Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
JMIR Res Protoc ; 10(10): e26816, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34528885

ABSTRACT

BACKGROUND: The number of solid organ transplants in Canada has increased 33% over the past decade. Hospital readmissions are common within the first year after transplant and are linked to increased morbidity and mortality. Nearly half of these admissions to the hospital appear to be preventable. Mobile health (mHealth) technologies hold promise to reduce admission to the hospital and improve patient outcomes, as they allow real-time monitoring and timely clinical intervention. OBJECTIVE: This study aims to determine whether an innovative mHealth intervention can reduce hospital readmission and unscheduled visits to the emergency department or transplant clinic. Our second objective is to assess the use of clinical and continuous ambulatory physiologic data to develop machine learning algorithms to predict the risk of infection, organ rejection, and early mortality in adult heart, kidney, and liver transplant recipients. METHODS: Remote Mobile Outpatient Monitoring in Transplant (Reboot) 2.0 is a two-phased single-center study to be conducted at the University Health Network in Toronto, Canada. Phase one will consist of a 1-year concealed randomized controlled trial of 400 adult heart, kidney, and liver transplant recipients. Participants will be randomized to receive either personalized communication using an mHealth app in addition to standard of care phone communication (intervention group) or standard of care communication only (control group). In phase two, the prior collected data set will be used to develop machine learning algorithms to identify early markers of rejection, infection, and graft dysfunction posttransplantation. The primary outcome will be a composite of any unscheduled hospital admission, visits to the emergency department or transplant clinic, following discharge from the index admission. Secondary outcomes will include patient-reported outcomes using validated self-administered questionnaires, 1-year graft survival rate, 1-year patient survival rate, and the number of standard of care phone voice messages. RESULTS: At the time of this paper's completion, no results are available. CONCLUSIONS: Building from previous work, this project will aim to leverage an innovative mHealth app to improve outcomes and reduce hospital readmission in adult solid organ transplant recipients. Additionally, the development of machine learning algorithms to better predict adverse health outcomes will allow for personalized medicine to tailor clinician-patient interactions and mitigate the health care burden of a growing patient population. TRIAL REGISTRATION: ClinicalTrials.gov NCT04721288; https://www.clinicaltrials.gov/ct2/show/NCT04721288. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): PRR1-10.2196/26816.

2.
Focus (Am Psychiatr Publ) ; 18(3): 277-284, 2020 Jul.
Article in English | MEDLINE | ID: mdl-33162864

ABSTRACT

Working with LGBTQ people regarding sex and relationship problems may be intimidating for the psychiatrist with little experience. This article provides an overview of common sex and relationship problems that can be encountered in clinical work with a focus on LGBTQ couples therapy. Topics include sex difficulties and their causes, drugs and alcohol, the effect of "the closet," discordant "outness" in couples, issues regarding sex roles, LGBTQ parenting, and issues arising when a member of a couple is transitioning.

3.
Plant Dis ; 104(8): 2060-2067, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32552394

ABSTRACT

We examine the economic costs of potato virus Y (PVY) and benefits to commercial potato growers from using screened seed. To do so, we use a quantile regression model to explore disease spread. We use this model to predict disease prevalence and corresponding losses in commercial potato operations with and without a screening and certification program in place. Our analysis suggests that this screening is very important; the amount of PVY in seed in the summer test is the strongest predictor of PVY in the winter test of the variables in our model. The amount of PVY in the seed can have major effects on commercial potato grower revenues and profitability. Using data and models from Idaho, a major purchaser of Montana seed, we estimate the annual benefit from Montana's program to Idaho to average $205 per acre or $22 million for the state.[Formula: see text] Copyright © 2020 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.


Subject(s)
Solanum tuberosum , Certification , Idaho , Montana , Plant Diseases , Seeds
4.
Plant Dis ; 103(9): 2179-2183, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31322494

ABSTRACT

Potato virus Y (PVY) is among the most economically impactful potato pathogens, yet the spread of PVY from infected seed potatoes within commercial potato fields has not been adequately studied. Test lots containing various seed-borne PVY levels were created by mixing different proportions of seed pieces from healthy and infected tubers drawn from the same seed source. These seed lots were planted in commercial potato fields near the Teton Seed Potato Management Area from 2010 to 2012. Regression analyses on data from these test plots produced models of the in-season spread of PVY originating from infected seed. Conventional ordinary least squares techniques were supplemented with the use of quantile regression; the resulting models indicate the significance of seed-borne PVY on end-of-season infection levels and highlight the need of seed potato buyers to review postharvest testing results.


Subject(s)
Plant Diseases , Plant Tubers , Potyvirus , Solanum tuberosum , Agriculture , Plant Diseases/prevention & control , Plant Diseases/virology , Plant Tubers/virology , Solanum tuberosum/virology
5.
J Med Regul ; 102(2): 7-12, 2016.
Article in English | MEDLINE | ID: mdl-27754500

ABSTRACT

Conversion therapies are any treatments, including individual talk therapy, behavioral (e.g. aversive stimuli), group therapy or milieu (e.g. "retreats or inpatient treatments" relying on all of the above methods) treatments, which attempt to change an individual's sexual orientation from homosexual to heterosexual. However these practices have been repudiated by major mental health organizations because of increasing evidence that they are ineffective and may cause harm to patients and their families who fail to change. At present, California, New Jersey, Oregon, Illinois, Washington, DC, and the Canadian Province of Ontario have passed legislation banning conversion therapy for minors and an increasing number of US States are considering similar bans. In April 2015, the Obama administration also called for a ban on conversion therapies for minors. The growing trend toward banning conversion therapies creates challenges for licensing boards and ethics committees, most of which are unfamiliar with the issues raised by complaints against conversion therapists. This paper reviews the history of conversion therapy practices as well as clinical, ethical and research issues they raise. With this information, state licensing boards, ethics committees and other regulatory bodies will be better able to adjudicate complaints from members of the public who have been exposed to conversion therapies.

6.
J Diabetes Investig ; 7 Suppl 1: 4-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27186348

ABSTRACT

The present review focuses initially on experimental studies that were designed to identify acid inhibitory factors, referred to as 'enterogastrones,' that ultimately led to the isolation of gastric inhibitory polypeptide (GIP), a 42-amino acid polypeptide. GIP was shown to inhibit acid secretion in animal models, as well as stimulating gastric somatostatin secretion. However, its role in human gastric physiology is unclear. Further studies showed that GIP strongly stimulated the secretion of insulin, in the presence of elevated glucose, and this 'incretin' action is now considered to be its most important; an alternative for the GIP acronym, glucose-dependent insulinotropic polypeptide, was therefore introduced. In the 1970s, GIP purified by conventional chromatography was shown by high-performance liquid chromatography to consist largely of GIP 1-42 and GIP 3-42. It was later shown that dipeptidyl peptidase 4 was a physiologically relevant enzyme responsible for this conversion, as well as the similar metabolism of the second incretin, glucagon-like peptide-1. Dipeptidyl peptidase-4 inhibitors are currently in use as type 2 diabetes therapeutics, and studies on islet transplantation in rodent models of type 1 diabetes have shown that dipeptidyl peptidase-4 inhibitor treatment reduces graft rejection. Additional studies on C-terminally shortened forms of GIP have shown that GIP 1-30 and a dipeptidyl peptidase-4-resistant form (D-Ala(2) GIP 1-30) are equipotent to the intact polypeptide in vitro, and administration of D-Ala(2) GIP 1-30 to diabetic rodents greatly improved glucose tolerance and reduced apoptotic cell death in islet ß-cells. There are probably therefore further clinically useful effects of GIP that require investigation.


Subject(s)
Gastric Acid/metabolism , Gastric Inhibitory Polypeptide/physiology , Animals , Diabetes Mellitus/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Gastric Inhibitory Polypeptide/history , History, 20th Century , Humans
7.
Hum Mol Genet ; 24(10): 3004, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25753258

ABSTRACT

In this article, Figure 2F was incorrect. The correct panel is shown below. The authors sincerely apologise for this error.

8.
Biochem J ; 451(2): 227-34, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23390957

ABSTRACT

The incretins, GIP (glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like peptide-1) are gastrointestinal hormones conferring a number of beneficial effects on ß-cell secretion, survival and proliferation. In a previous study, it was demonstrated that delayed rectifier channel protein Kv2.1 contributes to ß-cell apoptosis and that the prosurvival effects of incretins involve Kv2.1 PTMs (post-translational modifications), including phosphorylation and acetylation. Since Kv1.5 overexpression was also shown to stimulate ß-cell death, the present study was initiated in order to determine whether incretins modulate Kv1.5α-Kvß2 interaction via PTM and the mechanisms involved. GIP and GLP-1 reduced apoptosis in INS-1 ß-cells (clone 832/13) overexpressing Kv1.5, and RNAi (RNA interference)-mediated knockdown of endogenous Kv1.5 attenuated apoptotic ß-cell death. Both GIP and GLP-1 increased phosphorylation and acetylation of Kv1.5 and its Kvß2 protein subunit, leading to their enhanced interaction. Further studies demonstrated that CBP [CREB (cAMP-response-element-binding protein)-binding protein]/SirT1 mediated acetylation/deacetylation and interaction between Kvß2 and Kv1.5 in response to GIP or GLP-1. Incretin regulation of ß-cell function therefore involves the acetylation of multiple Kvα and Kvß subunits.


Subject(s)
CREB-Binding Protein/metabolism , Incretins/pharmacology , Insulin-Secreting Cells/metabolism , Kv1.5 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Sirtuin 1/metabolism , Acetylation/drug effects , Apoptosis/drug effects , CREB-Binding Protein/genetics , Cell Survival/drug effects , Cells, Cultured , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gene Knockdown Techniques , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Humans , Incretins/metabolism , Insulin-Secreting Cells/drug effects , Kv1.5 Potassium Channel/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational , Shaker Superfamily of Potassium Channels
9.
Diabetes ; 62(2): 471-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23002036

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone that also plays a regulatory role in fat metabolism. In 3T3-L1 cells, resistin was demonstrated to be a key mediator of GIP stimulation of lipoprotein lipase (LPL) activity, involving activation of protein kinase B (PKB) and reduced phosphorylation of liver kinase B1 (LKB1) and AMP-activated protein kinase (AMPK). The current study was initiated to determine whether resistin has additional roles in GIP-regulated adipocyte functions. Analysis of primary adipocytes isolated from Retn(-/-), Retn(+/-), and Retn(+/+) mice found that GIP stimulated the PKB/LKB1/AMPK/LPL pathway and fatty acid uptake only in Retn(+/+) adipocytes, suggesting that GIP signaling and/or GIP responsiveness were compromised in Retn(+/-) and Retn(-/-) adipocytes. GIP receptor (GIPR) protein and mRNA were decreased in Retn(+/-) and Retn(-/-) adipocytes, but resistin treatment rescued LPL responsiveness to GIP. In addition, genes encoding tumor necrosis factor (TNF), TNF receptor 2 (TNFR2), and the signaling proteins stress-activated protein kinase (SAPK)/Jun NH(2)-terminal kinase (JNK), were downregulated, and phosphorylated levels of SAPK/JNK/c-Jun were decreased in Retn(-/-) mice. Chromatin immunoprecipitation assays were used to identify a 12-O-tetradecanoylphorbol-13-acetate (TPA)-response element (TRE-III) responsible for c-Jun-mediated transcriptional activation of Gipr. Blunted GIP responsiveness in Retn(+/-) and Retn(-/-) adipocytes was therefore largely due to the greatly reduced GIPR expression associated with decreased c-Jun-mediated transcriptional activation of Gipr.


Subject(s)
Adipocytes/metabolism , Gastric Inhibitory Polypeptide/metabolism , Receptors, Gastrointestinal Hormone/biosynthesis , Resistin/metabolism , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Animals , Carcinogens/pharmacology , Cells, Cultured , Fatty Acids/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gene Expression Regulation , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/genetics , Resistin/genetics , Resistin/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
10.
PLoS One ; 7(7): e40156, 2012.
Article in English | MEDLINE | ID: mdl-22802954

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a gastrointestinal hormone that potentiates glucose-stimulated insulin secretion during a meal. Since GIP has also been shown to exert ß-cell prosurvival and adipocyte lipogenic effects in rodents, both GIP receptor agonists and antagonists have been considered as potential therapeutics in type 2 diabetes (T2DM). In the present study, we tested the hypothesis that chronically elevating GIP levels in a transgenic (Tg) mouse model would increase adipose tissue expansion and exert beneficial effects on glucose homeostasis. In contrast, although GIP Tg mice demonstrated enhanced ß-cell function, resulting in improved glucose tolerance and insulin sensitivity, they exhibited reduced diet-induced obesity. Adipose tissue macrophage infiltration and hepatic steatosis were both greatly reduced, and a number of genes involved in lipid metabolism/inflammatory signaling pathways were found to be down-regulated. Reduced adiposity in GIP Tg mice was associated with decreased energy intake, involving overexpression of hypothalamic GIP. Together, these studies suggest that, in the context of over-nutrition, transgenic GIP overexpression has the potential to improve hepatic and adipocyte function as well as glucose homeostasis.


Subject(s)
Fatty Liver/prevention & control , Gastric Inhibitory Polypeptide/biosynthesis , Glucose/metabolism , Homeostasis , Obesity/etiology , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Energy Metabolism , Gastric Inhibitory Polypeptide/pharmacology , Glucose Intolerance/metabolism , Glucose Tolerance Test , Insulin Resistance/physiology , Male , Mice , Mice, Transgenic , Obesity/metabolism
11.
PLoS One ; 7(3): e32675, 2012.
Article in English | MEDLINE | ID: mdl-22412906

ABSTRACT

In response to a meal, Glucose-dependent Insulinotropic Polypeptide (GIP) and Glucagon-like Peptide-1 (GLP-1) are released from gut endocrine cells into the circulation and interact with their cognate G-protein coupled receptors (GPCRs). Receptor activation results in tissue-selective pleiotropic responses that include augmentation of glucose-induced insulin secretion from pancreatic beta cells. N-glycosylation and receptor oligomerization are co-translational processes that are thought to regulate the exit of functional GPCRs from the ER and their maintenance at the plasma membrane. Despite the importance of these regulatory processes, their impact on functional expression of GIP and GLP-1 receptors has not been well studied. Like many family B GPCRs, both the GIP and GLP-1 receptors possess a large extracellular N-terminus with multiple consensus sites for Asn-linked (N)-glycosylation. Here, we show that each of these Asn residues is glycosylated when either human receptor is expressed in Chinese hamster ovary cells. N-glycosylation enhances cell surface expression and function in parallel but exerts stronger control over the GIP receptor than the GLP-1 receptor. N-glycosylation mainly lengthens receptor half-life by reducing degradation in the endoplasmic reticulum. N-glycosylation is also required for expression of the GIP receptor at the plasma membrane and efficient GIP potentiation of glucose-induced insulin secretion from the INS-1 pancreatic beta cell line. Functional expression of a GIP receptor mutant lacking N-glycosylation is rescued by co-expressed wild type GLP1 receptor, which, together with data obtained using Bioluminescence Resonance Energy Transfer, suggests formation of a GIP-GLP1 receptor heteromer.


Subject(s)
Protein Multimerization , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Consensus Sequence , Cyclic AMP/biosynthesis , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Glucagon-Like Peptide-1 Receptor , Glycosylation , Humans , Insulin/metabolism , Insulin Secretion , Ligands , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Mutation , Protein Isoforms , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/chemistry , Receptors, Glucagon/genetics , Sequence Alignment
12.
J Diabetes Investig ; 3(2): 96-106, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-24843552

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) was the first incretin to be identified. In addition to stimulating insulin secretion, GIP plays regulatory roles in the maintenance, growth and survival of pancreatic islets, as well as impacting on adipocyte function. The current review focuses on the intracellular signaling pathways by which GIP contributes to the regulation of ß-cell secretion and survival, and adipocyte differentiation and lipogenesis. Studies on signaling underlying the insulinotropic actions of the incretin hormones have largely been carried out with glucagon-like peptide-1. They have provided evidence for contributions by both protein kinase A (PKA) and exchange protein directly activated by cyclic adenosine monophosphate (EPAC2), and their probable role in GIP signaling is discussed. Recent studies have shown that inhibition of the kinase apoptosis signal-regulating kinase 1 (ASK1) by GIP plays a key role in reducing mitochondria-induced apoptosis in ß-cells through protein kinase B (PKB)-mediated pathways, and that GIP-induced post-translational modification of voltage- dependent K(+) (Kv) channels also contributes to its prosurvival role. Through regulation of gene expression, GIP tips the balance between pro- and anti-apoptotic members of the B-cell lymphoma-2 (Bcl-2) protein family towards ß-cell survival. GIP also plays important roles in the differentiation of pre-adipocytes to adipocytes, and in the regulation of lipoprotein lipase expression and lipogenesis. These events involve interactions between GIP, insulin and resistin signaling pathways. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2012.00196.x, 2012).

13.
J Lipid Res ; 52(4): 759-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21245029

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a gastrointestinal hormone that exerts insulinotropic and growth and survival effects on pancreatic ß-cells. Additionally, there is increasing evidence supporting an important role for GIP in the regulation of adipocyte metabolism. In the current study we examined the molecular mechanisms involved in the regulation of GIP receptor (GIPR) expression in 3T3-L1 cells. GIP acted synergistically with insulin to increase neutral lipid accumulation during progression of 3T3-L1 preadipocytes to the adipocyte phenotype. Both GIPR protein and mRNA expression increased during 3T3-L1 cell differentiation, and this increase was associated with upregulation of nuclear levels of sterol response element binding protein 1c (SREBP-1c) and peroxisome proliferator-activated receptor γ (PPARγ), as well as acetylation of histones H3/H4. The PPARγ receptor agonists LY171883 and rosiglitazone increased GIPR expression in differentiated 3T3-L1 adipocytes, whereas the antagonist GW9662 ablated expression. Additionally, both PPARγ and acetylated histones H3/H4 were shown to bind to a region of the GIPR promoter containing the peroxisome proliferator response element (PPRE). Knockdown of PPARγ in differentiated 3T3-L1 adipocytes, using RNA interference, reduced GIPR expression, supporting a functional regulatory role. Taken together, these studies show that GIP and insulin act in a synergistic manner on 3T3-L1 cell development and that adipocyte GIPR expression is upregulated through a mechanism involving interactions between PPARγ and a GIPR promoter region containing an acetylated histone region.


Subject(s)
Adipocytes/metabolism , Histones/metabolism , PPAR gamma/metabolism , Receptors, Gastrointestinal Hormone/metabolism , 3T3-L1 Cells , Acetylation , Adipocytes/cytology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Chromatin Immunoprecipitation , Immunoprecipitation , Mice , PPAR gamma/genetics , Promoter Regions, Genetic , Protein Binding , RNA Interference , Receptors, Gastrointestinal Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
14.
Vitam Horm ; 84: 21-79, 2010.
Article in English | MEDLINE | ID: mdl-21094896

ABSTRACT

The insulin secretory response to a meal results largely from glucose stimulation of the pancreatic islets and both direct and indirect (autonomic) glucose-dependent stimulation by incretin hormones released from the gastrointestinal tract. Two incretins, Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), have so far been identified. Localization of the cognate G protein-coupled receptors for GIP and GLP-1 revealed that they are present in numerous tissues in addition to the endocrine pancreas, including the gastrointestinal, cardiovascular, central nervous and autonomic nervous systems (ANSs), adipose tissue, and bone. At these sites, the incretin hormones exert a range of pleiotropic effects, many of which contribute to the integration of processes involved in the regulation of food intake, and nutrient and mineral processing and storage. From detailed studies at the cellular and molecular level, it is also evident that both incretin hormones act via multiple signal transduction pathways that regulate both acute and long-term cell function. Here, we provide an overview of current knowledge relating to the physiological roles of GIP and GLP-1, with specific emphasis on their modes of action on islet hormone secretion, ß-cell proliferation and survival, central and autonomic neuronal function, gastrointestinal motility, and glucose and lipid metabolism. However, it is emphasized that despite intensive research on the various body systems, in many cases there is uncertainty as to the pathways by which the incretins mediate their pleiotropic effects and only a rudimentary understanding of the underlying cellular mechanisms involved, and these are challenges for the future.


Subject(s)
Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/physiology , Insulin/physiology , Islets of Langerhans/physiology , Animals , Humans , Insulin/metabolism , Insulin Secretion , Receptors, Glucagon/physiology
15.
J Lipid Res ; 51(11): 3145-57, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20693566

ABSTRACT

GIP (glucose-dependent insulinotropic polypeptide) is a gastrointestinal hormone that regulates pancreatic islet function. Additionally, emerging evidence suggests an important physiological role for GIP in the regulation of adipocyte metabolism. In previous studies on the lipogenic effects of GIP, it was shown to increase adipocyte lipoprotein lipase (LPL) activity in both differentiated 3T3-L1 cells and human adipocytes through a pathway involving activation of protein kinase B (PKB)/Akt. In the current study, we examined the effects of GIP on LPL gene expression. GIP in the presence of insulin increased LPL gene expression in human adipocytes and LPL promoter activity in GIP receptor-expressing HEK-293 cells, and both effects were greatly reduced by the transcription inhibitor actinomycin D. Subsequent studies established that GIP increased phosphorylation of Serine 133 in cAMP-response element binding protein (CREB) and the nuclear localization of cAMP-responsive CREB coactivator 2 (TORC2) through a pathway involving phosphatidylinositol 3-kinase (PI3-K), PKB, and AMP-activated protein kinase (AMPK). However, in the presence of insulin, GIP failed to activate the cAMP/PKA pathway. Knockdown of CREB and TORC2 using RNA interference reduced LPL expression, supporting a functional regulatory role. GIP-induced phospho-CREB and TORC2 were shown to bind to a cAMP-response element (-II) site in the human LPL promoter and GIP increased protein-protein interactions of these two factors. The lipogenic effects of GIP in the presence of insulin are therefore at least partially mediated by upregulation of adipocyte LPL gene transcription through a pathway involving PI3-K/PKB/AMPK-dependent CREB/TORC2 activation.


Subject(s)
Adipocytes/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Lipoprotein Lipase/genetics , Transcription Factors/metabolism , Transcriptional Activation/drug effects , AMP-Activated Protein Kinases/metabolism , Active Transport, Cell Nucleus/drug effects , Adipocytes/cytology , Adipocytes/enzymology , Adipocytes/metabolism , Adult , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Female , HEK293 Cells , Humans , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transcription, Genetic/drug effects
16.
Diabetes ; 59(7): 1739-50, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20368408

ABSTRACT

OBJECTIVE: Treatment of NOD mice with the dipeptidyl peptidase-IV (DPP-IV) inhibitor sitagliptin preserved islet transplants through a pathway involving modulation of splenic CD4(+) T-cell migration. In the current study, effects of sitagliptin on migration of additional subsets of CD4(+) T-cells were examined and underlying molecular mechanisms were further defined. RESEARCH DESIGN AND METHODS: Effects of sitagliptin on migration of NOD mouse splenic, thymic, and lymph node CD4(+) T-cells were determined. Signaling modules involved in DPP-IV-, Sitagliptin- and incretin-mediated modulation of CD4(+) T-cell migration were studied using Western blot and Rac1 and nuclear factor-kappaB (NF-kappaB) activity assays. RESULTS: Migration of splenic and lymph node CD4(+) T-cells of diabetic NOD mice was reduced by sitagliptin treatment. In vitro treatment of splenic, but not thymic or lymph node CD4(+) T-cells, from nondiabetic NOD mice with soluble (s) DPP-IV increased migration. Sitagliptin abolished sDPP-IV effects on splenic CD4(+) T-cell migration, whereas incretins decreased migration of lymph node, but not splenic, CD4(+) T-cells. Splenic CD4(+) T-cells demonstrating increased in vitro migration in response to sDPP-IV and lymph node CD4(+) T-cells that were nonresponsive to incretins selectively infiltrated islets of NOD mice, after injection. Sitagliptin decreases migration of splenic CD4(+) T-cells through a pathway involving Rac1/vasodilator-stimulated phosphoprotein, whereas its inhibitory effects on the migration of lymph node CD4(+) T-cells involve incretin-activation of the NF-kappaB pathway. CONCLUSIONS: Benefits of sitagliptin treatment in diabetic NOD mice may be mediated through selective effects on subpopulations of T-cells that are related to autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cell Movement/drug effects , Dipeptidyl Peptidase 4/metabolism , Incretins/metabolism , Pyrazines/pharmacology , T-Lymphocyte Subsets/drug effects , Triazoles/pharmacology , Analysis of Variance , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred NOD , Signal Transduction/drug effects , Signal Transduction/immunology , Sitagliptin Phosphate , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/metabolism
17.
PLoS One ; 5(3): e9590, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20231880

ABSTRACT

AIMS: The gastrointestinal hormone GIP promotes pancreatic islet function and exerts pro-survival actions on cultured beta-cells. However, GIP also promotes lipogenesis, thus potentially restricting its therapeutic use. The current studies evaluated the effects of a truncated GIP analog, D-Ala(2)-GIP(1-30) (D-GIP(1-30)), on glucose homeostasis and beta-cell mass in rat models of diabetes. MATERIALS AND METHODS: The insulinotropic and pro-survival potency of D-GIP(1-30) was evaluated in perfused pancreas preparations and cultured INS-1 beta-cells, respectively, and receptor selectivity evaluated using wild type and GIP receptor knockout mice. Effects of D-GIP(1-30) on beta-cell function and glucose homeostasis, in vivo, were determined using Lean Zucker rats, obese Vancouver diabetic fatty rats, streptozotocin treated rats, and obese Zucker diabetic fatty rats, with effects on beta-cell mass determined in histological studies of pancreatic tissue. Lipogenic effects of D-GIP(1-30) were evaluated on cultured 3T3-L1 adipocytes. RESULTS: Acutely, D-GIP(1-30) improved glucose tolerance and insulin secretion. Chronic treatment with D-GIP(1-30) reduced levels of islet pro-apoptotic proteins in Vancouver diabetic fatty rats and preserved beta-cell mass in streptozotocin treated rats and Zucker diabetic fatty rats, resulting in improved insulin responses and glycemic control in each animal model, with no change in body weight. In in vitro studies, D-GIP(1-30) exhibited equivalent potency to GIP(1-42) on beta-cell function and survival, but greatly reduced action on lipoprotein lipase activity in 3T3-L1 adipocytes. CONCLUSIONS: These findings demonstrate that truncated forms of GIP exhibit potent anti-diabetic actions, without pro-obesity effects, and that the C-terminus contributes to the lipogenic actions of GIP.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis , Diabetes Mellitus, Experimental/drug therapy , Insulin-Secreting Cells/metabolism , Receptors, Gastrointestinal Hormone/agonists , 3T3 Cells , Adipocytes/cytology , Animals , Glucose/metabolism , Glucose Tolerance Test , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Zucker , Receptors, Gastrointestinal Hormone/chemistry
18.
J Biol Chem ; 284(44): 30372-82, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19748889

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) potentiates glucose-stimulated insulin secretion, insulin biosynthesis, and beta-cell proliferation and survival. In previous studies GIP was shown to promote beta-cell survival by modulating the activity of multiple signaling modules and regulating gene transcription of pro- and anti-apoptotic bcl-2 family proteins. We have now evaluated the mechanisms by which GIP regulates the dynamic interactions between cytoplasmic bcl-2 family members and the mitochondria in INS-1 cells during apoptosis induced by treatment with staurosporine (STS), an activator of the mitochondria-mediated apoptotic pathway. STS induced translocation of bad and bimEL, activation of mitochondrial bax, release of mitochondrial cytochrome c, cleavage of caspase-3, and apoptosis. Each response was significantly diminished by GIP. Using selective enzyme inhibitors, overexpression of dominant-negative Akt, and Akt siRNA, it was demonstrated that GIP promoted beta-cell survival via Akt-dependent suppression of p38 MAPK and JNK and that combined inhibition was sufficient to explain the entire pro-survival responses to GIP during STS treatment. This signaling pathway also explained the pro-survival effects of GIP on INS-1 cells exposed to two other promoters of stress: thapsigargin (endoplasmic reticulum stress) and etoposide (genotoxic stress). Importantly, we discovered that GIP suppressed p38 MAPK and JNK via Akt-mediated changes in the phosphorylation state of the apoptosis signal-regulating kinase 1 in INS-1 cells and human islets, resulting in inhibition of its activity. Inhibition of apoptosis by GIP is therefore mediated via a key pathway involving Akt-dependent inhibition of apoptosis signal-regulating kinase 1, which subsequently prevents the pro-apoptotic actions of p38 MAPK and JNK.


Subject(s)
Gastric Inhibitory Polypeptide/physiology , Insulin-Secreting Cells/cytology , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Apoptosis , Cell Survival , Cells, Cultured , Humans , Islets of Langerhans/cytology , Signal Transduction/physiology
19.
Am J Physiol Endocrinol Metab ; 297(3): E695-707, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19602585

ABSTRACT

In addition to its primary role in regulating glucose production from the liver, glucagon has many other actions, reflected by the wide tissue distribution of the glucagon receptor (Gcgr). To investigate the role of glucagon in the regulation of insulin secretion and whole body glucose homeostasis in vivo, we generated mice overexpressing the Gcgr specifically on pancreatic beta-cells (RIP-Gcgr). In vivo and in vitro insulin secretion in response to glucagon and glucose was increased 1.7- to 3.9-fold in RIP-Gcgr mice compared with controls. Consistent with the observed increase in insulin release in response to glucagon and glucose, the glucose excursion resulting from both a glucagon challenge and intraperitoneal glucose tolerance test (IPGTT) was significantly reduced in RIP-Gcgr mice compared with controls. However, RIP-Gcgr mice display similar glucose responses to an insulin challenge. beta-Cell mass and pancreatic insulin content were also increased (20 and 50%, respectively) in RIP-Gcgr mice compared with controls. When fed a high-fat diet (HFD), both control and RIP-Gcgr mice developed similar degrees of obesity and insulin resistance. However, the severity of both fasting hyperglycemia and impaired glucose tolerance (IGT) were reduced in RIP-Gcgr mice compared with controls. Furthermore, the insulin response of RIP-Gcgr mice to an IPGTT was twice that of controls when fed the HFD. These data indicate that increased pancreatic beta-cell expression of the Gcgr increased insulin secretion, pancreatic insulin content, beta-cell mass, and, when mice were fed a HFD, partially protected against hyperglycemia and IGT.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Receptors, Glucagon/genetics , Animals , Cell Proliferation , Cell Size , Cells, Cultured , Diet, Atherogenic , Female , Glucose Intolerance/genetics , Hyperglycemia/genetics , Insulin/metabolism , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Organ Specificity/genetics , Receptors, Glucagon/metabolism , Transfection
20.
Hum Mol Genet ; 18(13): 2388-99, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19386626

ABSTRACT

Recent human genetics studies have revealed that common variants of the TCF7L2 (T-cell factor 7-like 2, formerly known as TCF4) gene are strongly associated with type 2 diabetes mellitus (T2DM). We have shown that TCF7L2 expression in the beta-cells is correlated with function and survival of the insulin-producing pancreatic beta-cell. In order to understand how variations in TCF7L2 influence diabetes progression, we investigated its mechanism of action in the beta-cell. We show robust differences in TCF7L2 expression between healthy controls and models of T2DM. While mRNA levels were approximately 2-fold increased in isolated islets from the diabetic db/db mouse, the Vancouver Diabetic Fatty (VDF) Zucker rat and the high fat/high sucrose diet-treated mouse compared with the non-diabetic controls, protein levels were decreased. A similar decrease was observed in pancreatic sections from patients with T2DM. In parallel, expression of the receptors for glucagon-like peptide 1 (GLP-1R) and glucose-dependent insulinotropic polypeptide (GIP-R) was decreased in islets from humans with T2DM as well as in isolated human islets treated with siRNA to TCF7L2 (siTCF7L2). Also, insulin secretion stimulated by glucose, GLP-1 and GIP, but not KCl or cyclic adenosine monophosphate (cAMP) was impaired in siTCF7L2-treated isolated human islets. Loss of TCF7L2 resulted in decreased GLP-1 and GIP-stimulated AKT phosphorylation, and AKT-mediated Foxo-1 phosphorylation and nuclear exclusion. Our findings suggest that beta-cell function and survival are regulated through an interplay between TCF7L2 and GLP-1R/GIP-R expression and signaling in T2DM.


Subject(s)
Down-Regulation , Insulin-Secreting Cells/physiology , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/genetics , TCF Transcription Factors/metabolism , Aged , Animals , Female , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Humans , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rats , Rats, Zucker , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Signal Transduction , TCF Transcription Factors/genetics , Transcription Factor 7-Like 2 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...