Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.170
Filter
1.
Nat Nanotechnol ; 19(2): 237-245, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37813939

ABSTRACT

Insulin binds the insulin receptor (IR) and regulates anabolic processes in target tissues. Impaired IR signalling is associated with multiple diseases, including diabetes, cancer and neurodegenerative disorders. IRs have been reported to form nanoclusters at the cell membrane in several cell types, even in the absence of insulin binding. Here we exploit the nanoscale spatial organization of the IR to achieve controlled multivalent receptor activation. To control insulin nanoscale spatial organization and valency, we developed rod-like insulin-DNA origami nanostructures carrying different numbers of insulin molecules with defined spacings. Increasing the insulin valency per nanostructure markedly extended the residence time of insulin-DNA origami nanostructures at the receptors. Both insulin valency and spacing affected the levels of IR activation in adipocytes. Moreover, the multivalent insulin design associated with the highest levels of IR activation also induced insulin-mediated transcriptional responses more effectively than the corresponding monovalent insulin nanostructures. In an in vivo zebrafish model of diabetes, treatment with multivalent-but not monovalent-insulin nanostructures elicited a reduction in glucose levels. Our results show that the control of insulin multivalency and spatial organization with nanoscale precision modulates the IR responses, independent of the insulin concentration. Therefore, we propose insulin nanoscale organization as a design parameter in developing new insulin therapies.


Subject(s)
DNA , Nanostructures , Receptor, Insulin , Animals , Diabetes Mellitus/drug therapy , DNA/chemistry , Insulin , Nanostructures/chemistry , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Zebrafish
2.
Endocrinology ; 162(9)2021 09 01.
Article in English | MEDLINE | ID: mdl-33951176

ABSTRACT

Fibroblast growth factor (FGF) 21 is a member of the FGF family of proteins. The biological activity of FGF21 was first shown to induce insulin-independent glucose uptake in adipocytes through the GLUT1 transporter. Subsequently, it was shown to have effects on the liver to increase fatty acid oxidation. FGF21 treatment provides beneficial metabolic effects in both animal models and patients with obesity, type 2 diabetes mellitus (T2D) and/or fatty liver disease. In this paper, we revisited the original finding and found that insulin-independent glucose uptake in adipocytes is preserved in the presence of an insulin receptor antagonist. Using a 40-kDa PEGylated (PEG) and half-life extended form of FGF21 (FGF21-PEG), we extended these in vitro results to 2 different mouse models of diabetes. FGF21-PEG normalized plasma glucose in streptozotocin-treated mice, a model of type 1 diabetes (T1D), without restoring pancreatic ß-cell function. FGF21-PEG also normalized plasma glucose levels and improved glucose tolerance in mice chronically treated with an insulin competitive insulin receptor antagonist, a model of autoimmune/type-B insulin resistance. These data extend the pharmacological potential of FGF21 beyond the settings of T2D, fatty liver, and obesity.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Fibroblast Growth Factors/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , HEK293 Cells , Humans , Hyperglycemia/blood , Hyperglycemia/etiology , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Insulin/metabolism , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/blood , Obesity/complications , Obesity/pathology , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/drug effects , Receptor, Insulin/physiology , Streptozocin
3.
J Neurosci ; 41(11): 2360-2372, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33514676

ABSTRACT

Human fMRI studies show that insulin influences brain activity in regions that mediate reward and motivation, including the nucleus accumbens (NAc). Insulin receptors are expressed by NAc medium spiny neurons (MSNs), and studies of cultured cortical and hippocampal neurons suggest that insulin influences excitatory transmission via presynaptic and postsynaptic mechanisms. However, nothing is known about how insulin influences excitatory transmission in the NAc. Furthermore, insulin dysregulation accompanying obesity is linked to cognitive decline, depression, anxiety, and altered motivation that rely on NAc excitatory transmission. Using whole-cell patch-clamp and biochemical approaches, we determined how insulin affects NAc glutamatergic transmission in nonobese and obese male rats and the underlying mechanisms. We find that there are concentration-dependent, bidirectional effects of insulin on excitatory transmission, with insulin receptor activation increasing and IGF receptor activation decreasing NAc excitatory transmission. Increases in excitatory transmission were mediated by activation of postsynaptic insulin receptors located on MSNs. However, this effect was due to an increase in presynaptic glutamate release. This suggested feedback from MSNs to presynaptic terminals. In additional experiments, we found that insulin-induced increases in presynaptic glutamate release are mediated by opioid receptor-dependent disinhibition. Furthermore, obesity resulted in a loss of insulin receptor-mediated increases in excitatory transmission and a reduction in NAc insulin receptor surface expression, while preserving reductions in transmission mediated by IGF receptors. These results provide the first insights into how insulin influences excitatory transmission in the adult brain, and evidence for a previously unidentified form of opioid receptor-dependent disinhibition of NAc glutamatergic transmission.SIGNIFICANCE STATEMENT Data here provide the first insights into how insulin influences excitatory transmission in the adult brain, and identify previously unknown interactions between insulin receptor activation, opioids, and glutamatergic transmission. These data contribute to our fundamental understanding of insulin's influence on brain motivational systems and have implications for the use of insulin as a cognitive enhancer and for targeting of insulin receptors and IGF receptors to alter motivation.


Subject(s)
Endorphins/pharmacology , Glutamic Acid/metabolism , Insulin/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Receptor, Insulin/drug effects , Synaptic Transmission/drug effects , Animals , Diet, High-Fat , Male , Neurons/drug effects , Obesity/genetics , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/agonists , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D2/drug effects
4.
Nucleic Acids Res ; 49(2): 700-712, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33410883

ABSTRACT

Aptamers are single-stranded oligonucleotides that bind to a specific target with high affinity, and are widely applied in biomedical diagnostics and drug development. However, the use of aptamers has largely been limited to simple binders or inhibitors that interfere with the function of a target protein. Here, we show that an aptamer can also act as a positive allosteric modulator that enhances the activation of a receptor by stabilizing the binding of a ligand to that receptor. We developed an aptamer, named IR-A43, which binds to the insulin receptor, and confirmed that IR-A43 and insulin bind to the insulin receptor with mutual positive cooperativity. IR-A43 alone is inactive, but, in the presence of insulin, it potentiates autophosphorylation and downstream signaling of the insulin receptor. By using the species-specific activity of IR-A43 at the human insulin receptor, we demonstrate that residue Q272 in the cysteine-rich domain is directly involved in the insulin-enhancing activity of IR-A43. Therefore, we propose that the region containing residue Q272 is a hotspot that can be used to enhance insulin receptor activation. Moreover, our study implies that aptamers are promising reagents for the development of allosteric modulators that discriminate a specific conformation of a target receptor.


Subject(s)
Antigens, CD/drug effects , Aptamers, Nucleotide/pharmacology , Receptor, Insulin/drug effects , Allosteric Regulation , Animals , Antigens, CD/chemistry , Antigens, CD/metabolism , Cells, Cultured , Cricetinae , Glutamine/chemistry , Humans , Insulin/metabolism , Mice , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Protein Processing, Post-Translational , Rats , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , SELEX Aptamer Technique , Stimulation, Chemical
5.
Pharm Biol ; 58(1): 1199-1210, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33264567

ABSTRACT

CONTEXT: The Chinese herbal formula Heshouwu decoction (Heshouwuyin) has protective effects on testicular function in aging male rats, but the mechanism is unknown. OBJECTIVE: This study investigated whether Heshouwuyin affects the testicular function of aging rats by regulating the insulin/IGF signalling pathway. MATERIALS AND METHODS: Sixteen-month-old male Wistar rats in the Heshouwuyin group and the natural-aging group were orally administered Heshouwuyin granules (0.056 g/kg) or equivalent normal saline for 60 d. The testicular tissue of 12-month-old male Wistar rats was removed as a young control group (n = 10). The testicular tissue and spermatogenic cells were studied. RESULTS: The immunofluorescence results revealed that the insulin receptor (INSR)- (0.056 ± 0.00548), insulin receptor substrate 1(IRS1)- (0.251 ± 0.031), IRS2 (0.230 ± 0.019)- and insulin-like growth factor 1 (IGF1)-positive cell rate (0.33 ± 0.04) in the aging group was higher than that in the young control group (0.116 ± 0.011, 0.401 ± 0.0256, 0.427 ± 0.031, 0.56 ± 0.031; p < 0.01), and the IGF-binding protein 3 (IGFBP3)-positive cell rate (0.42 ± 0.024) was lower than that (0.06 ± 0.027) in the young group (p < 0.01). The intervention of Heshouwuyin reversed the above phenomena. The qPCR and immunoblot results were consistent with those of the immunofluorescence. The same results were obtained in spermatogenic cells. CONCLUSIONS: Our research shows that Heshouwuyin can regulate the insulin/IGF signalling pathway to improve testicular function, and provides an experimental basis for further clinical use.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gene Expression/drug effects , Insulin-Like Growth Factor I/drug effects , Insulin , Signal Transduction/drug effects , Spermatogenesis/drug effects , Spermatogenesis/genetics , Testis/drug effects , Animals , Cellular Senescence/drug effects , Insulin-Like Growth Factor Binding Protein 3/drug effects , Insulin-Like Growth Factor Binding Protein 3/metabolism , Male , Rats , Rats, Wistar , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Sertoli Cells/drug effects , Stem Cells/drug effects , Testis/cytology , Testis/metabolism
6.
Cells ; 9(5)2020 04 29.
Article in English | MEDLINE | ID: mdl-32365498

ABSTRACT

The insulin-like growth factor (IGF)-axis was implicated in cancer progression and identified as a clinically important therapeutic target. Several IGF-I receptor (IGF-IR) targeting drugs including humanized monoclonal antibodies have advanced to phase II/III clinical trials, but to date, have not progressed to clinical use, due, at least in part, to interference with insulin receptor signaling and compensatory signaling by the insulin receptor (IR) isoform A that can bind IGF-II and initiate mitogenic signaling. Here we briefly review the current state of IGF-targeting biologicals, discuss some factors that may be responsible for their poor performance in the clinic and outline the stepwise bioengineering and validation of an IGF-Trap-a novel anti-cancer therapeutic that could bypass these limitations. The IGF-Trap is a heterotetramer, consisting of the entire extracellular domain of the IGF-IR fused to the Fc portion of human IgG1. It binds human IGF-I and IGF-II with a three-log higher affinity than insulin and could inhibit IGF-IR driven cellular functions such as survival, proliferation and invasion in multiple carcinoma cell models in vitro. In vivo, the IGF-Trap has favorable pharmacokinetic properties and could markedly reduce metastatic outgrowth of colon and lung carcinoma cells in the liver, outperforming IGF-IR and ligand-binding monoclonal antibodies. Moreover, IGF-Trap dose-response profiles correlate with their bio-availability profiles, as measured by the IGF kinase receptor-activation (KIRA) assay, providing a novel, surrogate biomarker for drug efficacy. Our studies identify the IGF-Trap as a potent, safe, anti-cancer therapeutic that could overcome some of the obstacles encountered by IGF-targeting biologicals that have already been evaluated in clinical settings.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor, IGF Type 1/drug effects , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Antibodies, Monoclonal/metabolism , Humans , Insulin-Like Growth Factor I/drug effects , Pharmaceutical Preparations/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/drug effects
7.
Food Funct ; 11(2): 1455-1466, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-31974538

ABSTRACT

Both a diet rich in fructose and chronic stress exposure induce metabolic and cardiovascular disturbances. The aim of this study was to examine the effects of the fructose-rich diet and chronic stress, separately and in combination, on insulin signaling and molecules regulating glycogen synthesis and ion transport in the heart, and to reveal whether these effects coincide with changes in glucocorticoid receptor (GR) activation. Male Wistar rats were subjected to 10% fructose in drinking water and/or to chronic unpredictable stress for 9 weeks. Protein expression and/or phosphorylation of the insulin receptor (IR), protein tyrosine phosphatase 1B, insulin receptor substrate 1 (IRS1), protein kinase B (Akt), extracellular signal-regulated kinase 1/2 (ERK1/2), glycogen synthase kinase-3ß (GSK-3ß) and Na+/K+-ATPase α-subunits in cardiac tissue were analyzed by western blot. GR distribution between cytosolic and nuclear fractions was also analyzed. The fructose-rich diet decreased the level of pERK1/2 (Thr202/Tyr204) and pGSK-3ß (Ser9) independently of stress, while chronic stress increased the IRS1 content and prevented the fructose diet-induced decrease of the pAkt (Ser473) level. The fructose-rich diet in combination with chronic stress reduced the protein content of cardiac IR and attenuated IRS1 upregulation. Separate treatments increased the protein content of Na+/K+-ATPase α1- and α2-subunits, while after combined treatment the α2 content was at the control level and the α1 content was lower than the control level. The effect of combined treatment on cardiac IR and α2-subunit expression could be mediated by increased GR nuclear accumulation. Our study provides new insights into the effects of chronic stress and a combination of the fructose diet and chronic stress on the studied molecules in the heart.


Subject(s)
Fructose/pharmacology , Glycogen Synthase Kinase 3 beta/drug effects , Heart/drug effects , Receptor, Insulin/drug effects , Sodium-Potassium-Exchanging ATPase/drug effects , Animals , Diet , Glycogen Synthase Kinase 3 beta/metabolism , Male , Rats , Rats, Wistar , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Stress, Physiological
8.
Article in Chinese | MEDLINE | ID: mdl-31245954

ABSTRACT

OBJECTIVE: To observe the effects of AdipoRon orally on the functions of spleen and pancreas in type 2 diabetic mice, in order to present data for clinical application. METHODS: Forty C57/BL6 male mice were randomly divided into 2 groups: normal control group (n=10) and model group (n=30), the former group was fed normally, while the later group was fed with high fat and sugar for 4 weeks.After that, type 2 diabetes model was established in DM group induced by intraperitoneal injection of streptozotocin (STZ, 40 mg/kg).As type 2 diabetes model established successfully, the model mice were randomly divided into three groups (n=10): diabetes mellitus (DM) group, high dose of AdipoRon group (DM + H) and low dose of adiponRon group (DM + L).All the four groups were treated with saline, saline, AdipoRon at the doses of 20 mg/kg and 50 mg/kg by gavages respectively, once a day for 10 days.And then put them to death for collecting blood, pancreas and spleen.Pathological changes of pancreas were observed with a light microscope after HE staining.Protein contents of insulin receptor (INSR), insulin receptor substrate 1( IRS-1) and tumor necrosis factor-α(TNF-α) in pancreatic and spleen tissues were detected by ELISA.The protein level of phosphorylation insulin receptor substrate 1(p-IRS-1) in pancreas was determined by Western blot, and the expression of insulin mRNA in pancreas was tested by RT-PCR. RESULTS: Under the light microscope, it was visible that the pancreatic tissue in NC group was full and closely packed, and the islet was big.Pancreatic tissue of DM mice was incompact and the islet of DM mice was smaller than that of normal mice.As for the mice treated with AdipoRon orally, the pancreatic tissue was full and closely arranged, and the islet was slightly smaller.Compared with NC group, the levels of TNF-α in pancreas and spleen of DM group were increased markedly, the levels of INSR and IRS-1 were decreased, the spleen coefficient, p-IR-1 protein level and insulin mRNA expression in pancreas were decreased, all were significant statistically (P<0.05).Compared with DM group, the levels of TNF-α in pancreas and spleen of AdipoRon groups were decreased, the levels of INSR and IRS-1 in pancreas and spleen of AdipoRon groups were increased, while the spleen coefficient was increased (P<0.05).The p-IRS-1 protein level and insulin mRNA expression in pancreas in DM+H group were increased (P<0.05).Compared with DM + L group, the level of TNF-α was decreased, and the levels of INSR and IRS-1 were significantly increased (P<0.05) in DM + H group (P<0.05). CONCLUSION: Oral administration of AdipoRon can protect the spleen and pancreas of diabetic mice by decreasing the inflammatory response, up-regulating the expression of INSR, and increasing p-IRS-1 level in diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Piperidines , Spleen , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Inflammation , Insulin , Insulin Receptor Substrate Proteins/drug effects , Male , Mice , Pancreas , Piperidines/pharmacology , Random Allocation , Receptor, Insulin/drug effects , Spleen/drug effects
9.
Nutrients ; 11(3)2019 Mar 16.
Article in English | MEDLINE | ID: mdl-30884834

ABSTRACT

Individuals widely use non-nutritive sweeteners (NNS) in attempts to lower their overall daily caloric intake, lose weight, and sustain a healthy diet. There are insufficient scientific data that support the safety of consuming NNS. However, recent studies have suggested that NNS consumption can induce gut microbiota dysbiosis and promote glucose intolerance in healthy individuals that may result in the development of type 2 diabetes mellitus (T2DM). This sequence of events may result in changes in the gut microbiota composition through microRNA (miRNA)-mediated changes. The mechanism(s) by which miRNAs alter gene expression of different bacterial species provides a link between the consumption of NNS and the development of metabolic changes. Another potential mechanism that connects NNS to metabolic changes is the molecular crosstalk between the insulin receptor (IR) and G protein-coupled receptors (GPCRs). Here, we aim to highlight the role of NNS in obesity and discuss IR-GPCR crosstalk and miRNA-mediated changes, in the manipulation of the gut microbiota composition and T2DM pathogenesis.


Subject(s)
Diabetes Mellitus, Type 2/chemically induced , Dysbiosis/chemically induced , Metabolic Syndrome/chemically induced , MicroRNAs/drug effects , Non-Nutritive Sweeteners/adverse effects , Gastrointestinal Microbiome/drug effects , Humans , Obesity/metabolism , Receptor, Insulin/drug effects , Receptors, G-Protein-Coupled/drug effects
10.
Chemosphere ; 224: 71-76, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30818196

ABSTRACT

Fluoride is one of the common environmental pollutants. Internal exposure to fluoride is related to the lowered cognitive function and intelligence, particularly for children. Determination of protein content in brain tissue is a means to reflect the functional development of the central nervous system. Insulin and insulin receptor (IR) signaling systems are associated with cognitive ability. The present research focused on the assessment of the expressions of IR protein and mRNA in hippocampus and olfactory bulb (OB), as well as learning and memory ability of male Kunming mice. Mice were exposed to 50, 100, and 150 mg/L NaF for 90 continuous days. The results showed that learning and memory abilities as well as protein content of male mice brain was significantly decreased by fluoride. Fluoride could inhibit the protein and mRNA expressions of the IR in the hippocampus and OB of mice. IRs mainly distributed in the olfactory nerve layer of the outermost layer of the OB, and most distributed in the hippocampal cornu ammon 3 (CA3) region, followed by the dentate gyrus (DG) and cornu ammon 1 (CA1) regions. These findings suggested that inhibition of the IR protein and mRNA expressions in the hippocampus and OB by fluoride might in part affect learning and memory ability in male mice.


Subject(s)
Fluorides/toxicity , Hippocampus/physiopathology , Learning/drug effects , Olfactory Bulb/physiopathology , Receptor, Insulin/drug effects , Animals , Brain/metabolism , Fluorides/metabolism , Hippocampus/metabolism , Male , Memory/drug effects , Mice , Olfactory Bulb/metabolism , RNA, Messenger/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
11.
Am J Physiol Endocrinol Metab ; 316(4): E622-E634, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30668149

ABSTRACT

Excessive maternal high-fructose diet (HFD) during pregnancy and lactation has been reported to cause metabolic disorders in the offspring. Whether the infant's brain metabolism is disturbed by maternal HFD is largely unknown. Brain energy metabolism is elevated dramatically during fetal and postnatal development, whereby maternal nutrition is a key factor that determines cellular metabolism. Astrocytes, a nonneuronal cell type in the brain, are considered to support the high-energy demands of neurons by supplying lactate. In this study, the effects of maternal HFD on astrocytic glucose metabolism were investigated using hippocampal primary cultures of female infants. We found that glycolytic capacity and mitochondrial respiration and electron transport chain were suppressed by maternal HFD. Mitochondrial DNA copy number and mitochondrial transcription factor A expression were suppressed by maternal HFD. Western blots and immunofluorescent images further indicated that the glucose transporter 1 was downregulated whereas the insulin receptor-α, phospho-insulin receptor substrate-1 (Y612) and the p85 subunit of phosphatidylinositide 3-kinase were upregulated in the HFD group. Pioglitazone, which is known to increase astrocytic glucose metabolism, effectively reversed the suppressed glycolysis, and lactate release was restored. Moreover, pioglitazone also normalized oxidative phosphorylation with an increase of cytosolic ATP. Together, these results suggest that maternal HFD impairs astrocytic energy metabolic pathways that were reversed by pioglitazone.


Subject(s)
Astrocytes/drug effects , Dietary Sugars/pharmacology , Fructose/pharmacology , Glycolysis/drug effects , Hypoglycemic Agents/pharmacology , Oxidative Phosphorylation/drug effects , Pioglitazone/pharmacology , Animals , Astrocytes/metabolism , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Female , Fetal Development , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Primary Cell Culture , Rats , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
12.
Am J Physiol Endocrinol Metab ; 316(1): E16-E33, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30153063

ABSTRACT

Polycystic ovary syndrome (PCOS) is highly associated with cardiometabolic risk and the metabolic syndrome (MetS), predisposing women to increased risk of developing type 2 diabetes and cardiovascular disease. Metformin is commonly used to treat insulin resistance-glucose intolerance, and flutamide, an androgen receptor (AR) antagonist, is used to target hyperandrogenemia and dyslipidemia. Currently, the physiological mechanism of action of these treatments on androgen, lipidogenic, and insulin signaling pathways remains unclear in PCOS. The aim of this study was to investigate the effects and mechanisms of action of metformin and flutamide on plasma lipid-apolipoprotein (Apo)B-lipoprotein and insulin-glucose metabolism, and endocrine-reproductive indices in a PCOS-prone MetS rodent model. PCOS-prone rodents were treated with metformin (300 mg/kg body wt), flutamide (30 mg/kg body wt), or metformin + flutamide combination treatment for 6 wk. Metformin was shown to improve fasting insulin and HOMA-IR, whereas flutamide and combination treatment were shown to reduce plasma triglycerides, ApoB48, and ApoB100, and this was associated with decreased intestinal secretion of ApoB48/triglyceride. Flutamide and metformin were shown to reduce plasma androgen indices and to improve ovarian primary and preovulatory follicle frequency. Metformin treatment increased hepatic estrogen receptor (ER)α, and metformin-flutamide decreased intestinal AR and increased ERα mRNA expression. Metformin-flutamide treatment upregulated hepatic and intestinal insulin signaling, including insulin receptor, MAPK1, and AKT2. In conclusion, cardiometabolic risk factors, in particular ApoB-hypertriglyceridemia, are independently modulated via the AR, and understanding the contribution of AR and insulin-signaling pathways further may facilitate the development of targeted interventions in high-risk women with PCOS and MetS.


Subject(s)
Androgen Antagonists/pharmacology , Blood Glucose/drug effects , Estrogen Receptor alpha/drug effects , Flutamide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Metabolic Syndrome/metabolism , Metformin/pharmacology , Animals , Apolipoprotein B-100/drug effects , Apolipoprotein B-100/metabolism , Apolipoprotein B-48/drug effects , Apolipoprotein B-48/metabolism , Apolipoproteins B/drug effects , Apolipoproteins B/metabolism , Blood Glucose/metabolism , Cardiovascular Diseases , Disease Models, Animal , Estrogen Receptor alpha/genetics , Female , Follicular Phase , Insulin Resistance , Intestinal Mucosa/metabolism , Liver/drug effects , Liver/metabolism , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Ovarian Follicle/drug effects , Ovary/drug effects , Polycystic Ovary Syndrome/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger , Rats , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Risk , Triglycerides/metabolism
13.
Clin Pharmacol Ther ; 105(2): 417-425, 2019 02.
Article in English | MEDLINE | ID: mdl-30125349

ABSTRACT

The goal of this investigation was to examine clinical translation of glucose responsiveness of MK-2640, which is a novel insulin saccharide conjugate that can bind the insulin receptor or mannose receptor C type 1 (MRC1), the latter dependent upon glucose concentration. In a rising dose study in 36 healthy adults under euglycemic clamp conditions, rising exposures revealed saturation of MK-2640 clearance, likely due to saturation of clearance by MRC1. Potency of MK-2640 was ~25-fold reduced relative to regular human insulin. In a randomized, 2-period crossover trial in 16 subjects with type 1 diabetes mellitus to evaluate glucose-responsiveness of i.v. administered MK-2640, we were unable to demonstrate a glucose-dependent change in MK-2640 clearance, although a significant glucose-dependent augmentation of glucose infusion rate was observed. These pharmacokinetic (PK) and pharmacodynamic (PD) data provide crucial insights into next steps for developing an insulin saccharide conjugate as a clinically effective glucose-responsive insulin analog.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/analogs & derivatives , Administration, Intravenous , Adolescent , Adult , Antigens, CD/drug effects , Cross-Over Studies , Diabetes Mellitus, Type 1/blood , Dose-Response Relationship, Drug , Double-Blind Method , Female , Glucose Clamp Technique , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Insulin/adverse effects , Insulin/pharmacokinetics , Insulin/therapeutic use , Male , Middle Aged , Receptor, Insulin/drug effects , Young Adult
14.
Eur J Neurosci ; 49(6): 794-804, 2019 03.
Article in English | MEDLINE | ID: mdl-29791756

ABSTRACT

Diet influences dopamine transmission in motor- and reward-related basal ganglia circuitry. In part, this reflects diet-dependent regulation of circulating and brain insulin levels. Activation of striatal insulin receptors amplifies axonal dopamine release in brain slices, and regulates food preference in vivo. The effect of insulin on dopamine release is indirect, and requires striatal cholinergic interneurons that express insulin receptors. However, insulin also acts directly on dopamine axons to increase dopamine uptake by promoting dopamine transporter (DAT) surface expression, counteracting enhanced dopamine release. Here, we determined the functional consequences of acute insulin exposure and chronic diet-induced changes in insulin on DAT activity after evoked dopamine release in striatal slices from adult ad-libitum fed (AL) rats and mice, and food-restricted (FR) or high-fat/high-sugar obesogenic (OB) diet rats. Uptake kinetics were assessed by fitting evoked dopamine transients to the Michaelis-Menten equation and extracting Cpeak and Vmax . Insulin (30 nm) increased both parameters in the caudate putamen and nucleus accumbens core of AL rats in an insulin receptor- and PI3-kinase-dependent manner. A pure effect of insulin on uptake was unmasked using mice lacking striatal acetylcholine, in which increased Vmax caused a decrease in Cpeak . Diet also influenced Vmax , which was lower in FR vs. AL. The effects of insulin on Cpeak and Vmax were amplified by FR but blunted by OB, consistent with opposite consequences of these diets on insulin levels and insulin receptor sensitivity. Overall, these data reveal acute and chronic effects of insulin and diet on dopamine release and uptake that will influence brain reward pathways.


Subject(s)
Brain/metabolism , Diet, High-Fat , Dopamine/metabolism , Insulin/metabolism , Animals , Brain/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins/pharmacology , Insulin/pharmacology , Interneurons/drug effects , Interneurons/metabolism , Male , Nucleus Accumbens/drug effects , Rats, Sprague-Dawley , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism
15.
Eur J Neurosci ; 50(3): 2146-2155, 2019 08.
Article in English | MEDLINE | ID: mdl-30471157

ABSTRACT

Mesolimbic dopamine circuits, implicated in incentive motivation, are sensitive to changes in metabolic state such as weight loss and diet-induced obesity. These neurons are important targets for metabolic hormones such as leptin, glucagon-like peptide-1, ghrelin and insulin. Insulin receptors are located on dopamine neurons in the ventral tegmental area (VTA) and we have previously demonstrated that insulin induces long-term depression of excitatory synapses onto VTA dopamine neurons. While insulin can decrease dopamine concentration in somatodendritic regions, it can increase dopamine in striatal slices. Whether insulin directly targets the VTA to alter dopamine release in projection areas, such as the nucleus accumbens (NAc), remains unknown. The main goal of the present experiments was to examine NAc dopamine concentration following VTA administration of insulin. Using in vivo FSCV to detect rapid fluctuations in dopamine concentration, we showed that intra-VTA insulin via action at insulin receptors reduced pedunculopontine nucleus-evoked dopamine release in the NAc. Furthermore, intra-VTA insulin reduced cocaine-potentiated NAc dopamine. Finally, intra-VTA or intranasal insulin decreased locomotor responses to cocaine, an effect blocked by an intra-VTA administered insulin receptor antagonist. Together, these data demonstrate that mesolimbic dopaminergic projections are important targets of the metabolic hormone, insulin.


Subject(s)
Dopamine/metabolism , Insulin/pharmacology , Receptor, Insulin/drug effects , Ventral Tegmental Area/metabolism , Animals , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Insulin/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Nucleus Accumbens/drug effects , Receptor, Insulin/metabolism , Ventral Tegmental Area/drug effects
16.
Endocr Relat Cancer ; 26(2): 187-199, 2019 02.
Article in English | MEDLINE | ID: mdl-30422809

ABSTRACT

Ewing sarcoma family tumors (ESFTs) are a group of aggressive and highly metastatic tumors lacking efficient therapies. Insulin-like growth factor 1 receptor (IGF1R) blockade is one of the most efficient targeting therapy for ESFTs. However, the appliance is obstructed by drug resistance and disease recurrence due to the activation of insulin receptor (IR) signaling induced by IGF1R blockade. Herein ß-elemene, a compound derived from natural plants, exhibited a remarkable proliferation repression on ESFT cells, which was weakened by a caspase inhibitor Z-VAD. ß-elemene in combination with IGF1R inhibitors enhanced markedly the repression on cellular proliferation and mTOR activation by IGF1R inhibitors and suppressed the PI3K phosphorylation induced by IGF1R inhibitors. To investigate the mechanisms, we focused on the effects of ß-elemene on IR signaling pathway. ß-elemene significantly suppressed the insulin-driven cell growth and the activation of mTOR and PI3K in tumor cells, while the toxicity to normal hepatocytes was much lower. Further, the phosphorylation of IR was found to be suppressed notably by ß-elemene specifically in tumor cells other than normal hepatocytes. In addition, ß-elemene inhibited the growth of ESFT xenografts in vivo, and the phosphorylation of IR and S6 ribosomal protein was significantly repressed in the ß-elemene-treated xenografts. These data suggest that ß-elemene targets IR phosphorylation to inhibit the proliferation of tumor cells specifically and enhance the effects of IGF1R inhibitors. Thus, this study provides evidence for novel approaches by ß-elemene alone or in combination with IGF1R blockades in ESFTs and IR signaling hyperactivated tumors.


Subject(s)
Receptor, Insulin/drug effects , Sarcoma, Ewing/drug therapy , Sesquiterpenes/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Phosphorylation , Sarcoma, Ewing/mortality , Sesquiterpenes/pharmacology , Survival Rate
17.
Nat Commun ; 9(1): 5152, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30514845

ABSTRACT

Insulin and insulin-like signaling regulates a broad spectrum of growth and metabolic responses to a variety of internal and environmental stimuli. For example, the inhibition of insulin-like signaling in C. elegans mediates its response to both osmotic stress and starvation. We report that in response to osmotic stress the cytosolic sulfotransferase SSU-1 antagonizes insulin-like signaling and promotes developmental arrest. Both SSU-1 and the DAF-16 FOXO transcription factor, which is activated when insulin signaling is low, are needed to drive specific responses to reduced insulin-like signaling. We demonstrate that SSU-1 functions in a single pair of sensory neurons to control intercellular signaling via the nuclear hormone receptor NHR-1 and promote both the specific transcriptional response to osmotic stress and altered lysophosphatidylcholine metabolism. Our results show the requirement of a sulfotransferase-nuclear hormone receptor neurohormonal signaling pathway for some but not all consequences of reduced insulin-like signaling.


Subject(s)
Caenorhabditis elegans/metabolism , Nerve Tissue Proteins/drug effects , Neurotransmitter Agents/metabolism , Receptor, Insulin/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfotransferases/antagonists & inhibitors , Animals , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cloning, Molecular , Embryo, Nonmammalian , Embryonic Development/genetics , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Insulin/metabolism , Lysophosphatidylcholines/metabolism , Mutagenesis , Osmotic Pressure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sensory Receptor Cells/drug effects , Starvation , Stress, Physiological , Sulfotransferases/genetics , Sulfotransferases/metabolism
18.
Brain Res Bull ; 143: 27-35, 2018 10.
Article in English | MEDLINE | ID: mdl-30278200

ABSTRACT

Depression is associated with uncontrolled diabetes, which indicates a lack of insulin effect, yet the role of the insulin receptor in mediating depression is not clearly established because insulin receptors are not required for glucose entry into the brain. However, insulin receptors are important for brain function since insulin receptor knockout mice have depressive-like activity. Depression and cognitive problems are also associated with low insulin-like growth factor-1 (IGF-1) in the elderly. Rodent studies showed chronic IGF-1 administration had antidepressant-like (AD) activity. We asked if insulin in the brain might act through the IGF-1 receptor, as it does in some tissues. We used acute insulin or IGF-1 infusions into the 3rd ventricle (icv) in rats and tested animals in a forced swim test. We found that antidepressive-like behavior is mediated by insulin and IGF-1. Further, administration of the IGF-1 receptor antagonist JB-1 blocked the antidepressive-like activity of the insulin and IGF-1, indicating a strong relationship between insulin, IGF-1 and depression. Insulin acts at least partially through the IGF-1 receptor and is responsive to receptor antagonism. The model offers promise for future studies of the mechanism of depression, and therapy to increase insulin sensitivity and IGF-1 action including exercise and nutrition.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Receptor, IGF Type 1/drug effects , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Depression/etiology , Depressive Disorder/etiology , Fluoxetine/metabolism , Fluoxetine/pharmacology , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Motor Activity/drug effects , Oligopeptides/metabolism , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/pharmacology , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Swimming/psychology
19.
Diabetes ; 67(5): 946-959, 2018 05.
Article in English | MEDLINE | ID: mdl-29483182

ABSTRACT

Cardiovascular disease caused by atherosclerosis is the leading cause of mortality associated with type 2 diabetes and metabolic syndrome. Insulin therapy is often needed to improve glycemic control, but it does not clearly prevent atherosclerosis. Upon binding to the insulin receptor (IR), insulin activates distinct arms of downstream signaling. The IR-Akt arm is associated with blood glucose lowering and beneficial effects, whereas the IR-Erk arm might exert less desirable effects. We investigated whether selective activation of the IR-Akt arm, leaving the IR-Erk arm largely inactive, would result in protection from atherosclerosis in a mouse model of metabolic syndrome. The insulin mimetic peptide S597 lowered blood glucose and activated Akt in insulin target tissues, mimicking insulin's effects, but only weakly activated Erk and even prevented insulin-induced Erk activation. Strikingly, S597 retarded atherosclerotic lesion progression through a process associated with protection from leukocytosis, thereby reducing lesional accumulation of inflammatory Ly6Chi monocytes. S597-mediated protection from leukocytosis was accompanied by reduced numbers of the earliest bone marrow hematopoietic stem cells and reduced IR-Erk activity in hematopoietic stem cells. This study provides a conceptually novel treatment strategy for advanced atherosclerosis associated with metabolic syndrome and type 2 diabetes.


Subject(s)
Atherosclerosis/prevention & control , Blood Glucose/drug effects , MAP Kinase Signaling System/drug effects , Metabolic Syndrome/drug therapy , Peptides/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Receptor, Insulin/drug effects , Animals , Atherosclerosis/etiology , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/etiology , Diabetic Angiopathies/prevention & control , Disease Models, Animal , Male , Metabolic Syndrome/complications , Mice , Mice, Knockout , Monocytes , Plaque, Atherosclerotic , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/agonists , Receptor, Insulin/metabolism , Receptors, LDL/genetics , Signal Transduction
20.
Endocr Regul ; 52(1): 6-16, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29453923

ABSTRACT

OBJECTIVE: We investigated the effects of magnesium supplementation on glucose tolerance, insulin sensitivity, oxidative stress as well as the concentration of insulin receptor and glucose transporter-4 in streptozotocin-nicotinamide induced type-2 diabetic (T2D) rats. METHODS: Rats were divided into four groups designated as: 1) control (CTR); 2) diabetic untreated (DU); 3) diabetic treated with 1 mg of Mg/kg diet (Mg1-D); and 4) diabetic treated with 2 mg of Mg/kg diet (Mg2-D). T2D was induced with a single intraperitoneal (i.p.) injection of freshly prepared streptozotocin (55 mg/kg) aft er an initial i.p. injection of nicotinamide (120 mg/kg). Glucose tolerance, insulin sensitivity, lipid profile, malondialdehyde (MAD) and glutathione content, insulin receptors (INSR) and glucose transporter-4 (GLUT4), fasting insulin and glucose levels were measured, and insulin resistance index was calculated using the homeostatic model assessment of insulin resistance (HOMA-IR). RESULTS: Magnesium supplementation improved glucose tolerance and lowered blood glucose levels almost to the normal range. We also recorded a noticeable increase in insulin sensitivity in Mg-D groups when compared with DU rats. Lipid perturbations associated T2D were significantly attenuated by magnesium supplementation. Fasting glucose level was comparable to control values in the Mg-D groups while the HOMA-IR index was significantly lower compared with the DU rats. Magnesium reduced MDA but increased glutathione concentrations compared with DU group. Moreover, INSR and GLUT4 levels were elevated following magnesium supplementation in T2D rats. CONCLUSION: These findings demonstrate that magnesium may mediate effective metabolic control by stimulating the antioxidant defense, and increased levels of INSR and GLUT4 in diabetic rats.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Glucose Transporter Type 4/drug effects , Insulin Resistance , Magnesium/pharmacology , Receptor, Insulin/drug effects , Animals , Disease Models, Animal , Magnesium/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...