Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
PLoS One ; 13(11): e0206321, 2018.
Article in English | MEDLINE | ID: mdl-30439964

ABSTRACT

Stroke evolution is a highly dynamic but variable disease which makes clinical decision making difficult. Biomarker discovery programs intended to aid clinical decision making have however largely ignored the rapidity of stroke evolution. We have used gene array technology to determine blood mRNA expression changes over the first day after stroke in rats. Blood samples were collected from 8 male spontaneously hypertensive rats at 0, 1, 2, 3, 6 and 24h post stroke induction by middle cerebral artery occlusion. RNA was extracted from whole blood stabilized in PAXgene tubes and mRNA expression was detected by oligonucleotide Affymetrix microarray. Using a pairwise comparison model, 1932 genes were identified to vary significantly over time (p≤0.5x10(-7)) within 24h after stroke. Some of the top20 most changed genes are already known to be relevant to the ischemic stroke physiopathology (e.g. Il-1R, Nos2, Prok2). Cluster analysis showed multiple stereotyped and time dependent profiles of gene expression. Direction and rate of change of expression for some profiles varied dramatically during these 24h. Profiles with potential clinical utility including hyper acute or acute transient upregulation (with expression peaking from 2 to 6h after stroke and normalisation by 24h) were identified. We found that blood gene expression varies rapidly and stereotypically after stroke in rats. Previous researchers have often missed the optimum time for biomarker measurement. Temporally overlapping profiles have the potential to provide a biological "stroke clock" able to tell the clinician how far an individual stroke has evolved.


Subject(s)
Gene Expression Profiling , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/genetics , Animals , Biomarkers/blood , Infarction, Middle Cerebral Artery/surgery , RNA, Messenger/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Time Factors
2.
Mol Metab ; 5(3): 221-232, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26977394

ABSTRACT

OBJECTIVE: Muscle glucose storage and muscle glycogen synthase (gys1) defects have been associated with insulin resistance. As there are multiple mechanisms for insulin resistance, the specific role of glucose storage defects is not clear. The aim of this study was to examine the effects of muscle-specific gys1 deletion on glucose metabolism and exercise capacity. METHODS: Tamoxifen inducible and muscle specific gys-1 KO mice were generated using the Cre/loxP system. Mice were subjected to glucose tolerance tests, euglycemic/hyperinsulinemic clamps and exercise tests. RESULTS: gys1-KO mice showed ≥85% reduction in muscle gys1 mRNA and protein concentrations, 70% reduction in muscle glycogen levels, postprandial hyperglycaemia and hyperinsulinaemia and impaired glucose tolerance. Under insulin-stimulated conditions, gys1-KO mice displayed reduced glucose turnover and muscle glucose uptake, indicative of peripheral insulin resistance, as well as increased plasma and muscle lactate levels and reductions in muscle hexokinase II levels. gys1-KO mice also exhibited markedly reduced exercise and endurance capacity. CONCLUSIONS: Thus, muscle-specific gys1 deletion in adult mice results in glucose intolerance due to insulin resistance and reduced muscle glucose uptake as well as impaired exercise and endurance capacity. IN BRIEF: This study demonstrates why the body prioritises muscle glycogen storage over liver glycogen storage despite the critical role of the liver in supplying glucose to the brain in the fasting state and shows that glycogen deficiency results in impaired glucose metabolism and reduced exercise capacity.

3.
Int J Stroke ; 10(4): 636-44, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24206623

ABSTRACT

RATIONALE: Stroke and poststroke depression are common and have a profound and ongoing impact on an individual's quality of life. However, reliable biological correlates of poststroke depression and functional outcome have not been well established in humans. AIMS: Our aim is to identify biological factors, molecular and imaging, associated with poststroke depression and recovery that may be used to guide more targeted interventions. DESIGN: In a longitudinal cohort study of 200 stroke survivors, the START-STroke imAging pRevention and Treatment cohort, we will examine the relationship between gene expression, regulator proteins, depression, and functional outcome. Stroke survivors will be investigated at baseline, 24 h, three-days, three-months, and 12 months poststroke for blood-based biological associates and at days 3-7, three-months, and 12 months for depression and functional outcomes. A sub-group (n = 100), the PrePARE: Prediction and Prevention to Achieve optimal Recovery Endpoints after stroke cohort, will also be investigated for functional and structural changes in putative depression-related brain networks and for additional cognition and activity participation outcomes. Stroke severity, diet, and lifestyle factors that may influence depression will be monitored. The impact of depression on stroke outcomes and participation in previous life activities will be quantified. STUDY OUTCOMES: Clinical significance lies in the identification of biological factors associated with functional outcome to guide prevention and inform personalized and targeted treatments. Evidence of associations between depression, gene expression and regulator proteins, functional and structural brain changes, lifestyle and functional outcome will provide new insights for mechanism-based models of poststroke depression.


Subject(s)
Depressive Disorder/therapy , Stroke/psychology , Stroke/therapy , Brain/pathology , Depressive Disorder/etiology , Depressive Disorder/metabolism , Depressive Disorder/pathology , Diet , Gene Expression , Humans , Life Style , Longitudinal Studies , Prospective Studies , Psychiatric Status Rating Scales , Severity of Illness Index , Stroke/complications , Stroke/metabolism , Stroke/pathology , Time Factors , Treatment Outcome
4.
Diabetes ; 61(5): 1122-32, 2012 May.
Article in English | MEDLINE | ID: mdl-22517657

ABSTRACT

Liver fructose-1,6-bisphosphatase (FBPase) is a regulatory enzyme in gluconeogenesis that is elevated by obesity and dietary fat intake. Whether FBPase functions only to regulate glucose or has other metabolic consequences is not clear; therefore, the aim of this study was to determine the importance of liver FBPase in body weight regulation. To this end we performed comprehensive physiologic and biochemical assessments of energy balance in liver-specific transgenic FBPase mice and negative control littermates of both sexes. In addition, hepatic branch vagotomies and pharmacologic inhibition studies were performed to confirm the role of FBPase. Compared with negative littermates, liver-specific FBPase transgenic mice had 50% less adiposity and ate 15% less food but did not have altered energy expenditure. The reduced food consumption was associated with increased circulating leptin and cholecystokinin, elevated fatty acid oxidation, and 3-ß-hydroxybutyrate ketone levels, and reduced appetite-stimulating neuropeptides, neuropeptide Y and Agouti-related peptide. Hepatic branch vagotomy and direct pharmacologic inhibition of FBPase in transgenic mice both returned food intake and body weight to the negative littermates. This is the first study to identify liver FBPase as a previously unknown regulator of appetite and adiposity and describes a novel process by which the liver participates in body weight regulation.


Subject(s)
Adiposity/physiology , Appetite/physiology , Fructose-Bisphosphatase/metabolism , Liver/enzymology , Adiposity/genetics , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Appetite/genetics , Cholecystokinin/metabolism , Dose-Response Relationship, Drug , Eating , Energy Metabolism , Fatty Acids/metabolism , Female , Fructose-Bisphosphatase/antagonists & inhibitors , Fructose-Bisphosphatase/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Hydroxybutyrates , Ketone Bodies , Leptin/metabolism , Male , Mice , Mice, Transgenic , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Oxidation-Reduction , Peptide Fragments/genetics , Peptide Fragments/metabolism
5.
Am J Physiol Endocrinol Metab ; 295(5): E1132-41, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18780768

ABSTRACT

Increased endogenous glucose production (EGP) predominantly from the liver is a characteristic feature of type 2 diabetes, which positively correlates with fasting hyperglycemia. Gluconeogenesis is the biochemical pathway shown to significantly contribute to increased EGP in diabetes. Fructose-1,6-bisphosphatase (FBPase) is a regulated enzyme in gluconeogenesis that is increased in animal models of obesity and insulin resistance. However, whether a specific increase in liver FBPase can result in increased EGP has not been shown. The objective of this study was to determine the role of upregulated liver FBPase in glucose homeostasis. To achieve this goal, we generated human liver FBPase transgenic mice under the control of the transthyretin promoter, using insulator sequences to flank the transgene and protect it from site-of-integration effects. This resulted in a liver-specific model, as transgene expression was not detected in other tissues. Mice were studied under the following conditions: 1) at two ages (24 wk and 1 yr old), 2) after a 60% high-fat diet, and 3) when bred to homozygosity. Hemizygous transgenic mice had an approximately threefold increase in total liver FBPase mRNA with concomitant increases in FBPase protein and enzyme activity levels. After high-fat feeding, hemizygous transgenics were glucose intolerant compared with negative littermates (P < 0.02). Furthermore, when bred to homozygosity, chow-fed transgenic mice showed a 5.5-fold increase in liver FBPase levels and were glucose intolerant compared with negative littermates, with a significantly higher rate of EGP (P < 0.006). This is the first study to show that FBPase regulates EGP and whole body glucose homeostasis in a liver-specific transgenic model. Our homozygous transgenic model may be useful for testing human FBPase inhibitor compounds with the potential to treat patients with type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Fructose-Bisphosphatase/metabolism , Glucose/metabolism , Liver/metabolism , Animals , Body Weight/drug effects , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Eating/drug effects , Fructose-Bisphosphatase/genetics , Gene Expression , Glucose Intolerance , Glucose-6-Phosphatase/metabolism , Homozygote , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , Insulin/blood , Insulin Resistance , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Pyruvic Acid/metabolism
6.
Diabetes ; 57(7): 1887-95, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18375435

ABSTRACT

OBJECTIVE: Fructose-1,6-bisphosphatase (FBPase) is a gluconeogenic enzyme that is upregulated in islets or pancreatic beta-cell lines exposed to high fat. However, whether specific beta-cell upregulation of FBPase can impair insulin secretory function is not known. The objective of this study therefore is to determine whether a specific increase in islet beta-cell FBPase can result in reduced glucose-mediated insulin secretion. RESEARCH DESIGN AND METHODS: To test this hypothesis, we have generated three transgenic mouse lines overexpressing the human FBPase (huFBPase) gene specifically in pancreatic islet beta-cells. In addition, to investigate the biochemical mechanism by which elevated FBPase affects insulin secretion, we made two pancreatic beta-cell lines (MIN6) stably overexpressing huFBPase. RESULTS: FBPase transgenic mice showed reduced insulin secretion in response to an intravenous glucose bolus. Compared with the untransfected parental MIN6, FBPase-overexpressing cells showed a decreased cell proliferation rate and significantly depressed glucose-induced insulin secretion. These defects were associated with a decrease in the rate of glucose utilization, resulting in reduced cellular ATP levels. CONCLUSIONS: Taken together, these results suggest that upregulation of FBPase in pancreatic islet beta-cells, as occurs in states of lipid oversupply and type 2 diabetes, contributes to insulin secretory dysfunction.


Subject(s)
Fructose-Bisphosphatase/genetics , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Enhancer Elements, Genetic , Fatty Acids/pharmacology , Fructose-Bisphosphatase/metabolism , Gene Expression Regulation, Enzymologic , Humans , Insulin/genetics , Insulin Resistance , Insulin Secretion , Mice , Mice, Transgenic , Polymerase Chain Reaction , Promoter Regions, Genetic , Rats , Tissue Donors
7.
Lipids Health Dis ; 5: 29, 2006 Dec 17.
Article in English | MEDLINE | ID: mdl-17173705

ABSTRACT

Severe combined hyperlipidaemia has occasionally been associated with infiltration of tissues in addition to arteries and the skin. We report a woman with Type 2 diabetes mellitus (DM) and severe combined hyperlipidaemia who developed retinal lipid infiltration, resulting in blindness. A 61-year-old woman with a 15-year history of Type 2 DM was admitted following a two-week history of progressive visual loss. Examination identified lipid infiltration into the retina. Phenotypically she had severe combined hyperlipidaemia with elevated IDL cholesterol and a broad beta band on lipoprotein electrophoresis, raising the possibility of familial dysbetalipoproteinaemia. However, gene sequencing analysis indicated that the patient was homozygous for the E3/E3 allele of the ApoE gene with no mutations detected in either the coding region or intron-exon boundaries. Her lipid profile improved following dietary therapy and gemfibrozil treatment, but this had little effect on either her fundal appearances or her visual acuity. Type 2 DM plays a vital role both in allowing expression of severe combined hyperlipoproteinaemia, in addition to serving as a risk factor for complications such as tissue infiltration.


Subject(s)
Diabetes Mellitus, Type 2/complications , Hyperlipidemias/complications , Lipid Metabolism , Retina/metabolism , Retina/pathology , Aged , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Female , Hemorrhage , Humans , Hyperlipidemias/blood , Hyperlipidemias/metabolism , Lipids/blood
8.
Hum Mutat ; 27(5): 483-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16619235

ABSTRACT

We have characterized an unusual family with two different androgen receptor (AR) gene deletions, in which we propose a novel mechanism of deletion formation has occurred. Affected individuals have the X-linked disorder androgen insensitivity syndrome, and we previously showed that different family members have deletions of different exons of the AR gene. We have now fully sequenced the deletions from affected individuals, and confirmed the presence of different deletions in different affected family members. Most affected and heterozygote individuals have a 4,430-bp deletion of exon 5 that occurred between repeated GTGGCAT motifs in introns 4 and 5. One affected hemizygous individual has a 4,033-bp deletion of exons 6 and 7 that occurred between repeated CCTC motifs in introns 5 and 7. The intron 5 breakpoint junctions of the two deletions are only 11 bp apart. Surprisingly, the maternal grandmother of the original index case was found to be mosaic for both deletional events, as well as having the normal AR gene. Karyotyping ruled out 47,XXX trisomy, indicating triple mosaicism for the two different deleted AR alleles and a normal AR allele. This triple mosaicism must have occurred early in embryonic development, as both deletions were passed on to different children. Based on these findings, we propose a novel mechanism of deletion formation. We suggest that during AR gene replication, a double strand DNA break occurred in intron 5, and that a variant of replication slippage occurred on both newly synthesized strands between the repeat motifs of microhomology, leading to the formation of the two different AR gene deletions.


Subject(s)
DNA Repair , DNA Replication/physiology , Gene Deletion , Receptors, Androgen/genetics , Androgen-Insensitivity Syndrome/genetics , Chromosomes, Human, X , DNA Mutational Analysis , Exons , Female , Genetic Linkage , Heterozygote , Humans , Karyotyping , Male , Molecular Sequence Data , Mosaicism , Pedigree
9.
Endocrinology ; 147(6): 2764-72, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16497803

ABSTRACT

In type 2 diabetes, increased endogenous glucose production (EGP) as a result of elevated gluconeogenesis contributes to hyperglycemia. An increase in glycerol gluconeogenesis has led to the suggestion that, in obese human subjects with type 2 diabetes, there may be an increase in the activity of the gluconeogenic enzyme fructose-1,6-bisphosphatase (FBPase). The aim of this study was to generate transgenic mice that overexpress human liver FBPase in the liver and assess the consequences to whole-body and hepatic glucose metabolism. FBPase transgenic mice had significantly higher levels of transgene expression in the liver and, as a result, had increased FBPase protein and enzyme activity levels in the liver. This resulted in an increase in the rate of glycerol conversion to glucose but not in EGP. The increased expression of FBPase in the liver did not result in any significant differences compared with littermate control mice in insulin or glucose tolerance. Therefore, it appears that, on its own, an increase in FBPase does not lead to impaired regulation of EGP and hence does not affect whole-body glucose metabolism. This suggests that, for EGP to be increased, other factors associated with obesity are also required.


Subject(s)
Fructose-Bisphosphatase/physiology , Gluconeogenesis , Glycerol/metabolism , Liver/enzymology , Animals , Blood Glucose/analysis , Female , Fructose-Bisphosphatase/genetics , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/physiology
10.
J Mol Endocrinol ; 35(1): 39-48, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16087720

ABSTRACT

Type 2 diabetes is characterized by islet dysfunction resulting in hyperglycemia, which can then lead to further deterioration in islet function. A possible mechanism for hyperglycemia-induced islet dysfunction is the accumulation of advanced glycation end products (AGE). The DBA/2 mouse develops pancreatic islet dysfunction when exposed to a high glucose environment and/or obesity-induced insulin resistance. To determine the biochemical cause of dysfunction, DBA/2 and C57BL/6 control islets were incubated in 11.1 mM or 40 mM glucose in the absence or presence of the AGE inhibitor aminoguanidine (AG) for 10 days. Basal (2.8 mM glucose) insulin release was increased in both DBA/2 and C57BL/6 islets incubated with 40 mM vs 11.1 mM glucose for 10 days. Chronic exposure to hyperglycemia decreased glucose (20 mM)-stimulated insulin secretion in DBA/2 but not in C57BL/6 islets. AG significantly increased fold-induced insulin release in high glucose cultured DBA/2 mouse islets, but did not affect C57BL/6 islet function. DBA/2 islet glucokinase was significantly reduced following 40 mM glucose culture, compared with 11.1 mM glucose cultured DBA/2 islets and 40 mM glucose cultured C57BL/6 islets. Incubation of islets with AG resulted in a normalization of DBA/2 islet glucokinase levels. In conclusion, chronic high glucose-induced increases in AGE can result in islet dysfunction and this is associated with reduced glucokinase levels in a mouse model with susceptibility to islet failure.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/physiopathology , Glucokinase/metabolism , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/physiopathology , Animals , Base Sequence , Cell Survival , DNA/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Glucose/administration & dosage , Glucose Transporter Type 2 , Glycation End Products, Advanced/metabolism , Hexokinase/metabolism , In Vitro Techniques , Insulin/genetics , Insulin Secretion , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Monosaccharide Transport Proteins/metabolism , Proinsulin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL