Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 29(15): 2508-2522, 2020 08 29.
Article in English | MEDLINE | ID: mdl-32620959

ABSTRACT

Bardet-Biedl syndrome (BBS) is a pleiotropic autosomal recessive ciliopathy affecting multiple organs. The development of potential disease-modifying therapy for BBS will require concurrent targeting of multi-systemic manifestations. Here, we show for the first time that monosialodihexosylganglioside accumulates in Bbs2-/- cilia, indicating impairment of glycosphingolipid (GSL) metabolism in BBS. Consequently, we tested whether BBS pathology in Bbs2-/- mice can be reversed by targeting the underlying ciliary defect via reduction of GSL metabolism. Inhibition of GSL synthesis with the glucosylceramide synthase inhibitor Genz-667161 decreases the obesity, liver disease, retinal degeneration and olfaction defect in Bbs2-/- mice. These effects are secondary to preservation of ciliary structure and signaling, and stimulation of cellular differentiation. In conclusion, reduction of GSL metabolism resolves the multi-organ pathology of Bbs2-/- mice by directly preserving ciliary structure and function towards a normal phenotype. Since this approach does not rely on the correction of the underlying genetic mutation, it might translate successfully as a treatment for other ciliopathies.


Subject(s)
Bardet-Biedl Syndrome/genetics , Cilia/genetics , Ciliopathies/genetics , Proteins/genetics , Animals , Bardet-Biedl Syndrome/drug therapy , Bardet-Biedl Syndrome/pathology , Cell Differentiation/drug effects , Cilia/pathology , Ciliopathies/drug therapy , Ciliopathies/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gangliosides/biosynthesis , Gangliosides/genetics , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , Glycosphingolipids/biosynthesis , Glycosphingolipids/genetics , Mice, Knockout
2.
Crit Care Nurse ; 37(3): 42-48, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28572100

ABSTRACT

BACKGROUND: Catheter-associated urinary tract infections are preventable adverse outcomes that increase hospital morbidity, mortality, and costs. These infections are particularly prevalent in intensive care units. OBJECTIVES: To describe the success of an 18-bed neurological intensive care unit in using several nurse-implemented strategies that reduced the number of catheter-associated urinary tract infections. METHODS: A prospective, interventional design with application of evidence-based practices to reduce catheter-associated urinary tract infections was used. RESULTS: Before implementation of the strategies, 40 catheter-associated urinary tract infections were reported for 2012 and 38 for 2013. The standardized infection ratio was 2.04 for 2012 (95% CI, 1.456-2.775) and 2.34 (95% CI, 1.522-3.312) for 2013. After implementation of the strategies, significantly fewer catheter-associated urinary tract infections were reported. In 2014, a total of 15 infections were reported, and the standardized infection ratio was less than 1.0 (95% CI, 0.685-1.900). CONCLUSIONS: Application of current evidence-based practices resulted in a substantial decrease in the number of catheter-associated urinary tract infections and a lower standardized infection ratio. These findings support current recommendations for "bundling" to maximize outcomes.


Subject(s)
Catheter-Related Infections/etiology , Catheter-Related Infections/nursing , Catheters, Indwelling/adverse effects , Cross Infection/prevention & control , Urinary Catheterization/adverse effects , Urinary Tract Infections/complications , Urinary Tract Infections/nursing , Adult , Aged , Aged, 80 and over , Critical Care Nursing/standards , Female , Humans , Male , Middle Aged , Practice Guidelines as Topic , Prospective Studies
4.
Nutr Metab (Lond) ; 13: 17, 2016.
Article in English | MEDLINE | ID: mdl-26933443

ABSTRACT

BACKGROUND: The liver is an important site of fat oxidation, which participates in the metabolic regulation of food intake. We showed previously that mice with genetically inactivated Acads, encoding short-chain acyl-CoA dehydrogenase (SCAD), shift food consumption away from fat and toward carbohydrate when tested in a macronutrient choice paradigm. This phenotypic eating behavior suggests a link between fat oxidation and nutrient choice which may involve an energy sensing mechanism. To identify hepatic processes that could trigger energy-related signals, we have now performed transcriptional, metabolite and physiological analyses in Acads-/- mice following short-term (2 days) exposure to either high- or low-fat diet. METHODS AND RESULTS: Metabolite analysis revealed 25 acylcarnitine species that were altered by diet and/or genotype. Compared to wild-type mice, phosphorylated AMP-activated protein kinase was 40 % higher in Acads-/- mice after short-term high-fat diet, indicating a low ATP/AMP ratio. Metabolite analyses in isolated liver mitochondria from Acads-/- mice during ADP-linked respiration on butyrate demonstrated a reduced oxygen consumption rate (OCR) compared to wild-type, an effect that was not observed with succinate or palmitoylcarnitine substrates. Liver transcriptomic responses in Acads-/- mice fed high- vs. lowfat diet revealed increased RXR/PPARA signaling, up-regulation of lipid handling pathways (including beta and omega oxidation), and increased mRNA expression of Nfe2l2 target genes. CONCLUSIONS: Together, these results point to an oxidative shortage in this genetic model and support the hypothesis of a lower hepatic energy state associated with SCAD deficiency and high-fat diet.

5.
Mol Metab ; 4(10): 706-17, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26500842

ABSTRACT

OBJECTIVE: Leptin modulates food reward via central leptin receptor (LepRb) expressing neurons. Food reward requires stimulation of midbrain dopamine neurons and is modulated by central leptin action, but the exact central mechanisms remain unclear. Stimulatory and inhibitory leptin actions on dopamine neurons have been reported, e.g. by indirect actions on orexin neurons or via direct innervation of dopamine neurons in the ventral tegmental area. METHODS: We showed earlier that LepRb neurons in the lateral hypothalamus (LHA) co-express the inhibitory acting neuropeptide galanin (GAL-LepRb neurons). We studied the involvement of GAL-LepRb neurons to regulate nutrient reward in mice with selective LepRb deletion from galanin neurons (GAL-LepRb(KO) mice). RESULTS: We found that the rewarding value and preference for sucrose over fat was increased in GAL-LepRb(KO) mice compared to controls. LHA GAL-LepRb neurons innervate orexin neurons, but not the VTA. Further, expression of galanin and its receptor GalR1 are decreased in the LHA of GAL-LepRb(KO) mice, resulting in increased activation of orexin neurons. CONCLUSION: We suggest galanin as an important mediator of leptin action to modulate nutrient reward by inhibiting orexin neurons.

6.
Physiol Rep ; 3(2)2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25713331

ABSTRACT

Dcpp2, Prrt1, and Has1 are plausible candidate genes for the Mnic1 (macronutrient intake-carbohydrate) locus on mouse chromosome 17, based on their map positions and sequence variants, documented expression in salivary glands, and the important role of saliva in oral food processing and taste. We investigated the effects of genotype and diet on gene expression in salivary glands (parotid, submandibular, sublingual) of carbohydrate-preferring, C57BL6J.CAST/EiJ-17.1 subcongenic mice compared to fat-preferring wild-type C57BL/6J. To achieve accurate normalization of real-time quantitative RT-PCR data, we evaluated multiple reference genes to identify the most stably expressed control genes in salivary gland tissues, and then used geometric averaging to produce a reliable normalization factor. Gene expression was measured in mice fed different diets: (1) rodent chow, (2) macronutrient selection diets, (3) high-fat diet, and (4) low-fat diet. In addition, we measured salivary hyaluronan concentrations. All three genes showed strain differences in expression, in at least one major salivary gland, and diet effects were observed in two glands. Dcpp2 expression was limited primarily to sublingual gland, and strongly decreased in B6.CAST-17.1 subcongenic mice compared to wild-type B6, regardless of diet. In contrast, both genotype and diet affected Prrt1 and Has1 expression, in a gland-specific manner, for example, Prrt1 expression in the parotid gland alone was strongly reduced in both mouse strains when fed macronutrient selection diet compared to chow. Notably, we discovered an association between diet composition and salivary hyaluronan content. These results demonstrate robust effects of genetic background and diet composition on candidate gene expression in mouse salivary glands.

7.
PDA J Pharm Sci Technol ; 68(6): 651-60, 2014.
Article in English | MEDLINE | ID: mdl-25475640

ABSTRACT

Next-generation sequencing has been evaluated at Genzyme as a means of identifying bioreactor contaminants due to its capability for detection of known and novel microbial species. In this approach, data obtained from next-generation sequencing is used to interrogate databases containing genomic sequences and identities of potential adventitious agents. We describe here the use of this approach to help identify the causative agent of a bioreactor contamination. We also present the results of spiking experiments to establish the limits of detection for DNA viruses, RNA viruses, and bacteria, in a background of Chinese hamster ovary cells, a cell line used for production of many human therapeutics. Using Illumina sequencing-based detection, all of the viruses included in this study were detected at less than 1 copy per cell, and bacteria were detected at 0.001 copy per cell. Thus, next-generation sequencing-based detection of adventitious agents is a valuable approach that can fill a critical unmet need in the detection of known and novel microorganisms in biopharmaceutical manufacturing. LAY ABSTRACT: Because biological products are manufactured in cells, the living environment must be kept sterile. Any introduction of microorganisms into the culture vessel may affect the growth and other biological properties of the cells or contaminate the product. It is therefore important to monitor the culture for such contaminants, but many methods can only detect a specific microorganism. In this study, we show that next-generation sequencing-based detection is a sensitive and complementary approach that can potentially detect a wide range of organisms.


Subject(s)
Bacteria/genetics , Bacteriological Techniques , Biological Products/analysis , Biopharmaceutics/methods , Drug Contamination/prevention & control , High-Throughput Nucleotide Sequencing , Virology/methods , Viruses/genetics , Animals , Bacteriological Techniques/standards , Biopharmaceutics/standards , Bioreactors , CHO Cells , Cell Culture Techniques , Cricetulus , DNA, Bacterial/genetics , DNA, Viral/genetics , High-Throughput Nucleotide Sequencing/standards , Humans , Limit of Detection , RNA, Viral/genetics , Reference Standards , Virology/standards
8.
PLoS One ; 9(10): e110424, 2014.
Article in English | MEDLINE | ID: mdl-25330228

ABSTRACT

The specific genes regulating the quantitative variation in macronutrient preference and food intake are virtually unknown. We fine mapped a previously identified mouse chromosome 17 region harboring quantitative trait loci (QTL) with large effects on preferential macronutrient intake-carbohydrate (Mnic1), total kilcalories (Kcal2), and total food volume (Tfv1) using interval-specific strains. These loci were isolated in the [C57BL/6J.CAST/EiJ-17.1-(D17Mit19-D17Mit50); B6.CAST-17.1] strain, possessing a ∼ 40.1 Mb region of CAST DNA on the B6 genome. In a macronutrient selection paradigm, the B6.CAST-17.1 subcongenic mice eat 30% more calories from the carbohydrate-rich diet, ∼ 10% more total calories, and ∼ 9% more total food volume per body weight. In the current study, a cross between carbohydrate-preferring B6.CAST-17.1 and fat-preferring, inbred B6 mice was used to generate a subcongenic-derived F2 mapping population; genotypes were determined using a high-density, custom SNP panel. Genetic linkage analysis substantially reduced the 95% confidence interval for Mnic1 (encompassing Kcal2 and Tfv1) from 40.1 to 29.5 Mb and more precisely established its boundaries. Notably, no genetic linkage for self-selected fat intake was detected, underscoring the carbohydrate-specific effect of this locus. A second key finding was the separation of two energy balance QTLs: Mnic1/Kcal2/Tfv1 for food intake and a newly discovered locus regulating short term body weight gain. The Mnic1/Kcal2/Tfv1 QTL was further de-limited to 19.0 Mb, based on the absence of nutrient intake phenotypes in subcongenic HQ17IIa mice. Analyses of available sequence data and gene ontologies, along with comprehensive expression profiling in the hypothalamus of non-recombinant, cast/cast and b6/b6 F2 controls, focused our attention on candidates within the QTL interval. Zfp811, Zfp870, and Btnl6 showed differential expression and also contain stop codons, but have no known biology related to food intake regulation. The genes Decr2, Ppard and Agapt1 are more appealing candidates because of their involvement in lipid metabolism and down-regulation in carbohydrate-preferring animals.


Subject(s)
Carbohydrate Metabolism/genetics , Chromosome Mapping , Chromosomes, Mammalian/genetics , Energy Intake/genetics , Genetic Loci , Genome , Animals , Genetic Linkage , Mice , Polymorphism, Single Nucleotide , Quantitative Trait Loci
9.
PLoS One ; 7(8): e41709, 2012.
Article in English | MEDLINE | ID: mdl-22936979

ABSTRACT

BACKGROUND: How signals from fatty acid metabolism are translated into changes in food intake remains unclear. Previously we reported that mice with a genetic inactivation of Acads (acyl-coenzyme A dehydrogenase, short-chain), the enzyme responsible for mitochondrial beta-oxidation of C4-C6 short-chain fatty acids (SCFAs), shift consumption away from fat and toward carbohydrate when offered a choice between diets. In the current study, we sought to indentify candidate genes and pathways underlying the effects of SCFA oxidation deficiency on food intake in Acads-/- mice. METHODOLOGY/PRINCIPAL FINDINGS: We performed a transcriptional analysis of gene expression in brain tissue of Acads-/- and Acads+/+ mice fed either a high-fat (HF) or low-fat (LF) diet for 2 d. Ingenuity Pathway Analysis revealed three top-scoring pathways significantly modified by genotype or diet: oxidative phosphorylation, mitochondrial dysfunction, and CREB signaling in neurons. A comparison of statistically significant responses in HF Acads-/- vs. HF Acads+/+ (3917) and Acads+/+ HF vs. LF Acads+/+ (3879) revealed 2551 genes or approximately 65% in common between the two experimental comparisons. All but one of these genes were expressed in opposite direction with similar magnitude, demonstrating that HF-fed Acads-deficient mice display transcriptional responses that strongly resemble those of Acads+/+ mice fed LF diet. Intriguingly, genes involved in both AMP-kinase regulation and the neural control of food intake followed this pattern. Quantitative RT-PCR in hypothalamus confirmed the dysregulation of genes in these pathways. Western blotting showed an increase in hypothalamic AMP-kinase in Acads-/- mice and HF diet increased, a key protein in an energy-sensing cascade that responds to depletion of ATP. CONCLUSIONS: Our results suggest that the decreased beta-oxidation of short-chain fatty acids in Acads-deficient mice fed HF diet produces a state of energy deficiency in the brain and that AMP-kinase may be the cellular energy-sensing mechanism linking fatty acid oxidation to feeding behavior in this model.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Diet, High-Fat/adverse effects , Signal Transduction/drug effects , Acyl-CoA Dehydrogenase/genetics , Adenosine Triphosphate/metabolism , Animals , Brain , Fatty Acids/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
10.
Proc Nutr Soc ; 71(3): 390-400, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22617310

ABSTRACT

There is considerable disagreement regarding what constitutes a healthy diet. Ever since the influential work of Cannon and Richter, it was debated whether the 'wisdom of the body' will automatically direct us to the foods we need for healthy lives or whether we must carefully learn to eat the right foods, particularly in an environment of plenty. Although it is clear that strong mechanisms have evolved to prevent consumption of foods that have previously made us sick, it is less clear whether reciprocal mechanisms exist that reinforce the consumption of healthy diets. Here, we review recent progress in providing behavioural evidence for the regulation of intake and selection of proteins, carbohydrates and fats. We examine new developments in sensory physiology enabling recognition of macronutrients both pre- and post-ingestively. Finally, we propose a general model for central neural processing of nutrient-specific appetites. We suggest that the same basic neural circuitry responsible for the homoeostatic regulation of total energy intake is also used to control consumption of specific macro- and micronutrients. Similar to salt appetite, specific appetites for other micro- and macronutrients may be encoded by unique molecular changes in the hypothalamus. Gratification of such specific appetites is then accomplished by engaging the brain motivational system to assign the highest reward prediction to exteroceptive cues previously associated with consuming the missing ingredient. A better understanding of these nutrient-specific neural processes could help design drugs and behavioural strategies that promote healthier eating.


Subject(s)
Appetite Regulation/physiology , Diet/psychology , Energy Intake/physiology , Feeding Behavior/physiology , Food Preferences/physiology , Hypothalamus/metabolism , Sensation/physiology , Animals , Feeding Behavior/psychology , Food Preferences/psychology , Homeostasis , Humans , Motivation/physiology
11.
Mol Cancer Ther ; 10(3): 404-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21252283

ABSTRACT

sFLT01 is a novel fusion protein that consists of the VEGF/PlGF (placental growth factor) binding domain of human VEGFR1/Flt-1 (hVEGFR1) fused to the Fc portion of human IgG(1) through a polyglycine linker. It binds to both human VEGF (hVEGF) and human PlGF (hPlGF) and to mouse VEGF (mVEGF) and mouse PlGF (mPlGF). In vitro, sFLT01 inhibited the proliferation of human umbilical vein endothelial cells and pericytes stimulated by either hVEGF or hPlGF. In vivo, sFLT01 had robust and significant antitumor activity in numerous preclinical subcutaneous tumor models including H460 non-small cell lung carcinoma, HT29 colon carcinoma, Karpas 299 lymphoma, MOLM-13 AML (acute myeloid leukemia), 786-O, and RENCA renal cell carcinoma (RCC). sFLT01 also increased median survival in the orthotopic RENCA RCC model. sFLT01 had strong antiangiogenic activity and altered intratumoral microvessel density, blood vessel lumen size and perimeter, and vascular and vessel areas in RCC models. sFLT01 treatment resulted in fewer endothelial cells and pericytes within the tumor microenvironment. sFLT01 in combination with cyclophosphamide resulted in greater inhibition of tumor growth than either agent used alone as a monotherapy in the A673 Ewing's sarcoma model. Gene expression profiling indicated that the molecular changes in the A673 sarcoma tumors are similar to changes observed under hypoxic conditions. sFLT01 is an innovative fusion protein that possessed robust antitumor and antiangiogenic activities in preclinical cancer models. It is a dual targeting agent that neutralizes both VEGF and PlGF and, therefore, has potential as a next generation antiangiogenic therapeutic for oncology.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/blood supply , Neoplasms/drug therapy , Pregnancy Proteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Profiling , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Placenta Growth Factor , Tumor Microenvironment/drug effects
12.
Tissue Eng Part C Methods ; 16(6): 1367-75, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20345228

ABSTRACT

Methods for the lineage identification of cell or tissue-engineered therapeutics must provide a high degree of performance to confidently distinguish the intended cell type from other lineages that could be present in the finished product. For many applications, these methods also require rapid, high-throughput capability. In this work, methods for the identification of autologous cultured chondrocytes for implantation were investigated. A histological analysis confirmed that fibrous tissue occasionally present in biopsies procured for autologous chondrocyte implantation production comprised synovium. Chondrocyte and synovial cell cultures were then examined using a full transcriptome microarray analysis, which revealed cartilage link protein and microfibril-associated glycoprotein-2 (MAGP2) as the most differentially expressed transcripts between the culture types. Performance characteristics of gene expression assays formed by the analysis of cartilage link protein with normalization to either standard reference genes or to MAGP2 were evaluated. The results demonstrate that the MAGP2-based assay provided superior performance for the purpose of cell culture identification compared to assays using standard reference genes. The selectivity against synovial and heterogeneous samples provided by the novel assay suggests it as an appropriate lineage identification method for cell cultures derived from cartilage.


Subject(s)
Cell Lineage/genetics , Chondrocytes/cytology , Chondrocytes/metabolism , Contractile Proteins/genetics , Extracellular Matrix Proteins/genetics , Synovial Membrane/cytology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Lineage/physiology , Cell Separation/methods , Cells, Cultured , Chondrocytes/physiology , Contractile Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression/physiology , Gene Expression Profiling , Humans , Microarray Analysis , RNA Splicing Factors , Swine , Synovial Membrane/metabolism , Tissue Engineering/methods
13.
Mamm Genome ; 21(1-2): 52-63, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20033694

ABSTRACT

C57BL/6 J (B6) and CAST/EiJ (CAST), the inbred strain derived from M. musculus castaneus, differ in nutrient intake behaviors, including dietary fat and carbohydrate consumption in a two-diet-choice paradigm. Significant quantitative trait loci (QTLs) for carbohydrate (Mnic1) and total energy intake (Kcal2) are present between these strains on chromosome (Chr) 17. Here we report the refinement of the Chr 17 QTL in a subcongenic strain of the B6.CAST-( D17Mit19-D17Mit91 ) congenic mice described previously. This new subcongenic strain possesses CAST Chr 17 donor alleles from 4.8 to 45.4 Mb on a B6 background. Similar to CAST, the subcongenic mice exhibit increased carbohydrate and total calorie intake per body weight, while fat intake remains equivalent. Unexpectedly, this CAST genomic segment also confers two new physical activity phenotypes: 22% higher spontaneous physical activity levels and significantly increased voluntary wheel-running activity compared with the parental B6 strain. Overall, these data suggest that gene(s) involved in carbohydrate preference and increased physical activity are contained within the proximal region of Chr 17. Interval-specific microarray analysis in hypothalamus and skeletal muscle revealed differentially expressed genes within the subcongenic region, including neuropeptide W (Npw); glyoxalase I (Glo1); cytochrome P450, family 4, subfamily f, polypeptide 1 (Cyp4f15); phospholipase A2, group VII (Pla2g7); and phosphodiesterase 9a (Pde9a). This subcongenic strain offers a unique model for dissecting the contributions and possible interactions among genes controlling food intake and physical activity, key components of energy balance.


Subject(s)
Chromosomes, Mammalian , Dietary Carbohydrates/administration & dosage , Energy Intake/genetics , Motor Activity/genetics , Animals , Body Composition , Calorimetry, Indirect , Crosses, Genetic , Eating/genetics , Gene Expression Profiling , Mice , Mice, Congenic , Mice, Inbred C57BL , Motor Activity/physiology , Phenotype , Protein Array Analysis , Quantitative Trait Loci , Up-Regulation
14.
Am J Physiol Regul Integr Comp Physiol ; 294(2): R362-71, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077508

ABSTRACT

We demonstrated previously that food intake traits map to a quantitative trait locus (QTL) on proximal chromosome 17, which encompasses Glp1r (glucagon-like peptide 1 receptor), encoding an important modulator of gastric emptying. We then confirmed this QTL in a B6.CAST-17 congenic strain that consumed 27% more carbohydrate and 17% more total calories, yet similar fat calories, per body weight compared with the recipient C57BL/6J. The congenic strain also consumed greater food volume. The current aims were to 1) identify genetic linkage for total food volume in F(2) mice, 2) perform gene expression profiling in stomach of B6.CAST-17 congenic mice using oligonucleotide arrays, 3) test for allelic imbalance in Glp1r expression, 4) evaluate gastric emptying rate in parental and congenic mice, and 5) investigate a possible effect of genetic variation in Glp1r on gastric emptying. A genome scan revealed a single QTL for total food volume (Tfv1) (log of the odds ratio = 7.6), which was confirmed in B6.CAST-17 congenic mice. Glp1r exhibited allelic imbalance in stomach, which correlated with accelerated gastric emptying in parental CAST and congenic B6.CAST-17 mice. Moreover, congenic mice displayed an impaired gastric emptying response to exendin-(9-39). These results suggest that genetic variation in Glp1r contributes to the strain differences in gastric emptying rate.


Subject(s)
Gastric Emptying/genetics , Genetic Variation , Glucagon-Like Peptide 1/metabolism , Receptors, Glucagon/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Eating/genetics , Gastric Emptying/drug effects , Gene Expression/physiology , Gene Expression Profiling , Genomics , Mice , Mice, Congenic , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Phenotype , Polymorphism, Single Nucleotide , Receptors, Glucagon/metabolism
15.
J Nutrigenet Nutrigenomics ; 1(4): 155-71, 2008.
Article in English | MEDLINE | ID: mdl-19776624

ABSTRACT

BACKGROUND/AIMS: The genetic basis for ingestive behaviors is virtually unknown. Quantitative trait loci (QTLs) for carbohydrate and energy intake map to mouse chromosome 17 and were previously confirmed by a congenic strain bearing CAST/Ei (CAST) donor segment on the C57BL/6J (B6) background. METHODS: We used microarray technology to facilitate gene identification. Gene expression was compared between the B6.CAST-17 (BC-17) congenic and B6 strains in two diets: (1) chow, and (2) carbohydrate/protein vs. fat/protein. RESULTS: Within the QTL and unique to macronutrient selection, Agpat1 (acylglycerol-3-phosphate O-acyltransferase 1) was differentially expressed in hypothalamus. Irrespective of diet, the gene with the highest fold difference in congenic mice was trefoil factor 3 (Tff3) in liver. Several genes involved in fat metabolism were decreased in carbohydrate-preferring congenic mice, while genes associated with carbohydrate metabolism were increased. In particular, the glyoxalase pathway was enhanced including Glo1, Glo2, and dLDH. Higher expression of Glo1 mRNA in BC-17 congenic mice corresponded to increased protein expression revealed by Western blot, and to higher GLO1 activity in blood. CONCLUSION: These genes represent new candidates for nutrient intake phenotypes. We propose that increased GLO1 in the BC-17 strain supports its need to protect against dietary oxidants resulting from high carbohydrate intake.


Subject(s)
Carbohydrate Metabolism/genetics , Chromosomes, Mammalian/genetics , Energy Intake/genetics , Gene Expression Profiling , Quantitative Trait Loci , Animals , Food , Food Preferences/physiology , Mice , Mice, Congenic , Mice, Inbred C57BL , Models, Biological , Oligonucleotide Array Sequence Analysis , Phenotype , Signal Transduction/genetics , Transcriptional Activation/physiology
16.
Am J Physiol Regul Integr Comp Physiol ; 292(1): R207-16, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16946080

ABSTRACT

Quantitative trait loci (QTL) for carbohydrate (Mnic1) and total energy (Kcal2) intake on proximal mouse chromosome 17 were identified previously from a C57BL/6J (B6) X CAST/Ei (CAST) intercross. Here we report that a new congenic strain developed in our laboratory has confirmed this complex locus by recapitulating the original linked phenotypes: B6.CAST-17 homozygous congenic mice consumed more carbohydrate (27%) and total energy (17%) compared with littermate wild-type mice. Positional gene candidates with relevance to carbohydrate metabolism, glyoxalase I (Glo1) and glucagon-like peptide-1 receptor (Glp1r), were evaluated. Glo1 expression was upregulated in liver and hypothalamus of congenic mice when compared with B6 mice. Analyses of Glp1r mRNA and protein expression revealed tissue-specific strain differences in pancreas (congenic>B6) and stomach (B6>congenic). These results suggest the possibility of separate mechanisms for enhanced insulin synthesis and gastric accommodation in the presence of high carbohydrate intake and larger food volume, respectively. Sequence analysis of Glp1r found a G insert at nt position 1349, which results in earlier termination of the open reading frame, thus revealing an error in the public sequence. Consequently, the predicted length of GLP-1R is 463 aa compared with 489 aa, as previously reported. Also, we found a polymorphism in Glp1r between parental strains that alters the amino acid sequence. Variation in Glp1r could influence nutrient intake in this model through changes in the regulatory or protein coding regions of the gene. These congenic mice offer a powerful tool for investigating gene interactions in the control of food intake.


Subject(s)
Chromosomes, Mammalian/genetics , Energy Intake/genetics , Lactoylglutathione Lyase/genetics , Quantitative Trait Loci , Receptors, Glucagon/genetics , Animals , Blotting, Western , Body Weight/physiology , DNA/genetics , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Dietary Carbohydrates , Dietary Fats , Dietary Proteins , Genetic Variation , Glucagon-Like Peptide-1 Receptor , Mice , Mice, Congenic , Mice, Inbred C57BL , Microsatellite Repeats/genetics , Phenotype , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
17.
Prostate ; 67(1): 83-106, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17013881

ABSTRACT

BACKGROUND: A major focus of prostate cancer research has been to identify genes that are deregulated during tumor progression, potentially providing diagnostic markers and therapeutic targets. METHODS: We have employed serial analysis of gene expression (SAGE) and microarray hybridization to identify alterations that occur during malignant transformation in the Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model. Many of these alterations were validated by real-time PCR (rtPCR). RESULTS: We identified several hundred mRNAs that were deregulated. Cluster analysis of microarray profiles with samples from various stages of the disease demonstrated that androgen-independent (AI) primary tumors are similar to metastases; 180 transcripts have expression patterns suggesting an involvement in the genesis of late-stage tumors, and our data support a role for phospholipase A2 group IIA in the acquisition of their highly aggressive characteristics. CONCLUSIONS: Our analyses identified well-characterized genes that were previously known to be involved in prostate cancer, validating our study, and also uncovered transcripts that had not previously been implicated in prostate cancer progression.


Subject(s)
Adenocarcinoma/genetics , Androgens/genetics , Disease Models, Animal , Gene Expression Profiling , Genes, Neoplasm/physiology , Genetic Engineering/methods , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Adenocarcinoma/metabolism , Androgens/metabolism , Animals , Gene Expression Regulation, Neoplastic/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Prostatic Neoplasms/metabolism , Species Specificity
19.
Genomics ; 84(3): 497-510, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15498457

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in the PKD1 or PKD2 gene, but cellular mechanisms of cystogenesis remain unclear. In an attempt to display the array of cyst-specific molecules and to elucidate the disease pathway, we have performed comprehensive high-throughput expression analysis of normal and ADPKD epithelia in a two-step fashion. First, we generated expression profiles of normal and cystic epithelia derived from kidney and liver using serial analysis of gene expression (SAGE). We found 472 and 499 differentially expressed genes with fivefold difference in liver and kidney libraries, respectively. These genes encode growth factors, transcription factors, proteases, apoptotic factors, molecules involved in cell-extracellular matrix interactions, and ion channels. As a second step, we constructed a custom cDNA microarray using a subset of the differentially regulated genes identified by SAGE and interrogated ADPKD patient samples. Subsequently, a set of differentially expressed genes was refined to 26 up-regulated and 48 down-regulated genes with ap value of <0.01. This study may provide valuable insights into the pathophysiology of ADPKD and suggest potential therapeutic targets.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation , Genes/genetics , Kidney/metabolism , Liver/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Analysis of Variance , DNA Primers , Epithelium/metabolism , Gene Library , Humans , Oligonucleotide Array Sequence Analysis/methods , Reverse Transcriptase Polymerase Chain Reaction
20.
Biochem Biophys Res Commun ; 319(2): 518-24, 2004 Jun 25.
Article in English | MEDLINE | ID: mdl-15178437

ABSTRACT

The agouti-related protein (AgRP) is a powerful appetite modulator expressed in the hypothalamus and the adrenal gland and regulated by leptin. Here we report the robust expression of AgRP in epididymal fat and its upregulation in this tissue by feeding rather than by fasting. This was observed in both the obesity-susceptible C57BL/6J and the obesity-resistant CAST/Ei mouse strains. Surprisingly, AgRP expression was higher in the hypothalamus and the adrenal gland in the leaner and obesity-resistant CAST/Ei strain. In vitro leptin treatment upregulated endogenous AgRP in mouse hypothalamus and adrenal cells, after an acute 6-h exposure, but it downregulated AgRP after a long-term 60-h exposure. AgRP, on the other hand, upregulated its own endogenous expression in the hypothalamus and the adrenal cells and also upregulated endogenous leptin in the adrenal cells. These results reveal a novel feedback loop and reciprocal transcriptional regulation between AgRP and leptin centrally and peripherally.


Subject(s)
Leptin/metabolism , Proteins/metabolism , Adrenal Glands/metabolism , Agouti-Related Protein , Animals , Base Sequence , DNA Primers , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...