Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Harm Reduct J ; 18(1): 121, 2021 11 27.
Article in English | MEDLINE | ID: mdl-34838030

ABSTRACT

INTRODUCTION: Several published systematic reviews have examined the potential associations between e-cigarette use and cigarette smoking, but their methodological and/or reporting quality have not yet been assessed. This systematic quality review followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and AMSTAR (A MeaSurement Tool to Assess systematic Reviews) 2 to evaluate the quality of systematic reviews investigating potential associations between e-cigarette use and cigarette smoking. MATERIALS AND METHODS: PubMed/MEDLINE, Embase, and PsycINFO were searched from 01 January 2007 to 24 June 2020. Methodological quality was assessed using AMSTAR 2, and reporting quality was assessed using PRISMA guidelines. RESULTS: Of 331 potentially relevant systematic reviews, 20 met predefined inclusion criteria. Most reviews (n = 15; 75%) reported on e-cigarette use and cigarette smoking cessation, while three reported on e-cigarette use and cigarette smoking initiation (15%); and two reported on cigarette smoking initiation and cessation (10%). According to AMSTAR 2 guidelines, 18 of the 20 reviews (90%) were "critically low" in overall confidence of the results, while two were ranked "low." Additionally, reporting quality varied across the reviews, with only 60% reporting at least half of the PRISMA items. DISCUSSION: Methodological limitations were identified across reviews examining potential associations between e-cigarette use and cigarette smoking behaviors, indicating that findings from these reviews should be interpreted with caution. CONCLUSIONS: Future systematic reviews in this field should strive to adhere to AMSTAR 2 and PRISMA guidelines, to provide high quality syntheses of the available data with transparent and complete reporting.


Subject(s)
Cigarette Smoking , Electronic Nicotine Delivery Systems , Vaping , Humans , Research Report
2.
Adv Med Educ Pract ; 8: 63-73, 2017.
Article in English | MEDLINE | ID: mdl-28144171

ABSTRACT

The flipped classroom (FC) approach to teaching has been increasingly employed in undergraduate medical education in recent years. In FC applications, students are first exposed to content via online resources. Subsequent face-to-face class time can then be devoted to student-centered activities that promote active learning. Although the FC has been well received by students in other contexts, the perceptions of medical students regarding this innovation are unclear. This review serves as an early exploration into medical student perceptions of benefits and limitations of the FC. Medical students have generally expressed strong appreciation for the pre-class preparation activities (especially when facilitated by concise, readily accessed online tools) as well as for interactive, engaging small group classroom activities. Some students have expressed concerns with the FC and noted that suboptimal student preparation and insufficient direction and structure during active learning sessions may limit the student-centered benefits. Although students generally perceive that FC approaches can improve their learning and knowledge, this has not been conclusively shown via performances on assessment tools, which may be related to caveats with the assessment tools used. In any case, lifelong self-directed learning skills are perceived by medical students to be enhanced by the FC. In conclusion, medical students have generally expressed strong satisfaction with early applications of the FC to undergraduate medical education, and generally prefer this method to lecture-based instruction.

3.
J Mol Endocrinol ; 58(3): 127-139, 2017 04.
Article in English | MEDLINE | ID: mdl-28122818

ABSTRACT

Genome-wide association study (GWAS) data have linked the G6PC2 gene to variations in fasting blood glucose (FBG). G6PC2 encodes an islet-specific glucose-6-phosphatase catalytic subunit that forms a substrate cycle with the beta cell glucose sensor glucokinase. This cycle modulates the glucose sensitivity of insulin secretion and hence FBG. GWAS data have not linked G6PC2 to variations in body weight but we previously reported that female C57BL/6J G6pc2-knockout (KO) mice were lighter than wild-type littermates on both a chow and high-fat diet. The purpose of this study was to compare the effects of G6pc2 deletion on FBG and body weight in both chow-fed and high-fat-fed mice on two other genetic backgrounds. FBG was reduced in G6pc2 KO mice largely independent of gender, genetic background or diet. In contrast, the effect of G6pc2 deletion on body weight was markedly influenced by these variables. Deletion of G6pc2 conferred a marked protection against diet-induced obesity in male mixed genetic background mice, whereas in 129SvEv mice deletion of G6pc2 had no effect on body weight. G6pc2 deletion also reduced plasma cholesterol levels in a manner dependent on gender, genetic background and diet. An association between G6PC2 and plasma cholesterol was also observed in humans through electronic health record-derived phenotype analyses. These observations suggest that the action of G6PC2 on FBG is largely independent of the influences of environment, modifier genes or epigenetic events, whereas the action of G6PC2 on body weight and cholesterol are influenced by unknown variables.


Subject(s)
Body Weight/genetics , Cholesterol/blood , Gene Deletion , Genetic Association Studies , Glucose-6-Phosphatase/genetics , Animals , Blood Glucose , Diet, High-Fat , Fasting , Female , Gene Expression , Genetic Background , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pancreas/metabolism , Polymorphism, Single Nucleotide
4.
Transl Res ; 179: 183-198, 2017 01.
Article in English | MEDLINE | ID: mdl-27677687

ABSTRACT

The rise in new cases of type 1 diabetes (T1D) in genetically susceptible individuals over the past half century has been attributed to numerous environmental "triggers" or promoters such as enteroviruses, diet, and most recently, gut bacteria. No single cause has been identified in humans, likely because there are several pathways by which one can develop T1D. There is renewed attention to the role of the gut and its immune system in T1D pathogenesis based largely on recent animal studies demonstrating that altering the gut microbiota affects diabetes incidence. Although T1D patients display dysbiosis in the gut microbiome, it is unclear whether this is cause or effect. The heart of this question involves several moving parts including numerous risk genes, diet, viruses, gut microbiota, timing, and loss of immune tolerance to ß-cells. Most clinical trials have addressed only one aspect of this puzzle using some form of immune suppression, without much success. The key location where our genes meet and deal with the environment is the gastrointestinal tract. The influence of all of its major contents, including microbes, diet, and immune system, must be understood as part of the integrative biology of T1D before we can develop durable means of preventing, treating, or curing this disease. In the present review, we expand our previous gut-centric model based on recent developments in the field.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Gastrointestinal Tract/metabolism , Gene-Environment Interaction , Immunity/genetics , Pancreas/metabolism , Pancreas/pathology , Animals , Humans , Models, Biological
5.
PLoS One ; 11(9): e0162439, 2016.
Article in English | MEDLINE | ID: mdl-27611587

ABSTRACT

Elevated fasting blood glucose (FBG) has been associated with increased risk for development of type 2 diabetes. Single nucleotide polymorphisms (SNPs) in G6PC2 are the most important common determinants of variations in FBG in humans. Studies using G6pc2 knockout mice suggest that G6pc2 regulates the glucose sensitivity of insulin secretion. G6PC2 and the related G6PC1 and G6PC3 genes encode glucose-6-phosphatase catalytic subunits. This study describes a functional analysis of 22 non-synonymous G6PC2 SNPs, that alter amino acids that are conserved in human G6PC1, mouse G6pc1 and mouse G6pc2, with the goal of identifying variants that potentially affect G6PC2 activity/expression. Published data suggest strong conservation of catalytically important amino acids between all four proteins and the related G6PC3 isoform. Because human G6PC2 has very low glucose-6-phosphatase activity we used an indirect approach, examining the effect of these SNPs on mouse G6pc1 activity. Using a novel in situ functional assay for glucose-6-phosphatase activity we demonstrate that the amino acid changes associated with the human G6PC2 rs144254880 (Arg79Gln), rs149663725 (Gly114Arg) and rs2232326 (Ser324Pro) SNPs reduce mouse G6pc1 enzyme activity without affecting protein expression. The Arg79Gln variant alters an amino acid mutation of which, in G6PC1, has previously been shown to cause glycogen storage disease type 1a. We also demonstrate that the rs368382511 (Gly8Glu), rs138726309 (His177Tyr), rs2232323 (Tyr207Ser) rs374055555 (Arg293Trp), rs2232326 (Ser324Pro), rs137857125 (Pro313Leu) and rs2232327 (Pro340Leu) SNPs confer decreased G6PC2 protein expression. In summary, these studies identify multiple G6PC2 variants that have the potential to be associated with altered FBG in humans.


Subject(s)
Amino Acid Substitution , Gene Expression , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Mutation , Amino Acid Sequence , Animals , Blood Glucose , Cell Line , Codon , Conserved Sequence , Enzyme Activation , Fasting/blood , Glucose-6-Phosphatase/chemistry , Humans , Mice , Polymorphism, Single Nucleotide , Rats
6.
Endocrinology ; 157(8): 3002-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27300767

ABSTRACT

The glucose-6-phosphatase catalytic 2 (G6PC2) gene is expressed specifically in pancreatic islet beta cells. Genome-wide association studies have shown that single nucleotide polymorphisms in the G6PC2 gene are associated with variations in fasting blood glucose (FBG) but not fasting plasma insulin. Molecular analyses examining the functional effects of these single nucleotide polymorphisms demonstrate that elevated G6PC2 expression is associated with elevated FBG. Studies in mice complement these genome-wide association data and show that deletion of the G6pc2 gene lowers FBG without affecting fasting plasma insulin. This suggests that, together with glucokinase, G6PC2 forms a substrate cycle that determines the glucose sensitivity of insulin secretion. Because genome-wide association studies and mouse studies demonstrate that elevated G6PC2 expression raises FBG and because chronically elevated FBG is detrimental to human health, increasing the risk of type 2 diabetes, it is unclear why G6PC2 evolved. We show here that the synthetic glucocorticoid dexamethasone strongly induces human G6PC2 promoter activity and endogenous G6PC2 expression in isolated human islets. Acute treatment with dexamethasone selectively induces endogenous G6pc2 expression in 129SvEv but not C57BL/6J mouse pancreas and isolated islets. The difference is due to a single nucleotide polymorphism in the C57BL/6J G6pc2 promoter that abolishes glucocorticoid receptor binding. In 6-hour fasted, nonstressed 129SvEv mice, deletion of G6pc2 lowers FBG. In response to the stress of repeated physical restraint, which is associated with elevated plasma glucocorticoid levels, G6pc2 gene expression is induced and the difference in FBG between wild-type and knockout mice is enhanced. These data suggest that G6PC2 may have evolved to modulate FBG in response to stress.


Subject(s)
Blood Glucose/metabolism , Fasting/blood , Glucose-6-Phosphatase/physiology , Stress, Physiological , Animals , Cells, Cultured , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Glucose-6-Phosphatase/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas/drug effects , Pancreas/metabolism , Promoter Regions, Genetic/drug effects , Stress, Physiological/drug effects , Stress, Physiological/genetics
7.
Diabetes ; 64(12): 4135-47, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26370175

ABSTRACT

Cathelicidin antimicrobial peptide (CAMP) is a naturally occurring secreted peptide that is expressed in several organs with pleiotropic roles in immunomodulation, wound healing, and cell growth. We previously demonstrated that gut Camp expression is upregulated when type 1 diabetes-prone rats are protected from diabetes development. Unexpectedly, we have also identified novel CAMP expression in the pancreatic ß-cells of rats, mice, and humans. CAMP was present even in sterile rat embryo islets, germ-free adult rat islets, and neogenic tubular complexes. Camp gene expression was downregulated in young BBdp rat islets before the onset of insulitis compared with control BBc rats. CAMP treatment of dispersed islets resulted in a significant increase in intracellular calcium mobilization, an effect that was both delayed and blunted in the absence of extracellular calcium. Additionally, CAMP treatment promoted insulin and glucagon secretion from isolated rat islets. Thus, CAMP is a promoter of islet paracrine signaling that enhances islet function and glucoregulation. Finally, daily treatment with the CAMP/LL-37 peptide in vivo in BBdp rats resulted in enhanced ß-cell neogenesis and upregulation of potentially beneficial gut microbes. In particular, CAMP/LL-37 treatment shifted the abundance of specific bacterial populations, mitigating the gut dysbiosis observed in the BBdp rat. Taken together, these findings indicate a novel functional role for CAMP/LL-37 in islet biology and modification of gut microbiota.


Subject(s)
Cathelicidins/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Dysbiosis/drug therapy , Gastrointestinal Agents/therapeutic use , Hypoglycemic Agents/therapeutic use , Islets of Langerhans/drug effects , Peptide Fragments/therapeutic use , Regeneration/drug effects , Aged , Animals , Antimicrobial Cationic Peptides/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium Signaling/drug effects , Cathelicidins/pharmacology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/pathology , Dysbiosis/metabolism , Dysbiosis/microbiology , Dysbiosis/pathology , Gastrointestinal Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Gene Expression Profiling , Germ-Free Life , Glucagon/agonists , Glucagon/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/agonists , Insulin/metabolism , Insulin Secretion , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans/physiology , Jejunum/drug effects , Jejunum/metabolism , Jejunum/microbiology , Male , Mice, Inbred NOD , Peptide Fragments/pharmacology , Rats, Inbred Strains , Tissue Culture Techniques
8.
Nat Med ; 21(7): 769-76, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26076035

ABSTRACT

The peptide hormone urocortin3 (Ucn3) is abundantly expressed by mature beta cells, yet its physiological role is unknown. Here we demonstrate that Ucn3 is stored and co-released with insulin and potentiates glucose-stimulated somatostatin secretion via cognate receptors on delta cells. Further, we found that islets lacking endogenous Ucn3 have fewer delta cells, reduced somatostatin content, impaired somatostatin secretion, and exaggerated insulin release, and that these defects are rectified by treatment with synthetic Ucn3 in vitro. Our observations indicate that the paracrine actions of Ucn3 activate a negative feedback loop that promotes somatostatin release to ensure the timely reduction of insulin secretion upon normalization of plasma glucose. Moreover, Ucn3 is markedly depleted from beta cells in mouse and macaque models of diabetes and in human diabetic islets. This suggests that Ucn3 is a key contributor to stable glycemic control, whose reduction during diabetes aggravates glycemic volatility and contributes to the pathophysiology of this disease.


Subject(s)
Feedback, Physiological , Insulin/metabolism , Somatostatin/metabolism , Urocortins/metabolism , Adolescent , Adult , Aged , Animals , Child , Child, Preschool , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Female , Gene Expression Regulation , HEK293 Cells , Humans , Hyperglycemia/genetics , Hyperglycemia/pathology , Infant , Infant, Newborn , Insulin Secretion , Insulin-Secreting Cells/metabolism , Macaca , Male , Mice, Inbred C57BL , Middle Aged , Models, Biological , Paracrine Communication , Tissue Donors , Transcriptome/genetics , Urocortins/deficiency , Young Adult
9.
Diabetes ; 64(6): 2129-37, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25552595

ABSTRACT

A polymorphism located in the G6PC2 gene, which encodes an islet-specific glucose-6-phosphatase catalytic subunit, is the most important common determinant of variations in fasting blood glucose (FBG) levels in humans. Studies of G6pc2 knockout (KO) mice suggest that G6pc2 represents a negative regulator of basal glucose-stimulated insulin secretion (GSIS) that acts by hydrolyzing glucose-6-phosphate (G6P), thereby reducing glycolytic flux. However, this conclusion conflicts with the very low estimates for the rate of glucose cycling in pancreatic islets, as assessed using radioisotopes. We have reassessed the rate of glucose cycling in pancreatic islets using a novel stable isotope method. The data show much higher levels of glucose cycling than previously reported. In 5 mmol/L glucose, islets from C57BL/6J chow-fed mice cycled ∼16% of net glucose uptake. The cycling rate was further increased at 11 mmol/L glucose. Similar cycling rates were observed using islets from high fat-fed mice. Importantly, glucose cycling was abolished in G6pc2 KO mouse islets, confirming that G6pc2 opposes the action of the glucose sensor glucokinase by hydrolyzing G6P. The demonstration of high rates of glucose cycling in pancreatic islets explains why G6pc2 deletion enhances GSIS and why variants in G6PC2 affect FBG in humans.


Subject(s)
Glucose/metabolism , Islets of Langerhans/metabolism , Animals , Gas Chromatography-Mass Spectrometry , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , In Vitro Techniques , Isotope Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Am J Physiol Endocrinol Metab ; 307(1): E115-23, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24844258

ABSTRACT

Children exposed to a maternal Western-style diet in utero have an increased risk of developing type 2 diabetes. Understanding the mechanisms and an investigation of possible interventions are critical to reversing this phenomenon. We examined the impact of maternal Western-style diet consumption on the development of islet vascularization and innervation, both of which are critical to normal islet function, in fetal and juvenile offspring. Furthermore, we assessed whether improved dietary intake or resveratrol supplementation could ameliorate the harmful consequences of Western-style diet consumption during pregnancy. Adult female Japanese macaques were maintained on a control or Western-style diet for 4-7 yr. One cohort of dams was switched back onto a control diet, whereas another cohort received resveratrol supplementation throughout gestation. Pregnancies were terminated in the early third trimester by C-section, or offspring were born naturally and sent to necropsy at 1 yr of age. Western-style diet consumption resulted in impaired fetal islet capillary density and sympathetic islet innervation. Furthermore, this reduction in vascularization persisted in the juvenile offspring. This effect is independent of changes in the expression of key angiogenic markers. Diet reversal normalized islet vascularization to control offspring levels, whereas resveratrol supplementation caused a significant increase in capillary density above controls. These data provide a novel mechanism by which maternal Western-style diet consumption leads to increased susceptibility to type 2 diabetes in the offspring. Importantly, an improved maternal diet may mitigate these harmful effects. However, until the long-term consequences of increased vascularization can be determined, resveratrol use during pregnancy is not advised.


Subject(s)
Capillaries/growth & development , Diet , Islets of Langerhans/blood supply , Islets of Langerhans/innervation , Prenatal Nutritional Physiological Phenomena/physiology , Sympathetic Nervous System/growth & development , Animals , Female , Macaca mulatta , Male , Neovascularization, Physiologic/physiology , Pregnancy , Pregnancy, Animal
11.
Curr Opin Endocrinol Diabetes Obes ; 21(2): 89-94, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569549

ABSTRACT

PURPOSE OF THE REVIEW: Although rodent models provide insight into the mechanisms underlying type 2 diabetes mellitus (T2DM), they are limited in their translatability to humans. The nonhuman primate (NHP) shares important metabolic similarities with the human, making it an ideal model for the investigation of type 2 diabetes and use in preclinical trials. This review highlights the key contributions in the field over the last year using the NHP model. RECENT FINDINGS: The NHP has not only provided novel insight into the normal and pathological processes that occur within the islet, but has also allowed for the preclinical testing of novel pharmaceutical targets for obesity and T2DM. Particularly, administration of fibroblast growth factor-21 in the NHP resulted in weight loss and improvements in metabolic health, supporting rodent studies and recent clinical trials. In addition, the NHP was used to demonstrate that a novel melanocortin-4 receptor agonist did not cause adverse cardiovascular effects. Finally, this model has been used to provide evidence that glucagon-like peptide-1-based therapies do not induce pancreatitis in the healthy NHP. SUMMARY: The insight gained from studies using the NHP model has allowed for a better understanding of the processes driving T2DM and has promoted the development of well tolerated and effective treatments.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/agonists , Hypoglycemic Agents/pharmacology , Pancreatitis/chemically induced , Weight Loss/drug effects , Animals , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Disease Models, Animal , Female , Insulin-Secreting Cells , Macaca mulatta , Male
12.
FASEB J ; 28(6): 2466-77, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24563374

ABSTRACT

Resveratrol has been proposed as a potential therapeutic to improve metabolic health during pregnancy, yet little is known about the fetal effects of this maternal dietary supplement. We hypothesized that when administered to pregnant nonhuman primates (NHPs), resveratrol would increase uterine blood flow and mitigate the harmful consequences of maternal Western-style diet (WSD) consumption. NHPs were fed a WSD (36% fat) supplemented with 0.37% resveratrol throughout pregnancy. Outcomes were compared with cohorts fed WSD alone and control chow (14% fat) to distinguish between WSD and resveratrol-specific effects in these animals. In the early third trimester, uterine blood flow was measured by Doppler ultrasound before fetal delivery and tissue collection. Resveratrol resulted in 30% maternal weight loss and improved glucose tolerance, increased uterine artery volume blood flow, and decreased placental inflammation and liver triglyceride deposition. In addition, fetal pancreatic mass was enlarged by 42%, with a 12-fold increase in proliferation by Ki67 immunohistochemistry. These results demonstrate that resveratrol use during pregnancy yields improvements in maternal and placental phenotype with beneficial effects in the fetal liver but an unexplained and concerning alteration in fetal pancreatic development, which strongly cautions against the use of resveratrol by pregnant women.


Subject(s)
Fetal Development/drug effects , Stilbenes/adverse effects , Stilbenes/pharmacology , Animals , Contraindications , Diet/adverse effects , Dietary Supplements/adverse effects , Female , Fetus , Liver/drug effects , Liver/embryology , Macaca , Pancreas/drug effects , Pancreas/embryology , Placental Circulation/drug effects , Pregnancy , Regional Blood Flow/drug effects , Resveratrol , Stilbenes/blood , Triglycerides/blood , Uterus/blood supply
13.
Diabetes ; 62(5): 1547-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23274894

ABSTRACT

Elevated fasting blood glucose (FBG) is associated with increased risk for the development of type 2 diabetes and cardiovascular-associated mortality. Genome-wide association studies (GWAS) have linked polymorphisms in G6PC2 with variations in FBG and body fat, although not insulin sensitivity or glucose tolerance. G6PC2 encodes an islet-specific, endoplasmic reticulum-resident glucose-6-phosphatase catalytic subunit. A combination of in situ perfused pancreas, in vitro isolated islet, and in vivo analyses were used to explore the function of G6pc2 in mice. G6pc2 deletion had little effect on insulin sensitivity and glucose tolerance, whereas body fat was reduced in female G6pc2 knockout (KO) mice on both a chow and high-fat diet, observations that are all consistent with human GWAS data. G6pc2 deletion resulted in a leftward shift in the dose-response curve for glucose-stimulated insulin secretion (GSIS). As a consequence, under fasting conditions in which plasma insulin levels were identical, blood glucose levels were reduced in G6pc2 KO mice, again consistent with human GWAS data. Glucose-6-phosphatase activity was reduced, whereas basal cytoplasmic calcium levels were elevated in islets isolated from G6pc2 KO mice. These data suggest that G6pc2 represents a novel, negative regulator of basal GSIS that acts by hydrolyzing glucose-6-phosphate, thereby reducing glycolytic flux.


Subject(s)
Blood Glucose/analysis , Glucose-6-Phosphatase/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Proteins/metabolism , Adiposity , Animals , Calcium Signaling , Diet, High-Fat/adverse effects , Female , Glucose-6-Phosphatase/genetics , Heterozygote , Insulin Resistance , Insulin Secretion , Islets of Langerhans/metabolism , Kinetics , Male , Mice , Mice, Congenic , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Pancreas/metabolism , Proteins/genetics , Sex Characteristics
14.
PLoS One ; 7(7): e40972, 2012.
Article in English | MEDLINE | ID: mdl-22829903

ABSTRACT

OBJECTIVE: The SLC30A8 gene encodes the islet-specific transporter ZnT-8, which is hypothesized to provide zinc for insulin-crystal formation. A polymorphic variant in SLC30A8 is associated with altered susceptibility to type 2 diabetes. Several groups have examined the effect of global Slc30a8 gene deletion but the results have been highly variable, perhaps due to the mixed 129SvEv/C57BL/6J genetic background of the mice studied. We therefore sought to remove the conflicting effect of 129SvEv-specific modifier genes. METHODS: The impact of Slc30a8 deletion was examined in the context of the pure C57BL/6J genetic background. RESULTS: Male C57BL/6J Slc30a8 knockout (KO) mice had normal fasting insulin levels and no change in glucose-stimulated insulin secretion (GSIS) from isolated islets in marked contrast to the ∼50% and ∼35% decrease, respectively, in both parameters observed in male mixed genetic background Slc30a8 KO mice. This observation suggests that 129SvEv-specific modifier genes modulate the impact of Slc30a8 deletion. In contrast, female C57BL/6J Slc30a8 KO mice had reduced (∼20%) fasting insulin levels, though this was not associated with a change in fasting blood glucose (FBG), or GSIS from isolated islets. This observation indicates that gender also modulates the impact of Slc30a8 deletion, though the physiological explanation as to why impaired insulin secretion is not accompanied by elevated FBG is unclear. Neither male nor female C57BL/6J Slc30a8 KO mice showed impaired glucose tolerance. CONCLUSIONS: Our data suggest that, despite a marked reduction in islet zinc content, the absence of ZnT-8 does not have a substantial impact on mouse physiology.


Subject(s)
Cation Transport Proteins/metabolism , Fasting/blood , Insulin/blood , Animals , Blood Glucose/metabolism , Cation Transport Proteins/genetics , Female , Glucose Intolerance/blood , Glucose Intolerance/genetics , Insulin/genetics , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Factors , Zinc/metabolism , Zinc Transporter 8
15.
Mol Metab ; 2(1): 10-22, 2012.
Article in English | MEDLINE | ID: mdl-24024126

ABSTRACT

We investigated the impact of poor maternal nutrition and metabolic health on the development of islets of the nonhuman primate (NHP). Interestingly, fetal offspring of high fat diet (HFD) fed animals had normal total islet and ß cell mass; however, there was a significant reduction in α cell mass, and decreased expression of transcription factors involved in α cell differentiation. In juvenile animals all offspring maintained on a HFD during the postweaning period demonstrated increases in total islet mass, however, the control offspring displaying increased islet number, and HFD offspring displayed increased islet size. Finally, while control offspring had increases in α and ß cells, the HFD offspring had increases only in ß cell number. These studies indicate that consumption of a HFD diet during pregnancy in the NHP, independent of maternal metabolic health, causes long-term abnormalities in α cell plasticity that may contribute to chronic disease susceptibility.

16.
Diabetes ; 60(11): 2922-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21896930

ABSTRACT

OBJECTIVE: Islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), now known as G6PC2, is a major target of autoreactive T cells implicated in the pathogenesis of type 1 diabetes in both mice and humans. This study aimed to determine whether suppression of G6p2 gene expression might therefore prevent or delay disease progression. RESEARCH DESIGN AND METHODS: G6pc2(-/-) mice were generated on the NOD/ShiLtJ genetic background, and glycemia was monitored weekly up to 35 weeks of age to determine the onset and incidence of diabetes. The antigen specificity of CD8(+) T cells infiltrating islets from NOD/ShiLtJ G6pc2(+/+) and G6pc2(-/-) mice at 12 weeks was determined in parallel. RESULTS: The absence of G6pc2 did not affect the time of onset, incidence, or sex bias of type 1 diabetes in NOD/ShiLtJ mice. Insulitis was prominent in both groups, but whereas NOD/ShiLtJ G6pc2(+/+) islets contained CD8(+) T cells reactive to the G6pc2 NRP peptide, G6pc2 NRP-reactive T cells were absent in NOD/ShiLtJ G6pc2(-/-) islets. CONCLUSIONS: These results demonstrate that G6pc2 is an important driver for the selection and expansion of islet-reactive CD8(+) T cells infiltrating NOD/ShiLtJ islets. However, autoreactivity to G6pc2 is not essential for the emergence of autoimmune diabetes. The results remain consistent with previous studies indicating that insulin may be the primary autoimmune target, at least in NOD/ShiLtJ mice.


Subject(s)
Catalytic Domain , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Disease Progression , Gene Deletion , Glucose-6-Phosphatase/genetics , Islets of Langerhans/metabolism , Proteins/genetics , Animals , Autoantibodies/analysis , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Glucose-6-Phosphatase/chemistry , Humans , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Proteins/chemistry , Sex Characteristics
17.
J Mol Endocrinol ; 47(3): 251-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21798992

ABSTRACT

Genome-wide association studies have shown that a polymorphic variant in SLC30A8, which encodes zinc transporter-8, is associated with altered susceptibility to type 2 diabetes (T2D). This association is consistent with the observation that glucose-stimulated insulin secretion is decreased in islets isolated from Slc30a8 knockout mice. In this study, immunohistochemical staining was first used to show that SLC30A8 is expressed specifically in pancreatic islets. Fusion gene studies were then used to examine the molecular basis for the islet-specific expression of SLC30A8. The analysis of SLC30A8-luciferase expression in ßTC-3 cells revealed that the proximal promoter region, located between -6154 and -1, relative to the translation start site, was only active in stable but not transient transfections. VISTA analyses identified three regions in the SLC30A8 promoter and a region in SLC30A8 intron 2 that are conserved in the mouse Slc30a8 gene. Additional fusion gene experiments demonstrated that none of these Slc30a8 promoter regions exhibited enhancer activity when ligated to a heterologous promoter whereas the conserved region in SLC30A8 intron 2 conferred elevated reporter gene expression selectively in ßTC-3 but not in αTC-6 cells. Finally, the functional effects of a single nucleotide polymorphism (SNP), rs62510556, in this conserved intron 2 enhancer were investigated. Gel retardation studies showed that rs62510556 affects the binding of an unknown transcription factor and fusion gene analyses showed that it modulates enhancer activity. However, genetic analyses suggest that this SNP is not a causal variant that contributes to the association between SLC30A8 and T2D, at least in Europeans.


Subject(s)
Cation Transport Proteins/genetics , Enhancer Elements, Genetic , Introns , Promoter Regions, Genetic , Animals , Cation Transport Proteins/metabolism , Cells, Cultured , Conserved Sequence , Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation , Genes, Reporter , Glucagon-Secreting Cells/metabolism , Humans , Insulin-Secreting Cells/metabolism , Mice , Transcriptional Activation , Transfection , Zinc Transporter 8
18.
Biochem J ; 433(1): 95-105, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20942803

ABSTRACT

The SLC30A8 gene encodes the zinc transporter ZnT-8, which provides zinc for insulin-hexamer formation. Genome-wide association studies have shown that a polymorphic variant in SLC30A8 is associated with altered susceptibility to Type 2 diabetes and we recently reported that glucose-stimulated insulin secretion is decreased in islets isolated from Slc30a8-knockout mice. The present study examines the molecular basis for the islet-specific expression of Slc30a8. VISTA analyses identified two conserved regions in Slc30a8 introns 2 and 3, designated enhancers A and B respectively. Transfection experiments demonstrated that enhancer B confers elevated fusion gene expression in both ßTC-3 cells and αTC-6 cells. In contrast, enhancer A confers elevated fusion gene expression selectively in ßTC-3 and not αTC-6 cells. These data suggest that enhancer A is an islet ß-cell-specific enhancer and that the mechanisms controlling Slc30a8 expression in α- and ß-cells are overlapping, but distinct. Gel retardation and ChIP (chromatin immunoprecipitation) assays revealed that the islet-enriched transcription factor Pdx-1 binds enhancer A in vitro and in situ respectively. Mutation of two Pdx-1-binding sites in enhancer A markedly reduces fusion gene expression suggesting that this factor contributes to Slc30a8 expression in ß-cells, a conclusion consistent with developmental studies showing that restriction of Pdx-1 to pancreatic islet ß-cells correlates with the induction of Slc30a8 gene expression and ZnT-8 protein expression in vivo.


Subject(s)
Cation Transport Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Homeodomain Proteins/physiology , Islets of Langerhans/chemistry , Trans-Activators/physiology , Transcription, Genetic , Animals , Binding Sites , Introns/genetics , Islets of Langerhans/metabolism , Mice , Tissue Distribution , Transcription Factors , Zinc Transporter 8
19.
Biochem J ; 421(3): 371-6, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19450229

ABSTRACT

The Slc30a8 gene encodes the islet-specific zinc transporter ZnT-8, which provides zinc for insulin-hexamer formation. Polymorphic variants in amino acid residue 325 of human ZnT-8 are associated with altered susceptibility to Type 2 diabetes and ZnT-8 autoantibody epitope specificity changes in Type 1 diabetes. To assess the physiological importance of ZnT-8, mice carrying a Slc30a8 exon 3 deletion were analysed histologically and phenotyped for energy metabolism and pancreatic hormone secretion. No gross anatomical or behavioural changes or differences in body weight were observed between wild-type and ZnT-8-/- mice, and ZnT-8-/- mouse islets were indistinguishable from wild-type in terms of their numbers, size and cellular composition. However, total zinc content was markedly reduced in ZnT-8-/- mouse islets, as evaluated both by Timm's histochemical staining of pancreatic sections and direct measurements in isolated islets. Blood glucose levels were unchanged in 16-week-old, 6 h fasted animals of either gender; however, plasma insulin concentrations were reduced in both female (approximately 31%) and male (approximately 47%) ZnT-8-/- mice. Intraperitoneal glucose tolerance tests demonstrated no impairment in glucose clearance in male ZnT-8-/- mice, but glucose-stimulated insulin secretion from isolated islets was reduced approximately 33% relative to wild-type littermates. In summary, Slc30a8 gene deletion is accompanied by a modest impairment in insulin secretion without major alterations in glucose metabolism.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Diabetes Mellitus/metabolism , Insulin/metabolism , Sequence Deletion , Animals , Blood Glucose , Diabetes Mellitus/genetics , Female , Glucose Tolerance Test , Humans , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Zinc/metabolism , Zinc Transporter 8
SELECTION OF CITATIONS
SEARCH DETAIL
...