Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Clin Imaging Sci ; 12: 18, 2022.
Article in English | MEDLINE | ID: mdl-35510240

ABSTRACT

Following low anterior resection (LAR) of the colon, an image-guided assessment of the anastomosis for leak is typically performed using an enema via a rectal catheter, whether by CT or fluoroscopy. However, there is potential for poor assessment due to inappropriate catheter positioning as well as potential risk that the anastomosis becomes compromised by the balloon inflation. This article discusses the adaptation of a novel double-balloon catheter (originally designed by a member of our institution for use in pediatric intussusception reduction) for assessment of low rectal anastomoses. The goal of this technical note is to demonstrate our experience with this catheter, primarily through example cases, and explain its potential for optimizing colon distension, minimizing improper catheter placement, and potentially reducing the risk of iatrogenic anastomosis disruption.

2.
Acad Radiol ; 29(8): 1149-1156, 2022 08.
Article in English | MEDLINE | ID: mdl-34598868

ABSTRACT

RATIONALE AND OBJECTIVES: To date, no clinically useful classification system has been developed for reliably differentiating mucinous cystic neoplasm (MCN) from a benign hepatic cyst (BHC) in the liver. The objective was to use machine learning and a multi-center study design to develop and assess the performance of a novel classification system for predicting whether a hepatic cystic lesion represents MCN or BHC. MATERIALS AND METHODS: A multi-center cohort study identified 154 surgically resected hepatic cystic lesions in 154 subjects which were pathologic confirmed as MCN (43) or BHC (111). Readers at each institution recorded seven pre-determined imaging features previously identified as potential differentiating features from prior publications. The contribution of each of these features to differentiating MCN from BHC was assessed by machine learning to develop an optimal classification system. RESULTS: Although several of the assessed imaging features demonstrated statistical significance, only 3 imaging features were found by machine learning to significantly contribute to a potential classification system: (1) solid enhancing nodule (2) all septations arising from an external macro-lobulation (3) whether the lesion was solitary or one of multiple cystic liver lesions. The optimal classification system had only four categories and correctly identified 144/154 lesion (93.5%). CONCLUSION: This multi-center follow-up study was able to use machine learning to develop a highly accurate classification system for differentiation of hepatic MCN from BHC, which could be readily applied to clinical practice.


Subject(s)
Cysts , Pancreatic Neoplasms , Cohort Studies , Cysts/diagnostic imaging , Follow-Up Studies , Humans , Liver/diagnostic imaging , Liver/pathology , Liver Diseases , Machine Learning , Pancreatic Neoplasms/pathology
3.
J Clin Imaging Sci ; 11: 68, 2021.
Article in English | MEDLINE | ID: mdl-34992944

ABSTRACT

Dual-energy computed tomography (DECT) has become increasingly available and can be readily incorporated into clinical practice. Although DECT can provide a wide variety of spectral imaging reconstructions, most clinically valuable information is available from a limited number of standard image reconstructions including virtual non-contrast and iodine overlay. The combination of these standard reconstructions can be used for specific diagnostic tasks that provide added value over traditional CT protocols. In this pictorial essay, the added value of these standard reconstructed images will be demonstrated by case examples for diseases specifically related to the gastrointestinal system.

4.
J Comput Assist Tomogr ; 40(4): 557-9, 2016.
Article in English | MEDLINE | ID: mdl-27023858

ABSTRACT

The authors describe a case of a patient with a solitary hepatocellular carcinoma status post transarterial chemoembolization. Follow-up imaging was performed using dual-energy computed tomography. The study was performed with and without contrast and a virtual noncontrast data set was constructed from the postcontrast images. The evaluation of this patient status post transarterial chemoembolization with virtual noncontrast alone erroneously suggested enhancement and viable tumor. However, examination of true noncontrast images revealed these findings to be due to the subtraction of iodine in Ethiodol within the treated lesion.


Subject(s)
Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/methods , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/therapy , Radiography, Dual-Energy Scanned Projection/methods , Contrast Media , Ethiodized Oil , Humans , Male , Middle Aged , Tomography, X-Ray Computed , Treatment Outcome , User-Computer Interface
5.
Diabetes ; 59(10): 2662-71, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20622168

ABSTRACT

OBJECTIVE: Genome-wide association studies have identified a single nucleotide polymorphism (SNP), rs560887, located in a G6PC2 intron that is highly correlated with variations in fasting plasma glucose (FPG). G6PC2 encodes an islet-specific glucose-6-phosphatase catalytic subunit. This study examines the contribution of two G6PC2 promoter SNPs, rs13431652 and rs573225, to the association signal. RESEARCH DESIGN AND METHODS: We genotyped 9,532 normal FPG participants (FPG <6.1 mmol/l) for three G6PC2 SNPs, rs13431652 (distal promoter), rs573225 (proximal promoter), rs560887 (3rd intron). We used regression analyses adjusted for age, sex, and BMI to assess the association with FPG and haplotype analyses to assess comparative SNP contributions. Fusion gene and gel retardation analyses characterized the effect of rs13431652 and rs573225 on G6PC2 promoter activity and transcription factor binding. RESULTS: Genetic analyses provide evidence for a strong contribution of the promoter SNPs to FPG variability at the G6PC2 locus (rs13431652: ß = 0.075, P = 3.6 × 10(-35); rs573225 ß = 0.073 P = 3.6 × 10(-34)), in addition to rs560887 (ß = 0.071, P = 1.2 × 10(-31)). The rs13431652-A and rs573225-A alleles promote increased NF-Y and Foxa2 binding, respectively. The rs13431652-A allele is associated with increased FPG and elevated promoter activity, consistent with the function of G6PC2 in pancreatic islets. In contrast, the rs573225-A allele is associated with elevated FPG but reduced promoter activity. CONCLUSIONS: Genetic and in situ functional data support a potential role for rs13431652, but not rs573225, as a causative SNP linking G6PC2 to variations in FPG, though a causative role for rs573225 in vivo cannot be ruled out.


Subject(s)
Fasting , Glucose-6-Phosphatase/genetics , Metabolic Syndrome/genetics , Promoter Regions, Genetic , Adult , Blood Glucose/genetics , Child , Cohort Studies , DNA Primers , Female , Finland/epidemiology , Gene Expression Regulation , Genetic Variation , Humans , Insulin Resistance/genetics , Introns/genetics , Mothers , Organ Specificity , Polymorphism, Single Nucleotide , RNA/genetics , RNA/isolation & purification , Reference Values , White People/genetics
6.
Biochem J ; 417(2): 611-20, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18847435

ABSTRACT

The G6Pase (glucose-6-phosphatase catalytic subunit) catalyses the final step in the gluconeogenic and glycogenolytic pathways, the hydrolysis of glucose-6-phosphate to glucose. We show here that, in HepG2 hepatoma cells, EGF (epidermal growth factor) inhibits basal mouse G6Pase fusion gene transcription. Several studies have shown that insulin represses basal mouse G6Pase fusion gene transcription through FOXO1 (forkhead box O1), but Stoffel and colleagues have recently suggested that insulin can also regulate gene transcription through FOXA2 (forkhead box A2) [Wolfrum, Asilmaz, Luca, Friedman and Stoffel (2003) Proc. Natl. Acad. Sci. 100, 11624-11629]. A combined GR (glucocorticoid receptor)-FOXA2 binding site is located between -185 and -174 in the mouse G6Pase promoter overlapping two FOXO1 binding sites located between (-188 and -182) and (-174 and -168). Selective mutation of the FOXO1 binding sites reduced the effect of insulin, whereas mutation of the GR/FOXA2 binding site had no effect on the insulin response. In contrast, selective mutation of the FOXO1 and GR/FOXA2 binding sites both reduced the effect of EGF. The effect of these mutations was additive, since the combined mutation of both FOXO1 and GR/FOXA2 binding sites reduced the effect of EGF to a greater extent than the individual mutations. These results suggest that, in HepG2 cells, GR and/or FOXA2 are required for the inhibition of basal G6Pase gene transcription by EGF but not insulin. EGF also inhibits hepatic G6Pase gene expression in vivo, but in cultured hepatocytes EGF has the opposite effect of stimulating expression, an observation that may be explained by a switch in ErbB receptor sub-type expression following hepatocyte isolation.


Subject(s)
Biocatalysis , Epidermal Growth Factor/pharmacology , Glucose-6-Phosphatase/metabolism , Insulin/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Animals , Binding Sites , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Glucose-6-Phosphatase/genetics , Hepatocyte Nuclear Factor 1/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Subunits/metabolism , Rats
7.
J Mol Endocrinol ; 41(5): 315-28, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18753309

ABSTRACT

Islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP/G6PC2) is a major autoantigen in both mouse and human type 1 diabetes. IGRP is selectively expressed in islet beta cells and polymorphisms in the IGRP gene have recently been associated with variations in fasting blood glucose levels and cardiovascular-associated mortality in humans. Chromatin immunoprecipitation (ChIP) assays have shown that the IGRP promoter binds the islet-enriched transcription factors Pax-6 and BETA2. We show here, again using ChIP assays, that the IGRP promoter also binds the islet-enriched transcription factors MafA and Foxa2. Single binding sites for these factors were identified in the proximal IGRP promoter, mutation of which resulted in decreased IGRP fusion gene expression in betaTC-3, Hamster insulinoma tumor (HIT), and Min6 cells. ChiP assays have shown that the islet-enriched transcription factor Pdx-1 also binds the IGRP promoter, but mutational analysis of four Pdx-1 binding sites in the proximal IGRP promoter revealed surprisingly little effect of Pdx-1 binding on IGRP fusion gene expression in betaTC-3 cells. In contrast, in both HIT and Min6 cells mutation of these four Pdx-1 binding sites resulted in a approximately 50% reduction in fusion gene expression. These data suggest that the same group of islet-enriched transcription factors, namely Pdx-1, Pax-6, MafA, BETA2, and Foxa2, directly or indirectly regulate expression of the two major autoantigens in type 1 diabetes.


Subject(s)
Glucose-6-Phosphatase/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Islets of Langerhans/physiology , Maf Transcription Factors, Large/metabolism , Animals , Cell Line , Cell Line, Tumor , Cricetinae , DNA Mutational Analysis , Glucose-6-Phosphatase/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Islets of Langerhans/cytology , Maf Transcription Factors, Large/genetics , Mice , Promoter Regions, Genetic , Trans-Activators/genetics , Trans-Activators/metabolism
8.
Diabetes ; 57(1): 133-41, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942825

ABSTRACT

OBJECTIVE: Islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) is selectively expressed in islet beta-cells and is a major autoantigen in both mouse and human type 1 diabetes. This study describes the use of a combination of transgenic and transfection approaches to characterize the gene regions that confer the islet-specific expression of IGRP. RESEARCH DESIGN AND METHODS: Transgenic mice were generated containing the IGRP promoter sequence from -306, -911, or -3911 to +3 ligated to a LacZ reporter gene. Transgene expression was monitored by 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside staining of pancreatic tissue. RESULTS: In all the transgenic mice, robust LacZ expression was detected in newborn mouse islets, but expression became mosaic as animals aged, suggesting that additional elements are required for the maintenance of IGRP gene expression. VISTA analyses identified two conserved regions in the distal IGRP promoter and one in the third intron. Transfection experiments demonstrated that all three regions confer enhanced luciferase reporter gene expression in beta TC-3 cells when ligated to a minimal IGRP promoter. A transgene containing all three conserved regions was generated by using a bacterial recombination strategy to insert a LacZ cassette into exon 5 of the IGRP gene. Transgenic mice containing a 15-kbp fragment of the IGRP gene were then generated. This transgene conferred LacZ expression in newborn mouse islets; however, expression was still suppressed as animals aged. CONCLUSIONS: The data suggest that long-range enhancers 5' or 3' of the IGRP gene are required for the maintenance of IGRP gene expression in adult mice.


Subject(s)
Gene Expression Regulation , Glucose-6-Phosphatase/genetics , Islets of Langerhans/physiology , Proteins/genetics , Animals , Chromosomes, Artificial, Bacterial , Cloning, Molecular , Diabetes Mellitus, Type 1/genetics , Enhancer Elements, Genetic , Genes, Reporter , Luciferases/genetics , Mice , Mice, Transgenic , Promoter Regions, Genetic , Transfection , beta-Galactosidase/genetics
9.
Nature ; 443(7111): E10-1, 2006 Oct 05.
Article in English | MEDLINE | ID: mdl-17024043

ABSTRACT

Increased expression of the gene encoding the enzyme glucose-6-phosphatase (G6Pase) contributes to the increased production of glucose by the liver that occurs in individuals with diabetes. Puigserver et al. show that the transcription factor FOXO1 and the transcriptional co-activator PGC-1alpha act synergistically to stimulate the expression of genes in the gluconeogenesis pathway and propose that PGC-1alpha acts, in part, directly through FOXO1. Here we show that FOXO1 is neither required nor sufficient for the stimulation of G6Pase-luciferase fusion gene expression by PGC-1alpha. Our results indicate that the transcriptional interaction between FOXO1 and PGC-1alpha is indirect.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Gluconeogenesis/genetics , Glucose-6-Phosphatase/genetics , RNA-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Mice , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , Reproducibility of Results , Response Elements/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...