Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Hum Lact ; 40(1): 120-131, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38037896

RESUMEN

BACKGROUND: Breastfeeding behaviors and experiences exist on a continuum. What differentiates normal from dysfunctional is defined by frequency and severity. No current validated tool addresses the subjective experience of dyads with a predictive score that can be followed over time. RESEARCH AIM: To create and validate a self-report tool to assess breastfeeding and evaluate its ability to predict risk of breastfeeding dysfunction. METHODS: This study used a cross-sectional design to determine the validity of a novel instrument to assess breastfeeding dysfunction. We gave the initial questionnaire to 2085 breastfeeding dyads. We assessed content validity by comparison with other tools. We used exploratory factor analysis with varimax rotation for concept identification and Cronbach's alpha for internal consistency. We employed logistic regression to assess the tool's ability to differentiate between normal breastfeeding and breastfeeding dysfunction. RESULTS: Factor analysis mapped 17 questions to four concepts to create a score (FLIP; flow, latch, injury [to the nipple], and post-feed behavior). Internal consistency and reliability of the scores in these concepts were acceptable (Cronbach's alpha ≥ 0.087 for all measures). A logistic regression model that controlled for infant age, with a breastfeeding dysfunction risk classification threshold of 60%, yielded a correct classification of 88.7%, with 93.1% sensitivity, 64.6% specificity, and a 6.5% false positive rate. CONCLUSIONS: The FLIP score was determined to be a valid and reliable instrument for quantifying the severity of breastfeeding dysfunction in children under 1 year old. Further studies will assess its usefulness in the management of breastfeeding dysfunction.


Asunto(s)
Lactancia Materna , Frenillo Lingual , Lactante , Femenino , Niño , Humanos , Estudios Transversales , Reproducibilidad de los Resultados , Resultado del Tratamiento
2.
Front Genet ; 13: 932763, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36147501

RESUMEN

The clustering of mutations observed in cancer cells is reminiscent of the stress-induced mutagenesis (SIM) response in bacteria. Bacteria deploy SIM when faced with DNA double-strand breaks in the presence of conditions that elicit an SOS response. SIM employs DinB, the evolutionary precursor to human trans-lesion synthesis (TLS) error-prone polymerases, and results in mutations concentrated around DNA double-strand breaks with an abundance that decays with distance. We performed a quantitative study on single nucleotide variant calls for whole-genome sequencing data from 1950 tumors, non-inherited mutations from 129 normal samples, and acquired mutations in 3 cell line models of stress-induced adaptive mutation. We introduce statistical methods to identify mutational clusters, quantify their shapes and tease out the potential mechanism that produced them. Our results show that mutations in both normal and cancer samples are indeed clustered and have shapes indicative of SIM. Clusters in normal samples occur more often in the same genomic location across samples than in cancer suggesting loss of regulation over the mutational process during carcinogenesis. Additionally, the signatures of TLS contribute the most to mutational cluster formation in both patient samples as well as experimental models of SIM. Furthermore, a measure of cluster shape heterogeneity was associated with cancer patient survival with a hazard ratio of 5.744 (Cox Proportional Hazard Regression, 95% CI: 1.824-18.09). Our results support the conclusion that the ancient and evolutionary-conserved adaptive mutation response found in bacteria is a source of genomic instability in cancer. Biological adaptation through SIM might explain the ability of tumors to evolve in the face of strong selective pressures such as treatment and suggests that the conventional 'hit it hard' approaches to therapy could prove themselves counterproductive.

3.
Prog Biophys Mol Biol ; 165: 49-55, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34371024

RESUMEN

Cancer or cancer-like phenomena pervade multicellular life, implying deep evolutionary roots. Many of the hallmarks of cancer recapitulate unicellular modalities, suggesting that cancer initiation and progression represent a systematic reversion to simpler ancestral phenotypes in response to a stress or insult. This so-called atavism theory may be tested using phylostratigraphy, which can be used to assign ages to genes. Several research groups have confirmed that cancer cells tend to over-express evolutionary older genes, and rewire the architecture linking unicellular and multicellular gene networks. In addition, some of the elevated mutation rate - a well-known hallmark of cancer - is actually self-inflicted, driven by genes found to be homologs of the ancient SOS genes activated in stressed bacteria, and employed to evolve biological workarounds. These findings have obvious implications for therapy.


Asunto(s)
Neoplasias , Bacterias/genética , Evolución Biológica , Redes Reguladoras de Genes , Humanos , Neoplasias/genética , Fenotipo
4.
Bioessays ; 43(7): e2000305, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33984158

RESUMEN

It has long been recognized that cancer onset and progression represent a type of reversion to an ancestral quasi-unicellular phenotype. This general concept has been refined into the atavistic model of cancer that attempts to provide a quantitative analysis and testable predictions based on genomic data. Over the past decade, support for the multicellular-to-unicellular reversion predicted by the atavism model has come from phylostratigraphy. Here, we propose that cancer onset and progression involve more than a one-off multicellular-to-unicellular reversion, and are better described as a series of reversionary transitions. We make new predictions based on the chronology of the unicellular-eukaryote-to-multicellular-eukaryote transition. We also make new predictions based on three other evolutionary transitions that occurred in our lineage: eukaryogenesis, oxidative phosphorylation and the transition to adaptive immunity. We propose several modifications to current phylostratigraphy to improve age resolution to test these predictions. Also see the video abstract here: https://youtu.be/3unEu5JYJrQ.


Asunto(s)
Evolución Biológica , Neoplasias , Eucariontes , Células Eucariotas , Humanos , Neoplasias/genética , Fenotipo
5.
Comp Med ; 70(5): 358-367, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32753092

RESUMEN

In humans, abnormal thickening of the left ventricle of the heart clinically defines hypertrophic cardiomyopathy (HCM), a common inherited cardiovascular disorder that can precede a sudden cardiac death event. The wide range of clinical presentations in HCM obscures genetic variants that may influence an individual's susceptibility to sudden cardiac death. Although exon sequencing of major sarcomere genes can be used to detect high-impact causal mutations, this strategy is successful in only half of patient cases. The incidence of left ventricular hypertrophy (LVH) in a managed research colony of rhesus macaques provides an excellent comparative model in which to explore the genomic etiology of severe HCM and sudden cardiac death. Because no rhesus HCM-associated mutations have been reported, we used a next-generation genotyping assay that targets 7 sarcomeric rhesus genes within 63 genomic sites that are orthologous to human genomic regions known to harbor HCM disease variants. Amplicon sequencing was performed on 52 macaques with confirmed LVH and 42 unrelated, unaffected animals representing both the Indian and Chinese rhesus macaque subspecies. Bias-reduced logistic regression uncovered a risk haplotype in the rhesus MYBPC3 gene, which is frequently disrupted in both human and feline HCM; this haplotype implicates an intronic variant strongly associated with disease in either homozygous or carrier form. Our results highlight that leveraging evolutionary genomic data provides a unique, practical strategy for minimizing population bias in complex disease studies.


Asunto(s)
Cardiomiopatía Hipertrófica , Proteínas Portadoras , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/veterinaria , Proteínas Portadoras/genética , Gatos , Haplotipos , Humanos , Macaca mulatta/genética , Mutación
7.
PLoS One ; 12(4): e0176258, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28441401

RESUMEN

Cancer is sometimes depicted as a reversion to single cell behavior in cells adapted to live in a multicellular assembly. If this is the case, one would expect that mutation in cancer disrupts functional mechanisms that suppress cell-level traits detrimental to multicellularity. Such mechanisms should have evolved with or after the emergence of multicellularity. This leads to two related, but distinct hypotheses: 1) Somatic mutations in cancer will occur in genes that are younger than the emergence of multicellularity (1000 million years [MY]); and 2) genes that are frequently mutated in cancer and whose mutations are functionally important for the emergence of the cancer phenotype evolved within the past 1000 million years, and thus would exhibit an age distribution that is skewed to younger genes. In order to investigate these hypotheses we estimated the evolutionary ages of all human genes and then studied the probability of mutation and their biological function in relation to their age and genomic location for both normal germline and cancer contexts. We observed that under a model of uniform random mutation across the genome, controlled for gene size, genes less than 500 MY were more frequently mutated in both cases. Paradoxically, causal genes, defined in the COSMIC Cancer Gene Census, were depleted in this age group. When we used functional enrichment analysis to explain this unexpected result we discovered that COSMIC genes with recessive disease phenotypes were enriched for DNA repair and cell cycle control. The non-mutated genes in these pathways are orthologous to those underlying stress-induced mutation in bacteria, which results in the clustering of single nucleotide variations. COSMIC genes were less common in regions where the probability of observing mutational clusters is high, although they are approximately 2-fold more likely to harbor mutational clusters compared to other human genes. Our results suggest this ancient mutational response to stress that evolved among prokaryotes was co-opted to maintain diversity in the germline and immune system, while the original phenotype is restored in cancer. Reversion to a stress-induced mutational response is a hallmark of cancer that allows for effectively searching "protected" genome space where genes causally implicated in cancer are located and underlies the high adaptive potential and concomitant therapeutic resistance that is characteristic of cancer.


Asunto(s)
Mutación , Neoplasias/genética , Oncogenes , Animales , Ciclo Celular/genética , Reparación del ADN/genética , Bases de Datos Genéticas , Humanos , Fenotipo , Filogenia
8.
Sci Rep ; 6: 35227, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27731371

RESUMEN

In carcinogenesis, intercellular interactions within and between cell types are critical but remain poorly understood. We present a study on intercellular interactions between normal and premalignant epithelial cells and their functional relevance in the context of premalignant to malignant progression in Barrett's esophagus. Using whole transcriptome profiling we found that in the presence of normal epithelial cells, dysplastic cells but not normal cells, exhibit marked down-regulation of a number of key signaling pathways, including the transforming growth factor beta (TGFß) and epithelial growth factor (EGF). Functional assays revealed both cell types showed repressed proliferation and significant changes in motility (speed, displacement and directionality) as a result of interactions between the two cell types. Cellular interactions appear to be mediated through both direct cell-cell contact and secreted ligands. The findings of this study are important in that they reveal, for the first time, the effects of cellular communication on gene expression and cellular function between premalignant (dysplastic) epithelial cells and their normal counterparts.


Asunto(s)
Esófago de Barrett/patología , Regulación de la Expresión Génica , Transcripción Genética , Comunicación Celular , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Progresión de la Enfermedad , Factor de Crecimiento Epidérmico/metabolismo , Epitelio/patología , Humanos , Análisis de Secuencia de ARN , Transcriptoma , Factor de Crecimiento Transformador beta/metabolismo
9.
Sci Rep ; 6: 30593, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27503568

RESUMEN

The histone deacetylase (HDAC) inhibitor vorinostat has received significant attention in recent years as an 'epigenetic' drug used to treat solid tumors. However, its mechanisms of action are not entirely understood, particularly with regard to its interaction with the aberrations in 3D nuclear structure that accompany neoplastic progression. We investigated the impact of vorinostat on human esophageal epithelial cell lines derived from normal, metaplastic (pre-cancerous), and malignant tissue. Using a combination of novel optical computed tomography (CT)-based quantitative 3D absorption microscopy and conventional confocal fluorescence microscopy, we show that subjecting malignant cells to vorinostat preferentially alters their 3D nuclear architecture relative to non-cancerous cells. Optical CT (cell CT) imaging of fixed single cells showed that drug-treated cancer cells exhibit significant alterations in nuclear morphometry. Confocal microscopy revealed that vorinostat caused changes in the distribution of H3K9ac-marked euchromatin and H3K9me3-marked constitutive heterochromatin. Additionally, 3D immuno-FISH showed that drug-induced expression of the DNA repair gene MGMT was accompanied by spatial relocation toward the center of the nucleus in the nuclei of metaplastic but not in non-neoplastic cells. Our data suggest that vorinostat's differential modulation of 3D nuclear architecture in normal and abnormal cells could play a functional role in its anti-cancer action.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Neoplasias Esofágicas/metabolismo , Esófago/citología , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Línea Celular Tumoral , Núcleo Celular/química , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Esófago/efectos de los fármacos , Esófago/metabolismo , Esófago/patología , Histonas/metabolismo , Humanos , Imagenología Tridimensional , Microscopía Confocal , Microscopía Fluorescente , Vorinostat
11.
Cancer Cell ; 29(5): 723-736, 2016 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-27165744

RESUMEN

We describe a comprehensive genomic characterization of adrenocortical carcinoma (ACC). Using this dataset, we expand the catalogue of known ACC driver genes to include PRKAR1A, RPL22, TERF2, CCNE1, and NF1. Genome wide DNA copy-number analysis revealed frequent occurrence of massive DNA loss followed by whole-genome doubling (WGD), which was associated with aggressive clinical course, suggesting WGD is a hallmark of disease progression. Corroborating this hypothesis were increased TERT expression, decreased telomere length, and activation of cell-cycle programs. Integrated subtype analysis identified three ACC subtypes with distinct clinical outcome and molecular alterations which could be captured by a 68-CpG probe DNA-methylation signature, proposing a strategy for clinical stratification of patients based on molecular markers.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Genoma Humano/genética , Genómica/métodos , Adolescente , Neoplasias de la Corteza Suprarrenal/patología , Neoplasias de la Corteza Suprarrenal/terapia , Carcinoma Corticosuprarrenal/patología , Carcinoma Corticosuprarrenal/terapia , Adulto , Anciano , Anciano de 80 o más Años , Niño , Metilación de ADN , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación , Evaluación de Resultado en la Atención de Salud , Pronóstico , Adulto Joven
12.
PLoS One ; 11(3): e0150629, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963385

RESUMEN

CONTEXT: Adrenocortical carcinomas (ACC) are a rare tumor type with a poor five-year survival rate and limited treatment options. OBJECTIVE: Understanding of the molecular pathogenesis of this disease has been aided by genomic analyses highlighting alterations in TP53, WNT, and IGF signaling pathways. Further elucidation is needed to reveal therapeutically actionable targets in ACC. DESIGN: In this study, global DNA methylation levels were assessed by the Infinium HumanMethylation450 BeadChip Array on 18 ACC tumors and 6 normal adrenal tissues. A new, non-linear correlation approach, the discretization method, assessed the relationship between DNA methylation/gene expression across ACC tumors. RESULTS: This correlation analysis revealed epigenetic regulation of genes known to modulate TP53, WNT, and IGF signaling, as well as silencing of the tumor suppressor MARCKS, previously unreported in ACC. CONCLUSIONS: DNA methylation may regulate genes known to play a role in ACC pathogenesis as well as known tumor suppressors.


Asunto(s)
Neoplasias de la Corteza Suprarrenal , Carcinoma Corticosuprarrenal , Metilación de ADN , ADN de Neoplasias , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias , Transducción de Señal , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Carcinoma Corticosuprarrenal/genética , Carcinoma Corticosuprarrenal/metabolismo , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Humanos , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética
13.
Clin Transl Med ; 5(1): 1, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26754547

RESUMEN

BACKGROUND: Adrenocortical carcinoma (ACC) is an aggressive cancer with a 5 year survival rate of 20-30 %. Various factors have been implicated in the pathogenesis of ACC including dysregulation of the G2/M transition and aberrant activity of p53 and MDM2. Polo-like kinase 1 (PLK-1) negatively modulates p53 functioning, promotes MDM2 activity through its phosphorylation, and is involved in the G2/M transition. Gene expression profiling of 44 ACC samples showed that increased expression of PLK-1 in 29 % of ACC. Consequently, we examined PLK-1's role in the modulation of the p53 signaling pathway in adrenocortical cancer. METHODS: We used siRNA knock down PLK-1 and pharmacological inhibition of PLK-1 and MDM2 ACC cell lines SW-13 and H295R. We examined viability, protein expression, p53 transactivation, and induction of apoptosis. RESULTS: Knocking down expression of PLK-1 with siRNA or inhibition of PLK-1 by a small molecule inhibitor, BI-2536, resulted in a loss of viability of up to 70 % in the ACC cell lines H295R and SW-13. In xenograft models, BI-2536 demonstrated marked inhibition of growth of SW-13 with less inhibition of H295R. BI-2536 treatment resulted in a decrease in mutant p53 protein in SW-13 cells but had no effect on wild-type p53 protein levels in H295R cells. Additionally, inhibition of PLK-1 restored wild-type p53's transactivation and apoptotic functions in H295R cells, while these functions of mutant p53 were restored only to a smaller extent. Furthermore, inhibition of MDM2 with nutlin-3 reduced the viability of both the ACC cells and also reactivated wild-type p53's apoptotic function. Inhibition of PLK-1 sensitized the ACC cell lines to MDM2 inhibition and this dual inhibition resulted in an additive apoptotic response in H295R cells with wild-type p53. CONCLUSIONS: These preclinical studies suggest that targeting p53 through PLK-1 is an attractive chemotherapy strategy warranting further investigation in adrenocortical cancer.

14.
World J Surg ; 38(6): 1296-305, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24633422

RESUMEN

BACKGROUND: Recent advances in the treatment of cancer have focused on targeting genomic aberrations with selective therapeutic agents. In radioiodine resistant aggressive papillary thyroid cancers, there remain few effective therapeutic options. A 62-year-old man who underwent multiple operations for papillary thyroid cancer and whose metastases progressed despite standard treatments provided tumor tissue. METHODS: We analyzed tumor and whole blood DNA by whole genome sequencing, achieving 80× or greater coverage over 94 % of the exome and 90 % of the genome. We determined somatic mutations and structural alterations. RESULTS: We found a total of 57 somatic mutations in 55 genes of the cancer genome. There was notably a lack of mutations in NRAS and BRAF, and no RET/PTC rearrangement. There was a mutation in the TRAPP oncogene and a loss of heterozygosity of the p16, p18, and RB1 tumor suppressor genes. The oncogenic driver for this tumor is a translocation involving the genes for anaplastic lymphoma receptor tyrosine kinase (ALK) and echinoderm microtubule associated protein like 4 (EML4). The EML4-ALK translocation has been reported in approximately 5 % of lung cancers, as well as in pediatric neuroblastoma, and is a therapeutic target for crizotinib. CONCLUSIONS: This is the first report of the whole genomic sequencing of a papillary thyroid cancer in which we identified an EML4-ALK translocation of a TRAPP oncogene mutation. These findings suggest that this tumor has a more distinct oncogenesis than BRAF mutant papillary thyroid cancer. Whole genome sequencing can elucidate an oncogenic context and expose potential therapeutic vulnerabilities in rare cancers.


Asunto(s)
Carcinoma/genética , Proteínas de Ciclo Celular/genética , Proteínas Asociadas a Microtúbulos/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Pirazoles/administración & dosificación , Piridinas/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Serina Endopeptidasas/genética , Neoplasias de la Tiroides/genética , Administración Oral , Quinasa de Linfoma Anaplásico , Carcinoma/secundario , Carcinoma/cirugía , Carcinoma Papilar , Proteínas de Ciclo Celular/efectos de los fármacos , Mapeo Cromosómico , Crizotinib , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Masculino , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Mutación , Invasividad Neoplásica/patología , Recurrencia Local de Neoplasia/diagnóstico por imagen , Recurrencia Local de Neoplasia/genética , Proteínas Proto-Oncogénicas B-raf/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Tirosina Quinasas Receptoras/efectos de los fármacos , Medición de Riesgo , Serina Endopeptidasas/efectos de los fármacos , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/secundario , Neoplasias de la Tiroides/cirugía , Tiroidectomía/métodos , Factores de Tiempo , Tomografía Computarizada por Rayos X/métodos , Translocación Genética/genética , Resultado del Tratamiento
15.
J Child Neurol ; 29(9): NP81-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24141276

RESUMEN

Circulating biomarkers such as somatic chromosome mutations are novel diagnostic tools to detect cancer noninvasively. We describe focal deletions found in a patient with atypical teratoid rhabdoid tumor, a highly aggressive early childhood pediatric tumor. First, we used magnetic resonance imaging (MRI) and histopathology to study the tumor anatomy. Next, we used whole genome sequencing (Next Gen Sequencing) and Bioinformatics interrogation to discover the presence of 3 focal deletions in tumor tissue and 2 of these 3 focal deletions in patient's blood also. About 20% of the blood DNA sequencing reads matched the tumor DNA reads at the SMARCB1 gene locus. Circulating, tumor-specific DNA aberrations are a promising biomarker for atypical teratoid rhabdoid tumor patients. The high percentage of tumor DNA detected in blood indicates that either circulating brain tumor cells lyse in the blood or that contents of brain tumor cells traverse a possibly compromised blood-brain barrier in this patient.


Asunto(s)
Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/genética , Análisis Mutacional de ADN/métodos , Eliminación de Gen , Tumor Rabdoide/sangre , Tumor Rabdoide/genética , Biomarcadores/sangre , Encéfalo/patología , Encéfalo/cirugía , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Humanos , Imagen por Resonancia Magnética , Tumor Rabdoide/patología , Tumor Rabdoide/cirugía , Proteína SMARCB1 , Factores de Transcripción/genética
16.
Surgery ; 154(6): 1405-16; discussion 1416, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24238056

RESUMEN

BACKGROUND: Adrenocortical carcinoma (ACC) is associated with poor survival rates. The objective of the study was to analyze ACC gene expression profiling data for prognostic biomarkers and therapeutic targets. METHODS: We profiled 44 ACC and 4 normal adrenals on Affymetrix U133 Plus 2 expression microarrays. Pathway and transcriptional enrichment analysis was performed. Protein levels were determined by Western blot. Drug efficacy was assessed against ACC cell lines. Previously published expression datasets were analyzed for validation. RESULTS: Pathway enrichment analysis identified marked dysregulation of cyclin-dependent kinases and mitosis. Overexpression of PTTG1, which encodes securin, a negative regulator of p53, was identified as a marker of poor survival. Median survival for patients with tumors expressing high PTTG1 levels (log2 ratio of PTTG1 to average ß-actin <-3.04) was 1.8 years compared with 9.0 years if tumors expressed lower levels of PTTG1 (P < .0001). Analysis of a previously published dataset confirmed the association of high PTTG1 expression with a poor prognosis. Treatment of 2 ACC cell lines with vorinostat decreased securin levels and inhibited cell growth (median inhibition concentrations of 1.69 µmol/L and 0.891 µmol/L, for SW-13 and H295R, respectively). CONCLUSION: Overexpression of PTTG1 is correlated with poor survival in ACC. PTTG1/securin is a prognostic biomarker and warrants investigation as a therapeutic target.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Securina/genética , Neoplasias de la Corteza Suprarrenal/tratamiento farmacológico , Neoplasias de la Corteza Suprarrenal/metabolismo , Carcinoma Corticosuprarrenal/tratamiento farmacológico , Carcinoma Corticosuprarrenal/metabolismo , Adulto , Anciano , Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Persona de Mediana Edad , Pronóstico , Securina/antagonistas & inhibidores , Securina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Vorinostat , Adulto Joven
18.
PLoS One ; 7(1): e29230, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22242161

RESUMEN

BACKGROUND: Grading schemes for breast cancer diagnosis are predominantly based on pathologists' qualitative assessment of altered nuclear structure from 2D brightfield microscopy images. However, cells are three-dimensional (3D) objects with features that are inherently 3D and thus poorly characterized in 2D. Our goal is to quantitatively characterize nuclear structure in 3D, assess its variation with malignancy, and investigate whether such variation correlates with standard nuclear grading criteria. METHODOLOGY: We applied micro-optical computed tomographic imaging and automated 3D nuclear morphometry to quantify and compare morphological variations between human cell lines derived from normal, benign fibrocystic or malignant breast epithelium. To reproduce the appearance and contrast in clinical cytopathology images, we stained cells with hematoxylin and eosin and obtained 3D images of 150 individual stained cells of each cell type at sub-micron, isotropic resolution. Applying volumetric image analyses, we computed 42 3D morphological and textural descriptors of cellular and nuclear structure. PRINCIPAL FINDINGS: We observed four distinct nuclear shape categories, the predominant being a mushroom cap shape. Cell and nuclear volumes increased from normal to fibrocystic to metastatic type, but there was little difference in the volume ratio of nucleus to cytoplasm (N/C ratio) between the lines. Abnormal cell nuclei had more nucleoli, markedly higher density and clumpier chromatin organization compared to normal. Nuclei of non-tumorigenic, fibrocystic cells exhibited larger textural variations than metastatic cell nuclei. At p<0.0025 by ANOVA and Kruskal-Wallis tests, 90% of our computed descriptors statistically differentiated control from abnormal cell populations, but only 69% of these features statistically differentiated the fibrocystic from the metastatic cell populations. CONCLUSIONS: Our results provide a new perspective on nuclear structure variations associated with malignancy and point to the value of automated quantitative 3D nuclear morphometry as an objective tool to enable development of sensitive and specific nuclear grade classification in breast cancer diagnosis.


Asunto(s)
Neoplasias de la Mama/patología , Mama/patología , Células Epiteliales/patología , Enfermedad Fibroquística de la Mama/patología , Imagenología Tridimensional/métodos , Línea Celular , Núcleo Celular/patología , Femenino , Humanos , Metástasis de la Neoplasia
19.
Ann Surg ; 255(1): 140-6, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22156929

RESUMEN

BACKGROUND: Traditional drug discovery methods have a limited role in rare cancers. We hypothesized that molecular technology including gene expression profiling could expose novel targets for therapy in adrenocortical carcinoma (ACC), a rare and lethal cancer. SPARC (secreted protein acidic rich in cysteine) is an albumin-binding matrix-associated protein that is proposed to act as a mechanism for the increased efficacy of a nanoparticle albumin-bound preparation of the antimicrotubular drug Paclitaxel (nab-paclitaxel). METHODS: The transcriptomes of 19 ACC tumors and 4 normal adrenal glands were profiled on Affymetrix U133 Plus2 expression microarrays to identify genes representing potential therapeutic targets. Immunohistochemical analysis for target proteins was performed on 10 ACC, 6 benign adenomas, and 1 normal adrenal gland. Agents known to inhibit selected targets were tested in comparison with mitotane in the 2 ACC cell lines (H295R and SW-13) in vitro and in mouse xenografts. RESULTS: SPARC expression is increased in ACC samples by 1.56 ± 0.44 (µ ± SD) fold. Paclitaxel and nab-paclitaxel show in vitro inhibition of H295R and SW-13 cells at IC50 concentrations of 0.33 µM and 0.0078 µM for paclitaxel and 0.35 µM and 0.0087 µM for nab-paclitaxel compared with mitotane concentrations of 15.9 µM and 46.4 µM. In vivo nab-paclitaxel treatment shows a greater decrease in tumor weight in both xenograft models than mitotane. CONCLUSIONS: Biological insights garnered through expression profiling of ACC tumors suggest further investigation into the use of nab-paclitaxel for the treatment of ACC.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/patología , Adenoma Corticosuprarrenal/genética , Adenoma Corticosuprarrenal/patología , Antineoplásicos Fitogénicos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Perfilación de la Expresión Génica , Paclitaxel/farmacología , Paclitaxel Unido a Albúmina , Albúminas/farmacología , Animales , Antineoplásicos Hormonales/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Mitotano/farmacología , Nanopartículas , Trasplante de Neoplasias/patología , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Horm Cancer ; 2(6): 385-92, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22170383

RESUMEN

Standard chemotherapy for adrenocortical cancer currently is under evaluation in the context of the recently completed FIRM-ACT evaluating the combination of mitotane with either streptozocin or etoposide, cisplatin, and doxorubicin. New agents are eagerly sought by the ACC community that hopes to make progress against this deadly disease. Investigators have begun to dissect the molecular and genomic context of ACC with a goal of identifying potential novel therapeutic agents. One gene consistently overexpressed in ACC is insulin growth factor type 2. Targeting its receptor IGF1R has shown encouraging results in ACC cell lines and against murine xenografts. As a result, clinical trials to evaluate agents targeting the IGF1R have been done including mitotane and IMC-A12 (a monoclonal antibody) and the GALACCTIC trial that has just completed accrual to evaluate OSI-906, a small molecule IGF1R antagonist. On the horizon are other agents targeting other tyrosine kinases, including EGF and FGF, and novel strategies such as individualized tumor analysis to select treatment.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/tratamiento farmacológico , Carcinoma Corticosuprarrenal/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Terapia Molecular Dirigida/métodos , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/patología , Carcinoma Corticosuprarrenal/genética , Carcinoma Corticosuprarrenal/patología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Biomarcadores de Tumor , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Ratones , Mitotano/uso terapéutico , Medicina de Precisión , Receptor IGF Tipo 1/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...