Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Metab Eng ; 82: 274-285, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38428730

ABSTRACT

Rosavin is the characteristic component of Rhodiola rosea L., an important medicinal plant used widely in the world that has been reported to possess multiple biological activities. However, the endangered status of wild Rhodiola has limited the supply of rosavin. In this work, we successfully engineered an Escherichia coli strain to efficiently produce rosavin as an alternative production method. Firstly, cinnamate: CoA ligase from Hypericum calycinum, cinnamoyl-CoA reductase from Lolium perenne, and uridine diphosphate (UDP)-glycosyltransferase (UGT) from Bacillus subtilis (Bs-YjiC) were selected to improve the titer of rosin in E. coli. Subsequently, four UGTs from the UGT91R subfamily were identified to catalyze the formation of rosavin from rosin, with SlUGT91R1 from Solanum lycopersicum showing the highest activity level. Secondly, production of rosavin was achieved for the first time in E. coli by incorporating the SlUGT91R1 and UDP-arabinose pathway, including UDP-glucose dehydrogenase, UDP-xylose synthase, and UDP-xylose 4-epimerase, into the rosin-producing stain, and the titer reached 430.5 ± 91.4 mg/L. Thirdly, a two-step pathway derived from L-arabinose, composed of L-arabinokinase and UDP-sugar pyrophosphorylase, was developed in E. coli to further optimize the supply of the precursor UDP-arabinose. Furthermore, 1203.7 ± 32.1 mg/L of rosavin was produced from D-glucose and L-arabinose using shake-flask fermentation. Finally, the production of rosavin reached 7539.1 ± 228.7 mg/L by fed-batch fermentation in a 5-L bioreactor. Thus, the microbe-based production of rosavin shows great potential for commercialization. This work provides an effective strategy for the biosynthesis of other valuable natural products with arabinose-containing units from D-glucose and L-arabinose.


Subject(s)
Disaccharides , Glucose , Rhodiola , Glucose/genetics , Glucose/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Arabinose/metabolism , Rhodiola/genetics , Rhodiola/metabolism , Xylose/metabolism
2.
Molecules ; 26(5)2021 Mar 07.
Article in English | MEDLINE | ID: mdl-33800074

ABSTRACT

The present study aimed to investigate the effects of mungbean water extract (MWE) on insulin downstream signaling in insulin-resistant HepG2 cells. Whole seed mungbean was extracted using boiling water, mimicking a traditional cooking method. Vitexin and isovitexin were identified in MWE. The results showed that MWE inhibited protein tyrosine phosphatase (PTP)-1B (IC50 = 10 µg/mL), a negative regulator of insulin signaling. MWE enhanced cellular glucose uptake and altered expression of genes involved in glucose metabolism, including forkhead box O1 (FOXO1), phosphoenolpyruvate carboxykinase (PEPCK), and glycogen synthase kinase (GSK)-3ß in the insulin-resistant HepG2 cells. In addition, MWE inhibited both α-amylase (IC50 = 36.65 mg/mL) and α-glucosidase (IC50 = 3.07 mg/mL). MWE also inhibited the formation of advanced glycation end products (AGEs) (IC50 = 2.28 mg/mL). This is the first study to show that mungbean water extract increased cellular glucose uptake and improved insulin sensitivity of insulin-resistant HepG2 cells through PTP-1B inhibition and modulating the expression of genes related to glucose metabolism. This suggests that mungbean water extract has the potential to be a functional ingredient for diabetes.


Subject(s)
Enzyme Inhibitors/pharmacology , Plant Extracts/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Vigna/chemistry , Enzyme Inhibitors/chemistry , Flavonoids/analysis , Gene Expression Regulation/drug effects , Glucose/genetics , Glucose/pharmacokinetics , Glycation End Products, Advanced/drug effects , Glycation End Products, Advanced/metabolism , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , Hep G2 Cells , Humans , Insulin/pharmacology , Phenols/analysis , Plant Extracts/chemistry , Temperature , Water/chemistry , alpha-Amylases/antagonists & inhibitors
3.
Cancer Lett ; 506: 152-166, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33652086

ABSTRACT

Human glioblastoma (GBM) is one of the most feared primary malignant brain tumors. We investigated the effect of hyperbaric oxygen (HBO) on GBM patient-derived cells and on microglia cell biology (CHME-5). HBO administered to GBM cells inhibited cell proliferation, downregulated hypoxia-inducible factor 1 α (HIF-1α) expression, and induced glucose metabolism reprogramming (glucose rewiring). It also affected the ability of a cell to perpetuate its lineage, give rise to differentiated cells and interact with its environment to maintain a balance between quiescence, proliferation and regeneration (stemness features). Such an effect may be ascribable to an increase in intracellular ROS levels and to the triggering of inflammasome signaling by HBO itself through caspase1 activation. Moreover, the results obtained from the combination of HBO and radiotherapy (RT) clearly showed a radiosensitising effect of HBO on GBM cells grown in both 2D and 3D, and a radioprotective effect of HBO in CHME-5. In addition, the exposure of M0 microglia cells to exhausted medium or extracellular vesicles (EVs) of HBO-treated GBM cells upregulated the expression of pro-inflammatory cytokines IL1ß, IL6 and STAT1, whilst also downregulating the anti-inflammatory cytokine PPARγ. Collectively, these data provide a scientific rationale for the use of HBO in combination with RT for the treatment of patients with GBM.


Subject(s)
Glioblastoma/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-1beta/genetics , PPAR gamma/genetics , STAT1 Transcription Factor/genetics , Caspase 1/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Vesicles/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Glucose/genetics , Glucose/metabolism , Humans , Hyperbaric Oxygenation/adverse effects , Inflammasomes/drug effects , Interleukin-6/genetics , Microglia/drug effects , Microglia/pathology , Oxygen/pharmacology , Pressure , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
4.
Commun Biol ; 3(1): 514, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32948821

ABSTRACT

We demonstrate the mechanism by which C3G, a major dietary anthocyanin, regulates energy metabolism and insulin sensitivity. Oral administration of C3G reduced hepatic and plasma triglyceride levels, adiposity, and improved glucose tolerance in mice fed high-fat diet. Hepatic metabolomic analysis revealed that C3G shifted metabolite profiles towards fatty acid oxidation and ketogenesis. C3G increased glucose uptake in HepG2 cells and C2C12 myotubes and induced the rate of hepatic fatty acid oxidation. C3G directly interacted with and activated PPARs, with the highest affinity for PPARα. The ability of C3G to reduce plasma and hepatic triglycerides, glucose tolerance, and adiposity and to induce oxygen consumption and energy expenditure was abrogated in PPARα-deficient mice, suggesting that PPARα is the major target for C3G. These findings demonstrate that the dietary anthocyanin C3G activates PPARs, a master regulators of energy metabolism. C3G is an agonistic ligand of PPARs and stimulates fuel preference to fat.


Subject(s)
Anthocyanins/genetics , Mediator Complex Subunit 1/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Animals , Anthocyanins/pharmacology , Dietary Supplements , Energy Metabolism/genetics , Glucose/genetics , Hep G2 Cells , Humans , Insulin/genetics , Insulin/metabolism , Insulin Resistance/genetics , Lipid Metabolism/genetics , Liver/metabolism , Mice
5.
J Cell Physiol ; 233(9): 6878-6887, 2018 09.
Article in English | MEDLINE | ID: mdl-29319178

ABSTRACT

Aggrecan is a high molecular weight proteoglycan that plays a critical role in cartilage structure and the function of joints, providing intervertebral disc and cartilage with the ability to resist compressive loads. Aggrecan degradation in articular cartilage is a significant event in early-stage osteoarthritis (OA). Currently, no effective treatment exists for OA other than pain relief. Dextrose (D-glucose) prolotherapy has shown promising activity in the treatment of different musculoskeletal disorders, including OA. However, little is known about the molecular mechanism of the glucose effect in OA and on the regulation of chondrogenesis. We show for the first time that glucose upregulates aggrecan expression and subsequent chondrogenesis in ATDC5 cells. Moreover, we found that glucose-induced aggrecan expression is mediated through the protein kinase Cα (PKCα)- and p38-dependent pathway. As demonstrated by microRNA (miR) and luciferase analyses, the glucose-induced PKCα/p38 signaling axis is responsible for downregulating miR141-3p which targets to the 3'untranslated region of aggrecan. In summary, we show that glucose enhances chondrogenesis by upregulating aggrecan expression via the PKCα-p38-miR141-3p signaling pathway. This result provides new insights into the mechanism of glucose on chondrogenesis.


Subject(s)
Aggrecans/genetics , Chondrocytes/physiology , Glucose/genetics , MAP Kinase Signaling System/genetics , MicroRNAs/genetics , Protein Kinase C-alpha/genetics , Signal Transduction/genetics , 3' Untranslated Regions/genetics , Animals , Cartilage, Articular/physiology , Cells, Cultured , Chondrogenesis/genetics , Down-Regulation/genetics , Mice , Osteoarthritis/genetics , Up-Regulation/genetics
6.
PLoS One ; 12(9): e0185112, 2017.
Article in English | MEDLINE | ID: mdl-28945821

ABSTRACT

Floret, leaf, and root tissues were harvested from broccoli and collard cultivars and extracted to determine their glucosinolate and hydrolysis product profiles using high performance liquid chromatography and gas chromotography. Quinone reductase inducing bioactivity, an estimate of anti-cancer chemopreventive potential, of the extracts was measured using a hepa1c1c7 murine cell line. Extracts from root tissues were significantly different from other tissues and contained high levels of gluconasturtiin and glucoerucin. Targeted gene expression analysis on glucosinolate biosynthesis revealed that broccoli root tissue has elevated gene expression of AOP2 and low expression of FMOGS-OX homologs, essentially the opposite of what was observed in broccoli florets, which accumulated high levels of glucoraphanin. Broccoli floret tissue has significantly higher nitrile formation (%) and epithionitrile specifier protein gene expression than other tissues. This study provides basic information of the glucosinolate metabolome and transcriptome for various tissues of Brassica oleracea that maybe utilized as potential byproducts for the nutraceutical market.


Subject(s)
Anticarcinogenic Agents/metabolism , Brassica/genetics , Brassica/metabolism , Glucosinolates/genetics , Glucosinolates/metabolism , Anticarcinogenic Agents/analysis , Brassica/chemistry , Dietary Supplements/analysis , Flowering Tops/metabolism , Gene Expression Profiling , Genes, Plant , Glucose/analogs & derivatives , Glucose/analysis , Glucose/genetics , Glucose/metabolism , Glucosinolates/analysis , Humans , Hydrolysis , Imidoesters/analysis , Imidoesters/metabolism , Metabolome , Microfluidic Analytical Techniques , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Plant Leaves/metabolism , Plant Proteins/biosynthesis , Plant Roots/metabolism , RNA, Plant/genetics , RNA, Plant/metabolism , Tissue Distribution
7.
Funct Integr Genomics ; 17(4): 459-476, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28236275

ABSTRACT

Cold storage (2-4 °C) used in potato production to suppress diseases and sprouting during storage can result in cold-induced sweetening (CIS), where reducing sugars accumulate in tuber tissue leading to undesirable browning, production of bitter flavors, and increased levels of acrylamide with frying. Potato exhibits genetic and environmental variation in resistance to CIS. The current study profiles gene expression in post-harvest tubers before cold storage using transcriptome sequencing and identifies genes whose expression is predictive for CIS. A distance matrix for potato clones based on glucose levels after cold storage was constructed and compared to distance matrices constructed using RNA-seq gene expression data. Congruence between glucose and gene expression distance matrices was tested for each gene. Correlation between glucose and gene expression was also tested. Seventy-three genes were found that had significant p values in the congruence and correlation tests. Twelve genes from the list of 73 genes also had a high correlation between glucose and gene expression as measured by Nanostring nCounter. The gene annotations indicated functions in protein degradation, nematode resistance, auxin transport, and gibberellin response. These 12 genes were used to build models for prediction of CIS using multiple linear regression. Nine linear models were constructed that used different combinations of the 12 genes. An F-box protein, cellulose synthase, and a putative Lax auxin transporter gene were most frequently used. The findings of this study demonstrate the utility of gene expression profiles in predictive diagnostics for severity of CIS.


Subject(s)
Glucose/metabolism , Plant Proteins/genetics , Solanum tuberosum/genetics , Cold-Shock Response , Gene Expression Regulation, Plant , Glucose/genetics , Plant Proteins/metabolism , Solanum tuberosum/growth & development , Solanum tuberosum/metabolism
8.
J Biol Chem ; 291(42): 21869-21879, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27539851

ABSTRACT

Tumors rely on multiple nutrients to meet cellular bioenergetics and macromolecular synthesis demands of rapidly dividing cells. Although the role of glucose and glutamine in cancer metabolism is well understood, the relative contribution of acetate metabolism remains to be clarified. We show that glutamine supplementation is not sufficient to prevent loss of cell viability in a subset of glucose-deprived melanoma cells, but synergizes with acetate to support cell survival. Glucose-deprived melanoma cells depend on both oxidative phosphorylation and acetate metabolism for cell survival. Acetate supplementation significantly contributed to maintenance of ATP levels in glucose-starved cells. Unlike acetate, short chain fatty acids such as butyrate and propionate failed to prevent loss of cell viability from glucose deprivation. In vivo studies revealed that in addition to nucleo-cytoplasmic acetate assimilating enzyme ACSS2, mitochondrial ACSS1 was critical for melanoma tumor growth in mice. Our data indicate that acetate metabolism may be a potential therapeutic target for BRAF mutant melanoma.


Subject(s)
Acetates/metabolism , Glucose/metabolism , Melanoma/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Animals , Butyric Acid/metabolism , Cell Line, Tumor , Female , Glucose/genetics , Heterografts , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasm Transplantation , Oxidative Phosphorylation , Propionates/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
9.
J Inherit Metab Dis ; 39(2): 309-19, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26830550

ABSTRACT

Tetrahydrobiopterin (BH4) is an essential cofactor for the aromatic amino acid hydroxylases, alkylglycerol monooxygenase, and nitric oxide synthases (NOS). Inborn errors of BH4 metabolism lead to severe insufficiency of brain monoamine neurotransmitters while augmentation of BH4 by supplementation or stimulation of its biosynthesis is thought to ameliorate endothelial NOS (eNOS) dysfunction, to protect from (cardio-) vascular disease and/or prevent obesity and development of the metabolic syndrome. We have previously reported that homozygous knock-out mice for the 6-pyruvolytetrahydropterin synthase (PTPS; Pts-ko/ko) mice with no BH4 biosynthesis die after birth. Here we generated a Pts-knock-in (Pts-ki) allele expressing the murine PTPS-p.Arg15Cys with low residual activity (15% of wild-type in vitro) and investigated homozygous (Pts-ki/ki) and compound heterozygous (Pts-ki/ko) mutants. All mice showed normal viability and depending on the severity of the Pts alleles exhibited up to 90% reduction of PTPS activity concomitant with neopterin elevation and mild reduction of total biopterin while blood L-phenylalanine and brain monoamine neurotransmitters were unaffected. Yet, adult mutant mice with compromised PTPS activity (i.e., Pts-ki/ko, Pts-ki/ki or Pts-ko/wt) had increased body weight and elevated intra-abdominal fat. Comprehensive phenotyping of Pts-ki/ki mice revealed alterations in energy metabolism with proportionally higher fat content but lower lean mass, and increased blood glucose and cholesterol. Transcriptome analysis indicated changes in glucose and lipid metabolism. Furthermore, differentially expressed genes associated with obesity, weight loss, hepatic steatosis, and insulin sensitivity were consistent with the observed phenotypic alterations. We conclude that reduced PTPS activity concomitant with mildly compromised BH4-biosynthesis leads to abnormal body fat distribution and abdominal obesity at least in mice. This study associates a novel single gene mutation with monogenic forms of obesity.


Subject(s)
Adipose Tissue/metabolism , Biopterins/analogs & derivatives , Body Fat Distribution , Obesity, Abdominal/genetics , Phosphorus-Oxygen Lyases/genetics , Alleles , Animals , Biopterins/biosynthesis , Biopterins/genetics , Body Weight/genetics , Cholesterol/genetics , Female , Genotype , Glucose/genetics , Heterozygote , Homozygote , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III/genetics , Phenylalanine/genetics , Transcriptome/genetics
10.
J Biol Rhythms ; 29(5): 332-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25252711

ABSTRACT

Birds seasonally switch from one life history state (LHS) to another to maximize their fitness. Accordingly, they exhibit distinct differences in their physiological and behavioral phenotypes between seasons. Possible molecular mechanisms underlying changes through the seasons have scarcely been examined in migratory birds. The present study measured key genes suggested to be involved in the metabolic regulation of 4 photoperiodically induced seasonal LHSs in a long-distance migratory songbird, the blackheaded bunting (Emberiza melanocephala). Buntings were held under short days (8 h light:16 h darkness, 8L:16D), during which they maintained the winter nonmigratory phenotype. Then they were exposed for several weeks to long days (13L:11D). Differences in the activity-rest pattern, body fattening and weight gain, testis size, organ (heart, intestine) weights, and blood glucose and triglyceride levels confirmed that buntings sequentially exhibited spring migration-linked premigratory, migratory, and postmigratory LHSs under long days. The mRNA levels of circadian genes involved in metabolism (Bmal1, Clock, Npas2, Rorα, and Rev-erbα) and of genes that encode for proteins/enzymes involved in the regulation of glucose (Sirt1, FoxO1, Glut1, and Pygl) and lipids (Hmg-CoA; Pparα, Pparγ; Fasn and Acaca) showed LHS-dependent changes in their light-dark expression patterns in the hypothalamus and liver. These initial results on genetic regulation of metabolism in a migratory species extend the idea that the transitions between LHSs in a seasonal species are accomplished by changes at multiple regulatory levels. Thus, these findings promise new insights into the mechanism(s) of adaptation to seasons in higher vertebrates.


Subject(s)
Animal Migration/physiology , Circadian Rhythm/genetics , Hypothalamus/physiology , Liver/physiology , Songbirds/physiology , Adaptation, Physiological/physiology , Animals , Gene Expression , Glucose/genetics , Glucose/metabolism , Hypothalamus/metabolism , Light , Male , Photoperiod , RNA, Messenger/genetics , RNA, Messenger/metabolism , Seasons , Songbirds/genetics , Songbirds/metabolism , Testis/metabolism , Testis/physiology
11.
J Clin Invest ; 124(5): 2193-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24667636

ABSTRACT

The majority of mammalian cells have nonmotile primary cilia on their surface that act as antenna-like sensory organelles. Genetic defects that result in ciliary dysfunction are associated with obesity in humans and rodents, which suggests that functional cilia are important for controlling energy balance. Here we demonstrated that neuronal cilia lengths were selectively reduced in hypothalami of obese mice with leptin deficiency and leptin resistance. Treatment of N1 hypothalamic neuron cells with leptin stimulated cilia assembly via inhibition of the tumor suppressors PTEN and glycogen synthase kinase 3ß (GSK3ß). Induction of short cilia in the hypothalamus of adult mice increased food intake and decreased energy expenditure, leading to a positive energy balance. Moreover, mice with short hypothalamic cilia exhibited attenuated anorectic responses to leptin, insulin, and glucose, which indicates that leptin-induced cilia assembly is essential for sensing these satiety signals by hypothalamic neurons. These data suggest that leptin governs the sensitivity of hypothalamic neurons to metabolic signals by controlling the length of the cell's antenna.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/metabolism , Neurons/metabolism , Animals , Anorexia/genetics , Anorexia/metabolism , Cell Line , Cilia/genetics , Cilia/metabolism , Glucose/genetics , Glucose/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hypothalamus/cytology , Insulin/genetics , Insulin/metabolism , Leptin , Mice , Mice, Knockout , Neurons/cytology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
12.
J Cereb Blood Flow Metab ; 34(2): 339-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24301293

ABSTRACT

Hypothalamic glucose detection participates in maintaining glycemic balance, food intake, and thermogenesis. Although hypothalamic neurons are the executive cells involved in these responses, there is increasing evidence that astrocytes participate in glucose sensing (GS); however, it is unknown whether astroglial networking is required for glucose sensitivity. Astroglial connexins 30 and 43 (Cx30 and Cx43) form hexameric channels, which are apposed in gap junctions, allowing for the intercellular transfer of small molecules such as glucose throughout the astroglial networks. Here, we hypothesized that hypothalamic glucose sensitivity requires these connexins. First, we showed that both Cxs are enriched in the rat hypothalamus, with highly concentrated Cx43 expression around blood vessels of the mediobasal hypothalamus (MBH). Both fasting and high glycemic levels rapidly altered the protein levels of MBH astroglial connexins, suggesting cross talk within the MBH between glycemic status and the connexins' ability to dispatch glucose. Finally, the inhibition of MBH Cx43 (by transient RNA interference) attenuated hypothalamic glucose sensitivity in rats, which was demonstrated by a pronounced decreased insulin secretion in response to a brain glucose challenge. These results illustrate that astroglial connexins contribute to hypothalamic GS.


Subject(s)
Astrocytes/metabolism , Connexin 43/metabolism , Connexins/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Nerve Tissue Proteins/metabolism , Animals , Astrocytes/cytology , Connexin 30 , Connexin 43/genetics , Connexins/genetics , Fasting/metabolism , Glucose/genetics , Hypothalamus/cytology , Insulin Secretion , Male , Nerve Tissue Proteins/genetics , RNA Interference , Rats , Rats, Wistar
13.
Metab Eng ; 19: 116-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23938029

ABSTRACT

Most central metabolic pathways such as glycolysis, fatty acid synthesis, and the TCA cycle have complementary pathways that run in the reverse direction to allow flexible storage and utilization of resources. However, the glyoxylate shunt, which allows for the synthesis of four-carbon TCA cycle intermediates from acetyl-CoA, has not been found to be reversible to date. As a result, glucose can only be converted to acetyl-CoA via the decarboxylation of the three-carbon molecule pyruvate in heterotrophs. A reverse glyoxylate shunt (rGS) could be extended into a pathway that converts C4 carboxylates into two molecules of acetyl-CoA without loss of CO2. Here, as a proof of concept, we engineered in Escherichia coli such a pathway to convert malate and succinate to oxaloacetate and two molecules of acetyl-CoA. We introduced ATP-coupled heterologous enzymes at the thermodynamically unfavorable steps to drive the pathway in the desired direction. This synthetic pathway in essence reverses the glyoxylate shunt at the expense of ATP. When integrated with central metabolism, this pathway has the potential to increase the carbon yield of acetate and biofuels from many carbon sources in heterotrophic microorganisms, and could be the basis of novel carbon fixation cycles.


Subject(s)
Citric Acid Cycle , Escherichia coli/metabolism , Glucose/metabolism , Glyoxylates/metabolism , Metabolic Engineering , Oxaloacetic Acid/metabolism , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/metabolism , Escherichia coli/genetics , Glucose/genetics , Malates/metabolism , Succinic Acid/metabolism
14.
J Biol Chem ; 288(15): 10722-35, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23457303

ABSTRACT

Sirt1 is a NAD(+)-dependent class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, emerging evidence suggests that neuronal Sirt1 activity plays a role in the central regulation of energy balance and glucose metabolism. To assess this idea, we generated Sirt1 neuron-specific knockout (SINKO) mice. On both standard chow and HFD, SINKO mice were more insulin sensitive than Sirt1(f/f) mice. Thus, SINKO mice had lower fasting insulin levels, improved glucose tolerance and insulin tolerance, and enhanced systemic insulin sensitivity during hyperinsulinemic euglycemic clamp studies. Hypothalamic insulin sensitivity of SINKO mice was also increased over controls, as assessed by hypothalamic activation of PI3K, phosphorylation of Akt and FoxO1 following systemic insulin injection. Intracerebroventricular injection of insulin led to a greater systemic effect to improve glucose tolerance and insulin sensitivity in SINKO mice compared with controls. In line with the in vivo results, insulin-induced AKT and FoxO1 phosphorylation were potentiated by inhibition of Sirt1 in a cultured hypothalamic cell line. Mechanistically, this effect was traced to a reduced effect of Sirt1 to directly deacetylate and repress IRS-1 function. The enhanced central insulin signaling in SINKO mice was accompanied by increased insulin receptor signal transduction in liver, muscle, and adipose tissue. In summary, we conclude that neuronal Sirt1 negatively regulates hypothalamic insulin signaling, leading to systemic insulin resistance. Interventions that reduce neuronal Sirt1 activity have the potential to improve systemic insulin action and limit weight gain on an obesigenic diet.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Nerve Tissue Proteins/metabolism , Sirtuin 1/metabolism , Animals , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucose/genetics , Glucose/metabolism , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin/genetics , Insulin/pharmacology , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Organ Specificity , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sirtuin 1/genetics
15.
J Biol Chem ; 287(50): 42324-32, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23074218

ABSTRACT

A scientific explanation for the beneficial role of vitamin D supplementation in the lowering of glycemia in diabetes remains to be determined. This study examined the biochemical mechanism by which vitamin D supplementation regulates glucose metabolism in diabetes. 3T3L1 adipocytes were treated with high glucose (HG, 25 mm) in the presence or absence of 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) (25, 50 nm), the active form of vitamin D. 1,25(OH)(2)D(3) treatment caused significant up-regulation of GLUT4 total protein expression and its translocation to cell surface, and an increase in glucose uptake as well as glucose utilization in HG-treated cells. 1,25(OH)(2)D(3) also caused cystathionine-γ-lyase (CSE) activation and H(2)S formation in HG-treated adipocytes. The effect of 1,25(OH)(2)D(3) on GLUT4 translocation, glucose utilization, and H(2)S formation was prevented by propargylglycine, an inhibitor of CSE that catalyzes H(2)S formation. Studies using antisense CSE also demonstrated the inhibition of GLUT4 translocation as well as glucose uptake and utilization in 1,25(OH)(2)D(3)-supplemented CSE-siRNA-transfected adipocytes compared with controls. 1,25(OH)(2)D(3) treatment along with insulin enhanced GLUT4 translocation and glucose utilization compared with either insulin or 1,25(OH)(2)D(3) alone in HG-treated adipocytes. 1,25(OH)(2)D(3) supplementation also inhibited monocyte chemoattractant protein-1 and stimulated adiponectin secretion in HG-treated adipocytes, and this positive effect was prevented in propargylglycine-treated or CSE-knockdown adipocytes. This is the first report to demonstrate that 1,25(OH)(2)D(3) up-regulates GLUT4 translocation and glucose utilization and decreases inflammatory markers, which is mediated by CSE activation and H(2)S formation in adipocytes. This study provides evidence for a novel molecular mechanism by which 1,25(OH)(2)D(3) can up-regulate the GLUT4 translocation essential for maintenance of glucose metabolism.


Subject(s)
Adipocytes/metabolism , Calcitriol/pharmacology , Cystathionine gamma-Lyase/metabolism , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Hydrogen Sulfide/metabolism , Up-Regulation/drug effects , Vitamins/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cystathionine gamma-Lyase/genetics , Enzyme Activation/drug effects , Glucose/genetics , Glucose Transporter Type 4/genetics , Mice , Protein Transport/drug effects , Protein Transport/physiology , Up-Regulation/physiology
16.
Sci Rep ; 2: 693, 2012.
Article in English | MEDLINE | ID: mdl-23012647

ABSTRACT

In settings of increased insulin demand, failure to expand pancreatic ß-cells mass leads to diabetes. Genome-wide scans of diabetic populations have uncovered several genes associated with susceptibility to type 2 diabetes and a number of them are part of the Wnt signaling. ß-Catenin, a Wnt downstream effector participates in pancreatic development, however, little is known about its action in mature ß-cells. Deletion of ß-Catenin in Pdx1 pancreatic progenitors leads to a decreased ß-cell mass and impaired glucose tolerance. Surprisingly, loss of ß-catenin made these mice resistant to high fat diet because of their increased energy expenditure and insulin sensitivity due to hyperactivity. The complexity of this phenotype was also explained in part by ectopic expression of Cre recombinase in the hypothalamus. Our data implicates ß-Catenin in the regulation of metabolism and energy homeostasis and suggest that Wnt signaling modulates the susceptibility to diabetes by acting on different tissues.


Subject(s)
Energy Metabolism/physiology , Glucose/metabolism , Homeostasis/physiology , Insulin-Secreting Cells/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Gene Deletion , Glucose/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Insulin/genetics , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Stem Cells/cytology , Trans-Activators/genetics , Trans-Activators/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
17.
Pesqui. vet. bras ; 32(5): 463-469, maio 2012. ilus, graf
Article in Portuguese | LILACS | ID: lil-626488

ABSTRACT

O objetivo principal da nossa pesquisa foi avaliar o potencial de diferenciação osteogênica de células-tronco mesenquimais (MSC) obtidas da medula óssea do cão. As MSC foram separadas pelo método Ficoll e cultivadas sob duas condições distintas: DMEM baixa glicose ou DMEM/F12, ambos contendo L-glutamina, 20% de SFB e antibióticos. Marcadores de MSC foram testados, confirmando células CD44+ e CD34- através da citometria de fluxo. Para a diferenciação osteogênica, as células foram submetidas a quatro diferentes condições: Grupo 1, as mesmas condições utilizadas para a cultura de células primárias com os meios DMEM baixa glicose suplementado; Grupo 2, as mesmas condições do Grupo 1, mais os indutores de diferenciação dexametasona, ácido ascórbico e b-glicerolfosfato; Grupo 3, células cultivadas com meios DMEM/F12 suplementado; e Grupo 4, nas mesmas condições que no Grupo 3, mais indutores de diferenciação de dexametasona, ácido ascórbico e b-glicerolfosfato. A diferenciação celular foi confirmada através da coloração com alizarin red e da imunomarcação com o anticorpo SP7/Osterix. Nós observamos através da coloração com alizarin red que o depósito de cálcio foi mais evidente nas células cultivadas em DMEM/F12. Além disso, usando a imunomarcação com o anticorpo SP/7Osterix obtivemos positividade em 1:6 células para o Meio DMEM/F12 comparada com 1:12 para o meio DMEM-baixa glicose. Com base nos nossos resultados concluímos que o meio DMEM/F12 é mais eficiente para a indução da diferenciação de células-tronco mesenquimais caninas em promotores osteogênicos. Este efeito provavelmente ocorre em decorrência da maior quantidade de glicose neste meio, bem como da presença de diversos aminoácidos.


The aim of our research was to evaluate the potential for osteogenic differentiation of mesenchimal stem cells (MSC) obtained from dog bone marrow. The MSC were separated using the Ficoll method and cultured under two different conditions: DMEM low glucose or DMEM/F12, both containing L-glutamine, 20% of FBS and antibiotics. MSC markers were tested, confirming CD44+ and CD34- cells with flow cytometry. For osteogenic differentiation, cells were submitted to four different conditions: Group 1, same conditions used for primary cell culture with DMEM supplemented media; Group 2, same conditions of Group 1 plus differentiation inductors Dexametazone, ascorbic acid and β-glicerolphosphate. Group 3, Cells cultured with supplemented DMEM/F12 media, and Group 4, same conditions as in Group 3 plus differentiation inductors Dexametazone, ascorbic acid and β-glicerolphosphate. The cellular differentiation was confirmed using alizarin red and imunostaining with SP7/Osterix antibody. We observed by alizarin staining that calcium deposit was more evident in cells cultivated in DMEM/F12.Furthermore, by SP/7Osterix antibody immunostaining we obtained 1:6 positive cells when using DMEM/F12 compared with 1:12 for low-glucose DMEM. Based on our results, we conclude that the medium DMEM/F12 is more efficient for induction of differentiation of mesenchymal stem cells in canine osteogenic progenitors. This effect is probably due to the greater amount of glucose in the medium and the presence of various amino acids.


Subject(s)
Animals , Dogs , Dogs/genetics , Mesenchymal Stem Cells/cytology , Bone Marrow/physiology , Osteogenesis/genetics , Glucose/genetics , Culture Media/isolation & purification , Cell Culture Techniques/veterinary
18.
J Biol Chem ; 286(39): 34164-74, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21813650

ABSTRACT

Glutamate dehydrogenase (GDH) catalyzes the oxidative deamination of L-glutamate and, in animals, is extensively regulated by a number of metabolites. Gain of function mutations in GDH that abrogate GTP inhibition cause the hyperinsulinism/hyperammonemia syndrome (HHS), resulting in increased pancreatic ß-cell responsiveness to leucine and susceptibility to hypoglycemia following high protein meals. We have previously shown that two of the polyphenols from green tea (epigallocatechin gallate (EGCG) and epicatechin gallate (ECG)) inhibit GDH in vitro and that EGCG blocks GDH-mediated insulin secretion in wild type rat islets. Using structural and site-directed mutagenesis studies, we demonstrate that ECG binds to the same site as the allosteric regulator, ADP. Perifusion assays using pancreatic islets from transgenic mice expressing a human HHS form of GDH demonstrate that the hyperresponse to glutamine caused by dysregulated GDH is blocked by the addition of EGCG. As observed in HHS patients, these transgenic mice are hypersensitive to amino acid feeding, and this is abrogated by oral administration of EGCG prior to challenge. Finally, the low basal blood glucose level in the HHS mouse model is improved upon chronic administration of EGCG. These results suggest that this common natural product or some derivative thereof may prove useful in controlling this genetic disorder. Of broader clinical implication is that other groups have shown that restriction of glutamine catabolism via these GDH inhibitors can be useful in treating various tumors. This HHS transgenic mouse model offers a highly useful means to test these agents in vivo.


Subject(s)
Adenosine Diphosphate/metabolism , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Catechin/analogs & derivatives , Glutamate Dehydrogenase/metabolism , Islets of Langerhans/metabolism , Tea/chemistry , Adenosine Diphosphate/genetics , Animals , Anticarcinogenic Agents/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Catalytic Domain/genetics , Catechin/chemistry , Catechin/pharmacology , Glucose/genetics , Glucose/metabolism , Glutamate Dehydrogenase/genetics , Glutamine/genetics , Glutamine/metabolism , Humans , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Rats
19.
Diabetes ; 60(3): 735-45, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21266325

ABSTRACT

OBJECTIVE: AMP-activated protein kinase (AMPK) signaling acts as a sensor of nutrients and hormones in the hypothalamus, thereby regulating whole-body energy homeostasis. Deletion of Ampkα2 in pro-opiomelanocortin (POMC) neurons causes obesity and defective neuronal glucose sensing. LKB1, the Peutz-Jeghers syndrome gene product, and Ca(2+)-calmodulin-dependent protein kinase kinase ß (CaMKKß) are key upstream activators of AMPK. This study aimed to determine their role in POMC neurons upon energy and glucose homeostasis regulation. RESEARCH DESIGN AND METHODS: Mice lacking either Camkkß or Lkb1 in POMC neurons were generated, and physiological, electrophysiological, and molecular biology studies were performed. RESULTS: Deletion of Camkkß in POMC neurons does not alter energy homeostasis or glucose metabolism. In contrast, female mice lacking Lkb1 in POMC neurons (PomcLkb1KO) display glucose intolerance, insulin resistance, impaired suppression of hepatic glucose production, and altered expression of hepatic metabolic genes. The underlying cellular defect in PomcLkb1KO mice involves a reduction in melanocortin tone caused by decreased α-melanocyte-stimulating hormone secretion. However, Lkb1-deficient POMC neurons showed normal glucose sensing, and body weight was unchanged in PomcLkb1KO mice. CONCLUSIONS: Our findings demonstrate that LKB1 in hypothalamic POMC neurons plays a key role in the central regulation of peripheral glucose metabolism but not body-weight control. This phenotype contrasts with that seen in mice lacking AMPK in POMC neurons with defects in body-weight regulation but not glucose homeostasis, which suggests that LKB1 plays additional functions distinct from activating AMPK in POMC neurons.


Subject(s)
Glucose/metabolism , Homeostasis/genetics , Hypothalamus/metabolism , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Analysis of Variance , Animals , Area Under Curve , Body Weight/genetics , Cell Count , Eating/genetics , Electrophysiology , Energy Metabolism/genetics , Female , Glucose/genetics , Glucose Clamp Technique , Immunohistochemistry , Insulin Resistance/genetics , Male , Mice , Mice, Transgenic , Pro-Opiomelanocortin/genetics , Protein Serine-Threonine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Statistics, Nonparametric
20.
J Nutr Biochem ; 22(2): 179-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20970313

ABSTRACT

Peroxisome-proliferator-activated receptor γ (PPARγ) plays a critical role in regulation of adipocyte differentiation and insulin sensitivity. To become functional, PPARγ must be activated by binding an appropriate ligand. Polyunsaturated fatty acids (PUFA) are potential ligands for PPARγ. The current experiment was designed to determine the potential for PUFA, particularly eicosapentaenoic acid and docosahexaenoic acid, to activate the function of porcine PPARγ in vivo. Transgenic mice, expressing porcine PPARγ in skeletal muscle were generated and fed with a high-saturated fat (beef tallow) or high-unsaturated fat (fish oil) diet for 4 months. When transgenic mice were fed a fish oil supplemented diet, the expression of adipogenic and glucose uptake genes was increased, leading to reduced plasma glucose concentration. The PPARγ transgene increased the expression of Glut4 in the muscle. This result suggests that there was increased glucose utilization and, therefore, a reduced blood glucose concentration in the transgenic mice. Also, the plasma adiponectin was elevated by fish oil treatment, suggesting a role of adiponectin in mediating the PUFA effect. These results suggest that PUFA may serve as a natural regulator of glucose uptake in vivo and these effects are mainly through PPARγ function.


Subject(s)
Dietary Fats, Unsaturated/metabolism , Fish Oils/metabolism , Glucose/genetics , Lipid Metabolism/genetics , PPAR gamma/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Animals , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Gene Expression Regulation , Glucose/metabolism , Insulin Resistance , Male , Mice , Mice, Transgenic , Muscle, Skeletal/chemistry , PPAR gamma/genetics , Swine
SELECTION OF CITATIONS
SEARCH DETAIL