RESULTS: There was no difference in terms of age, gender, Child-Pugh score, MELD score, proportion of malignance, operation time, and total amount of transfused blood. Complete thrombectomy was successful in 73.2% (41/56), partial thrombectomy in 26.8% (15/56), and one case needed jump graft for portal vein reconstruction. Among patients with partial thrombectomy, when the PV velocity was above 20 cm/s, the remnant thrombus disappeared in 46%. The rate of PV complication was statistically not different (8.9% vs 3.4%, P=.062). Five-year survival of mild PVT was 69.3%, 60.6% for severe PVT, and 80.4% for no-PVT (P=.059). CONCLUSIONS: Eversion thrombectomy by modified technique is feasible in most cases of PVT. Good long-term outcome may be expected in LDLT with PVT.


Asunto(s)
Enfermedad Hepática en Estado Terminal/complicaciones , Trasplante de Hígado/métodos , Donadores Vivos , Procedimientos de Cirugía Plástica/métodos , Vena Porta , Trombectomía/métodos , Trombosis de la Vena/etiología , Aloinjertos , Enfermedad Hepática en Estado Terminal/cirugía , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Trombosis de la Vena/diagnóstico , Trombosis de la Vena/cirugía
9.
Breast Cancer Res ; 17: 33, 2015 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-25888415

RESUMEN

INTRODUCTION: Olaparib, a poly (ADP-ribose) polymerase (PARP) inhibitor, has been found to have therapeutic potential for treating cancers associated with impaired DNA repair capabilities, particularly those with deficiencies in the homologous recombination repair (HRR) pathway. Histone deacetylases (HDACs) are important for enabling functional HRR of DNA by regulating the expression of HRR-related genes and promoting the accurate assembly of HRR-directed sub-nuclear foci. Thus, HDAC inhibitors have recently emerged as a therapeutic agent for treating cancer by inhibiting DNA repair. Based on this, HDAC inhibition could be predicted to enhance the anti-tumor effect of PARP inhibitors in cancer cells by blocking the HRR pathway. METHODS: We determined whether suberoylanilide hydroxamic acid (SAHA), a HDAC inhibitor, could enhance the anti-tumor effects of olaparib on breast cancer cell lines using a cytotoxic assay, cell cycle analysis, and Western blotting. We evaluated how exposure to SAHA affects the expression of HRR-associated genes. The accumulation of DNA double strand breaks (DSBs) induced by combination treatment was assessed. Induction of autophagy was monitored by imaging green fluorescent protein-tagged microtubule-associated protein 1A/1B-light chain 3 (LC3) expression following co-treatment with olaparib and SAHA. These in vitro data were validated in vivo using a human breast cancer xenograft model. RESULTS: Triple-negative breast cancer cell (TNBC) lines showed heterogeneous responses to the PARP and HDAC inhibitors. Co-administration of olaparib and SAHA synergistically inhibited the growth of TNBC cells that expressed functional Phosphatase and tensin homolog (PTEN). This effect was associated with down-regulation of the proliferative signaling pathway, increased apoptotic and autophagic cell death, and accumulation of DNA damage. The combined anti-tumor effect of olaparib and SAHA was also observed in a xenograft model. These data suggest that PTEN expression in TNBC cells can sensitize the cell response to simultaneous inhibition of PARP and HDAC both in vitro and in vivo. CONCLUSION: Our findings suggest that expression of functional PTEN may serve as a biomarker for selecting TNBC patients that would favorably respond to a combination of olaparib with SAHA. This provides a strong rationale for treating TNBC patients with PTEN expression with a combination therapy consisting of olaparib and SAHA.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Apoptosis , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Femenino , Expresión Génica , Humanos , Ratones , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Blood ; 121(22): 4575-85, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23610375

RESUMEN

Erythropoiesis is dependent on the lineage-specific transcription factors Gata1, Tal1, and Klf1. Several erythroid genes have been shown to require all 3 factors for their expression, suggesting that they function synergistically; however, there is little direct evidence for widespread cooperation. Gata1 and Tal1 can assemble within higher-order protein complexes (Ldb1 complexes) that include the adapter molecules Lmo2 and Ldb1. Ldb1 proteins are capable of coassociation, and long-range Ldb1-mediated oligomerization of enhancer- and promoter-bound Ldb1 complexes has been shown to be required for ß-globin gene expression. In this study, we generated a genomewide map of Ldb1 complex binding sites that revealed widespread binding at erythroid genes and at known erythroid enhancer elements. Ldb1 complex binding sites frequently colocalized with Klf1 binding sites and with consensus binding motifs for other erythroid transcription factors. Transcriptomic analysis demonstrated a strong correlation between Ldb1 complex binding and Ldb1 dependency for gene expression and identified a large cohort of genes coregulated by Ldb1 complexes and Klf1. Together, these results provide a foundation for defining the mechanism and scope of Ldb1 complex activity during erythropoiesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Eritroides/metabolismo , Factor de Transcripción GATA1/genética , Proteínas con Dominio LIM/genética , Transcripción Genética/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Células Eritroides/citología , Eritropoyesis/genética , Eritropoyesis/fisiología , Factor de Transcripción GATA1/metabolismo , Regulación de la Expresión Génica/fisiología , Prueba de Complementación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas con Dominio LIM/metabolismo , Leucemia Eritroblástica Aguda , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda
11.
Clin Transplant ; 28(1): 141-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24372624

RESUMEN

Liver transplantation (LT) is performed in patients with hepatocellular carcinoma (HCC), but recurrent HCC after LT remains a problem. We retrospectively reviewed the data from 63 patients with recurrent HCC who underwent LT at a single institution between September 1996 and March 2011 to determine the prognosis of patients with recurrent HCC after LT. A survival analysis was performed with the preoperative data, histological findings, patterns of recurrence, and treatment methods. Univariate and multivariate analyses were performed to determine the factors associated with early (<1 yr) cancer-related death. The independent prognostic factors, according to the multivariate analysis, were recurrence within six months (hazards ratio [HR] = 4.557, p = 0.021) and initial multiple-organ involvement (HR = 5.494, p = 0.015). The survival rates of patients differed according to the treatment type. The combined treatment with local and systemic treatment resulted in increased survival even in patients with HCC recurrences involving multiple organs.


Asunto(s)
Carcinoma Hepatocelular/cirugía , Neoplasias Hepáticas/cirugía , Recurrencia Local de Neoplasia/diagnóstico , Adulto , Anciano , Carcinoma Hepatocelular/mortalidad , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Hepáticas/mortalidad , Trasplante de Hígado , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia
12.
Cent Eur J Immunol ; 39(1): 96-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26155107

RESUMEN

OBJECTIVES: The ImmuKnow assay (cylex Inc., Columbia, MD) has been reported to measure the global immune monitoring tool for organ transplantation recipients. We assess immuKnow ATP values in stable kidney transplant patients. MATERIAL AND METHODS: Patients who were kidney transplanted between September 2008 and May 2011 were enrolled in the prospective serial ImmuKnow assay study. The criteria of inclusion were living donor kidney transplantation (KT), no evidence of hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, and PRA less than 50%. ImmuKnow assay monitoring was performed at one day before operation, post operative weeks 4, 8, 12, 16, 20, 24, 36 and 52. We excluded patients who had undergone infectious syndrome or rejection episodes during the follow-up period. RESULTS: Among 71 patients who were enrolled in prospective serial ImmuKnow assay monitoring, 37 patients were proven to stable KT patients during the follow-up period. Two hundred and twenty-four samples from 37 patients were collected. ImmuKnow value and immunosuppression drug level were compared in post operative weeks 4, 8, 12, 16, 20, 24, 36 and 52. The value of ImmuKnow assay was significantly different depending on the length of time after transplant (p = 0.038). Interestingly, the pre-transplant ImmuKnow values were lower than those of the immediate post-transplant period. CONCLUSIONS: The ImmuKnow value of stable KT recipients is different according to "time after transplant". Therefore, "time after transplant" should be considered when applying an ImmuKnow assay in clinical practice.

13.
Biochim Biophys Acta ; 1819(8): 885-91, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22609543

RESUMEN

Carbonic anhydrase 1 (Car1), an early specific marker of the erythroid differentiation, has been used to distinguish fetal and adult erythroid cells since its production closely follows the γ- to ß-globin transition, but the molecular mechanism underlying transcriptional regulation of Car1 is unclear. Here, we show that Car1 mRNA decreases significantly when erythroid differentiation is induced in MEL cells. The Ldb1 protein complex including GATA1/SCL/LMO2 binds to the Car1 promoter in uninduced cells and reduced enrichment of the complex during differentiation correlates with loss of Car1 expression. Knockdown of Ldb1 results in a reduction of Ser2 phosphorylated RNA Pol II and Cdk9 at the Car1 promoter region, suggesting that Ldb1 is required for recruitment of Pol II as well as the transcription regulator P-TEFb to enhance elongation of Car1 transcripts. Taken together, these data show that Ldb1 forms a regulatory complex to maintain Car1 expression in erythroid cells.


Asunto(s)
Anhidrasa Carbónica I , Proteínas de Unión al ADN , Células Eritroides , Globinas , Proteínas con Dominio LIM , Animales , Anhidrasa Carbónica I/genética , Anhidrasa Carbónica I/metabolismo , Diferenciación Celular , Línea Celular , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Globinas/química , Globinas/genética , Globinas/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones , Fosforilación , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas
14.
Clin Transplant ; 27(5): E597-604, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24093615

RESUMEN

The purpose of this study was to identify the factors associated with the recurrence of hepatitis B virus (HBV) following liver transplantation (LT) for HBV-related disease and to recognize the outcome of treatment for HBV recurrence with oral nucleos(t)ide analogues. Six hundred and sixty-seven LTs were performed for HBsAg-positive adult patients in our institute from 1996 to 2010. HBV prophylaxis was performed by hepatitis B immunoglobulin (HBIG) monotherapy or HBIG and entecavir combination therapy. There were 63 cases (11.4%) of HBV recurrences during a median follow-up of 51 months. The median time to HBV recurrence was 22 months. A preoperative HBV DNA load of more than 10(5) IU/mL, HBIG monotherapy, and hepatocellular carcinoma in the explant liver were independent risk factors for HBV recurrence following LT in multivariate analysis. Patient survival at 10 yr was 54.2% for HBV-recurrent patients. Among patients with HBV recurrence, HBsAg seroclearance was achieved in 13 patients (20.6%), but HBsAg seroclearance did not affect survival in these patients after the recurrence of HBV (p = 0.28). The recurrence of HBV led to graft failure in six cases. HBV recurrence should be prevented by strict management of pre-transplant HBV viremia and an effective post-transplant HBV prophylaxis.


Asunto(s)
Antivirales/uso terapéutico , Virus de la Hepatitis B/patogenicidad , Hepatitis B/tratamiento farmacológico , Trasplante de Hígado , Prevención Secundaria , Adulto , Carcinoma Hepatocelular/complicaciones , ADN Viral/genética , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Guanina/análogos & derivados , Guanina/uso terapéutico , Hepatitis B/diagnóstico , Hepatitis B/virología , Virus de la Hepatitis B/genética , Humanos , Inmunoglobulinas/uso terapéutico , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/complicaciones , Masculino , Registros Médicos , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia
15.
Cell Rep ; 42(7): 112778, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37453058

RESUMEN

The regulatory effect of non-coding large-scale structural variations (SVs) on proto-oncogene activation remains unclear. This study investigated SV-mediated gene dysregulation by profiling 3D cancer genome maps from 40 patients with colorectal cancer (CRC). We developed a machine learning-based method for spatial characterization of the altered 3D cancer genome. This revealed a frequent establishment of "de novo chromatin contacts" that can span multiple topologically associating domains (TADs) in addition to the canonical TAD fusion/shuffle model. Using this information, we precisely identified super-enhancer (SE)-hijacking and its clonal characteristics. Clonal SE-hijacking genes, such as TOP2B, are recurrently associated with cell-cycle/DNA-processing functions, which can potentially be used as CRC prognostic markers. Oncogene activation and increased drug resistance due to SE-hijacking were validated by reconstructing the patient's SV using CRISPR-Cas9. Collectively, the spatial and clonality-resolved analysis of the 3D cancer genome reveals regulatory principles of large-scale SVs in oncogene activation and their clinical implications.


Asunto(s)
Neoplasias Colorrectales , Genoma , Humanos , Pronóstico , Cromatina , ADN , Neoplasias Colorrectales/genética
16.
BMB Rep ; 56(10): 563-568, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37574809

RESUMEN

DNA methylation regulates gene expression and contributes to tumorigenesis in the early stages of cancer. In colorectal cancer (CRC), CpG island methylator phenotype (CIMP) is recognized as a distinct subset that is associated with specific molecular and clinical features. In this study, we investigated the genomewide DNA methylation patterns among patients with CRC. The methylation data of 1 unmatched normal, 142 adjacent normal, and 294 tumor samples were analyzed. We identified 40,003 differentially methylated positions with 6,933 (79.8%) hypermethylated and 16,145 (51.6%) hypomethylated probes in the genic region. Hypermethylated probes were predominantly found in promoter-like regions, CpG islands, and N shore sites; hypomethylated probes were enriched in open-sea regions. CRC tumors were categorized into three CIMP subgroups, with 90 (30.6%) in the CIMP-high (CIMP-H), 115 (39.1%) in the CIMP-low (CIMP-L), and 89 (30.3%) in the non-CIMP group. The CIMP-H group was associated with microsatellite instabilityhigh tumors, hypermethylation of MLH1, older age, and rightsided tumors. Our results showed that genome-wide methylation analyses classified patients with CRC into three subgroups according to CIMP levels, with clinical and molecular features consistent with previous data. [BMB Reports 2023; 56(10): 563-568].


Asunto(s)
Neoplasias Colorrectales , Metilación de ADN , Humanos , Metilación de ADN/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Islas de CpG/genética , Fenotipo , Epigénesis Genética/genética , República de Corea
17.
Exp Mol Med ; 55(11): 2445-2460, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37907748

RESUMEN

Cell-free DNA (cfDNA) sequencing has demonstrated great potential for early cancer detection. However, most large-scale studies have focused only on either targeted methylation sites or whole-genome sequencing, limiting comprehensive analysis that integrates both epigenetic and genetic signatures. In this study, we present a platform that enables simultaneous analysis of whole-genome methylation, copy number, and fragmentomic patterns of cfDNA in a single assay. Using a total of 950 plasma (361 healthy and 589 cancer) and 240 tissue samples, we demonstrate that a multifeature cancer signature ensemble (CSE) classifier integrating all features outperforms single-feature classifiers. At 95.2% specificity, the cancer detection sensitivity with methylation, copy number, and fragmentomic models was 77.2%, 61.4%, and 60.5%, respectively, but sensitivity was significantly increased to 88.9% with the CSE classifier (p value < 0.0001). For tissue of origin, the CSE classifier enhanced the accuracy beyond the methylation classifier, from 74.3% to 76.4%. Overall, this work proves the utility of a signature ensemble integrating epigenetic and genetic information for accurate cancer detection.


Asunto(s)
Ácidos Nucleicos Libres de Células , Neoplasias , Humanos , Detección Precoz del Cáncer , Variaciones en el Número de Copia de ADN , Neoplasias/diagnóstico , Neoplasias/genética , Metilación de ADN , Biomarcadores de Tumor/genética
18.
Lab Invest ; 92(7): 1033-44, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22508389

RESUMEN

Epiregulin (EREG) induces cell growth by binding to the epidermal growth factor receptor (EGFR). Expression of EREG affects sensitivity to cetuximab a chimeric monoclonal antibody that inhibits the EGFR signaling pathway. The mechanism through which EREG is regulated is largely unknown, but a methyl-array study previously performed by our group revealed that EREG is methylated in gastric cancer cells. In this study, we found that EREG gene expression was low in 7 out of 11 gastric cancer cells and this downregulation was mediated by aberrant CpG methylation of the EREG promoter. Treatment with 5-aza-CdR restored EREG expression and demethylated CpG sites in the EREG promoter. Compared with DNA methyltransferase 1 (DNMT1), knock-down of DNA methyltransferase 3b (DNMT3b) significantly increased the expression of EREG and led to the demethylation of specific CpG sites in the EREG promoter, suggesting that DNMT3b primarily regulates CpG methylation and silencing of the EREG gene. EREG methylation was observed in 30% (4/13) of human primary gastric tumor tissues we evaluated. In addition to DNA methylation, results from a chromatin immunoprecipitation assay demonstrated that transcriptional levels of EREG were associated with the enrichment of active histone marks (H3K4me3 and AcH3) and of a repressive mark (H3K27me2). Treatment with 5-aza-CdR dynamically increased the low occupancy of H3K4me3 and AcH3, while decreasing the high enrichment of H3K27me2, indicating that dynamic histone modifications contribute to EREG regulation in addition to DNA methylation. Finally, the combination of 5-aza-CdR and cetuximab exerted a synergistic anti-proliferative effect on gastric cancer cells. Taken together, the results of our study showed for the first time that EREG is epigenetically silenced in gastric cancer cells by aberrant DNA methylation and histone modification.


Asunto(s)
Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/genética , Epigénesis Genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Cetuximab , Islas de CpG , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Decitabina , Epigénesis Genética/efectos de los fármacos , Epirregulina , Receptores ErbB/metabolismo , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Neoplasias Gástricas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , ADN Metiltransferasa 3B
19.
Ann Surg Oncol ; 19(7): 2246-55, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22402812

RESUMEN

BACKGROUND: Reports on the risk factors of peritoneal recurrence (PR) after liver resection for hepatocellular carcinoma are lacking. We examined the risk factors of PR after hepatectomy and the outcome of resected PR at our institution. METHODS: We retrospectively reviewed the data from 1,222 patients who underwent hepatectomies for hepatocellular carcinoma in Samsung Medical Center from January 2006 to August 2010. We identified patients with PR and studied the risk factors and outcomes of resected PR. RESULTS: The rate of PR was 3.0% (n=36). The mean±SD age of patients was 54.0±10.2 years. Among those with PR, 23 patients (63.9%) had unresectable disease and 13 patients (36.1%) had resectable disease. Multivariate analysis found that tumor size>50 mm, presence of microvascular invasion, bile duct invasion, and positive margins were significant risk factors of PR after liver resection. The median overall survival (OS) for resectable PR was 33.0 (28.0-61.6) months compared to 14.0 (6.8-21.2) months for unresectable PR (P=0.009). Cox regression analysis demonstrated that resected PR [hazard ratio (HR) 0.042, P = 0.001] and interval between hepatectomy and PR (>6 months) (HR 0.195, P=0.016) were positive prognostic factors for OS, while alfa-fetoprotein>200 ng/dl at detection of PR (HR 11.321, P=0.015) and serosal involvement of primary hepatocellular carcinoma (HR 25.616, P=0.007) were negative prognostic factors for OS. CONCLUSIONS: We found that tumor size>50 mm, presence of microvascular invasion, bile duct invasion, and positive resection margins were significant risk factors of PR after liver resection. Selected patients with resected PR had significantly better OS.


Asunto(s)
Carcinoma Hepatocelular/mortalidad , Hepatectomía/mortalidad , Neoplasias Hepáticas/mortalidad , Recurrencia Local de Neoplasia/mortalidad , Neoplasias Peritoneales/mortalidad , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/cirugía , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Estadificación de Neoplasias , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/cirugía , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia
20.
Blood ; 116(13): 2356-64, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20570862

RESUMEN

Ldb1 and erythroid partners SCL, GATA-1, and LMO2 form a complex that is required to establish spatial proximity between the ß-globin locus control region and gene and for transcription activation during erythroid differentiation. Here we show that Ldb1 controls gene expression at multiple levels. Ldb1 stabilizes its erythroid complex partners on ß-globin chromatin, even though it is not one of the DNA-binding components. In addition, Ldb1 is necessary for enrichment of key transcriptional components in the locus, including P-TEFb, which phosphorylates Ser2 of the RNA polymerase C-terminal domain for efficient elongation. Furthermore, reduction of Ldb1 results in the inability of the locus to migrate away from the nuclear periphery, which is necessary to achieve robust transcription of ß-globin in nuclear transcription factories. Ldb1 contributes these critical functions at both embryonic and adult stages of globin gene expression. These results implicate Ldb1 as a factor that facilitates nuclear relocation for transcription activation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Eritropoyesis/fisiología , Globinas beta/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Factor de Transcripción GATA1/química , Factor de Transcripción GATA1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas con Dominio LIM , Región de Control de Posición , Metaloproteínas/química , Metaloproteínas/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Complejos Multiproteicos , Fosforilación , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas , Estabilidad Proteica , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Serina/química , Proteína 1 de la Leucemia Linfocítica T Aguda , Globinas beta/deficiencia , Globinas beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA