Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Biomark Res ; 4: 20, 2016.
Article in English | MEDLINE | ID: mdl-27795830

ABSTRACT

Pancreatic cancer is characterized as a disease with low survival and high mortality because of no effective diagnostic and therapeutic strategies available in clinic. Conventional clinical diagnostic methods including serum markers and radiological imaging (CT, MRI, EUS, etc.) often fail to detect precancerous or early stage lesions. Development of effective biomarkers is unmet for reduction of mortality of pancreatic cancer. MicroRNAs (miRNAs) are a group of small non-protein-coding RNAs playing roles in regulation of cell physiology including tumorigenesis, apoptotic escape, proliferation, invasion, epithelial-mesenchymal transition (EMT), metastasis and chemoresistance. Various altered signaling pathways involving in molecular pathogenesis of pancreatic cancer are mediated by miRNAs as a role of either oncogenes or tumor suppressors. Among biomarkers developed including protein, metabolites, DNA, RNA, epigenetic mutation, miRNAs are superior because of its unique chemical property. Recent study suggests that miRNAs may be promising biomarkers used for early detection of pancreatic cancer. This review will update the progression made in early detection of pancreatic cancer.

2.
Diabetes ; 65(10): 2876-87, 2016 10.
Article in English | MEDLINE | ID: mdl-27385159

ABSTRACT

Hepatic steatosis is associated with increased insulin resistance and tricarboxylic acid (TCA) cycle flux, but decreased ketogenesis and pyruvate dehydrogenase complex (PDC) flux. This study examined whether hepatic PDC activation by inhibition of pyruvate dehydrogenase kinase 2 (PDK2) ameliorates these metabolic abnormalities. Wild-type mice fed a high-fat diet exhibited hepatic steatosis, insulin resistance, and increased levels of pyruvate, TCA cycle intermediates, and malonyl-CoA but reduced ketogenesis and PDC activity due to PDK2 induction. Hepatic PDC activation by PDK2 inhibition attenuated hepatic steatosis, improved hepatic insulin sensitivity, reduced hepatic glucose production, increased capacity for ß-oxidation and ketogenesis, and decreased the capacity for lipogenesis. These results were attributed to altered enzymatic capacities and a reduction in TCA anaplerosis that limited the availability of oxaloacetate for the TCA cycle, which promoted ketogenesis. The current study reports that increasing hepatic PDC activity by inhibition of PDK2 ameliorates hepatic steatosis and insulin sensitivity by regulating TCA cycle anaplerosis and ketogenesis. The findings suggest PDK2 is a potential therapeutic target for nonalcoholic fatty liver disease.


Subject(s)
Fatty Liver/enzymology , Fatty Liver/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Citric Acid Cycle/genetics , Citric Acid Cycle/physiology , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Glucose/metabolism , Insulin Resistance , Lipogenesis/physiology , Liver/metabolism , Liver/pathology , Male , Malonyl Coenzyme A/metabolism , Mice , Mice, Knockout , Oxaloacetic Acid/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/metabolism , Pyruvic Acid/metabolism
3.
J Clin Invest ; 125(4): 1579-90, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25798621

ABSTRACT

Metabolic reprogramming is implicated in macrophage activation, but the underlying mechanisms are poorly understood. Here, we demonstrate that the NOTCH1 pathway dictates activation of M1 phenotypes in isolated mouse hepatic macrophages (HMacs) and in a murine macrophage cell line by coupling transcriptional upregulation of M1 genes with metabolic upregulation of mitochondrial oxidative phosphorylation and ROS (mtROS) to augment induction of M1 genes. Enhanced mitochondrial glucose oxidation was achieved by increased recruitment of the NOTCH1 intracellular domain (NICD1) to nuclear and mitochondrial genes that encode respiratory chain components and by NOTCH-dependent induction of pyruvate dehydrogenase phosphatase 1 (Pdp1) expression, pyruvate dehydrogenase activity, and glucose flux to the TCA cycle. As such, inhibition of the NOTCH pathway or Pdp1 knockdown abrogated glucose oxidation, mtROS, and M1 gene expression. Conditional NOTCH1 deficiency in the myeloid lineage attenuated HMac M1 activation and inflammation in a murine model of alcoholic steatohepatitis and markedly reduced lethality following endotoxin-mediated fulminant hepatitis in mice. In vivo monocyte tracking further demonstrated the requirement of NOTCH1 for the migration of blood monocytes into the liver and subsequent M1 differentiation. Together, these results reveal that NOTCH1 promotes reprogramming of mitochondrial metabolism for M1 macrophage activation.


Subject(s)
Inflammation/immunology , Macrophage Activation/physiology , Mitochondria/metabolism , Receptor, Notch1/physiology , Signal Transduction/physiology , Animals , Cell Line , Electron Transport/genetics , Endotoxemia/complications , Fatty Liver, Alcoholic/immunology , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Feedback, Physiological , Gene Expression Regulation , Glucose/metabolism , Inflammation/metabolism , Liver Failure, Acute/etiology , Liver Failure, Acute/immunology , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Macrophage Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Nitric Oxide/metabolism , Oxidative Phosphorylation , Protein Structure, Tertiary , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/antagonists & inhibitors , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/genetics , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Reactive Oxygen Species/metabolism , Receptor, Notch1/deficiency , Transcription, Genetic , Up-Regulation
4.
Endocrinology ; 154(1): 102-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23183174

ABSTRACT

Enhanced de novo lipogenesis (DNL), an adult hepatic adaption, is seen with high carbohydrate or low-fat diets. We hypothesized that ad libitum intake after prenatal calorie restriction will result in adult-onset glucose intolerance and enhanced DNL with modified lipid metabolic gene expression profile. Stable isotopes were used in 15-month-old adult male rat offspring exposed to prenatal (IUGR), pre- and postnatal (IPGR), or postnatal (PNGR) caloric restriction vs. controls (CON). IUGR vs. CON were heavier with hepatomegaly but unchanged visceral white adipose tissue (WAT), glucose intolerant with reduced glucose-stimulated insulin secretion (GSIS), pancreatic ß-cell mass, and total glucose clearance rate but unsuppressed hepatic glucose production. Liver glucose transporter (Glut) 1 and DNL increased with decreased hepatic acetyl-CoA carboxylase (ACC) and fatty acid synthase but increased WAT fatty acid transport protein-1 and peroxisomal proliferator-activated receptor-γ, resistin, and visfatin gene expression. In contrast, PNGR and IPGR were lighter, had reduced visceral WAT, and were glucose tolerant with unchanged hepatic glucose production but with increased GSIS, ß-cell mass, glucose clearance rate, and WAT insulin receptor. Hepatic Glut1 and DNL were also increased in lean IPGR and PNGR with increased hepatic ACC, phosphorylated ACC, and pAMPK and reduced WAT fatty acid transport protein-1, peroxisomal proliferator-activated receptor-γ, and ACCα. We conclude the following: 1) the heavy, glucose-intolerant and insulin-resistant IUGR adult phenotype is ameliorated by postnatal caloric restriction; 2) increased DNL paralleling hepatic Glut1 is a biomarker of exposure to early caloric restriction rather than the adult metabolic status; 3) hepatic lipid enzyme expression reflects GSIS rather than DNL; and 4) WAT gene expression reflects an obesogenic vs. lean phenotype.


Subject(s)
Caloric Restriction , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose/metabolism , Lipid Metabolism/physiology , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adipose Tissue, White/metabolism , Animals , Animals, Newborn , Blood Glucose/metabolism , Blotting, Western , Chromatography, Gas , Drinking/genetics , Drinking/physiology , Eating/genetics , Eating/physiology , Female , Insulin/metabolism , Male , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
5.
PPAR Res ; 2012: 604216, 2012.
Article in English | MEDLINE | ID: mdl-22829803

ABSTRACT

Prenatally administered rosiglitazone (RGZ) is effective in enhancing lung maturity; however, its long-term safety remains unknown. This study aimed to determine the effects of prenatally administered RGZ on the metabolic phenotype of adult rats. Methods. Pregnant Sprague-Dawley rat dams were administered either placebo or RGZ at embryonic days 18 and 19. Between 12 and 20 weeks of age, the rats underwent glucose and insulin tolerance tests and de novo fatty acid synthesis assays. The lungs, liver, skeletal muscle, and fat tissue were processed by Western hybridization for peroxisome proliferator-activated receptor (PPAR)γ, adipose differentiation-related protein (ADRP), and surfactant proteins B (SPB) and C (SPC). Plasma was assayed for triglycerides, cholesterol, insulin, glucagon, and troponin-I levels. Lungs were also morphometrically analyzed. Results. Insulin and glucose challenges, de novo fatty acid synthesis, and all serum assays revealed no differences among all groups. Western hybridization for PPARγ, ADRP, SPB, and SPC in lung, liver, muscle, and fat tissue showed equal levels. Histologic analyses showed a similar number of alveoli and septal thickness in all experimental groups. Conclusions. When administered prenatally, RGZ does not affect long-term fetal programming and may be safe for enhancing fetal lung maturation.

6.
Endocr Pract ; 18(3): e52-6, 2012.
Article in English | MEDLINE | ID: mdl-22548943

ABSTRACT

OBJECTIVE: To describe the uncommon presentation of hyperinsulinism in an 8-year-old boy. METHODS: We describe the patient's clinical findings, results from biochemical and imaging studies, surgical approach, and outcome. The discussion encompasses a review of literature that provided the basis for the diagnostic and surgical approach applied to this patient's case. RESULTS: An obese 8.5-year-old boy initially presented with hypoglycemic seizures after initiation of dietary changes to treat obesity. Biochemical analysis indicated hyperinsulinism. Endoscopic ultrasonography showed no pancreatic lesions suggestive of insulinoma. Genetic studies identified no known mutations in the ABCC8, KCNJ11, GCK, or GLUD1 genes. Selective arterial calcium stimulation and hepatic venous sampling did not document a focal source for hyperinsulinism in the pancreas, and positron emission tomography with 18-fluoro-L-3,4-dihydroxyphenylalanine showed diffusely increased uptake in the pancreas. The patient ultimately required partial pancreatectomy because of continued hypoglycemia while taking diazoxide and octreotide. Intraoperative glucose monitoring directed the extent of surgical resection. A 45% pancreatectomy was performed, which resolved the hypoglycemia but led to impaired glucose tolerance after surgery. CONCLUSION: The unusual presentation of hyperinsulinism in childhood required a personalized approach to diagnosis and surgical management using intraoperative glucose monitoring that resulted in a conservative pancreatectomy.


Subject(s)
Hyperinsulinism/etiology , Insulinoma/diagnosis , Insulinoma/surgery , Organ Sparing Treatments , Pancreatectomy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/surgery , Child , Diagnosis, Differential , Diet, Reducing/adverse effects , Glucose Intolerance/etiology , Humans , Hyperinsulinism/physiopathology , Hyperinsulinism/prevention & control , Hypoglycemia/etiology , Hypoglycemia/physiopathology , Hypoglycemia/prevention & control , Insulinoma/complications , Insulinoma/physiopathology , Male , Obesity/complications , Obesity/diet therapy , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/physiopathology , Seizures/etiology , Seizures/prevention & control , Treatment Outcome
7.
Biochem J ; 443(3): 829-39, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22360721

ABSTRACT

The importance of PDHK (pyruvate dehydrogenase kinase) 2 and 4 in regulation of the PDH complex (pyruvate dehydrogenase complex) was assessed in single- and double-knockout mice. PDHK2 deficiency caused higher PDH complex activity and lower blood glucose levels in the fed, but not the fasted, state. PDHK4 deficiency caused similar effects, but only after fasting. Double deficiency intensified these effects in both the fed and fasted states. PDHK2 deficiency had no effect on glucose tolerance, PDHK4 deficiency produced only a modest effect, but double deficiency caused a marked improvement and also induced lower insulin levels and increased insulin sensitivity. In spite of these beneficial effects, the double-knockout mice were more sensitive than wild-type and single-knockout mice to long-term fasting, succumbing to hypoglycaemia, ketoacidosis and hypothermia. Stable isotope flux analysis indicated that hypoglycaemia was due to a reduced rate of gluconeogenesis and that slightly more glucose was converted into ketone bodies in the double-knockout mice. The findings establish that PDHK2 is more important in the fed state, PDHK4 is more important in the fasted state, and survival during long-term fasting depends upon regulation of the PDH complex by both PDHK2 and PDHK4.


Subject(s)
Fasting , Hypothermia/etiology , Isoenzymes/metabolism , Ketosis/etiology , Protein Serine-Threonine Kinases/metabolism , Animals , Gas Chromatography-Mass Spectrometry , Isoenzymes/genetics , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
8.
Neonatology ; 101(3): 217-24, 2012.
Article in English | MEDLINE | ID: mdl-22076469

ABSTRACT

BACKGROUND: Rosiglitazone (RGZ), a peroxisome proliferator-activated receptor-γ (PPARγ) agonist, significantly enhances lung maturation without affecting blood biochemical and metabolic profiles in the newborn period. However, whether this exposure to RGZ in neonatal life alters the adult metabolic phenotype is not known. OBJECTIVE: To determine the effects of early postnatal administration of RGZ on the young adult metabolic phenotype. METHODS: Newborn rat pups were administered either saline or RGZ for the first 7 days of life. At 11-14 weeks, glucose and insulin tolerance tests and deuterium labeling were performed. Blood and tissues were analyzed for various metabolic parameters. RESULTS: Overall, there was no effect of early postnatal RGZ administration on young adult body weight, glucose and insulin tolerance, plasma cholesterol and triglyceride profiles, insulin, glucagon, cardiac troponin, fatty acid synthesis, or tissue adipogenic differentiation. CONCLUSIONS: Treatment with RGZ in early neonatal life does not alter later developmental metabolic programming or lead to an altered metabolic phenotype in the young adult, further re-enforcing the safety of PPARγ agonists as a novel lung-protective strategy.


Subject(s)
Hypoglycemic Agents/pharmacology , Lung/drug effects , Metabolic Networks and Pathways/drug effects , Thiazolidinediones/pharmacology , Animals , Animals, Newborn , Biomarkers/metabolism , Clinical Chemistry Tests , Disease Models, Animal , Glucose Tolerance Test , Insulin/blood , Insulin Resistance , Lung/metabolism , Lung/pathology , PPAR gamma/agonists , Phenotype , Rats , Rosiglitazone
9.
J Lipid Res ; 52(12): 2226-2233, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21957203

ABSTRACT

Cardiolipin (CL) is a unique phospholipid (PL) found in the mitochondria of mammalian cells. CL remodeling is accompanied by turnover of its fatty acid acyl groups. Abnormalities in CL remodeling have been found in Barth's syndrome, diabetes, and obesity. The objective of this study was to determine nonessential fatty acid turnover in CL and phosphatidylethanolamine (PE) in the rat heart in vivo. Sprague-Dawley rats were fed either a regular chow or a high-fat diet for 15 weeks, and consumed 6% deuterium-enriched drinking water as a tracer for 14 days. CL and PE were extracted from cardiac tissue and isolated by TLC. Fatty acids from CL, PE, and plasma were analyzed by GC/MS for deuterium incorporation. Results showed oleate and vaccenate turnover were the highest in CL whereas palmitate and stearate turnover were low. Among the nonessential fatty acids in PE, turnover of stearate and vaccenate were the highest. The high turnover rate in vaccenate was unexpected, because vaccenate previously had no known metabolic or physiologic function. In conclusion, the similarly high turnover rates of both oleate and vaccenate readily suggest that remodeling is an important functional aspect of PL metabolism in CL.


Subject(s)
Cardiolipins/chemistry , Cardiolipins/metabolism , Fatty Acids, Nonesterified/metabolism , Myocardium/metabolism , Acetates/metabolism , Animals , Deuterium/chemistry , Diet, High-Fat , Gene Expression Regulation, Enzymologic , Kinetics , Male , Phosphatidylethanolamines/metabolism , Rats , Rats, Sprague-Dawley
10.
Clin Biochem ; 43(1-2): 198-207, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19747474

ABSTRACT

OBJECTIVE: The impact of increased fructose consumption on carbohydrate metabolism is a topic of current interest, but determination of serum level has been hindered due to low concentration and interference from serum glucose. We are reporting a method for the quantification of glucose and fructose in clinical samples using gas chromatography/mass spectrometry (GC/MS). The accuracy and precision of GC/MS and an enzymatic assay were compared. DESIGN AND METHODS: Mass spectrometry fragmentation patterns of methyloxime peracetate derivatized aldose and ketose were determined. Unique fragments for glucose and fructose were used for quantitative analysis using isotope labeled recovery standards. RESULTS: Methyloxime peracetate derivatives of glucose and fructose showed characteristic loss of acetate (M-60) or ketene (M-42) under chemical ionization (CI). Under electron impact (EI) ionization, a unique C1-C2 fragment of glucose was formed, while a C1-C3 fragment was formed from keto-hexoses. These unique fragments were used in the quantitative assay of glucose and fructose in clinical samples. In clinical samples, the GC/MS assay has a lower limit of detection than that of the enzymatic assay. In plasma samples from patients evaluated for diabetes the average serum glucose and fructose were 6.19+/-2.72 mM and 46+/- 25.22 microM. Fructose concentrations in many of these samples were below the limit of detection of the enzymatic method. CONCLUSION: Derivatization of aldose and ketose monosaccharides to their respective O-methyloxime acetates for GC/MS analysis is a facile method for determination of serum/plasma glucose and fructose samples.


Subject(s)
Carbohydrate Metabolism , Fructose/blood , Gas Chromatography-Mass Spectrometry/methods , Glucose/metabolism , Dietary Carbohydrates/metabolism , Fructose/chemistry , Gas Chromatography-Mass Spectrometry/instrumentation , Glucose/chemistry , Humans
11.
J Cereb Blood Flow Metab ; 27(9): 1593-602, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17293841

ABSTRACT

Patients with traumatic brain injury (TBI) routinely exhibit cerebral glucose uptake in excess of that expected by the low levels of oxygen consumption and lactate production. This brings into question the metabolic fate of glucose. Prior studies have shown increased flux through the pentose phosphate cycle (PPC) during cellular stress. This study assessed the PPC after TBI in humans. [1,2-(13)C(2)]glucose was infused for 60 mins in six consented, severe-TBI patients (GCS<9) and six control subjects. Arterial and jugular bulb blood sampled during infusion was analyzed for (13)C-labeled isotopomers of lactate by gas chromatography/mass spectroscopy. The product of lactate concentration and fractional abundance of isotopomers was used to determine blood concentration of each isotopomer. The difference of jugular and arterial concentrations determined cerebral contribution. The formula PPC=(m1/m2)/(3+(m1/m2)) was used to calculate PPC flux relative to glycolysis. There was enrichment of [1,2-(13)C(2)]glucose in arterial-venous blood (enrichment averaged 16.6% in TBI subjects and 28.2% in controls) and incorporation of (13)C-label into lactate, showing metabolism of labeled substrate. The PPC was increased in TBI patients relative to controls (19.6 versus 6.9%, respectively; P=0.002) and was excellent for distinguishing the groups (AUC=0.944, P<0.0001). No correlations were found between PPC and other clinical parameters, although PPC was highest in patients studied within 48 h of injury (averaging 33% versus 13% in others; P=0.0006). This elevation in the PPC in the acute period after severe TBI likely represents a shunting of substrate into alternative biochemical pathways that may be critical for preventing secondary injury and initiating recovery.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Glucose/metabolism , Pentose Phosphate Pathway/physiology , Adolescent , Adult , Aged , Blood Glucose/analysis , Carbon Radioisotopes , Cerebrovascular Circulation/physiology , Female , Humans , Male , Middle Aged
12.
Diabetes ; 55(12): 3372-80, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17130482

ABSTRACT

Despite altered regulation of insulin signaling, Pten(+/-) heterodeficient standard diet-fed mice, approximately 4 months old, exhibit normal fasting glucose and insulin levels. We report here a stable isotope flux phenotyping study of this "silent" phenotype, in which tissue-specific insulin effects in whole-body Pten(+/-)-deficient mice were dissected in vivo. Flux phenotyping showed gain of function in Pten(+/-) mice, seen as increased peripheral glucose disposal, and compensation by a metabolic feedback mechanism that 1) decreases hepatic glucose recycling via suppression of glucokinase expression in the basal state to preserve hepatic glucose production and 2) increases hepatic responsiveness in the fasted-to-fed transition. In Pten(+/-) mice, hepatic gene expression of glucokinase was 10-fold less than wild-type (Pten(+/+)) mice in the fasted state and reached Pten(+/+) values in the fed state. Glucose-6-phosphatase expression was the same for Pten(+/-) and Pten(+/+) mice in the fasted state, and its expression for Pten(+/-) was 25% of Pten(+/+) in the fed state. This study demonstrates how intra- and interorgan flux compensations can preserve glucose homeostasis (despite a specific gene defect that accelerates glucose disposal) and how flux phenotyping can dissect these tissue-specific flux compensations in mice presenting with a "silent" phenotype.


Subject(s)
Liver/physiology , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Eating , Fasting , Gene Expression Regulation, Enzymologic , Glucokinase/genetics , Glucose Tolerance Test , Glucose-6-Phosphatase/genetics , Insulin/pharmacology , Lipolysis , Mice
13.
Diabetes ; 55(12): 3429-38, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17130489

ABSTRACT

Fatty liver is a common feature of both obesity and lipodystrophy, reflecting compromised adipose tissue function. The lipin-deficient fatty liver dystrophy (fld) mouse is an exception, as there is lipodystrophy without a fatty liver. Using a combination of indirect calorimetry and stable-isotope flux phenotyping, we determined that fld mice exhibit abnormal fuel utilization throughout the diurnal cycle, with increased glucose oxidation near the end of the fasting period and increased fatty acid oxidation during the feeding period. The mechanisms underlying these alterations include a twofold increase compared with wild-type mice in tissue glycogen storage during the fed state, a 40% reduction in hepatic glucose production in the fasted state, and a 27-fold increase in de novo fatty acid synthesis in liver during the fed state. Thus, the inability to store energy in adipose tissue in the fld mouse leads to a compensatory increase in glycogen storage for use during the fasting period and reliance upon hepatic fatty acid synthesis to provide fuel for peripheral tissues during the fed state. The increase in hepatic fatty acid synthesis and peripheral utilization provides a potential mechanism to ameliorate fatty liver in the fld that would otherwise occur as a consequence of adipose tissue dysfunction.


Subject(s)
Circadian Rhythm , Energy Metabolism , Nuclear Proteins/deficiency , Animals , Base Sequence , Calorimetry, Indirect , DNA Primers , Fatty Acid Synthases/metabolism , Fatty Liver/genetics , Glycogen/metabolism , Liver/enzymology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Proteins/genetics , Phosphatidate Phosphatase
14.
Am J Physiol Endocrinol Metab ; 290(6): E1218-26, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16449299

ABSTRACT

We studied glucose metabolic adaptations in the intrauterine growth-restricted (IUGR) rat offspring to decipher glucose homeostasis in metabolic programming. Glucose futile cycling (GFC), which is altered when there is imbalance between glucose production and utilization, was studied during a glucose tolerance test (GTT) in 2-day-old (n = 8), 2-mo-old (n = 22), and 15-mo-old (n = 22) female rat offspring. The IUGR rats exposed to either prenatal (CM/SP, n = 5 per age), postnatal (SM/CP, n = 6), or pre- and postnatal (SM/SP, n = 6) nutrient restriction were compared with age-matched controls (CM/CP, n = 5). At 2 days, IUGR pups (SP) were smaller and glucose intolerant and had increased hepatic glucose production and increased glucose disposal (P < 0.01) compared with controls (CP). At 2 mo, the GTT, glucose clearance, and GFC did not change. However, a decline in hepatic glucose-6-phosphatase (P < 0.05) and fructose-1,6-biphosphatase (P < 0.05) enzyme activities in the IUGR offspring was detected. At 15 mo, prenatal nutrient restriction (CM/SP) resulted in greater weight gain (P < 0.01) and hyperinsulinemia (P < 0.001) compared with postnatal nutrient restriction (SM/CP). A decline in GFC in the face of a normal GTT occurred in both the prenatal (CM/SP, P < 0.01) and postnatal calorie (SM/CP, P < 0.03) and growth-restricted offspring. The IUGR offspring with pre- and postnatal nutrient restriction (SM/SP) were smaller, hypoinsulinemic (P < 0.03), and hypoleptinemic (P < 0.03), with no change in GTT, hepatic glucose production, GFC, or glucose clearance. We conclude that there is pre- and postnatal programming that affects the postnatal compensatory adaptation of GFC and disposal initiated by changes in circulating insulin concentrations, thereby determining hepatic insulin sensitivity in a phenotype-specific manner.


Subject(s)
Caloric Restriction/adverse effects , Fetal Growth Retardation/metabolism , Glucose/metabolism , Lactation/physiology , Animals , Animals, Newborn , Animals, Suckling , Body Weight , Caloric Restriction/methods , Female , Glucose-6-Phosphatase/metabolism , Homeostasis , Rats , Rats, Sprague-Dawley , Time Factors
15.
JPEN J Parenter Enteral Nutr ; 29(2): 81-6, 2005.
Article in English | MEDLINE | ID: mdl-15772384

ABSTRACT

BACKGROUND: Low-birth-weight (LBW) infants have high energy requirements and are dependent on high fat intake to maintain adequate postnatal growth. Fat energy is transported in plasma as triglycerides, which are either derived from the diet or from de novo lipogenesis (DNL). It is our hypothesis that DNL plays an important physiologic role in adapting to exclusive breast milk (EBM) feeding or to parenteral nutrition (PN). METHODS: We studied hepatic de novo lipogenesis in 14 LBW (<34-week gestation) appropriate for gestational age and receiving either EBM feedings or full PN support. Stable isotope tracer [2-(13)C] acetate was administered for 72 hours to achieve an estimated 10% enrichment of daily fat intake. Fatty acids were extracted from plasma for gas chromatography-mass spectrometry analyses. RESULTS: Percent new synthesis of palmitate was 13.1% +/- 2.5% in the EBM group and 14.9% +/- 0.7% in the PN group (NS), stearate was 11.1% +/- 2.7% in the EBM group and 10.6% +/- 14% in the PN group (NS) and cholesterol was 12.7% +/- 2.1% in the EBM group and 17.4% +/- 4.6% in the PN group (NS) after 72 hours of tracer administration (mean +/- SEM). The plasma lipid fatty acid composition in palmitate, oleate, and stearate with intake of 3.6 +/- 0.6 g/kg/d of IV lipids (ILs) was similar to EBM-feeding infants taking 6.3 +/- 0.13 g/kg/d of fat. CONCLUSIONS: De novo lipogenesis is active in stable LBW infants maintaining standard postnatal growth. Hepatic DNL permits newborn infants to meet the fat energy needs of peripheral tissues for growth and storage and to maintain plasma fatty acid composition in adaptation to different dietary fat intake.


Subject(s)
Breast Feeding , Infant, Low Birth Weight/metabolism , Infant, Low Birth Weight/physiology , Lipids/biosynthesis , Liver/metabolism , Parenteral Nutrition , Adaptation, Physiological , Birth Weight , Carbon Isotopes , Dietary Fats/administration & dosage , Energy Metabolism/physiology , Fatty Acids/analysis , Female , Gas Chromatography-Mass Spectrometry , Gestational Age , Humans , Infant, Low Birth Weight/growth & development , Infant, Newborn , Lipid Metabolism , Male , Nutritional Requirements , Weight Gain
16.
Biochem J ; 381(Pt 1): 287-94, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15032751

ABSTRACT

Recent studies in metabolic profiling have underscored the importance of the concept of a metabolic network of pathways with special functional characteristics that differ from those of simple reaction sequences. The characterization of metabolic functions requires the simultaneous measurement of substrate fluxes of interconnecting pathways. Here we present a novel stable isotope method by which the forward and reverse fluxes of the futile cycles of the hepatic glucose metabolic network are simultaneously determined. Unlike previous radio-isotope methods, a single tracer [1,2-13C2]D-glucose and mass isotopomer analysis is used. Changes in fluxes of substrate cycles, in response to several gluconeogenic substrates, in isolated fasted hepatocytes from male Wistar rats were measured simultaneously. Incubation with these substrates resulted in a change in glucose-6-phosphatase/glucokinase and glycolytic/gluconeogenic flux ratios. Different net redistributions of intermediates in the glucose network were observed, resulting in distinct metabolic phenotypes of the fasted hepatocytes in response to each substrate condition. Our experimental observations show that the constraints of concentrations of shared intermediates, and enzyme kinetics of intersecting pathways of the metabolic network determine substrate redistribution throughout the network when it is perturbed. These results support the systems-biology notion that network analysis provides an integrated view of the physiological state. Interaction between metabolic intermediates and glycolytic/gluconeogenic pathways is a basic element of cross-talk in hepatocytes, and may explain some of the difficulties in genotype and phenotype correlation.


Subject(s)
Fasting/physiology , Glucose/metabolism , Hepatocytes/chemistry , Hepatocytes/metabolism , Animals , Carbon Isotopes/metabolism , Citric Acid Cycle/genetics , Citric Acid Cycle/physiology , Gas Chromatography-Mass Spectrometry/methods , Gas Chromatography-Mass Spectrometry/statistics & numerical data , Genotype , Glucokinase/metabolism , Gluconeogenesis/genetics , Gluconeogenesis/physiology , Glucose/chemistry , Glucose-6-Phosphatase/metabolism , Glycogen/biosynthesis , Glycogen/chemistry , Glycolysis/genetics , Glycolysis/physiology , Hepatocytes/enzymology , Lactic Acid/biosynthesis , Lactic Acid/chemistry , Male , Phenotype , Rats , Rats, Wistar , Substrate Cycling/genetics , Substrate Cycling/physiology
17.
Endocrinology ; 145(3): 1087-95, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14670991

ABSTRACT

Our previous work led to the hypothesis that peroxisomal proliferator-activated receptor alpha (PPAR alpha) modulates insulin action in a compensatory fashion for hepatic glucose balance vs. peripheral glucose disposal. Therefore, we have examined the expression of insulin-dependent gluconeogenic/glycolytic/pentose cycle enzymes and compared these to insulin responsiveness for peripheral vs. hepatic substrate flux and futile cycling in the PPAR alpha knockout mouse. Hepatic gluconeogenic flux, glucose absorption, clearance and recycling, as well as in vivo glucose disposal were evaluated using new mass isotopomer methods. Insulin-dependent gluconeogenic/glycolytic/pentose cycle enzyme expression and glucose futile cycling were diminished; however, glucose disappearance was increased. This supports the hypothesis of hepatic insulin resistance and increased peripheral glucose uptake as compensatory events secondary to the decrease in fatty acid oxidation characteristic of the PPAR alpha knockout. We conclude that 1) the loss of PPAR alpha results in lower expression levels and diminished response to meal regulation for gluconeogenic/glycolytic enzyme expression; and 2) consequently, substrate/futile cycling of glucose is decreased when PPAR alpha is absent despite increased gluconeogenesis. The compensatory changes in liver and peripheral tissue substrate flux and the resultant adaptation for enzyme expression in the liver to have a diminished insulin dependence reflect the loosely linked correlation between phenotype and genotype in hepatic glucose metabolism.


Subject(s)
Energy Metabolism/physiology , Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Blood Glucose/metabolism , Carbon/metabolism , Carbon Isotopes , Fatty Acids/metabolism , Gene Expression , Gluconeogenesis/physiology , Glycolysis/physiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pentose Phosphate Pathway/physiology
18.
Anal Biochem ; 315(2): 238-46, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12689833

ABSTRACT

Increased glucose cycling between glucose and glucose-6-phosphate is characteristic of insulin resistance and hyperglycemia seen with Type II diabetes. Traditionally, glucose cycling is determined by the difference between hepatic glucose output measured with separate [2-3H]glucose and [6-3H]glucose infusions. We demonstrate a novel method for determining hepatic glucose recycling from an intraperitoneal glucose tolerance test (IPGTT). A single tracer, [1, 2-13C(2)]glucose (a M2 glucose isotopomer), was administered at 1mg/g body weight to 4-month-old C57BL/6 mice. Hepatic glucose recycling was monitored by the appearance of a plasma M1 isotopomer of glucose, which is produced by the action of the pentose cycle on the M2 glucose isotopomer in the liver. The initial M2 enrichment was 56% and decreased to 13% at the end of 3 h, and the M1 enrichment peaked at 2 h. The ratio of plasma M1/M2 glucose increased linearly with time to approximately 25%, and the regression of the M1/M2 ratio against time gives a slope, termed the in vivo glucose-dependent futile recycling rate constant k(HR). k(HR) estimates glucose/glucose-6-phosphate futile cycling, along with glucose recycling through the pentose cycle. These observations demonstrate complex substrate cycling during an IPGTT using a single stable isotope tracer.


Subject(s)
Glucose Tolerance Test , Glucose/metabolism , Liver/metabolism , Animals , Blood Glucose/analysis , Blotting, Western , Carbon Isotopes , Gas Chromatography-Mass Spectrometry , Glucose/administration & dosage , Glycogen/metabolism , Glycolysis , Injections, Intraperitoneal , Insulin/blood , Insulin/metabolism , Isotope Labeling , Kinetics , Male , Mice , Mice, Inbred C57BL , Time Factors
19.
J Biol Chem ; 277(52): 50237-44, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12176975

ABSTRACT

The hypoglycemia seen in the fasting PPARalpha null mouse is thought to be due to impaired liver fatty acid beta-oxidation. The etiology of hypoglycemia in the PPARalpha null mouse was determined via stable isotope studies. Glucose, lactate, and glycerol flux was assessed in the fasted and fed states in 4-month-old PPARalpha null mice and in C57BL/6 WT maintained on standard chow using a new protocol for flux assessment in the fasted and fed states. Hepatic glucose production (HGP) and glucose carbon recycling were estimated using [U-(13)C(6)]glucose, and HGP, lactate, and glycerol turnover was estimated utilizing either [U-(13)C(3)]lactate or [2-(13)C]glycerol infused subcutaneously via Alza miniosmotic pumps. At the end of a 17-h fast, HGP was higher in the PPARalpha null mice than in WT by 37% (p < 0.01). However, recycling of glucose carbon from lactate back to glucose was lower in the PPARalpha null than in WT (39% versus 51%, p < 0.02). The lack of conversion of lactate to glucose was confirmed using an [U-(13)C(3)]lactate infusion. In the fasted state, HGP from lactate and lactate production were decreased by 65 and 55%, respectively (p < 0.05) in PPARalpha null mice. In contrast, when [2-(13)C]glycerol was infused, glycerol production and HGP from glycerol increased by 80 and 250%, respectively (p < 0.01), in the fasted state of PPARalpha null mice. The increased HGP from glycerol was not suppressed in the fed state. While little change was evident for phosphoenolpyruvate carboxykinase (PEPCK) expression, pyruvate kinase expression was decreased 16-fold in fasted PPARalpha null mice as compared with the wild-type control. The fasted and fed insulin levels were comparable, but blood glucose levels were lower in the PPARalpha null mice than in controls. In conclusion, PPARalpha receptor function creates a setpoint for a metabolic network that regulates the rate and route of HGP in the fasted and fed states, in part, by controlling the flux of glycerol and lactate between the triose-phosphate and pyruvate/lactate pools.


Subject(s)
Gluconeogenesis/physiology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Base Sequence , Blood Glucose/metabolism , Carbon Isotopes , DNA Primers , Fasting , Gene Expression Regulation, Enzymologic , Glucose/metabolism , Glycerol/administration & dosage , Glycerol/metabolism , Homeostasis , Insulin/blood , Lactic Acid/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Phosphoenolpyruvate Carboxykinase (GTP) , Pyruvate Kinase/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
20.
Am J Physiol Endocrinol Metab ; 282(3): E507-13, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11832351

ABSTRACT

De novo lipogenesis and dietary fat uptake are two major sources of fatty acid deposits in fat of obese animals. To determine the relative contribution of fatty acids from these two sources in obesity, we have determined the distribution of c16 and c18 fatty acids of triglycerides in plasma, liver, and epididymal fat pad of Zucker diabetic fatty (ZDF) rats and their lean littermates (ZL) under two isocaloric dietary fat conditions. Lipogenesis was also determined using the deuterated water method. Conversion of palmitate to stearate and stearate to oleate was calculated from the deuterium incorporation by use of the tracer dilution principle. In the ZL rat, lipogenesis was suppressed from 70 to 24%, conversion of palmitate to stearate from 86 to 78%, and conversion of stearate to oleate from 56 to 7% in response to an increase in the dietary fat-to-carbohydrate ratio. The results suggest that suppression of fatty acid synthase and stearoyl-CoA desaturase activities is a normal adaptive mechanism to a high-fat diet. In contrast, de novo lipogenesis, chain elongation, and desaturation were not suppressed by dietary fat in the ZDF rat. The lack of ability to adapt to a high-fat diet resulted in a higher plasma triglyceride concentration and excessive fat accumulation from both diet and de novo synthesis in the ZDF rat.


Subject(s)
Diabetes Mellitus/metabolism , Lipids/biosynthesis , Oleic Acid/biosynthesis , Stearic Acids/metabolism , Adipose Tissue/metabolism , Animals , Deuterium , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Epididymis , Fatty Acid Synthases/metabolism , Fatty Acids/blood , Fatty Acids/metabolism , Liver/metabolism , Male , Obesity , Palmitic Acid/metabolism , Rats , Rats, Zucker , Stearoyl-CoA Desaturase/metabolism , Triglycerides/analysis , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...