Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 651: 62-69, 2023 04 09.
Article in English | MEDLINE | ID: mdl-36791500

ABSTRACT

Obesity is a major risk factor for type 2 diabetes, coronary heart disease, and strok. These diseases are associated with profound alterations in gene expression in metabolic tissues. Epigenetic-mediated regulation of gene expression is one mechanism through which environmental factors, such as diet, modify gene expression and disease predisposition. However, epigenetic control of gene expression in obesity and insulin resistance is not fully characterized. We discovered that liver-specific stearoyl-CoA desaturase-1 (Scd1) knockout mice (LKO) fed a high-carbohydrate low-fat diet exhibit dramatic changes in hepatic gene expression and metabolites of the folate cycle and one-carbon metabolism respectively for the synthesis of S-adenosylmethionine (SAM). LKO mice show an increased ratio of S-adenosylmethionine to S-adenosylhomocysteine, a marker for increased cellular methylation capacity. Furthermore, expression of DNA and histone methyltransferase genes is up-regulated while the mRNA and protein levels of the non-DNA methyltransferases including phosphatidylethanolamine methyltransferase (PEMT), Betaine homocysteine methyltransferase (Bhmt), and the SAM-utilizing enzymes such as glycine-N-methyltransferase (Gnmt) and guanidinoacetate methyltransferase (Gamt) are generally down-regulated. Feeding LKO mice a high carbohydrate diet supplemented with triolein, but not tristearin, and increased endogenous hepatic synthesis of oleate but not palmitoleate in Scd1 global knockout mice normalized one carbon gene expression and metabolite levels. Additionally, changes in one carbon gene expression are independent of the PGC-1α-mediated ER stress response previously reported in the LKO mice. Together, these results highlight the important role of oleate in maintaining one-carbon cycle homeostasis and point to observed changes in one-carbon metabolism as a novel mediator of the Scd1 deficiency-induced liver phenotype.


Subject(s)
Diabetes Mellitus, Type 2 , Oleic Acid , Mice , Animals , Oleic Acid/metabolism , S-Adenosylmethionine/metabolism , Diabetes Mellitus, Type 2/metabolism , Liver/metabolism , Carbohydrates , Mice, Knockout , Obesity/metabolism , Carbon/metabolism , Phosphatidylethanolamine N-Methyltransferase/metabolism
2.
Science ; 375(6579): 460-463, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35084962

ABSTRACT

Hibernation is a mammalian strategy that uses metabolic plasticity to reduce energy demands and enable long-term fasting. Fasting mitigates winter food scarcity but eliminates dietary nitrogen, jeopardizing body protein balance. Here, we reveal gut microbiome-mediated urea nitrogen recycling in hibernating thirteen-lined ground squirrels (Ictidomys tridecemlineatus). Ureolytic gut microbes incorporate urea nitrogen into metabolites that are absorbed by the host, with the nitrogen reincorporated into the squirrel's protein pool. Urea nitrogen recycling is greatest after prolonged fasting in late winter, when urea transporter abundance in gut tissue and urease gene abundance in the microbiome are highest. These results reveal a functional role for the gut microbiome during hibernation and suggest mechanisms by which urea nitrogen recycling may contribute to protein balance in other monogastric animals.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Hibernation , Nitrogen/metabolism , Sciuridae/metabolism , Sciuridae/microbiology , Urea/metabolism , Animals , Cecum/metabolism , Cecum/microbiology , Fasting , Female , Liver/metabolism , Male , Protein Biosynthesis , Seasons , Symbiosis , Urea/blood , Urease/genetics , Urease/metabolism
3.
Int J Mol Sci ; 20(16)2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31434215

ABSTRACT

3-Iodothyronamine (T1AM) and its synthetic analog SG-2 are rapidly emerging as promising drivers of cellular metabolic reprogramming. Our recent research indicates that in obese mice a sub-chronic low dose T1AM treatment increased lipolysis, associated with significant weight loss independent of food consumption. The specific cellular mechanism of T1AM's lipolytic effect and its site of action remains unknown. First, to study the mechanism used by T1AM to gain entry into cells, we synthesized a fluoro-labeled version of T1AM (FL-T1AM) by conjugating it to rhodamine (TRITC) and analyzed its cellular uptake and localization in 3T3-L1 mouse adipocytes. Cell imaging using confocal microscopy revealed a rapid intercellular uptake of FL-T1AM into mitochondria without localization to the lipid droplet or nucleus of mature adipocytes. Treatment of 3T3-L1 adipocytes with T1AM and SG-2 resulted in decreased lipid accumulation, the latter showing a significantly higher potency than T1AM (10 µM vs. 20 µM, respectively). We further examined the effects of T1AM and SG-2 on liver HepG2 cells. A significant decrease in lipid accumulation was observed in HepG2 cells treated with T1AM or SG-2, due to increased lipolytic activity. This was confirmed by accumulation of glycerol in the culture media and through activation of the AMPK/ACC signaling pathways.


Subject(s)
Thyronines/pharmacology , 3T3-L1 Cells , Animals , Cellular Reprogramming/drug effects , Glycerol/metabolism , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lipolysis/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism
4.
Am J Physiol Regul Integr Comp Physiol ; 316(6): R764-R775, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30969844

ABSTRACT

It is well established that hibernating mammals rely predominantly on lipid stores to fuel metabolism throughout the hibernation season. However, it is unclear if other endogenous fuels contribute to the rapid, ~400-fold increase in metabolic rate during the early phase of arousal from torpor. To investigate this issue, we used cavity ring-down spectroscopy, a technique that provides a real-time indication of fuel use by measuring the ratio of 13C to 12C in the exhaled CO2 of arousing 13-lined ground squirrels (Ictidomys tridecemlineatus). We used infrared thermography to simultaneously measure ventilation and surface temperature change in various body regions, and we interpreted these data in light of changing plasma metabolite abundances at multiple stages of arousal from torpor. We found that hibernating squirrels use a combination of lipids and, likely, carbohydrates to fuel the initial ~60 min of arousal before switching to predominantly lipid oxidation. This fuel switch coincided with times of maximal rates of ventilation and rewarming of different body surface regions, including brown adipose tissue. Infrared thermography revealed zonal rewarming, whereby the brown adipose tissue region was the first to warm, followed by the thoracic and head regions and, finally, the posterior half of the body. Consistent with the results from cavity ring-down spectroscopy, plasma metabolite dynamics during early arousal suggested a large reliance on fatty acids, with a contribution from carbohydrates and glycerol. Because of their high oxidative flux rates and efficient O2 use, carbohydrates might be an advantageous metabolic fuel during the early phase of arousal, when metabolic demands are high but ventilation rates and, thus, O2 supply are relatively low.


Subject(s)
Arousal , Energy Metabolism , Hibernation , Pulmonary Ventilation , Sciuridae/physiology , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Biomarkers/blood , Carbohydrate Metabolism , Female , Lipid Metabolism , Male , Oxidation-Reduction , Sciuridae/metabolism , Time Factors
5.
Sci Rep ; 9(1): 371, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30674908

ABSTRACT

We assessed adverse effects of external sublethal exposure of Deepwater Horizon, Mississippi Canyon 252 oil on plasma and liver metabolome profiles of the double-crested cormorant (Phalacrocorax auritus), a large (1.5 to 3.0 kg) diving waterbird common in the Gulf of Mexico. Metabolomics analysis of avian plasma showed significant negative effects on avian metabolic profiles, in some cases after only two external exposures (26 g cumulative) to oil. We observed significant (p < 0.05) changes in intermediate metabolites of energy metabolism and fatty acid and amino acid metabolic pathways in cormorants after repeated exposure to oil. Exposure to oil increased several metabolites (glycine, betaine, serine and methionine) that are essential to the one-carbon metabolism pathway. Lipid metabolism was affected, causing an increase in production of ketone bodies, suggesting lipids were used as an alternative energy source for energy production in oil exposed birds. In addition, metabolites associated with hepatic bile acid metabolism were affected by oil exposure which was correlated with changes observed in bile acids in exposed birds. These changes at the most basic level of phenotypic expression caused by sublethal exposure to oil can have effects that would be detrimental to reproduction, migration, and survival in avian species.


Subject(s)
Birds/metabolism , Environmental Exposure/adverse effects , Metabolome/drug effects , Petroleum Pollution/adverse effects , Water Pollutants, Chemical/adverse effects , Animals , Energy Metabolism , Gulf of Mexico , Liver/metabolism , Metabolomics/methods
6.
Molecules ; 23(10)2018 Oct 03.
Article in English | MEDLINE | ID: mdl-30282936

ABSTRACT

Taste signaling is a complex process that is linked to obesity and its associated metabolic syndromes. The sweet taste is mediated through a heterodimeric G protein coupled receptor (GPCR) in a species-specific manner and at multi-tissue specific levels. The sweet receptor recognizes a large number of ligands with structural and functional diversities to modulate different amplitudes of downstream signaling pathway(s). The human sweet-taste receptor has been extremely difficult to study by biophysical methods due to the difficulty in producing large homogeneous quantities of the taste-receptor protein and the lack of reliable in vitro assays to precisely measure productive ligand binding modes that lead to activation of the receptor protein. We report here a multimodal high throughput assay to monitor ligand binding, receptor stability and conformational changes to model the molecular ligand-receptor interactions. We applied saturation transfer difference nuclear magnetic resonance spectroscopy (STD-NMR) complemented by differential scanning calorimetry (DSC), circular dichroism (CD) spectroscopy, and intrinsic fluorescence spectroscopy (IF) to characterize binding interactions. Our method using complementary NMR and biophysical analysis is advantageous to study the mechanism of ligand binding and signaling processes in other GPCRs.


Subject(s)
Receptors, G-Protein-Coupled/genetics , Sweetening Agents/chemistry , Taste/genetics , Animals , Humans , Ligands , Mice , Protein Binding , Protein Conformation/drug effects , Protein Domains , Receptors, G-Protein-Coupled/chemistry , Sweetening Agents/administration & dosage , Taste/drug effects
7.
Int J Mol Sci ; 19(5)2018 May 22.
Article in English | MEDLINE | ID: mdl-29786646

ABSTRACT

Obesity is a complex disease associated with environmental and genetic factors. 3-Iodothyronamine (T1AM) has revealed great potential as an effective weight loss drug. We used metabolomics and associated transcriptional gene and protein expression analysis to investigate the tissue specific metabolic reprogramming effects of subchronic T1AM treatment at two pharmacological daily doses (10 and 25 mg/kg) on targeted metabolic pathways. Multi-analytical results indicated that T1AM at 25 mg/kg can act as a novel master regulator of both glucose and lipid metabolism in mice through sirtuin-mediated pathways. In liver, we observed an increased gene and protein expression of Sirt6 (a master gene regulator of glucose) and Gck (glucose kinase) and a decreased expression of Sirt4 (a negative regulator of fatty acids oxidation (FAO)), whereas in white adipose tissue only Sirt6 was increased. Metabolomics analysis supported physiological changes at both doses with most increases in FAO, glycolysis indicators and the mitochondrial substrate, at the highest dose of T1AM. Together our results suggest that T1AM acts through sirtuin-mediated pathways to metabolically reprogram fatty acid and glucose metabolism possibly through small molecules signaling. Our novel mechanistic findings indicate that T1AM has a great potential as a drug for the treatment of obesity and possibly diabetes.


Subject(s)
Anti-Obesity Agents/pharmacology , Mitochondrial Proteins/genetics , Obesity/metabolism , Sirtuins/genetics , Thyronines/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Anti-Obesity Agents/therapeutic use , Fatty Acids/metabolism , Female , Germinal Center Kinases , Glucose/metabolism , Glycolysis , Liver/drug effects , Liver/metabolism , Mice , Mitochondrial Proteins/metabolism , Obesity/drug therapy , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sirtuins/metabolism , Thyronines/therapeutic use
8.
Nutr Res ; 45: 52-62, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29037332

ABSTRACT

Polyunsaturated fatty acid (PUFA)-rich diets are thought to provide beneficial effects toward metabolic health in part through their bioactive properties. We hypothesized that increasing PUFA intake in mice would increase peroxisome proliferator activated receptor delta (PPARδ) expression and activity, and we sought to examine the effect of different PUFA-enriched oils on muscle PPARδ expression. One of the oils we tested was cottonseed oil (CSO) which is primarily linoleic acid (53%) and palmitic acid (24%). Mice fed a CSO-enriched diet (50% energy from fat) displayed no change in muscle PPARδ expression; however, in the liver, it was consistently elevated along with its transcriptional coactivator Pgc-1. Male mice were fed chow or CSO-, saturated fat (SFA)-, or linoleic acid (18:2)-enriched diets that were matched for macronutrient content for 4 weeks. There were no differences in food intake, body weight, fasting glucose, glucose tolerance, or energy expenditure between chow- and CSO-fed mice, whereas SFA-fed mice had increased fat mass and 18:2-fed mice were less glucose tolerant. Metabolomic analyses revealed that the livers of CSO-fed mice closely matched those of chow-fed but significantly differed from SFA- and 18:2-enriched groups. Fatty acid composition of the diets and livers revealed an impairment in desaturase activity and the presence of dihydrosterculic acid (DHSA) in the CSO-fed mice. The effect of DHSA on PPARδ and stearoyl-CoA desaturase-1 expression mimicked that of the CSO-fed mice. Taken together, these data suggest that DHSA from CSO may be an effective means to increase PPARδ expression with concomitant suppression of liver stearoyl-CoA desaturase-1 activity.


Subject(s)
Cottonseed Oil/chemistry , Diet, High-Fat , Fatty Acids/pharmacology , Liver/metabolism , PPAR delta/analysis , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Energy Metabolism , Fatty Acids/analysis , Lipid Metabolism/drug effects , Liver/chemistry , Male , Metabolomics , Mice , Mice, Inbred C57BL , Muscle, Skeletal/chemistry
9.
Annu Rev Nutr ; 37: 477-500, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28715992

ABSTRACT

Animals that undergo seasonal cycles of feeding and fasting have adaptations that maintain integrity of organ systems when dietary nutrients are lacking. Food deprivation also challenges the gut microbiota, which relies heavily on host diet for metabolic substrates and the gastrointestinal tract, which is influenced by enteral nutrients and microbial activity. Winter fasting in hibernators shifts the microbiota to favor taxa with the capacity to degrade and utilize host-derived substrates and disfavor taxa that prefer complex plant polysaccharides. Microbiome alterations may contribute to hibernation-induced changes in the intestinal immune system, epithelial barrier function, and other host features that are affected by microbial short-chain fatty acids and other metabolites. Understanding mechanisms by which the hibernator host and its gut symbionts adapt to the altered nutritional landscape during winter fasting may provide insights into protective mechanisms that are compromised when nonhibernating species, such as humans, undergo long periods of enteral nutrient deprivation.


Subject(s)
Food , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/physiology , Hibernation/physiology , Periodicity , Symbiosis , Animals , Diet , Gastrointestinal Tract/metabolism
10.
FEBS Open Bio ; 7(4): 587-601, 2017 04.
Article in English | MEDLINE | ID: mdl-28396842

ABSTRACT

3-Iodothyronamine (T1AM) is a structural analog of thyroid hormone that has been demonstrated to have potent affects on numerous physiological systems. Most studies on T1AM have explored its effects in healthy functional systems; while its potential therapeutic uses and safety, and efficacy in pathological conditions are largely unknown. We sought to evaluate the effects of T1AM and its structural analog SG-2 on cancer cell growth and viability. We analyzed the cytotoxicity of these analogs on MCF7 breast cancer cells, HepG2 hepatocellular cancer cells as well as normal control cells using primary human foreskin fibroblasts and mouse preadipocytes control cells. The cytotoxicity of T1AM and SG-2 was determined by cell growth curves, and validated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide cell viability assays. Cellular uptake analysis was conducted using confocal microscopy. Real-time (RT)-PCR was conducted to identify gene pathways affected by SG-2 in cancer cells. The IC 50 of T1AM was approximately double the concentration of its analog SG-2 in cancer cells. Cytotoxicity studies on normal cells revealed that IC 50 concentrations of SG-2 in cancer cells had no significant impact on cell viability in these cell types. Cell-imaging experiments demonstrated rapid uptake and localization to the mitochondrial membrane. T1AM and SG-2 are able to reduce cancer cell growth and viability. These findings support the potential for use of these compounds and related analogs for their antiproliferation properties in cancer cells.

11.
Physiol Rep ; 5(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-28082426

ABSTRACT

Complex diseases such as polycystic ovary syndrome (PCOS) are associated with intricate pathophysiological, hormonal, and metabolic feedbacks that make their early diagnosis challenging, thus increasing the prevalence risks for obesity, cardiovascular, and fatty liver diseases. To explore the crosstalk between endocrine and lipid metabolic pathways, we administered 3-iodothyronamine (T1AM), a natural analog of thyroid hormone, in a mouse model of PCOS and analyzed plasma and tissue extracts using multidisciplinary omics and biochemical approaches. T1AM administration induces a profound tissue-specific antilipogenic effect in liver and muscle by lowering gene expression of key regulators of lipid metabolism, PTP1B and PLIN2, significantly increasing metabolites (glucogenic, amino acids, carnitine, and citrate) levels, while enhancing protection against oxidative stress. In contrast, T1AM has an opposing effect on the regulation of estrogenic pathways in the ovary by upregulating STAR, CYP11A1, and CYP17A1. Biochemical measurements provide further evidence of significant reduction in liver cholesterol and triglycerides in post-T1AM treatment. Our results shed light onto tissue-specific metabolic vs. hormonal pathway interactions, thus illuminating the intricacies within the pathophysiology of PCOS This study opens up new avenues to design drugs for targeted therapeutics to improve quality of life in complex metabolic diseases.


Subject(s)
Gene Expression/drug effects , Lipid Metabolism/genetics , Metabolic Networks and Pathways/drug effects , Polycystic Ovary Syndrome/drug therapy , Thyronines/administration & dosage , Animals , Cholesterol/metabolism , Female , Gene Expression/genetics , Liver/metabolism , Magnetic Resonance Spectroscopy , Metabolic Networks and Pathways/genetics , Metabolomics/methods , Mice , Muscles/metabolism , Obesity/metabolism , Ovary/metabolism , Oxidative Stress/drug effects , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Quality of Life , Thyronines/metabolism , Thyronines/pharmacology , Triglycerides/metabolism
12.
Protein Sci ; 25(3): 711-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26701738

ABSTRACT

Brazzein (Brz) is a small (54 amino acid residue) sweet tasting protein with physical and taste properties superior to other non-carbohydrate sweeteners. In an investigation of sequence-dependent functional properties of the protein, we used NMR spectroscopy to determine the three-dimensional structures and dynamic properties of two Brz variants: one with a single-site substitution (D40K), which is three-fold sweeter than wild-type Brz, and one with a two-residue insertion between residues 18 and 19 (ins18 RI19 ), which is devoid of sweetness. Although the three-dimensional folds of the two variants were very similar to wild-type Brz, they exhibited local conformational and dynamic differences. The D40K substitution abolished the strong inter-stand H-bond between the side chains of residues Gln46 and Asp40 present in wild-type Brz and increased the flexibility of the protein especially at the mutation site. This increased flexibility presumably allows this site to interact more strongly with the G-protein coupled human sweet receptor. On the other hand, the Arg-Ile insertion within Loop9-19 leads to distortion of this loop and stiffening of the adjacent site whose flexibility appears to be required for productive interaction with the sweet receptor.


Subject(s)
Magnoliopsida/metabolism , Plant Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Sweetening Agents/metabolism , Humans , Magnoliopsida/chemistry , Magnoliopsida/genetics , Models, Molecular , Mutation , Nuclear Magnetic Resonance, Biomolecular , Plant Proteins/chemistry , Plant Proteins/genetics , Protein Subunits/metabolism , Sweetening Agents/chemistry , Taste
13.
J Proteome Res ; 14(8): 3284-91, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26076986

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with metabolic and endocrine disorders in women of reproductive age. The etiology of PCOS is still unknown. Mice prenatally treated with glucocorticoids exhibit metabolic disturbances that are similar to those seen in women with PCOS. We used an untargeted nuclear magnetic resonance (NMR)-based metabolomics approach to understand the metabolic changes occurring in the plasma and kidney over time in female glucocorticoid-treated (GC-treated) mice. There are significant changes in plasma amino acid levels (valine, tyrosine, and proline) and their intermediates (2-hydroxybutyrate, 4-aminobutyrate, and taurine), whereas in kidneys, the TCA cycle metabolism (citrate, fumarate, and succinate) and the pentose phosphate (PP) pathway products (inosine and uracil) are significantly altered (p < 0.05) from 8 to 16 weeks of age. Levels of NADH, NAD(+), NAD(+)/NADH, and NADH redox in kidneys indicate increased mitochondrial oxidative stress from 8 to 16 weeks in GC-treated mice. These results indicate that altered metabolic substrates in the plasma and kidneys of treated mice are associated with altered amino acid metabolism, increased cytoplasmic PP, and increased mitochondrial activity, leading to a more oxidized state. This study identifies biomarkers associated with metabolic dysfunction in kidney mitochondria of a prenatal gluococorticoid-treated mouse model of PCOS that may be used as early predictive biomarkers of oxidative stress in the PCOS metabolic disorder in women.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Metabolic Diseases/metabolism , Metabolomics/methods , Mitochondria/metabolism , Oxidative Stress , Polycystic Ovary Syndrome/metabolism , Amino Acids/blood , Amino Acids/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Citric Acid Cycle , Disease Models, Animal , Female , Glucocorticoids , Humans , Hydroxybutyrates/blood , Hydroxybutyrates/metabolism , Kidney/metabolism , Kidney/pathology , Metabolic Diseases/blood , Metabolic Diseases/chemically induced , Metabolome , Mice , NAD/metabolism , Pentose Phosphate Pathway , Polycystic Ovary Syndrome/blood
14.
J Biomol NMR ; 62(4): 481-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25900069

ABSTRACT

The computationally demanding nature of automated NMR structure determination necessitates a delicate balancing of factors that include the time complexity of data collection, the computational complexity of chemical shift assignments, and selection of proper optimization steps. During the past two decades the computational and algorithmic aspects of several discrete steps of the process have been addressed. Although no single comprehensive solution has emerged, the incorporation of a validation protocol has gained recognition as a necessary step for a robust automated approach. The need for validation becomes even more pronounced in cases of proteins with higher structural complexity, where potentially larger errors generated at each step can propagate and accumulate in the process of structure calculation, thereby significantly degrading the efficacy of any software framework. This paper introduces a complete framework for protein structure determination with NMR--from data acquisition to the structure determination. The aim is twofold: to simplify the structure determination process for non-NMR experts whenever feasible, while maintaining flexibility by providing a set of modules that validate each step, and to enable the assessment of error propagations. This framework, called NMRFAM-SDF (NMRFAM-Structure Determination Framework), and its various components are available for download from the NMRFAM website (http://nmrfam.wisc.edu/software.htm).


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Proteins/chemistry , Software , Carbon-13 Magnetic Resonance Spectroscopy , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Conformation , Web Browser , Workflow
15.
Curr Metabolomics ; 2(4): 269-278, 2014.
Article in English | MEDLINE | ID: mdl-24765590

ABSTRACT

Polycystic ovary syndrome (PCOS), a common female endocrinopathy, is a complex metabolic syndrome of enhanced weight gain. The goal of this pilot study was to evaluate metabolic differences between normal (n=10) and PCOS (n=10) women via breath carbon isotope ratio, urinary nitrogen and nuclear magnetic resonance (NMR)-determined serum metabolites. Breath carbon stable isotopes measured by cavity ring down spectroscopy (CRDS) indicated diminished (p<0.030) lipid use as a metabolic substrate during overnight fasting in PCOS compared to normal women. Accompanying urinary analyses showed a trending correlation (p<0.057) between overnight total nitrogen and circulating testosterone in PCOS women, alone. Serum analyzed by NMR spectroscopy following overnight, fast and at 2 h following an oral glucose tolerance test showed that a transient elevation in blood glucose levels decreased circulating levels of lipid, glucose and amino acid metabolic intermediates (acetone, 2-oxocaporate, 2-aminobutyrate, pyruvate, formate, and sarcosine) in PCOS women, whereas the 2 h glucose challenge led to increases in the same intermediates in normal women. These pilot data suggest that PCOS-related inflexibility in fasting-related switching between lipid and carbohydrate/protein utilization for carbon metabolism may contribute to enhanced weight gain.

16.
Exp Biol Med (Maywood) ; 239(2): 151-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24302559

ABSTRACT

This study used an optical technique to measure the effects of treating low (10 mg/kg) and high (25 mg/kg) doses of 3-iodothyronamine (T1AM) on the metabolism in the kidney and heart of mice. The ratio of two intrinsic fluorophores in tissue, (NADH/FAD), called the NADH redox ratio (NADH RR), is a marker of the metabolic state of the tissue. A cryofluorescence imaging instrument was used to provide a quantitative assessment of NADH RR in both kidneys and hearts in mice treated with 3-iodothyronamine. We compared those results to corresponding tissues in control mice. In the kidneys of mice treated with a high dose T1AM, the mean values of the maximum projection of NADH RR were 2.6 ± 0.6 compared to 3.20 ± 0.03 in control mice, indicating a 19% (± 0.4) significant increase in oxidative stress (OS) in the high dose-treated kidneys (P = 0.047). However, kidneys treated with a low dose of T1AM showed no difference in NADH RR compared to the kidneys of control mice. Furthermore, low versus high dose treatment of T1AM showed different responses in the heart than in the kidneys. The mean value of the maximum projection of NADH RR in the heart changed from 3.0 ± 0.3 to 3.2 ± 0.6 for the low dose and the high dose T1AM-treated mice, respectively, as compared to 2.8 ± 0.7 in control mice. These values correspond to a 9% (±0.5) (P = 0.045) and 14% (±0.5) (P = 0.008) significant increase in NADH RR in the T1AM-treated hearts, indicating that the high dose T1AM-treated tissues have reduced OS compared to the low dose-treated tissues or the control tissues. These results suggest that while T1AM at a high dose increases oxidative response in kidneys, it has a protective effect in the heart and may exert its effect through alternative pathways at different doses and at tissue specific levels.


Subject(s)
Mitochondria/drug effects , Oxidative Stress , Thyronines/pharmacology , Animals , Animals, Outbred Strains , Female , Heart/drug effects , Kidney/drug effects , Kidney/metabolism , Mice , Mitochondria/metabolism , Myocardium/metabolism , Optical Imaging , Oxidation-Reduction/drug effects
17.
J Biol Chem ; 288(45): 32475-32489, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24068707

ABSTRACT

G protein-coupled receptors mediate responses to a myriad of ligands, some of which regulate adipocyte differentiation and metabolism. The sweet taste receptors T1R2 and T1R3 are G protein-coupled receptors that function as carbohydrate sensors in taste buds, gut, and pancreas. Here we report that sweet taste receptors T1R2 and T1R3 are expressed throughout adipogenesis and in adipose tissues. Treatment of mouse and human precursor cells with artificial sweeteners, saccharin and acesulfame potassium, enhanced adipogenesis. Saccharin treatment of 3T3-L1 cells and primary mesenchymal stem cells rapidly stimulated phosphorylation of Akt and downstream targets with functions in adipogenesis such as cAMP-response element-binding protein and FOXO1; however, increased expression of peroxisome proliferator-activated receptor γ and CCAAT/enhancer-binding protein α was not observed until relatively late in differentiation. Saccharin-stimulated Akt phosphorylation at Thr-308 occurred within 5 min, was phosphatidylinositol 3-kinase-dependent, and occurred in the presence of high concentrations of insulin and dexamethasone; phosphorylation of Ser-473 occurred more gradually. Surprisingly, neither saccharin-stimulated adipogenesis nor Thr-308 phosphorylation was dependent on expression of T1R2 and/or T1R3, although Ser-473 phosphorylation was impaired in T1R2/T1R3 double knock-out precursors. In mature adipocytes, artificial sweetener treatment suppressed lipolysis even in the presence of forskolin, and lipolytic responses were correlated with phosphorylation of hormone-sensitive lipase. Suppression of lipolysis by saccharin in adipocytes was also independent of T1R2 and T1R3. These results suggest that some artificial sweeteners have previously uncharacterized metabolic effects on adipocyte differentiation and metabolism and that effects of artificial sweeteners on adipose tissue biology may be largely independent of the classical sweet taste receptors, T1R2 and T1R3.


Subject(s)
Adipocytes/metabolism , Adipogenesis/drug effects , Lipolysis/drug effects , Receptors, G-Protein-Coupled/metabolism , Saccharin/pharmacology , Stem Cells/metabolism , Sweetening Agents/adverse effects , 3T3-L1 Cells , Adipogenesis/genetics , Adjuvants, Immunologic/pharmacology , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/drug effects , Colforsin/pharmacology , Cyclic AMP/genetics , Cyclic AMP/metabolism , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Lipolysis/genetics , Male , Mice , Middle Aged , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/genetics , Sterol Esterase/genetics , Sterol Esterase/metabolism , Sweetening Agents/pharmacokinetics
18.
Proteins ; 81(6): 919-25, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23349025

ABSTRACT

The sweet protein brazzein, a member of the Csßα fold family, contains four disulfide bonds that lend a high degree of thermal and pH stability to its structure. Nevertheless, a variable temperature study has revealed that the protein undergoes a local, reversible conformational change between 37 and 3°C with a midpoint about 27°C that changes the orientations and side-chain hydrogen bond partners of Tyr8 and Tyr11. To test the functional significance of this effect, we used NMR saturation transfer to investigate the interaction between brazzein and the amino terminal domain of the sweet receptor subunit T1R2; the results showed a stronger interaction at 7°C than at 37°C. Thus the low temperature conformation, which alters the orientations of two loops known to be critical for the sweetness of brazzein, may represent the bound state of brazzein in the complex with the human sweet receptor.


Subject(s)
Brassicaceae/chemistry , Plant Proteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Sweetening Agents/chemistry , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
19.
Mol Cell ; 49(1): 186-99, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23201123

ABSTRACT

Calorie restriction (CR) extends life span in diverse species. Mitochondria play a key role in CR adaptation; however, the molecular details remain elusive. We developed and applied a quantitative mass spectrometry method to probe the liver mitochondrial acetyl-proteome during CR versus control diet in mice that were wild-type or lacked the protein deacetylase SIRT3. Quantification of 3,285 acetylation sites-2,193 from mitochondrial proteins-rendered a comprehensive atlas of the acetyl-proteome and enabled global site-specific, relative acetyl occupancy measurements between all four experimental conditions. Bioinformatic and biochemical analyses provided additional support for the effects of specific acetylation on mitochondrial protein function. Our results (1) reveal widespread reprogramming of mitochondrial protein acetylation in response to CR and SIRT3, (2) identify three biochemically distinct classes of acetylation sites, and (3) provide evidence that SIRT3 is a prominent regulator in CR adaptation by coordinately deacetylating proteins involved in diverse pathways of metabolism and mitochondrial maintenance.


Subject(s)
Caloric Restriction , Mitochondrial Proteins/metabolism , Proteome/metabolism , Sirtuin 3/physiology , Acetyl Coenzyme A/metabolism , Acetylation , Adaptation, Physiological , Amino Acid Motifs , Amino Acid Sequence , Amino Acids/metabolism , Animals , Carbohydrate Metabolism , Cells, Cultured , Chromatography, Ion Exchange , Cluster Analysis , Consensus Sequence , Gene Expression , Genes, Mitochondrial , Liver/metabolism , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/isolation & purification , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Processing, Post-Translational , Proteome/chemistry , Proteome/isolation & purification , Sirtuin 3/chemistry , Sirtuin 3/isolation & purification , Sirtuin 3/metabolism , Staining and Labeling , Tandem Mass Spectrometry
20.
Methods Mol Biol ; 914: 47-63, 2012.
Article in English | MEDLINE | ID: mdl-22976022

ABSTRACT

Detection of weak ligand binding to membrane-spanning proteins, such as receptor proteins at low physiological concentrations, poses serious experimental challenges. Saturation transfer difference nuclear magnetic resonance (STD-NMR) spectroscopy offers an excellent way to surmount these problems. As the name suggests, magnetization transferred from the receptor to its bound ligand is measured by directly observing NMR signals from the ligand itself. Low-power irradiation is applied to a (1)H NMR spectral region containing protein signals but no ligand signals. This irradiation spreads quickly throughout the membrane protein by the process of spin diffusion and saturates all protein (1)H NMR signals. (1)H NMR signals from a ligand bound transiently to the membrane protein become saturated and, upon dissociation, serve to decrease the intensity of the (1)H NMR signals measured from the pool of free ligand. The experiment is repeated with the irradiation pulse placed outside the spectral region of protein and ligand, a condition that does not lead to saturation transfer to the ligand. The two resulting spectra are subtracted to yield the difference spectrum. As an illustration of the methodology, we review here STD-NMR experiments designed to investigate binding of ligands to the human sweet taste receptor, a member of the large family of G-protein-coupled receptors. Sweetener molecules bind to the sweet receptor with low affinity but high specificity and lead to a variety of physiological responses.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Membrane Proteins/metabolism , Animals , Glucose/metabolism , HEK293 Cells , Humans , Ligands , Mice , Models, Biological , Protein Binding , Receptors, Cell Surface/metabolism , Statistics as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...