Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Heliyon ; 9(6): e17429, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37416648

ABSTRACT

In new research growth long after glow material is a potential candidate due to its physical properties, chemical stability and wide application in modern solid-state lightning (LED), display devices, dosimetry and sensors. A cerium doped strontium aluminate phosphor (SrAl2O4:Ce3+) was synthesized by conventional solid-state reaction method. The crystal structure and morphology of phosphors, while doping rare earth metal and lithium metal ion was investigated by using X-ray diffraction, Raman spectroscopy and field emission scanning electron microscopy. Fourier transformed infrared spectrum results of the synthesized phosphor composition conforms the characteristic vibration bands of synthesized phosphor. Surface composition analysis of the prepared samples was examined using X-ray photoelectron spectroscopy. Photoluminescence emission band observed at ∼420 nm, ∼490 nm and ∼610 nm region under the excitation wavelength of 256 nm. Wight light emission was confirmed using the Commission Internationale de L'Eclairage (CIE) chromatic coordinate graph. The correlated colour temperature (CCT) value of 0.5% Ce3+ doped SAO of phosphors was calculated is in the range of 1543 K, which is indicated the synthesized phosphors performance as warm white light source. The obtained phosphor has a high dielectric constant and low loss tangent which is useful to optoelectronic devices.

2.
J Biol Chem ; 295(40): 13753-13768, 2020 10 02.
Article in English | MEDLINE | ID: mdl-32727846

ABSTRACT

The micropeptide adropin encoded by the clock-controlled energy homeostasis-associated gene is implicated in the regulation of glucose metabolism. However, its links to rhythms of nutrient intake, energy balance, and metabolic control remain poorly defined. Using surveys of Gene Expression Omnibus data sets, we confirm that fasting suppresses liver adropin expression in lean C57BL/6J (B6) mice. However, circadian rhythm data are inconsistent. In lean mice, caloric restriction (CR) induces bouts of compulsive binge feeding separated by prolonged fasting intervals, increasing NAD-dependent deacetylase sirtuin-1 signaling important for glucose and lipid metabolism regulation. CR up-regulates adropin expression and induces rhythms correlating with cellular stress-response pathways. Furthermore, adropin expression correlates positively with phosphoenolpyruvate carboxokinase-1 (Pck1) expression, suggesting a link with gluconeogenesis. Our previous data suggest that adropin suppresses gluconeogenesis in hepatocytes. Liver-specific adropin knockout (LAdrKO) mice exhibit increased glucose excursions following pyruvate injections, indicating increased gluconeogenesis. Gluconeogenesis is also increased in primary cultured hepatocytes derived from LAdrKO mice. Analysis of circulating insulin levels and liver expression of fasting-responsive cAMP-dependent protein kinase A (PKA) signaling pathways also suggests enhanced responses in LAdrKO mice during a glucagon tolerance test (250 µg/kg intraperitoneally). Fasting-associated changes in PKA signaling are attenuated in transgenic mice constitutively expressing adropin and in fasting mice treated acutely with adropin peptide. In summary, hepatic adropin expression is regulated by nutrient- and clock-dependent extrahepatic signals. CR induces pronounced postprandial peaks in hepatic adropin expression. Rhythms of hepatic adropin expression appear to link energy balance and cellular stress to the intracellular signal transduction pathways that drive the liver fasting response.


Subject(s)
Caloric Restriction , Fasting , Gene Expression Regulation , Hepatocytes/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Liver/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gluconeogenesis/genetics , Hepatocytes/cytology , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Liver/cytology , Mice , Mice, Knockout , Phosphoenolpyruvate Carboxykinase (GTP)/biosynthesis , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Signal Transduction/genetics
3.
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.

4.
Cancer Res ; 74(3): 908-20, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24285728

ABSTRACT

Myc oncoproteins induce genes driving aerobic glycolysis, including lactate dehydrogenase-A that generates lactate. Here, we report that Myc controls transcription of the lactate transporter SLC16A1/MCT1 and that elevated MCT1 levels are manifest in premalignant and neoplastic Eµ-Myc transgenic B cells and in human malignancies with MYC or MYCN involvement. Notably, disrupting MCT1 function leads to an accumulation of intracellular lactate that rapidly disables tumor cell growth and glycolysis, provoking marked alterations in glycolytic intermediates, reductions in glucose transport, and in levels of ATP, NADPH, and ultimately, glutathione (GSH). Reductions in GSH then lead to increases in hydrogen peroxide, mitochondrial damage, and ultimately, cell death. Finally, forcing glycolysis by metformin treatment augments this response and the efficacy of MCT1 inhibitors, suggesting an attractive combination therapy for MYC/MCT1-expressing malignancies.


Subject(s)
Glutathione/biosynthesis , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/genetics , Proto-Oncogene Proteins c-myc/metabolism , Symporters/genetics , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Glycolysis/genetics , Homeostasis/drug effects , Humans , Hydrogen Peroxide/pharmacology , Metformin/pharmacology , Mice , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Symporters/antagonists & inhibitors , Symporters/metabolism , Transcription, Genetic
5.
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
6.
ACS Chem Biol ; 6(3): 218-22, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21090593

ABSTRACT

Several nuclear receptors (NRs) are still character-ized as orphan receptors because ligands have not yet been identified for these proteins. The retinoic acid receptor-related receptors (RORs) have no well-defined physiological ligands. Here, we describe the identification of a selective RORα synthetic ligand, SR3335 (ML-176). SR3335 directly binds to RORα, but not other RORs, and functions as a selective partial inverse agonist of RORα in cell-based assays. Furthermore, SR3335 suppresses the expression of endogenous RORα target genes in HepG2 involved in hepatic gluconeogenesis including glucose-6-phosphatase and phosphoenolpyruvate carboxykinase. Pharmacokinetic studies indicate that SR3335 displays reasonable exposure following an ip injection into mice. We assess the ability of SR3335 to suppress gluconeogenesis in vivo using a diet-induced obesity (DIO) mouse model where the mice where treated with 15 mg/kg b.i.d., ip for 6 days followed by a pyruvate tolerance test. SR3335-treated mice displayed lower plasma glucose levels following the pyruvate challenge consistent with suppression of gluconeogenesis. Thus, we have identified the first selective synthetic RORα inverse agonist, and this compound can be utilized as a chemical tool to probe the function of this receptor both in vitro and in vivo. Additionally, our data suggests that RORα inverse agonists may hold utility for suppression of elevated hepatic glucose production in type 2 diabetics.


Subject(s)
Drug Inverse Agonism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Sulfonamides/pharmacology , Thiophenes/pharmacology , Cells, Cultured , Humans , Ligands , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Thiophenes/chemical synthesis , Thiophenes/chemistry
7.
FASEB J ; 24(3): 862-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19837866

ABSTRACT

Melanocortin-3 receptors (Mc3rs) in the central nervous system are involved in expression of anticipatory rhythms and synchronizing clocks maintaining circadian rhythms during restricted feeding (RF) [mice housed under a 12-h light-dark cycle with lights on between zeitgeber time (ZT) 0 to ZT12 fed 60% of normal calories between ZT7 and ZT11]. Because the systems governing circadian rhythms are important for adaptation to RF, we investigated whether Mc3rs are required for metabolic adaption to RF. Mc3r(-/-) mice subjected to RF exhibited normal weight loss; however, they developed hyperinsulinemia, glucose intolerance, increased expression of lipogenic genes, and increased ketogenesis relative to controls. Rhythmic expression of transcription factors regulating liver clock activity and energy metabolism (Bmal1, Rev-erbalpha, Pgc1, Foxo1, Hnf4alpha, and Pck1) was severely compromised in Mc3r(-/-) mice during RF. Inhibition of neural melanocortin receptors by agouti-related peptide also attenuated rhythmicity in the hepatic expression of these genes during RF. Collectively, these data suggest that neural Mc3rs are important for adapting metabolism and maintaining rhythms of liver metabolism during periods when feeding is restricted to the light cycle.-Sutton, G. M., Begriche, K., Kumar, K. G., Gimble, J. M., Perez-Tilve, D., Nogueiras, R., McMillan, R. P., Hulver, M. W., Tschöp, M. H., Butler, A. A. Central nervous system melanocortin-3 receptors are required for synchronizing metabolism during entrainment to restricted feeding during the light cycle.


Subject(s)
Caloric Restriction , Central Nervous System/metabolism , Photoperiod , Receptor, Melanocortin, Type 3/physiology , Agouti-Related Protein/pharmacology , Animals , Central Nervous System/radiation effects , Electrophoresis, Polyacrylamide Gel , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acids/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Glucose Intolerance/genetics , Glucose Tolerance Test , Hyperinsulinism/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Mutant Strains , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/genetics
8.
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
9.
Blood ; 114(18): 3803-12, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19721009

ABSTRACT

As the expanding obese population grows older, their successful immunologic aging will be critical to enhancing the health span. Obesity increases risk of infections and cancer, suggesting adverse effects on immune surveillance. Here, we report that obesity compromises the mechanisms regulating T-cell generation by inducing premature thymic involution. Diet-induced obesity reduced thymocyte counts and significantly increased apoptosis of developing T-cell populations. Obesity accelerated the age-related reduction of T-cell receptor (TCR) excision circle bearing peripheral lymphocytes, an index of recently generated T cells from thymus. Consistent with reduced thymopoiesis, dietary obesity led to reduction in peripheral naive T cells with increased frequency of effector-memory cells. Defects in thymopoiesis in obese mice were related with decrease in the lymphoid-primed multipotent progenitor (Lin-Sca1+Kit+ Flt3+) as well as common lymphoid progenitor (Lin-Sca1+CD117(lo)CD127+) pools. The TCR spectratyping analysis showed that obesity compromised V-beta TCR repertoire diversity. Furthermore, the obesity induced by melanocortin 4 receptor deficiency also constricted the T-cell repertoire diversity, recapitulating the thymic defects observed with diet-induced obesity. In middle-aged humans, progressive adiposity with or without type 2 diabetes also compromised thymic output. Collectively, these findings establish that obesity constricts T-cell diversity by accelerating age-related thymic involution.


Subject(s)
Aging/immunology , Multipotent Stem Cells/immunology , Obesity/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Adiposity/genetics , Adiposity/immunology , Aging/pathology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/pathology , Humans , Immunologic Memory/immunology , Immunologic Surveillance/immunology , Infections/genetics , Infections/immunology , Infections/pathology , Mice , Mice, Knockout , Middle Aged , Multipotent Stem Cells/pathology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/immunology , Receptors, Antigen, T-Cell/immunology , Risk Factors , T-Lymphocytes/pathology , Thymus Gland/pathology
10.
Peptides ; 30(10): 1892-900, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19646498

ABSTRACT

Melanocortin receptor agonists act in the brain to regulate food intake and body weight and, independently of these actions, affect insulin sensitivity. These experiments investigated the function of novel non-selective melanocortin receptor agonists (BIM-22493, BIM-22511) that cross the blood-brain barrier when administered peripherally. Treatment of diet induced obese C57BL/6J (B6) mice with melanocortin agonists administered peripherally improved obesity, hyperinsulinemia (approximately 50%) and fatty liver disease. Specificity of function was determined using B6 melanocortin-3 and melanocortin-4 receptor knockout mice (MC3RKO, MC4RKO). Chow fed MC4RKO but not MC3RKO used for these tests exhibited obesity, hyperinsulinemia and severe hepatosteatosis associated with increased expression of insulin-stimulated genes involved in lipogenesis. Reduced food intake associated with acute BIM-22493 treatment, and weight loss associated with 14 days of treatment with BIM-22511, required functional MC4R but not MC3R. However, while 14 days of treatment with BIM-22511 did not affect body weight and even increased cumulative food intake in MC4RKO, a significant reduction (approximately 50%) in fasting insulin was still observed. Despite lowering insulin, chronic treatment with BIM-22511 did not improve hepatosteatosis in MC4RKO, and did not affect hepatic lipogenic gene expression. Together, these results demonstrate that peripherally administered melanocortin receptor agonists regulate body weight, liver metabolism and glucose homeostasis through independent pathways. MC4R are necessary for melanocortin agonist-induced weight loss and improvements in liver metabolism, but are not required for improvements in hyperinsulinemia. Agonists with activity at MC4R improve glucose homeostasis at least partially by causing weight loss, however other melanocortin receptors may have potential for treating aberrations in glucose homeostasis associated with obesity.


Subject(s)
Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/genetics , alpha-MSH/analogs & derivatives , Animals , Diet , Eating , Energy Metabolism , Female , Glucose/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/metabolism , Weight Loss , alpha-MSH/pharmacology
11.
Cell Metab ; 8(6): 468-81, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19041763

ABSTRACT

Obesity and nutrient homeostasis are linked by mechanisms that are not fully elucidated. Here we describe a secreted protein, adropin, encoded by a gene, Energy Homeostasis Associated (Enho), expressed in liver and brain. Liver Enho expression is regulated by nutrition: lean C57BL/6J mice fed high-fat diet (HFD) exhibited a rapid increase, while fasting reduced expression compared to controls. However, liver Enho expression declines with diet-induced obesity (DIO) associated with 3 months of HFD or with genetically induced obesity, suggesting an association with metabolic disorders in the obese state. In DIO mice, transgenic overexpression or systemic adropin treatment attenuated hepatosteatosis and insulin resistance independently of effects on adiposity or food intake. Adropin regulated expression of hepatic lipogenic genes and adipose tissue peroxisome proliferator-activated receptor gamma, a major regulator of lipogenesis. Adropin may therefore be a factor governing glucose and lipid homeostasis, which protects against hepatosteatosis and hyperinsulinemia associated with obesity.


Subject(s)
Blood Proteins/physiology , Energy Metabolism , Lipid Metabolism , Proteins/physiology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Amino Acid Sequence , Animals , Base Sequence , Benzoates/chemistry , Benzoates/metabolism , Benzylamines/chemistry , Benzylamines/metabolism , Blood Proteins/genetics , Blood Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins/agonists , DNA-Binding Proteins/metabolism , Fasting , Fatty Liver/metabolism , Female , Humans , Intercellular Signaling Peptides and Proteins , Leptin/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Obesity/genetics , Obesity/metabolism , Orphan Nuclear Receptors , Peptides , Proteins/genetics , Proteins/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism
12.
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
13.
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
14.
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
15.
Immunogenetics ; 55(12): 811-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14770272

ABSTRACT

The complement system helps in the lysis of invading pathogens and modulates the inflammatory as well as the humoral and cellular immune responses. C5 mediates many potent inflammatory and cytolytic events after proteolytic activation by complement convertase enzymes. Hence, to investigate the role of pig C5 ( pC5) as a candidate gene for disease resistance in pigs, the complete cDNA of pC5 was sequenced, screened for single nucleotide polymorphisms (SNPs), and an association analysis with various immunological parameters measured in F2 animals of a pig resource population based on a cross of Duroc and Berlin miniature pigs (DUMI) was carried out. In total, 5,422 bp of pC5 cDNA was sequenced, which codes for the 1,677-amino-acid precursor of C5. Four polymorphic sites were detected, one of which was segregating in the DUMI population in three genotypic patterns: AA, AC and CC. Classical (CH50) and alternative (AH50) complement activities, C3c levels, haptoglobin (HP) acute phase protein levels, and antibody titers against Mycoplasma (Mk) and Aujesky (ADV) vaccines were measured in the resource population. Association analysis between C5 and the immunological parameters was carried out using repeated measures mixed and general linear model analysis. The homozygote AA was found to be significantly different from the other two genotypes with respect to AH50 and CH50, whereas genotype CC was found to be significantly different from the other genotypes for C3c and HP levels. No significant difference could be seen between genotypes for antibody titers against vaccinations. Association of C5 with complement activity traits and acute phase proteins promotes pC5 as a candidate gene for innate disease resistance.


Subject(s)
Complement C5/genetics , Quantitative Trait Loci/immunology , Swine/immunology , Acute-Phase Proteins/metabolism , Animals , Base Sequence , Chromosome Segregation , Cloning, Molecular , Complement C3c/immunology , Complement Hemolytic Activity Assay , Complement Pathway, Alternative/immunology , Complement Pathway, Classical/immunology , DNA, Complementary/analysis , Female , Genotype , Haptoglobins/metabolism , Male , Molecular Sequence Data , Phenotype , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...