Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 14(5): 437-45, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23563689

RESUMEN

How hematopoietic stem cells (HSCs) coordinate the regulation of opposing cellular mechanisms such as self-renewal and differentiation commitment remains unclear. Here we identified the transcription factor and chromatin remodeler Satb1 as a critical regulator of HSC fate. HSCs lacking Satb1 had defective self-renewal, were less quiescent and showed accelerated lineage commitment, which resulted in progressive depletion of functional HSCs. The enhanced commitment was caused by less symmetric self-renewal and more symmetric differentiation divisions of Satb1-deficient HSCs. Satb1 simultaneously repressed sets of genes encoding molecules involved in HSC activation and cellular polarity, including Numb and Myc, which encode two key factors for the specification of stem-cell fate. Thus, Satb1 is a regulator that promotes HSC quiescence and represses lineage commitment.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Polaridad Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(3): 1666-1677, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31911474

RESUMEN

Major efforts are underway to identify agents that can potentiate effects of immune checkpoint inhibition. Here, we show that ascorbic acid (AA) treatment caused genomewide demethylation and enhanced expression of endogenous retroviral elements in lymphoma cells. AA also increased 5-hydroxymethylcytosine (5hmC) levels of CD8+ T cells and enhanced their cytotoxic activity in a lymphoma coculture system. High-dose AA treatment synergized with anti-PD1 therapy in a syngeneic lymphoma mouse model, resulting in marked inhibition of tumor growth compared with either agent alone. Analysis of the intratumoral epigenome revealed increased 5hmC with AA treatment, consistent with in vitro findings. Analysis of the tumor immune microenvironment revealed that AA strikingly increased intratumoral infiltration of CD8+ T cells and macrophages, suggesting enhanced tumor immune recognition. The combination treatment markedly enhanced intratumoral infiltration of macrophages and CD8+ T lymphocytes, granzyme B production by cytotoxic cells (cytotoxic T cells and natural killer cells), and interleukin 12 production by antigen-presenting cells compared with single-agent anti-PD1. These data indicate that AA potentiates anti-PD1 checkpoint inhibition through synergistic mechanisms. The study provides compelling rationale for testing combinations of high-dose AA and anti-PD1 agents in patients with aggressive B cell lymphoma as well as in preclinical models of other malignancies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Ácido Ascórbico/administración & dosificación , Linfoma/tratamiento farmacológico , 5-Metilcitosina/análogos & derivados , Animales , Antígeno B7-H1 , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Granzimas , Inmunoterapia , Ratones , Ratones Endogámicos BALB C , Receptor de Muerte Celular Programada 1/efectos de los fármacos , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral/efectos de los fármacos
3.
Genome Res ; 27(11): 1830-1842, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28986391

RESUMEN

Transcriptional deregulation of oncogenic pathways is a hallmark of cancer and can be due to epigenetic alterations. 5-Hydroxymethylcytosine (5-hmC) is an epigenetic modification that has not been studied in pancreatic cancer. Genome-wide analysis of 5-hmC-enriched loci with hmC-seal was conducted in a cohort of low-passage pancreatic cancer cell lines, primary patient-derived xenografts, and pancreatic controls and revealed strikingly altered patterns in neoplastic tissues. Differentially hydroxymethylated regions preferentially affected known regulatory regions of the genome, specifically overlapping with known H3K4me1 enhancers. Furthermore, base pair resolution analysis of cytosine methylation and hydroxymethylation with oxidative bisulfite sequencing was conducted and correlated with chromatin accessibility by ATAC-seq and gene expression by RNA-seq in pancreatic cancer and control samples. 5-hmC was specifically enriched at open regions of chromatin, and gain of 5-hmC was correlated with up-regulation of the cognate transcripts, including many oncogenic pathways implicated in pancreatic neoplasia, such as MYC, KRAS, VEGFA, and BRD4 Specifically, BRD4 was overexpressed and acquired 5-hmC at enhancer regions in the majority of neoplastic samples. Functionally, acquisition of 5-hmC at BRD4 promoter was associated with increase in transcript expression in reporter assays and primary samples. Furthermore, blockade of BRD4 inhibited pancreatic cancer growth in vivo. In summary, redistribution of 5-hmC and preferential enrichment at oncogenic enhancers is a novel regulatory mechanism in human pancreatic cancer.


Asunto(s)
5-Metilcitosina/análogos & derivados , Neoplasias Pancreáticas/genética , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de Secuencia de ARN/métodos , 5-Metilcitosina/metabolismo , Animales , Línea Celular Tumoral , Epigénesis Genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Histonas/metabolismo , Humanos , Ratones , Trasplante de Neoplasias , Modelación Específica para el Paciente
4.
Blood ; 132(14): 1507-1518, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30104217

RESUMEN

Adult T-cell leukemia lymphoma (ATLL) is a rare T cell neoplasm that is endemic in Japanese, Caribbean, and Latin American populations. Most North American ATLL patients are of Caribbean descent and are characterized by high rates of chemo-refractory disease and worse prognosis compared with Japanese ATLL. To determine genomic differences between these 2 cohorts, we performed targeted exon sequencing on 30 North American ATLL patients and compared the results with the Japanese ATLL cases. Although the frequency of TP53 mutations was comparable, the mutation frequency in epigenetic and histone modifying genes (57%) was significantly higher, whereas the mutation frequency in JAK/STAT and T-cell receptor/NF-κB pathway genes was significantly lower. The most common type of epigenetic mutation is that affecting EP300 (20%). As a category, epigenetic mutations were associated with adverse prognosis. Dissimilarities with the Japanese cases were also revealed by RNA sequencing analysis of 9 primary patient samples. ATLL samples with a mutated EP300 gene have decreased total and acetyl p53 protein and a transcriptional signature reminiscent of p53-mutated cancers. Most importantly, decitabine has highly selective single-agent activity in the EP300-mutated ATLL samples, suggesting that decitabine treatment induces a synthetic lethal phenotype in EP300-mutated ATLL cells. In conclusion, we demonstrate that North American ATLL has a distinct genomic landscape that is characterized by frequent epigenetic mutations that are targetable preclinically with DNA methyltransferase inhibitors.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Decitabina/uso terapéutico , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Leucemia-Linfoma de Células T del Adulto/genética , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Proteína p300 Asociada a E1A/genética , Epigénesis Genética , Femenino , Humanos , Japón/epidemiología , Leucemia-Linfoma de Células T del Adulto/diagnóstico , Leucemia-Linfoma de Células T del Adulto/epidemiología , Masculino , Persona de Mediana Edad , Tasa de Mutación , Pronóstico , Transcriptoma , Proteína p53 Supresora de Tumor/genética , Estados Unidos/epidemiología
5.
J Biol Chem ; 292(3): 837-846, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-27909050

RESUMEN

Clear cell renal cell carcinoma (CCRCC) is an incurable malignancy in advanced stages and needs newer therapeutic targets. Transcriptomic analysis of CCRCCs and matched microdissected renal tubular controls revealed overexpression of NOTCH ligands and receptors in tumor tissues. Examination of the TCGA RNA-seq data set also revealed widespread activation of NOTCH pathway in a large cohort of CCRCC samples. Samples with NOTCH pathway activation were also clinically distinct and were associated with better overall survival. Parallel DNA methylation and copy number analysis demonstrated that both genetic and epigenetic alterations led to NOTCH pathway activation in CCRCC. NOTCH ligand JAGGED1 was overexpressed and associated with loss of CpG methylation of H3K4me1-associated enhancer regions. JAGGED2 was also overexpressed and associated with gene amplification in distinct CCRCC samples. Transgenic expression of intracellular NOTCH1 in mice with tubule-specific deletion of VHL led to dysplastic hyperproliferation of tubular epithelial cells, confirming the procarcinogenic role of NOTCH in vivo Alteration of cell cycle pathways was seen in murine renal tubular cells with NOTCH overexpression, and molecular similarity to human tumors was observed, demonstrating that human CCRCC recapitulates features and gene expression changes observed in mice with transgenic overexpression of the Notch intracellular domain. Treatment with the γ-secretase inhibitor LY3039478 led to inhibition of CCRCC cells in vitro and in vivo In summary, these data reveal the mechanistic basis of NOTCH pathway activation in CCRCC and demonstrate this pathway to a potential therapeutic target.


Asunto(s)
Neoplasias Renales/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Carcinoma de Células Renales , Islas de CpG , Metilación de ADN , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Inhibidores de Proteasas/farmacología , Receptor Notch1/genética
6.
Proc Natl Acad Sci U S A ; 112(46): E6359-68, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578796

RESUMEN

Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in -7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.


Asunto(s)
Eritroblastos/metabolismo , Proteínas Activadoras de GTPasa/biosíntesis , Regulación de la Expresión Génica , Síndromes Mielodisplásicos/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Eritroblastos/patología , Femenino , Proteínas Activadoras de GTPasa/genética , Humanos , Masculino , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
7.
Blood ; 125(8): e1-13, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25573988

RESUMEN

Macrophage polarization between the M2 (repair, protumorigenic) and M1 (inflammatory) phenotypes is seen as a continuum of states. The detailed transcriptional events and signals downstream of colony-stimulating factor 1 receptor (CSF-1R) that contributes to amplification of the M2 phenotype and suppression of the M1 phenotype are largely unknown. Macrophage CSF-1R pTyr-721 signaling promotes cell motility and enhancement of tumor cell invasion in vitro. Combining analysis of cellular systems for CSF-1R gain of function and loss of function with bioinformatic analysis of the macrophage CSF-1R pTyr-721-regulated transcriptome, we uncovered microRNA-21 (miR-21) as a downstream molecular switch controlling macrophage activation and identified extracellular signal-regulated kinase1/2 and nuclear factor-κB as CSF-1R pTyr-721-regulated signaling nodes. We show that CSF-1R pTyr-721 signaling suppresses the inflammatory phenotype, predominantly by induction of miR-21. Profiling of the miR-21-regulated messenger RNAs revealed that 80% of the CSF-1-regulated canonical miR-21 targets are proinflammatory molecules. Additionally, miR-21 positively regulates M2 marker expression. Moreover, miR-21 feeds back to positively regulate its own expression and to limit CSF-1R-mediated activation of extracellular signal-regulated kinase1/2 and nuclear factor-κB. Consistent with an anti-inflammatory role of miRNA-21, intraperitoneal injection of mice with a miRNA-21 inhibitor increases the recruitment of inflammatory monocytes and enhances the peritoneal monocyte/macrophage response to lipopolysaccharide. These results identify the CSF-1R-regulated miR-21 network that modulates macrophage polarization.


Asunto(s)
Inflamación/genética , Macrófagos Peritoneales/inmunología , MicroARNs/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/fisiología , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Polaridad Celular/genética , Polaridad Celular/inmunología , Células Cultivadas , Redes Reguladoras de Genes , Inflamación/inmunología , Activación de Macrófagos/genética , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
8.
Blood ; 125(20): 3144-52, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25810490

RESUMEN

Acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are associated with disease-initiating stem cells that are not eliminated by conventional therapies. Novel therapeutic targets against preleukemic stem cells need to be identified for potentially curative strategies. We conducted parallel transcriptional analysis of highly fractionated stem and progenitor populations in MDS, AML, and control samples and found interleukin 8 (IL8) to be consistently overexpressed in patient samples. The receptor for IL8, CXCR2, was also significantly increased in MDS CD34(+) cells from a large clinical cohort and was predictive of increased transfusion dependence. High CXCR2 expression was also an adverse prognostic factor in The Cancer Genome Atlas AML cohort, further pointing to the critical role of the IL8-CXCR2 axis in AML/MDS. Functionally, CXCR2 inhibition by knockdown and pharmacologic approaches led to a significant reduction in proliferation in several leukemic cell lines and primary MDS/AML samples via induction of G0/G1 cell cycle arrest. Importantly, inhibition of CXCR2 selectively inhibited immature hematopoietic stem cells from MDS/AML samples without an effect on healthy controls. CXCR2 knockdown also impaired leukemic growth in vivo. Together, these studies demonstrate that the IL8 receptor CXCR2 is an adverse prognostic factor in MDS/AML and is a potential therapeutic target against immature leukemic stem cell-enriched cell fractions in MDS and AML.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Interleucina-8/metabolismo , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicos/metabolismo , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Análisis por Conglomerados , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Interleucina-8/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Ratones , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/mortalidad , Células Madre Neoplásicas/metabolismo , Pronóstico , Receptores de Interleucina-8B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Contemp Dent Pract ; 18(5): 405-409, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28512281

RESUMEN

INTRODUCTION: Replacement of missing teeth by dental implants is one of the most common methods employed these days. Because of significant advancement in the design of implants and modifications in the procedure of dental implant surgery, the survival rate of the dental implants has reached up to approximately 95%. Osseointegration is one of the important factors affecting the survival of dental implants. Apart from these, the body's physiologic alterations can also predispose the dental implants for failure. Diabetes is one such metabolic disease characterized by abnormal or delayed wound healing. Hence, we assessed the clinicomicrobial and salivary profile of diabetic patients undergoing rehabilitation by dental implants. MATERIALS AND METHODS: This study included diabetic patients who underwent dental implant surgeries for prosthetic rehabilitation. Follow-up records of the patients' up to 1 year were maintained. Various clinicoradiographic and periodontal parameters were measured at various time intervals during follow-up time; 25 mL of salivary and blood sample was taken from all the subjects and was sent to the laboratories for assessment of various salivary biomarkers. All the results were analyzed by Statistical Package for the Social Sciences software. RESULTS: The mean level of interleukin-p at baseline time was found to be 2.38 and 2.21 in diabetic group and control group respectively. While comparing the levels of osteoprotegerin in both study groups, a significant correlation was obtained. In diabetic and control group, 62 and 61 years was the mean age of the patients respectively. No significant correlation was obtained while comparing the microbial flora of diabetic and control group. CONCLUSION: In both diabetic and nondiabetic patients, similar microbial, salivary marker, and clinicoradiological patterns were seen. CLINICAL SIGNIFICANCE: Diabetic patients who maintain their body's metabolic rate show similar success rate of dental implants as seen in nondiabetic patients.


Asunto(s)
Implantes Dentales , Diabetes Mellitus Tipo 2/fisiopatología , Oseointegración , Adulto , Biomarcadores/análisis , Implantación Dental Endoósea , Fracaso de la Restauración Dental , Femenino , Humanos , Interleucinas/análisis , Estudios Longitudinales , Masculino , Metaloproteinasas de la Matriz/análisis , Persona de Mediana Edad , Osteoprotegerina/análisis , Saliva/química , Saliva/microbiología , Análisis de Supervivencia
10.
J Contemp Dent Pract ; 17(3): 253-7, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27207207

RESUMEN

BACKGROUND: It has been since a long time that the use of base-metal alloys has significantly increased in the area of prosthetic science. One of the factors contributing for the increasing use of base-metal alloys is the increasing and high cost factor of noble metals. Although numerous materials have been tried since the recent past, Cobalt-Chromium (CoCr) alloys still form the most commonly used ones because of the numerous advantages they offer. Titanium alloys are also significantly being used in this field because of their high resistance strength and high corrosion resistance. Therefore, we aimed to evaluate the flexure strength and fatigue resistance of clasps made up of CoCr alloys and Nickel-titanium (NiTi) alloys. MATERIALS AND METHODS: We evaluated the retentive forces of CoCr and NiTi clasps two different sizes engaging retentive undercuts of different depths (0.25 and 0.50 mm). Mandibluar second premolar was prepared to frame the prosthesis for missing mandibular first and second molar. A total of 50 clasps were included in the study out of which 30 were NiTi clasps and 30 CoCr clasps. We evaluated the loading of the force at the rate of half Newton(N)/second at the central part of the specimen until fracturing of the specimen occurred and finally measurement of the modulus of elasticity was also done. Statistical analysis was carried out; unpaired "t" test was used for evaluating the level of significance. RESULTS: The mean load needed for CoCr clasps was 0.7450 and for NiTi clasp was 0.6140 Kgf for producing a deflection of 0.25 mm. As far as flexibility is concerned, more value was seen in NiTi group than CoCr group. For deflecting the clasp up to 0.50 mm, the mean load needed for CoCr clasps was 1.4102 and for NiTi clasp was 0.8260 Kgf. The results were statistically significant. While measuring the flexibility, more value (p < 0.05) was seen in NiTi group than in CoCr group. NiTi alloy clasps had mean flexural strength of 1640 MPa and modulus of elasticity of 32 GPa. CONCLUSION: For producing deflection of 0.25 and 0.50 mm, the mean loading force was higher for NiTi alloys. Although being increasingly used these days, NiTi alloys have limited use as compared with CoCr alloys due to high cost of titanium and certain other limitations.


Asunto(s)
Aleaciones de Cromo , Abrazadera Dental , Dentadura Parcial Removible , Níquel , Titanio , Humanos
11.
Blood ; 121(15): 2875-81, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23390194

RESUMEN

Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis that leads to peripheral cytopenias. We observed that SMAD7, a negative regulator of transforming growth factor-beta (TGF-ß) receptor-I kinase, is markedly reduced in MDS and leads to ineffective hematopoiesis by overactivation of TGF-ß signaling. To determine the cause of SMAD7 reduction in MDS, we analyzed the 3'UTR of the gene and determined that it contains a highly conserved putative binding site for microRNA-21. We observed significantly elevated levels of miR-21 in MDS marrow samples when compared with age-matched controls. miR-21 was shown to directly bind to the 3'UTR of SMAD7 and reduce its expression in hematopoietic cells. Next, we tested the role of miR-21 in regulating TGF-ß signaling in a TGF-ß-overexpressing transgenic mouse model that develops progressive anemia and dysplasia and thus serves as a model of human bone marrow failure. Treatment with a chemically modified miR-21 inhibitor led to significant increases in hematocrit and led to an increase in SMAD7 expression in vivo. Inhibition of miR-21 also led to an increase in erythroid colony formation from primary MDS bone marrow progenitors, demonstrating its ability in stimulating hematopoiesis in vitro. Taken together, these studies demonstrate the role of miR-21 in regulating overactivated TGF-ß signaling in MDS.


Asunto(s)
Hematopoyesis/genética , MicroARNs/genética , Síndromes Mielodisplásicos/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta1/genética , Regiones no Traducidas 3'/genética , Anciano , Anciano de 80 o más Años , Animales , Sitios de Unión/genética , Células de la Médula Ósea/metabolismo , Línea Celular , Células Cultivadas , Femenino , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células K562 , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Persona de Mediana Edad , Mutación , Síndromes Mielodisplásicos/metabolismo , Proteína smad7/genética
12.
J Immunol ; 190(6): 2966-75, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23408834

RESUMEN

Epigenetic changes play important roles in carcinogenesis and influence initial steps in neoplastic transformation by altering genome stability and regulating gene expression. To characterize epigenomic changes during the transformation of normal plasma cells to myeloma, we modified the HpaII tiny fragment enrichment by ligation-mediated PCR assay to work with small numbers of purified primary marrow plasma cells. The nano-HpaII tiny fragment enrichment by ligation-mediated PCR assay was used to analyze the methylome of CD138(+) cells from 56 subjects representing premalignant (monoclonal gammopathy of uncertain significance), early, and advanced stages of myeloma, as well as healthy controls. Plasma cells from premalignant and early stages of myeloma were characterized by striking, widespread hypomethylation. Gene-specific hypermethylation was seen to occur in the advanced stages, and cell lines representative of relapsed cases were found to be sensitive to decitabine. Aberrant demethylation in monoclonal gammopathy of uncertain significance occurred primarily in CpG islands, whereas differentially methylated loci in cases of myeloma occurred predominantly outside of CpG islands and affected distinct sets of gene pathways, demonstrating qualitative epigenetic differences between premalignant and malignant stages. Examination of the methylation machinery revealed that the methyltransferase, DNMT3A, was aberrantly hypermethylated and underexpressed, but not mutated in myeloma. DNMT3A underexpression was also associated with adverse overall survival in a large cohort of patients, providing insights into genesis of hypomethylation in myeloma. These results demonstrate widespread, stage-specific epigenetic changes during myelomagenesis and suggest that early demethylation can be a potential contributor to genome instability seen in myeloma. We also identify DNMT3A expression as a novel prognostic biomarker and suggest that relapsed cases can be therapeutically targeted by hypomethylating agents.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Metilación de ADN/genética , Metilación de ADN/inmunología , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Transformación Celular Neoplásica/genética , Estudios de Cohortes , Diagnóstico Precoz , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Mieloma Múltiple/patología , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa , Recurrencia , Inducción de Remisión , Reproducibilidad de los Resultados , Sindecano-1/biosíntesis , Sindecano-1/genética , Células Tumorales Cultivadas
13.
Nucleic Acids Res ; 41(16): e157, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23861445

RESUMEN

5-hydroxymethylcytosine (5-hmC) is a recently discovered epigenetic modification that is altered in cancers. Genome-wide assays for 5-hmC determination are needed as many of the techniques for 5-methylcytosine (5-mC) determination, including methyl-sensitive restriction digestion and bisulfite sequencing cannot distinguish between 5-mC and 5-hmC. Glycosylation of 5-hmC residues by beta-glucosyl transferase (ß-GT) can make CCGG residues insensitive to digestion by MspI. Restriction digestion by HpaII, MspI or MspI after ß-GT conversion, followed by adapter ligation, massive parallel sequencing and custom bioinformatic analysis allowed us determine distribution of 5-mC and 5-hmC at single base pair resolution at MspI restriction sites. The resulting HpaII tiny fragment Enrichment by Ligation-mediated PCR with ß-GT (HELP-GT) assay identified 5-hmC loci that were validated at global level by liquid chromatography-mass spectrometry (LC-MS) and the locus-specific level by quantitative reverse transcriptase polymerase chain reaction of 5-hmC pull-down DNA. Hydroxymethylation at both promoter and intragenic locations correlated positively with gene expression. Analysis of pancreatic cancer samples revealed striking redistribution of 5-hmC sites in cancer cells and demonstrated enrichment of this modification at many oncogenic promoters such as GATA6. The HELP-GT assay allowed global determination of 5-hmC and 5-mC from low amounts of DNA and with the use of modest sequencing resources. Redistribution of 5-hmC seen in cancer highlights the importance of determination of this modification in conjugation with conventional methylome analysis.


Asunto(s)
Citosina/análogos & derivados , ADN de Neoplasias/química , 5-Metilcitosina/análisis , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Citosina/análisis , Citosina/metabolismo , Expresión Génica , Genoma Humano , Genómica/métodos , Glicosiltransferasas/metabolismo , Humanos , Ratones , Neoplasias Pancreáticas/genética , Reacción en Cadena de la Polimerasa
14.
Gut ; 63(6): 881-90, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24000294

RESUMEN

OBJECTIVES: Long non-coding RNAs (lncRNA) have been shown to play important roles in the development and progression of cancer. However, functional lncRNAs and their downstream mechanisms are largely unknown in the molecular pathogenesis of oesophageal adenocarcinoma (EAC) and its progression. DESIGN: lncRNAs that are abnormally upregulated in EACs were identified by RNA-sequencing analysis, followed by quantitative RT-PCR (qRTPCR) validation using tissues from 25 EAC patients. Cell biological assays in combination with small interfering RNA-mediated knockdown were performed in order to probe the functional relevance of these lncRNAs. RESULTS: We discovered that a lncRNA, HNF1A-AS1, is markedly upregulated in human primary EACs relative to their corresponding normal oesophageal tissues (mean fold change 10.6, p<0.01). We further discovered that HNF1A-AS1 knockdown significantly inhibited cell proliferation and anchorage-independent growth, suppressed S-phase entry, and inhibited cell migration and invasion in multiple in vitro EAC models (p<0.05). A gene ontological analysis revealed that HNF1A-AS1 knockdown preferentially affected genes that are linked to assembly of chromatin and the nucleosome, a mechanism essential to cell cycle progression. The well known cancer-related lncRNA, H19, was the gene most markedly inhibited by HNF1A-AS1 knockdown. Consistent to this finding, there was a significant positive correlation between HNF1A-AS1 and H19 expression in primary EACs (p<0.01). CONCLUSIONS: We have discovered abnormal upregulation of a lncRNA, HNF1A-AS1, in human EAC. Our findings suggest that dysregulation of HNF1A-AS1 participates in oesophageal tumorigenesis, and that this participation may be mediated, at least in part, by modulation of chromatin and nucleosome assembly as well as by H19 induction.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Esofágicas/genética , Expresión Génica , ARN Largo no Codificante/genética , ARN Largo no Codificante/fisiología , Adenocarcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Neoplasias Esofágicas/patología , Técnicas de Silenciamiento del Gen , Humanos , ARN Interferente Pequeño , Puntos de Control de la Fase S del Ciclo Celular/genética , Regulación hacia Arriba
15.
J Biol Chem ; 288(13): 8805-14, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23306203

RESUMEN

Differentiation of hematopoietic stem cells to red cells requires coordinated expression of numerous erythroid genes and is characterized by nuclear condensation and extrusion during terminal development. To understand the regulatory mechanisms governing these widespread phenotypic changes, we conducted a high resolution methylomic and transcriptomic analysis of six major stages of human erythroid differentiation. We observed widespread epigenetic differences between early and late stages of erythropoiesis with progressive loss of methylation being the dominant change during differentiation. Gene bodies, intergenic regions, and CpG shores were preferentially demethylated during erythropoiesis. Epigenetic changes at transcription factor binding sites correlated significantly with changes in gene expression and were enriched for binding motifs for SCL, MYB, GATA, and other factors not previously implicated in erythropoiesis. Demethylation at gene promoters was associated with increased expression of genes, whereas epigenetic changes at gene bodies correlated inversely with gene expression. Important gene networks encoding erythrocyte membrane proteins, surface receptors, and heme synthesis proteins were found to be regulated by DNA methylation. Furthermore, integrative analysis enabled us to identify novel, potential regulatory areas of the genome as evident by epigenetic changes in a predicted PU.1 binding site in intron 1 of the GATA1 gene. This intronic site was found to be conserved across species and was validated to be a novel PU.1 binding site by quantitative ChIP in erythroid cells. Altogether, our study provides a comprehensive analysis of methylomic and transcriptomic changes during erythroid differentiation and demonstrates that human terminal erythropoiesis is surprisingly associated with hypomethylation of the genome.


Asunto(s)
Eritropoyesis/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Antígenos CD34/biosíntesis , Sitios de Unión , Diferenciación Celular , Islas de CpG , Metilación de ADN , Epigénesis Genética , Epigenómica , Eritrocitos/citología , Citometría de Flujo/métodos , Genoma Humano , Genómica , Humanos , Intrones , Metilación , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre/química
16.
Gastroenterology ; 144(5): 956-966.e4, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23333711

RESUMEN

BACKGROUND & AIMS: Alterations in methylation of protein-coding genes are associated with Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC). Dysregulation of noncoding RNAs occurs during carcinogenesis but has never been studied in BE or EAC. We applied high-resolution methylome analysis to identify changes at genomic regions that encode noncoding RNAs in BE and EAC. METHODS: We analyzed methylation of 1.8 million CpG sites using massively parallel sequencing-based HELP tagging in matched EAC, BE, and normal esophageal tissues. We also analyzed human EAC (OE33, SKGT4, and FLO-1) and normal (HEEpic) esophageal cells. RESULTS: BE and EAC exhibited genome-wide hypomethylation, significantly affecting intragenic and repetitive genomic elements as well as noncoding regions. These methylation changes targeted small and long noncoding regions, discriminating normal from matched BE or EAC tissues. One long noncoding RNA, AFAP1-AS1, was extremely hypomethylated and overexpressed in BE and EAC tissues and EAC cells. Its silencing by small interfering RNA inhibited proliferation and colony-forming ability, induced apoptosis, and reduced EAC cell migration and invasion without altering the expression of its protein-coding counterpart, AFAP1. CONCLUSIONS: BE and EAC exhibit reduced methylation that includes noncoding regions. Methylation of the long noncoding RNA AFAP1-AS1 is reduced in BE and EAC, and its expression inhibits cancer-related biologic functions of EAC cells.


Asunto(s)
Adenocarcinoma/genética , Esófago de Barrett/genética , ADN de Neoplasias/genética , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Microfilamentos/genética , ARN Largo no Codificante/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Esófago de Barrett/metabolismo , Esófago de Barrett/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Metilación de ADN , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Humanos , Proteínas de Microfilamentos/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética
17.
Blood ; 120(10): 2076-86, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22753872

RESUMEN

Even though hematopoietic stem cell (HSC) dysfunction is presumed in myelodysplastic syndrome (MDS), the exact nature of quantitative and qualitative alterations is unknown. We conducted a study of phenotypic and molecular alterations in highly fractionated stem and progenitor populations in a variety of MDS subtypes. We observed an expansion of the phenotypically primitive long-term HSCs (lineage(-)/CD34(+)/CD38(-)/CD90(+)) in MDS, which was most pronounced in higher-risk cases. These MDS HSCs demonstrated dysplastic clonogenic activity. Examination of progenitors revealed that lower-risk MDS is characterized by expansion of phenotypic common myeloid progenitors, whereas higher-risk cases revealed expansion of granulocyte-monocyte progenitors. Genome-wide analysis of sorted MDS HSCs revealed widespread methylomic and transcriptomic alterations. STAT3 was an aberrantly hypomethylated and overexpressed target that was validated in an independent cohort and found to be functionally relevant in MDS HSCs. FISH analysis demonstrated that a very high percentage of MDS HSC (92% ± 4%) carry cytogenetic abnormalities. Longitudinal analysis in a patient treated with 5-azacytidine revealed that karyotypically abnormal HSCs persist even during complete morphologic remission and that expansion of clonotypic HSCs precedes clinical relapse. This study demonstrates that stem and progenitor cells in MDS are characterized by stage-specific expansions and contain epigenetic and genetic alterations.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 7/genética , Células Madre Hematopoyéticas , Síndromes Mielodisplásicos/genética , Factor de Transcripción STAT3/genética , Antígenos CD/genética , Antígenos CD/metabolismo , Azacitidina/administración & dosificación , Estudios de Casos y Controles , Linaje de la Célula , Metilación de ADN , Epigénesis Genética , Citometría de Flujo , Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Cariotipificación , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/patología , Cultivo Primario de Células , Recurrencia , Factor de Transcripción STAT3/metabolismo
18.
PLoS Genet ; 7(3): e1001356, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21483804

RESUMEN

Although a combination of genomic and epigenetic alterations are implicated in the multistep transformation of normal squamous esophageal epithelium to Barrett esophagus, dysplasia, and adenocarcinoma, the combinatorial effect of these changes is unknown. By integrating genome-wide DNA methylation, copy number, and transcriptomic datasets obtained from endoscopic biopsies of neoplastic progression within the same individual, we are uniquely able to define the molecular events associated progression of Barrett esophagus. We find that the previously reported global hypomethylation phenomenon in cancer has its origins at the earliest stages of epithelial carcinogenesis. Promoter hypomethylation synergizes with gene amplification and leads to significant upregulation of a chr4q21 chemokine cluster and other transcripts during Barrett neoplasia. In contrast, gene-specific hypermethylation is observed at a restricted number of loci and, in combination with hemi-allelic deletions, leads to downregulatation of selected transcripts during multistep progression. We also observe that epigenetic regulation during epithelial carcinogenesis is not restricted to traditionally defined "CpG islands," but may also occur through a mechanism of differential methylation outside of these regions. Finally, validation of novel upregulated targets (CXCL1 and 3, GATA6, and DMBT1) in a larger independent panel of samples confirms the utility of integrative analysis in cancer biomarker discovery.


Asunto(s)
Esófago de Barrett/genética , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/genética , Quimiocinas/genética , Metilación de ADN , Neoplasias Esofágicas/genética , Amplificación de Genes , Esófago de Barrett/patología , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocinas/metabolismo , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Proteínas de Unión al ADN , Neoplasias Esofágicas/patología , Factor de Transcripción GATA6/genética , Factor de Transcripción GATA6/metabolismo , Perfilación de la Expresión Génica , Humanos , Estadificación de Neoplasias , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor
19.
Genes Chromosomes Cancer ; 52(12): 1123-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24123713

RESUMEN

Barrett's epithelium (BE) is a premalignant condition resulting from chronic gastroesophageal reflux that may progress to esophageal adenocarcinoma (EAC). Early intervention holds promise in preventing BE progression. However, identification of high-risk BE patients remains challenging due to inadequate biomarkers for early diagnosis. We investigated the effect of prolonged chronic acid and bile exposure on transcriptome, methylome, and mutatome of cells in an in-vitro BE carcinogenesis (BEC) model. Twenty weeks acid and bile exposed cells from the BEC model (BEC20w) were compared with their naïve predecessors HiSeq Illumina based RNA sequencing was performed on RNA from both the cells for gene expression and mutational analysis. HELP Tagging Assay was performed for DNA methylation analysis. Ingenuity pathway, Gene Ontology, and KEGG PATHWAY analyses were then performed on datasets. Widespread aberrant genetic and epigenetic changes were observed in the BEC20w cells. Combinatorial analyses revealed 433 from a total of 863 downregulated genes had accompanying hypermethylation of promoters. Simultaneously, 690 genes from a total of 1,492 were upregulated with accompanying promoter hypomethylation. In addition, 763 mutations were identified on 637 genes. Ingenuity pathway analysis, Gene Ontology, and KEGG PATHWAY analyses associated the genetic and epigenetic changes in BEC20w cells with cellular and biological functions. Integration of high resolution comparative analyses of naïve BAR-T and BEC20w cells revealed striking genetic and epigenetic changes induced by chronic acid and bile exposure that may disrupt normal cellular functions and promote carcinogenesis. This novel study reveals several potential targets for future biomarkers and therapeutic development.


Asunto(s)
Esófago de Barrett/genética , Bilis/metabolismo , Carcinogénesis/genética , Epigénesis Genética , Ácido Gástrico/metabolismo , Esófago de Barrett/metabolismo , Esófago de Barrett/patología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Células Cultivadas , Metilación de ADN , Ácido Glicoquenodesoxicólico/farmacología , Humanos , Ácido Clorhídrico/farmacología , Concentración de Iones de Hidrógeno , Mutación , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ARN/métodos , Transcriptoma
20.
Technol Health Care ; 31(1): 247-258, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36031920

RESUMEN

BACKGROUND: Advances in digital dentistry lead to use of three-dimensional (3D) printed resin denture teeth. Fracture toughness of these teeth must be assessed. OBJECTIVE: This study aimed to compare the chipping and indirect tensile fracture resistance of denture teeth fabricated by 3D printing technique with traditionally fabricated resin denture teeth. METHOD: Four groups (Gr) were made (n= 50/group): Gr-1 3D printed denture teeth (denture teeth; Formlabs Inc., Somerville, MA, USA), Gr-2 SR-Orthosit-PE (Ivoclar Vivadent AG), Gr-3 Portrait IPN (Dentsply Sirona), Gr-4 Pala Premium 8 (Heraeus Kulzer GmbH). Stereolithography 3D printing was used to create the methacrylate-based photopolymerized resin teeth models and remaining group teeth were collected commercially. A 1 mm/min chipping and indirect tensile fracture speed tests were carried out till fracture occurred. The data so obtained were statistically analysed using one-way analysis of variance with Tukey's honestly significant difference multiple comparisons test (p< 0.05). At the end of the test, the fractured areas of the specimens were evaluated by the chief researcher to assess the fracture pattern of the teeth. RESULTS: The indirect tensile fracture values of the 3D printed teeth were more than that of Pala Premium-8 and SR-Orthosit-PE but it was lower than that of Portrait IPN teeth. In chipping test, buccal chipping of the loaded cusp was seen in 3D printed resin without distortion and in indirect tensile test in 3D printed resin teeth, line of fracture emerges near the loading point propagates from the inner incline of both cusps and extends cervically, unlike in other groups where first deformation occurs then fracture. CONCLUSION: Prosthetic teeth fabricated by the 3D printing technique using printable resin material provide adequate fracture resistance as denture teeth.


Asunto(s)
Dentaduras , Impresión Tridimensional , Humanos , Ensayo de Materiales , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA