Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619103

ABSTRACT

We evaluated the potential for a monoclonal antibody antagonist of the glucagon receptor (Ab-4) to maintain glucose homeostasis in type 1 diabetic rodents. We noted durable and sustained improvements in glycemia which persist long after treatment withdrawal. Ab-4 promoted ß-cell survival and enhanced the recovery of insulin+ islet mass with concomitant increases in circulating insulin and C peptide. In PANIC-ATTAC mice, an inducible model of ß-cell apoptosis which allows for robust assessment of ß-cell regeneration following caspase-8-induced diabetes, Ab-4 drove a 6.7-fold increase in ß-cell mass. Lineage tracing suggests that this restoration of functional insulin-producing cells was at least partially driven by α-cell-to-ß-cell conversion. Following hyperglycemic onset in nonobese diabetic (NOD) mice, Ab-4 treatment promoted improvements in C-peptide levels and insulin+ islet mass was dramatically increased. Lastly, diabetic mice receiving human islet xenografts showed stable improvements in glycemic control and increased human insulin secretion.


Subject(s)
Antibodies, Monoclonal/pharmacology , Diabetes Mellitus, Experimental/therapy , Glucagon-Secreting Cells/drug effects , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Receptors, Glucagon/antagonists & inhibitors , Animals , Blood Glucose/metabolism , C-Peptide/metabolism , Cell Lineage/drug effects , Cell Transdifferentiation/drug effects , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Gene Expression , Glucagon/antagonists & inhibitors , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Islets of Langerhans Transplantation , Mice , Mice, Inbred NOD , Organ Size/drug effects , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Treatment Outcome
2.
J Biol Chem ; 294(8): 2935-2946, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30587572

ABSTRACT

Estrogen hormones play an important role in controlling glucose homeostasis and pancreatic ß-cell function. Despite the significance of estrogen hormones for regulation of glucose metabolism, little is known about the roles of endogenous estrogen metabolites in modulating pancreatic ß-cell function. In this study, we evaluated the effects of major natural estrogen metabolites, catechol estrogens, on insulin secretion in pancreatic ß-cells. We show that catechol estrogens, hydroxylated at positions C2 and C4 of the steroid A ring, rapidly potentiated glucose-induced insulin secretion via a nongenomic mechanism. 2-Hydroxyestrone, the most abundant endogenous estrogen metabolite, was more efficacious in stimulating insulin secretion than any other tested catechol estrogens. In insulin-secreting cells, catechol estrogens produced rapid activation of calcium influx and elevation in cytosolic free calcium. Catechol estrogens also generated sustained elevations in cytosolic free calcium and evoked inward ion current in HEK293 cells expressing the transient receptor potential A1 (TRPA1) cation channel. Calcium influx and insulin secretion stimulated by estrogen metabolites were dependent on the TRPA1 activity and inhibited with the channel-specific pharmacological antagonists or the siRNA. Our results suggest the role of estrogen metabolism in a direct regulation of TRPA1 activity with potential implications for metabolic diseases.


Subject(s)
Estrogens, Catechol/pharmacology , Gene Expression Regulation/drug effects , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , TRPA1 Cation Channel/metabolism , Animals , Cells, Cultured , Glucose/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Rats
4.
Genes Chromosomes Cancer ; 58(4): 208-218, 2019 04.
Article in English | MEDLINE | ID: mdl-30382602

ABSTRACT

MicroRNAs are small noncoding RNAs that modulate gene expression either directly, by impairing the stability and/or translation of transcripts that contain their specific target sequence, or indirectly through the targeting of transcripts that encode transcription factors, factors implicated in signal transduction pathways, or epigenetic regulators. Abnormal expression of micro-RNAs has been found in nearly all types of pathologies, including cancers. MiR-155 has been the first microRNA to be implicated in the regulation of the innate and adaptative immune responses, and its expression is either increased or decreased in a variety of liquid and solid malignancies. In this review, we examine the oncogenic and antitumor potentials of miR-155, with special emphasize on its dose-dependent effects. We describe the impact of miR-155 levels on antitumor activity of lymphocytes and myeloid cells. We discuss miR-155 dose-dependent effects in leukemias and analyze results showing that miR-155 intermediate levels tend to be detrimental, whereas high levels of miR-155 expression usually prove beneficial. We also examine the beneficial effects of high levels of miR-155 expression in solid tumors. We discuss the possible causal involvement of miR-155 in leukemias and dementia in individuals with Down's syndrome. We finally propose that increasing miR-155 levels in immune cells might increase the efficiency of newly developed cancer immunotherapies, due to miR-155 ability to target transcripts encoding immune checkpoints such as cytotoxic T lymphocyte antigen-4 or programmed death-ligand 1.


Subject(s)
Carcinogenesis/genetics , Leukemia/genetics , MicroRNAs/genetics , Tumor Escape/genetics , Animals , Carcinogenesis/immunology , Down Syndrome/genetics , Down Syndrome/immunology , Humans , Immunotherapy/methods , Leukemia/immunology , Leukemia/therapy , MicroRNAs/metabolism
5.
Am J Physiol Endocrinol Metab ; 305(10): E1319-26, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24085034

ABSTRACT

Extracellular ATP released from pancreatic ß-cells acts as a potent insulinotropic agent through activation of P2 purinergic receptors. Ectonucleotidases, a family of membrane-bound nucleotide-metabolizing enzymes, regulate extracellular ATP levels by degrading ATP and related nucleotides. Ectonucleotidase activity affects the relative proportion of ATP and its metabolites, which in turn will impact the level of purinergic receptor stimulation exerted by extracellular ATP. Therefore, we investigated the expression and role of ectonucleotidases in pancreatic ß-cells. Of the ectonucleotidases studied, only ENTPD3 (gene encoding the NTPDase3 enzyme) mRNA was detected at fairly abundant levels in human and mouse pancreatic islets as well as in insulin-secreting MIN6 cells. ARL67156, a selective ectonucleotidase inhibitor, blocked degradation of extracellular ATP that was added to MIN6 cells. The compound also decreased degradation of endogenous ATP released from cells. Measurements of insulin secretion in MIN6 cells as well as in mouse and human pancreatic islets demonstrated that ARL67156 potentiated glucose-dependent insulin secretion. Downregulation of NTPDase3 expression in MIN6 cells with the specific siRNA replicated the effects of ARL67156 on extracellular ATP hydrolysis and insulin secretion. Our results demonstrate that NTPDase3 is the major ectonucleotidase in pancreatic ß-cells in multiple species and that it modulates insulin secretion by controlling activation of purinergic receptors.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/enzymology , Insulin/metabolism , Pyrophosphatases/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glucose/pharmacology , Humans , Insulin Secretion , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Pyrophosphatases/analysis , Pyrophosphatases/antagonists & inhibitors , RNA, Messenger/analysis , RNA, Messenger/metabolism , Tissue Distribution
6.
Nat Commun ; 14(1): 2533, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37137910

ABSTRACT

We identify biomarkers for disease progression in three type 2 diabetes cohorts encompassing 2,973 individuals across three molecular classes, metabolites, lipids and proteins. Homocitrulline, isoleucine and 2-aminoadipic acid, eight triacylglycerol species, and lowered sphingomyelin 42:2;2 levels are predictive of faster progression towards insulin requirement. Of ~1,300 proteins examined in two cohorts, levels of GDF15/MIC-1, IL-18Ra, CRELD1, NogoR, FAS, and ENPP7 are associated with faster progression, whilst SMAC/DIABLO, SPOCK1 and HEMK2 predict lower progression rates. In an external replication, proteins and lipids are associated with diabetes incidence and prevalence. NogoR/RTN4R injection improved glucose tolerance in high fat-fed male mice but impaired it in male db/db mice. High NogoR levels led to islet cell apoptosis, and IL-18R antagonised inflammatory IL-18 signalling towards nuclear factor kappa-B in vitro. This comprehensive, multi-disciplinary approach thus identifies biomarkers with potential prognostic utility, provides evidence for possible disease mechanisms, and identifies potential therapeutic avenues to slow diabetes progression.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Mice , Animals , Male , Diabetes Mellitus, Type 2/metabolism , Blood Glucose/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Lipids , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Extracellular Matrix Proteins/metabolism
7.
Am J Physiol Endocrinol Metab ; 303(12): E1469-78, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23074242

ABSTRACT

The GPR119 receptor plays an important role in the secretion of incretin hormones in response to nutrient consumption. We have studied the ability of an array of naturally occurring endocannabinoid-like lipids to activate GPR119 and have identified several lipid receptor agonists. The most potent receptor agonists identified were three N-acylethanolamines: oleoylethanolamine (OEA), palmitoleoylethanolamine, and linoleylethanolamine (LEA), all of which displayed similar potency in activating GPR119. Another lipid, 2-oleoylglycerol (2-OG), also activated GPR119 receptor but with significantly lower potency. Endogenous levels of endocannabinoid-like lipids were measured in intestine in fasted and refed mice. Of the lipid GPR119 agonists studied, the intestinal levels of only OEA, LEA, and 2-OG increased significantly upon refeeding. Intestinal levels of OEA and LEA in the fasted mice were low. In the fed state, OEA levels only moderately increased, whereas LEA levels rose drastically. 2-OG was the most abundant of the three GPR119 agonists in intestine, and its levels were radically elevated in fed mice. Our data suggest that, in lean mice, 2-OG and LEA may serve as physiologically relevant endogenous GPR119 agonists that mediate receptor activation upon nutrient uptake.


Subject(s)
Cannabinoid Receptor Agonists/metabolism , Endocannabinoids/metabolism , Receptors, G-Protein-Coupled/agonists , Amides , Animals , Cannabinoid Receptor Agonists/chemistry , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Cell Line , Endocannabinoids/antagonists & inhibitors , Endocrine Cells/drug effects , Endocrine Cells/metabolism , Ethanolamines/antagonists & inhibitors , Ethanolamines/metabolism , Fasting/metabolism , Glucagon-Like Peptide 1/metabolism , Glycerides/antagonists & inhibitors , Glycerides/metabolism , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Oleic Acids/antagonists & inhibitors , Oleic Acids/metabolism , Organ Specificity , Palmitic Acids/antagonists & inhibitors , Palmitic Acids/metabolism , Random Allocation , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Thinness/metabolism , Up-Regulation
8.
Front Mol Neurosci ; 15: 788301, 2022.
Article in English | MEDLINE | ID: mdl-35185466

ABSTRACT

Aortic aneurism open repair surgery can cause spinal cord (SC) injury with 5-15% of patients developing paraparesis or paraplegia. Using a mouse model of transient aortic cross-clamping (ACC), we have previously found that the expression of proinflammatory microRNA miR-155 increases in motoneurons (MNs) and endothelial cells (ECs) of ischemic SCs, and that global miR-155 deletion decreases the percentage of paraplegia by 37.4% at 48-h post-ACC. Here, we investigated the cell-specific contribution of miR-155 in choline acetyltransferase-positive (ChAT+) neurons (that include all MNs of the SC) and ECs to SC injury after ACC. Mice lacking miR-155 in ChAT+ neurons (MN-miR-155-KO mice) developed 24.6% less paraplegia than control mice at 48-h post-ACC. In contrast, mice lacking miR-155 in ECs (ECs-miR-155-KO mice) experienced the same percentage of paraplegia as control mice, despite presenting smaller central cord edema. Unexpectedly, mice overexpressing miR-155 in ChAT+ neurons were less likely than control mice to develop early paraplegia during the first day post-ACC, however they reached the same percentage of paraplegia at 48-h. In addition, all mice overexpressing miR-155 in ECs (ECs-miR-155-KI mice) were paraplegic at 48-h post-ACC. Altogether, our results suggest that miR-155 activity in ChAT+ neurons protects the SC against ischemic injury during the first day post-ACC before becoming deleterious during the second day, which indicates that early and late paraplegias arise from different molecular malfunctions. These results point to the need to develop specific protective therapeutics aimed at inhibiting both the early and late deleterious events after open repair surgery of aortic aneurisms.

9.
J Neuropathol Exp Neurol ; 80(10): 895-911, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34534333

ABSTRACT

Spinal cord ischemic injury and paralysis are devastating complications after open surgical repair of thoracoabdominal aortic aneurysms. Preclinical models have been developed to simulate the clinical paradigm to better understand the neuropathophysiology and develop therapeutic treatment. Neuropathological findings in the preclinical models have not been comprehensively examined before. This systematic review studies the past 40 years of the histological findings after open surgical repair in preclinical models. Our main finding is that damage is predominantly in the grey matter of the spinal cord, although white matter damage in the spinal cord is also reported. Future research needs to examine the neuropathological findings in preclinical models after endovascular repair, a newer type of surgical repair used to treat aortic aneurysms.


Subject(s)
Aortic Aneurysm, Abdominal/pathology , Disease Models, Animal , Reperfusion Injury/pathology , Spinal Cord/blood supply , Spinal Cord/pathology , Animals , Aortic Aneurysm, Abdominal/complications , Aortic Aneurysm, Abdominal/surgery , Constriction , Dogs , Gray Matter/pathology , Humans , Mice , Papio , Rabbits , Rats , Reperfusion Injury/etiology , Sheep , Species Specificity , Spinal Cord Injuries/etiology , Spinal Cord Injuries/pathology , Swine
10.
Assay Drug Dev Technol ; 19(1): 27-37, 2021 01.
Article in English | MEDLINE | ID: mdl-33164547

ABSTRACT

Phenotypic screening is a neoclassical approach for drug discovery. We conducted phenotypic screening for insulin secretion enhancing agents using INS-1E insulinoma cells as a model system for pancreatic beta-cells. A principal regulator of insulin secretion in beta-cells is the metabolically regulated potassium channel Kir6.2/SUR1 complex. To characterize hit compounds, we developed an assay to quantify endogenous potassium channel activity in INS-1E cells. We quantified ligand-regulated potassium channel activity in INS-1E cells using fluorescence imaging and thallium flux. Potassium channel activity was metabolically regulated and coupled to insulin secretion. The pharmacology of channel opening agents (diazoxide) and closing agents (sulfonylureas) was used to validate the applicability of the assay. A precise high-throughput assay was enabled, and phenotypic screening hits were triaged to enable a higher likelihood of discovering chemical matter with novel and useful mechanisms of action.


Subject(s)
Diazoxide/pharmacology , Insulin-Secreting Cells/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Secretagogues/pharmacology , Sulfonylurea Compounds/pharmacology , Sulfonylurea Receptors/metabolism , Cells, Cultured , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Optical Imaging , Phenotype
11.
J Clin Invest ; 131(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34907913

ABSTRACT

Type 2 diabetes (T2D) is associated with defective insulin secretion and reduced ß cell mass. Available treatments provide a temporary reprieve, but secondary failure rates are high, making insulin supplementation necessary. Reversibility of ß cell failure is a key translational question. Here, we reverse engineered and interrogated pancreatic islet-specific regulatory networks to discover T2D-specific subpopulations characterized by metabolic inflexibility and endocrine progenitor/stem cell features. Single-cell gain- and loss-of-function and glucose-induced Ca2+ flux analyses of top candidate master regulatory (MR) proteins in islet cells validated transcription factor BACH2 and associated epigenetic effectors as key drivers of T2D cell states. BACH2 knockout in T2D islets reversed cellular features of the disease, restoring a nondiabetic phenotype. BACH2-immunoreactive islet cells increased approximately 4-fold in diabetic patients, confirming the algorithmic prediction of clinically relevant subpopulations. Treatment with a BACH inhibitor lowered glycemia and increased plasma insulin levels in diabetic mice, and restored insulin secretion in diabetic mice and human islets. The findings suggest that T2D-specific populations of failing ß cells can be reversed and indicate pathways for pharmacological intervention, including via BACH2 inhibition.


Subject(s)
Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic-Leucine Zipper Transcription Factors/metabolism , Calcium Signaling , Diabetes Mellitus, Type 2/metabolism , Epigenesis, Genetic , Insulin-Secreting Cells/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , HEK293 Cells , Humans
12.
Sci Rep ; 11(1): 7834, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33837260

ABSTRACT

Both endovascular repair (EVR) and open repair (OR) surgery of thoraco-abdominal aortic aneurysms cause spinal cord (SC) injury that can lead to paraparesis or paraplegia. It has been assumed that mechanisms responsible for SC damage after EVR are similar to those after OR. This pilot study compared the pathophysiology of SC injury after EVR versus OR using a newly developed EVR dog model. An increasing number of stents similar to those used in patients were inserted in the aorta of three dogs to ensure thoracic or thoracic plus lumbar coverage. The aorta of OR dogs was cross-clamped for 45 min. Behavior assessment demonstrated unique patterns of proprioceptive ataxia and evolving paraparesis in EVR versus irreversible paraplegia in OR. MRI showed posterior signal in lumbar SC after EVR versus central cord edema after OR. Histopathology showed white matter edema in L3-L5 localized to the dorsal column medial lemniscus area associated with loss of myelin basic protein but not neurons after EVR, versus massive neuronal loss in the gray matter in L3-L5 after OR. Metabolome analysis demonstrates a distinctive chemical fingerprint of cellular processes in both interventions. Our results call for the development of new therapeutics tailored to these distinct pathophysiologic findings.


Subject(s)
Aortic Aneurysm, Thoracic/surgery , Blood Vessel Prosthesis Implantation/adverse effects , Blood Vessel Prosthesis/adverse effects , Endovascular Procedures/adverse effects , Endovascular Procedures/methods , Postoperative Complications/etiology , Spinal Cord Injuries/etiology , Stents/adverse effects , Animals , Behavior, Animal , Computed Tomography Angiography/methods , Disease Models, Animal , Dogs , Magnetic Resonance Imaging/methods , Male , Metabolome , Paraplegia/etiology , Pilot Projects , Postoperative Complications/diagnostic imaging , Spinal Cord/diagnostic imaging , Spinal Cord Injuries/diagnostic imaging , Treatment Outcome
13.
J Endocr Soc ; 3(7): 1423-1444, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31286109

ABSTRACT

Oxytocin (OXT) has been shown to suppress appetite, induce weight loss, and improve glycemic control and lipid metabolism in several species, including humans, monkeys, and rodents. However, OXT's short half-life in circulation and lack of receptor selectivity limit its application and efficacy. In this study, we report an OXT peptide analog (OXTGly) that is potent and selective for the OXT receptor (OXTR). OXT, but not OXTGly, activated vasopressin receptors in vitro and acutely increased blood pressure in vivo when administered IP. OXT suppressed food intake in mice, whereas OXTGly had a moderate effect on food intake when administered IP or intracerebroventricularly. Both OXT (IP) and OXTGly (IP) improved glycemic control in glucose tolerance tests. Additionally, both OXT (IP) and OXTGly (IP) stimulated insulin, glucagon-like peptide 1, and glucagon secretion in mice. We generated lipid-conjugated OXT (acylated-OXT) and OXTGly (acylated-OXTGly) and demonstrated that these molecules have significantly extended half-lives in vivo. Compared with OXT, 2-week treatment of diet-induced obese mice with acylated-OXT [subcutaneous(ly) (SC)] resulted in enhanced body weight reduction, an improved lipid profile, and gene expression changes consistent with increased lipolysis and decreased gluconeogenesis. Treatment with acylated-OXTGly (SC) also resulted in a statistically significant weight loss, albeit to a lesser degree compared with acylated-OXT treatment. In conclusion, we demonstrate that selective activation of the OXTR pathway results in both acute and chronic metabolic benefits, whereas potential activation of vasopressin receptors by nonselective OXT analogs causes physiological stress that contributes to additional weight loss.

14.
Mol Metab ; 11: 205-211, 2018 05.
Article in English | MEDLINE | ID: mdl-29506910

ABSTRACT

OBJECTIVE: GPR142 agonists are being pursued as novel diabetes therapies by virtue of their insulin secretagogue effects. But it is undetermined whether GPR142's functions in pancreatic islets are limited to regulating insulin secretion. The current study expands research on its action. METHODS AND RESULTS: We demonstrated by in situ hybridization and immunostaining that GPR142 is expressed not only in ß cells but also in a subset of α cells. Stimulation of GPR142 by a selective agonist increased glucagon secretion in both human and mouse islets. More importantly, the GPR142 agonist also potentiated glucagon-like peptide-1 (GLP-1) production and its release from islets through a mechanism that involves upregulation of prohormone convertase 1/3 expression. Strikingly, stimulation of insulin secretion and increase in insulin content via GPR142 engagement requires intact GLP-1 receptor signaling. Furthermore, GPR142 agonist increased ß cell proliferation and protected both mouse and human islets against stress-induced apoptosis. CONCLUSIONS: Collectively, we provide here evidence that local GLP-1 release from α cells defines GPR142's beneficial effects on improving ß cell function and mass, and we propose that GPR142 agonism may have translatable and durable efficacy for the treatment of type 2 diabetes.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Proprotein Convertase 1/metabolism
15.
Endocrinology ; 148(4): 1843-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17194744

ABSTRACT

Liver X receptors (LXRs) form functional heterodimers with the retinoid X receptors (RXRs) and regulate cholesterol, lipid, and glucose metabolism. We demonstrated previously that activation of LXR modulates insulin secretion in MIN6 cells and pancreatic islets. In this study we investigated the effects of the LXR agonist T0901317 and the RXR agonist 9-cis-retinoic acid (9cRA) on cell proliferation and apoptosis in MIN6 cells. Whereas T0901317 showed no effect on proliferation of MIN6 cells, combination of T0901317 with 9cRA inhibited cell proliferation. Flow cytometry analysis of cell cycle demonstrated that activation of LXR/RXR prevented MIN6 cells from G1 to G2 phase progression. Combination of T0901317 and 9cRA increased apoptosis rate and caspase-3/7 activity in MIN6 cells. Moreover, T0901317 or its combination with 9cRA significantly increased the cell susceptibility to free fatty acid- and cytokine-induced apoptosis. Treatment of MIN6 cells with LXR and RXR agonists produced a strong increase in expression of mothers against decapentaplegic homolog 3, a protein known to inhibit cell cycle G1/S phase progression and induce apoptosis. In isolated rat islets, the effect of palmitic acid on caspase-3/7 activity was increased with T0901317 alone and even more with the combination of T0901317 and 9cRA. Thus, activation of LXR/RXR signaling inhibits cell proliferation and induces apoptosis in pancreatic beta-cells.


Subject(s)
Apoptosis , Cell Proliferation , DNA-Binding Proteins/metabolism , Insulin-Secreting Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoid X Receptors/metabolism , Animals , Caspases/metabolism , Cells, Cultured , DNA-Binding Proteins/agonists , Hydrocarbons, Fluorinated , Insulin-Secreting Cells/drug effects , Liver X Receptors , Male , Mice , Orphan Nuclear Receptors , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/agonists , Retinoid X Receptors/agonists , Sulfonamides/pharmacology , Transcriptional Activation
16.
Diabetes ; 55(9): 2470-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936195

ABSTRACT

Fibroblast growth factor-21 (FGF-21) is a recently discovered metabolic regulator. Here, we investigated the effects of FGF-21 in the pancreatic beta-cell. In rat islets and INS-1E cells, FGF-21 activated extracellular signal-regulated kinase 1/2 and Akt signaling pathways. In islets isolated from healthy rats, FGF-21 increased insulin mRNA and protein levels but did not potentiate glucose-induced insulin secretion. Islets and INS-1E cells treated with FGF-21 were partially protected from glucolipotoxicity and cytokine-induced apoptosis. In islets isolated from diabetic rodents, FGF-21 treatment increased islet insulin content and glucose-induced insulin secretion. Short-term treatment of normal or db/db mice with FGF-21 lowered plasma levels of insulin and improved glucose clearance compared with vehicle after oral glucose tolerance testing. Constant infusion of FGF-21 for 8 weeks in db/db mice nearly normalized fed blood glucose levels and increased plasma insulin levels. Immunohistochemistry of pancreata from db/db mice showed a substantial increase in the intensity of insulin staining in islets from FGF-21-treated animals as well as a higher number of islets per pancreas section and of insulin-positive cells per islet compared with control. No effect of FGF-21 was observed on islet cell proliferation. In conclusion, preservation of beta-cell function and survival by FGF-21 may contribute to the beneficial effects of this protein on glucose homeostasis observed in diabetic animals.


Subject(s)
Fibroblast Growth Factors/pharmacology , Insulin-Secreting Cells/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Diabetes Mellitus, Type 2/metabolism , Glucose Tolerance Test , Insulin/biosynthesis , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulinoma/metabolism , Male , Membrane Proteins/metabolism , Mice , Phosphorylation , Rats , Signal Transduction/drug effects
17.
Endocrinology ; 158(11): 3859-3873, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28938487

ABSTRACT

Incretin and insulin responses to nutrient loads are suppressed in persons with diabetes, resulting in decreased glycemic control. Agents including sulfonylureas and dipeptidyl peptidase-4 inhibitors (DPP4i) partially reverse these effects and provide therapeutic benefit; however, their modes of action limit efficacy. Because somatostatin (SST) has been shown to suppress insulin and glucagonlike peptide-1 (GLP-1) secretion through the Gi-coupled SST receptor 5 (SSTR5) isoform in vitro, antagonism of SSTR5 may improve glycemic control via intervention in both pathways. Here, we show that a potent and selective SSTR5 antagonist reverses the blunting effects of SST on insulin secretion from isolated human islets, and demonstrate that SSTR5 antagonism affords increased levels of systemic GLP-1 in vivo. Knocking out Sstr5 in mice provided a similar increase in systemic GLP-1 levels, which were not increased further by treatment with the antagonist. Treatment of mice with the SSTR5 antagonist in combination with a DPP4i resulted in increases in systemic GLP-1 levels that were more than additive and resulted in greater glycemic control compared with either agent alone. In isolated human islets, the SSTR5 antagonist completely reversed the inhibitory effect of exogenous SST-14 on insulin secretion. Taken together, these data suggest that SSTR5 antagonism should increase circulating GLP-1 levels and stimulate insulin secretion (directly and via GLP-1) in humans, improving glycemic control in patients with diabetes.


Subject(s)
Benzoates/pharmacology , Glucagon-Like Peptide 1/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Receptors, Somatostatin/antagonists & inhibitors , Spiro Compounds/pharmacology , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , HEK293 Cells , Humans , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptors, Somatostatin/genetics , Secretory Pathway/drug effects
18.
Endocrinology ; 147(8): 3898-905, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16644917

ABSTRACT

Liver X receptors (LXRalpha and LXRbeta) regulate glucose and lipid metabolism. Pancreatic beta-cells and INS-1E insulinoma cells express only the LXRbeta isoform. Activation of LXRbeta with the synthetic agonist T0901317 increased glucose-induced insulin secretion and insulin content, whereas deletion of the receptor in LXRbeta knockout mice severely blunted insulin secretion. Analysis of gene expression in LXR agonist-treated INS-1E cells and islets from LXRbeta-deficient mice revealed that LXRbeta positively regulated expression of ATP-binding cassette transporter A1 (ABCA1), sterol regulatory element-binding protein 1 (SREBP-1), insulin, PDX-1, glucokinase, and glucose transporter 2 (Glut2). Down-regulation of SREBP-1 expression with the specific small interfering RNA blocked basal and LXRbeta-induced expression of pancreatic duodenal homeobox 1 (PDX-1), insulin, and Glut2 genes. SREBP-1 small interfering RNA also prevented an increase in insulin secretion and insulin content induced by T0901317. Moreover, 5-(tetradecyloxy)-2-furoic acid, an inhibitor of the SREBP-1 target gene acetyl-coenzyme A carboxylase, blocked T0901317-induced stimulation of insulin secretion. In conclusion, activation of LXRbeta in pancreatic beta-cells increases insulin secretion and insulin mRNA expression via SREBP-1-regulated pathway. These data support the role of LXRbeta, SREBP-1, and cataplerosis/anaplerosis pathways in the control of insulin secretion in pancreatic beta-cells.


Subject(s)
DNA-Binding Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Alternative Splicing , Animals , Cell Line, Tumor , DNA-Binding Proteins/agonists , Gene Expression Regulation/physiology , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hydrocarbons, Fluorinated , Insulin Secretion , Insulinoma , Islets of Langerhans/cytology , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Pancreatic Neoplasms , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Cytoplasmic and Nuclear/agonists , Sterol Regulatory Element Binding Protein 1/genetics , Sulfonamides/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism
19.
PLoS One ; 11(6): e0157298, 2016.
Article in English | MEDLINE | ID: mdl-27322810

ABSTRACT

GPR142, a putative amino acid receptor, is expressed in pancreatic islets and the gastrointestinal tract, but the ligand affinity and physiological role of this receptor remain obscure. In this study, we show that in addition to L-Tryptophan, GPR142 signaling is also activated by L-Phenylalanine but not by other naturally occurring amino acids. Furthermore, we show that Tryptophan and a synthetic GPR142 agonist increase insulin and incretin hormones and improve glucose disposal in mice in a GPR142-dependent manner. In contrast, Phenylalanine improves in vivo glucose disposal independently of GPR142. Noteworthy, refeeding-induced elevations in insulin and glucose-dependent insulinotropic polypeptide are blunted in Gpr142 null mice. In conclusion, these findings demonstrate GPR142 is a Tryptophan receptor critically required for insulin and incretin hormone regulation and suggest GPR142 agonists may be effective therapies that leverage amino acid sensing pathways for the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Phenylalanine/metabolism , Receptors, G-Protein-Coupled/genetics , Tryptophan/metabolism , Animals , Blood Glucose , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Glucose/genetics , Humans , Incretins/genetics , Incretins/metabolism , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Phenylalanine/administration & dosage , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/drug effects , Tryptophan/administration & dosage
20.
Endocrinology ; 146(9): 3696-701, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15919746

ABSTRACT

The glucose-sensing enzyme glucokinase (GK) plays a key role in glucose metabolism. We report here the effects of a novel glucokinase activator, LY2121260. The activator enhanced GK activity via binding to the allosteric site located in the hinge region of the enzyme. LY2121260 stimulated insulin secretion in a glucose-dependent manner in pancreatic beta-cells and increased glucose use in rat hepatocytes. In addition, incubation of beta-cells with the GK activator resulted in increased GK protein levels, suggesting that enhanced insulin secretion on chronic treatment with a GK activator may be due to not only changed enzyme kinetics but also elevated enzyme levels. Animals treated with LY2121260 showed an improved glucose tolerance after oral glucose challenge. These results support the concept that GK activators represent a new class of compounds that increase both insulin secretion and hepatic glucose use and in doing so may prove to be effective agents for the control of blood glucose levels in patients with type 2 diabetes.


Subject(s)
Enzyme Activators/pharmacology , Glucokinase/metabolism , Hepatocytes/drug effects , Islets of Langerhans/drug effects , Sulfones/pharmacology , Thiazoles/pharmacology , Animals , Blood Glucose/drug effects , Cells, Cultured , Crystallography , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Glucokinase/chemistry , Hepatocytes/cytology , Hepatocytes/enzymology , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/enzymology , Male , Protein Structure, Tertiary , Rats , Rats, Wistar , Sulfones/chemistry , Thiazoles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL