Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.170
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
J Pathol ; 245(1): 85-100, 2018 05.
Article in English | MEDLINE | ID: mdl-29435980

ABSTRACT

Alzheimer's disease (AD) is a devastating neurological disorder that still lacks an effective treatment, and this has stimulated an intense pursuit of disease-modifying therapeutics. Given the increasingly recognized link between AD and defective brain insulin signaling, we investigated the actions of liraglutide, a glucagon-like peptide-1 (GLP-1) analog marketed for treatment of type 2 diabetes, in experimental models of AD. Insulin receptor pathology is an important feature of AD brains that impairs the neuroprotective actions of central insulin signaling. Here, we show that liraglutide prevented the loss of brain insulin receptors and synapses, and reversed memory impairment induced by AD-linked amyloid-ß oligomers (AßOs) in mice. Using hippocampal neuronal cultures, we determined that the mechanism of neuroprotection by liraglutide involves activation of the PKA signaling pathway. Infusion of AßOs into the lateral cerebral ventricle of non-human primates (NHPs) led to marked loss of insulin receptors and synapses in brain regions related to memory. Systemic treatment of NHPs with liraglutide provided partial protection, decreasing AD-related insulin receptor, synaptic, and tau pathology in specific brain regions. Synapse damage and elimination are amongst the earliest known pathological changes and the best correlates of memory impairment in AD. The results illuminate mechanisms of neuroprotection by liraglutide, and indicate that GLP-1 receptor activation may be harnessed to protect brain insulin receptors and synapses in AD. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cognitive Dysfunction/drug therapy , Liraglutide/pharmacology , Memory/drug effects , Receptor, Insulin/drug effects , Synapses/pathology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Hippocampus/drug effects , Hypoglycemic Agents/pharmacology , Male , Mice , Receptor, Insulin/metabolism , Synapses/drug effects
9.
Acta Pharmacol Sin ; 39(6): 923-929, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29168473

ABSTRACT

Graves' disease (GD) is the leading cause of hyperthyroidism, and the majority of GD patients eventually develop disorders of glucose handling, which further affects their quality of life. Yangxin Tongmai formula (YTF) is modified from a famous formula of traditional Chinese medicine for the treatment of cardiovascular diseases. In this study we investigated the potential effects of YTF in the treatment of pediatric GD patients with impaired glucose tolerance. Forty pediatric GD patients and 20 healthy children were recruited for this clinical study. Based on the glucose tolerance, the GD patients were divided into two groups: 20 patients displayed impaired glucose tolerance, while the other 20 patients displayed normal glucose tolerance. YTF was orally administered for 60 days. YTF administration significantly ameliorated the abnormal glucose tolerance and insulin sensitivity in the GD patients with impaired glucose tolerance. To determine the molecular mechanisms of this observation, the number of plasma insulin receptors was determined by ELISA. Before treatment, the fasting and postprandial levels of the insulin receptor were significantly lower in patients with impaired glucose tolerance compared with those in patients with normal glucose tolerance and healthy children. After YTF treatment, both the fasting and the postprandial circulating insulin receptor levels were upregulated, and close to those in healthy children. Therefore, YTF is a potential effective treatment to enhance glucose handling in GD children with impaired glucose tolerance.


Subject(s)
Antigens, CD/drug effects , Blood Glucose/drug effects , Drugs, Chinese Herbal/therapeutic use , Glucose Intolerance/drug therapy , Graves Disease/complications , Hypoglycemic Agents/therapeutic use , Receptor, Insulin/drug effects , Adolescent , Age Factors , Antigens, CD/blood , Biomarkers/blood , Blood Glucose/metabolism , Child , China , Drugs, Chinese Herbal/adverse effects , Female , Glucose Intolerance/blood , Glucose Intolerance/diagnosis , Glucose Intolerance/etiology , Graves Disease/diagnosis , Humans , Hypoglycemic Agents/adverse effects , Insulin Resistance , Male , Receptor, Insulin/blood , Time Factors , Treatment Outcome , Up-Regulation
10.
J Neurosci ; 36(41): 10683-10695, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27733618

ABSTRACT

HIV-1 infection of the brain causes the neurodegenerative syndrome HIV-associated neurocognitive disorders (HAND), for which there is no specific treatment. Herein, we investigated the actions of insulin using ex vivo and in vivo models of HAND. Increased neuroinflammatory gene expression was observed in brains from patients with HIV/AIDS. The insulin receptor was detected on both neurons and glia, but its expression was unaffected by HIV-1 infection. Insulin treatment of HIV-infected primary human microglia suppressed supernatant HIV-1 p24 levels, reduced CXCL10 and IL-6 transcript levels, and induced peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. Insulin treatment of primary human neurons prevented HIV-1 Vpr-mediated cell process retraction and death. In feline immunodeficiency virus (FIV) infected cats, daily intranasal insulin treatment (20.0 IU/200 µl for 6 weeks) reduced CXCL10, IL-6, and FIV RNA detection in brain, although PPAR-γ in glia was increased compared with PBS-treated FIV+ control animals. These molecular changes were accompanied by diminished glial activation in cerebral cortex and white matter of insulin-treated FIV+ animals, with associated preservation of cortical neurons. Neuronal counts in parietal cortex, striatum, and hippocampus were higher in the FIV+/insulin-treated group compared with the FIV+/PBS-treated group. Moreover, intranasal insulin treatment improved neurobehavioral performance, including both memory and motor functions, in FIV+ animals. Therefore, insulin exerted ex vivo and in vivo antiviral, anti-inflammatory, and neuroprotective effects in models of HAND, representing a new therapeutic option for patients with inflammatory or infectious neurodegenerative disorders including HAND. SIGNIFICANCE STATEMENT: HIV-associated neurocognitive disorders (HAND) represent a spectrum disorder of neurocognitive dysfunctions resulting from HIV-1 infection. Although the exact mechanisms causing HAND are unknown, productive HIV-1 infection in the brain with associated neuroinflammation is a potential pathogenic mechanism resulting in neuronal damage and death. We report that, in HIV-infected microglia cultures, insulin treatment led to reduced viral replication and inflammatory gene expression. In addition, intranasal insulin treatment of experimentally feline immunodeficiency virus-infected animals resulted in improved motor and memory performances. We show that insulin restored expression of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-γ), which is suppressed by HIV-1 replication. Our findings indicate a unique function for insulin in improving neurological outcomes in lentiviral infections, implicating insulin as a therapeutic intervention for HAND.


Subject(s)
AIDS Dementia Complex/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Neuritis/prevention & control , Neurodegenerative Diseases/prevention & control , Neurons/pathology , Neuroprotective Agents/therapeutic use , Administration, Intranasal , Animals , Cats , Cell Death/drug effects , Female , HIV-1 , Human Immunodeficiency Virus Proteins/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Immunodeficiency Virus, Feline , Insulin/administration & dosage , Lentivirus Infections/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Pregnancy , Receptor, Insulin/drug effects
11.
Adv Exp Med Biol ; 975 Pt 1: 271-279, 2017.
Article in English | MEDLINE | ID: mdl-28849462

ABSTRACT

In this study we examined the role of chronic taurine supplementation on plasma glucose homeostasis and brain excitability through activation of the insulin receptor. FVB/NJ male mice were supplemented with taurine in drinking water (0.05% w/v) for 4 weeks and subjected to a glucose tolerance test (7.5 mg/kg BW) after 12 h fasting. We found that taurine-fed mice were slightly hypoglycemic prior to glucose injection and showed significantly reduced plasma glucose at 30 and 60 min post-glucose injection when compared to control mice. Previously, we reported that taurine supplementation induces biochemical changes that target the GABAergic system. Those studies show that taurine-fed mice are hyperexcitable, have reduced GABAA receptors expression and increased GAD and somatostatin expression in the brain. In this study, we found that taurine-fed mice had a significant increase in insulin receptor (IR) immuno-reactivity in the pancreas and all brain regions examined. At the mRNA level, we found that the IR showed differential regional expression. Surprisingly, we found that neurons express the gene for insulin and that taurine had a significant role in regulating insulin gene expression. We propose that increased insulin production and secretion in taurine-fed mice cause an increase activation of the central IR and may be partially responsible for the increased neuronal excitability observed in taurine supplemented mice. Furthermore, the high levels of neuronal insulin expression and its regulation by taurine implicates taurine in the regulation of metabolic homeostasis.


Subject(s)
Blood Glucose/drug effects , Gene Expression Regulation/drug effects , Neurons/drug effects , Receptor, Insulin/drug effects , Taurine/pharmacology , Animals , Dietary Supplements , Homeostasis/drug effects , Male , Mice , Receptor, Insulin/biosynthesis
12.
Am J Physiol Endocrinol Metab ; 310(6): E440-51, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26714846

ABSTRACT

Neuregulin (NRG) is an EGF-related growth factor that binds to the tyrosine kinase receptors ErbB3 and ErbB4, thus inducing tissue development and muscle glucose utilization during contraction. Here, we analyzed whether NRG has systemic effects regulating glycemia in control and type 2 diabetic rats. To this end, recombinant NRG (rNRG) was injected into Zucker diabetic fatty (ZDF) rats and their respective lean littermates 15 min before a glucose tolerance test (GTT) was performed. rNRG enhanced glucose tolerance without promoting the activation of the insulin receptor (IR) or insulin receptor substrates (IRS) in muscle and liver. However, in control rats, rNRG induced the phosphorylation of protein kinase B (PKB) and glycogen synthase kinase-3 (GSK-3) in liver but not in muscle. In liver, rNRG increased ErbB3 tyrosine phosphorylation and its binding to phosphatidylinositol 3-kinase (PI3K), thus indicating that rNRG activates the ErbB3/PI3K/PKB signaling pathway. rNRG increased glycogen content in liver but not in muscle. rNRG also increased the content of fructose-2,6-bisphosphate (Fru-2,6-P2), an activator of hepatic glycolysis, and lactate in liver but not in muscle. Increases in lactate were abrogated by wortmannin, a PI3K inhibitor, in incubated hepatocytes. The liver of ZDF rats showed a reduced content of ErbB3 receptors, entailing a minor stimulation of the rNRG-induced PKB/GSK-3 cascade and resulting in unaltered hepatic glycogen content. Nonetheless, rNRG increased hepatic Fru-2,6-P2 and augmented lactate both in liver and in plasma of diabetic rats. As a whole, rNRG improved response to the GTT in both control and diabetic rats by enhancing hepatic glucose utilization.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/metabolism , Liver/drug effects , Muscle, Skeletal/drug effects , Neuregulins/pharmacology , Animals , Blood Glucose/metabolism , Case-Control Studies , Fructosediphosphates/metabolism , Glucose/metabolism , Glucose Tolerance Test , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Insulin , Insulin Receptor Substrate Proteins/drug effects , Insulin Receptor Substrate Proteins/metabolism , Lactic Acid/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinase/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Zucker , Receptor, ErbB-3/drug effects , Receptor, ErbB-3/metabolism , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism
13.
Eur J Neurosci ; 44(8): 2557-2568, 2016 10.
Article in English | MEDLINE | ID: mdl-27543746

ABSTRACT

Insulin serves as a link between the metabolic and reproductive systems, communicating energy availability to the hypothalamus and enabling reproductive mechanisms. Adult Suffolk ewes prenatally exposed to testosterone (T) display an array of reproductive and metabolic dysfunctions similar to those seen in women with polycystic ovarian syndrome (PCOS), including insulin resistance. Moreover, prenatal T treatment alters neuropeptide expression in KNDy (co-expressing kisspeptin, neurokinin B/dynorphin) and agouti-related peptide (AgRP) neurons in the arcuate nucleus, two populations that play key roles in the control of reproduction and metabolism, respectively. In this study, we determined whether prenatal T treatment also altered insulin receptors in KNDy and AgRP neurons, as well as in preoptic area (POA) kisspeptin, pro-opiomelanocortin (POMC), and gonadotropin-releasing hormone (GnRH) neurons of the adult sheep brain. Immunofluorescent detection of the beta subunit of insulin receptor (IRß) revealed that KNDy, AgRP and POMC neurons, but not GnRH or POA kisspeptin neurons, colocalize IRß in control females. Moreover, prenatal T treatment decreased the percentage of KNDy and AgRP neurons that colocalized IRß, consistent with reduced insulin sensitivity. Administration of the anti-androgen drug, Flutamide, during prenatal T treatment, prevented the reduction in IRß colocalization in AgRP, but not in KNDy neurons, suggesting that these effects are programmed by androgenic and oestrogenic actions, respectively. These findings provide novel insight into the effects of prenatal T treatment on hypothalamic insulin sensitivity and raise the possibility that decreased insulin receptors, specifically within KNDy and AgRP neurons, may contribute to the PCOS-like phenotype of this animal model.


Subject(s)
Dynorphins/metabolism , Hypothalamus/drug effects , Kisspeptins/metabolism , Neurokinin B/metabolism , Receptor, Insulin/drug effects , Testosterone/pharmacology , Aging , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Neurons/drug effects , Neurons/metabolism , Receptor, Insulin/metabolism , Sheep
14.
J Pharmacol Exp Ther ; 357(2): 367-74, 2016 May.
Article in English | MEDLINE | ID: mdl-26994072

ABSTRACT

Phenobarbital (PB) antagonized insulin to inactivate the insulin receptor and attenuated the insulin receptor downstream protein kinase B (AKT)-forkhead box protein O1 and extracellular signal-regulated kinase 1/2 signals in mouse primary hepatocytes and HepG2 cells. Hepatic AKT began dephosphorylation in an early stage of PB treatment, and blood glucose levels transiently increased in both wild-type and constitutive androstane receptor (CAR) knockout (KO) mice. On the other hand, blood glucose levels increased in wild-type mice, but not KO mice, in later stages of PB treatment. As a result, PB, acting as an insulin receptor antagonist, elicited CAR-independent increases and CAR-dependent decreases of blood glucose levels at these different stages of treatment, respectively. Reciprocally, insulin activation of the insulin receptor repressed CAR activation and induction of its target CYP2B6 gene in HepG2 cells. Thus, PB and insulin cross-talk through the insulin receptor to regulate glucose and drug metabolism reciprocally.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Phenobarbital/pharmacology , Receptor, Insulin/drug effects , Receptors, G-Protein-Coupled/agonists , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Blood Glucose/metabolism , Cytochrome P-450 CYP2B6/drug effects , Cytochrome P450 Family 2 , Hepatocytes/drug effects , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk/drug effects , Receptors, Calcium-Sensing , Receptors, G-Protein-Coupled/genetics , Steroid Hydroxylases/metabolism , Transfection
15.
Nutr Metab Cardiovasc Dis ; 26(7): 614-622, 2016 07.
Article in English | MEDLINE | ID: mdl-27052924

ABSTRACT

We recently observed that free fatty acids impair the stimulation of glucose transport into cardiomyocytes in response to either insulin or metabolic stress. In vivo, fatty acids for the myocardium are mostly obtained from triglyceride-rich lipoproteins (chylomicrons and Very Low-Density Lipoproteins). We therefore determined whether exposure of cardiac myocytes to VLDL resulted in impaired basal and stimulated glucose transport. Primary adult rat cardiac myocytes were chronically exposed to VLDL before glucose uptake was measured in response to insulin or metabolic stress, provoked by the mitochondrial ATP synthase inhibitor oligomycin. Exposure of cardiac myocytes to VLDL reduced both insulin-and oligomycin-stimulated glucose uptake. The reduction of glucose uptake was associated with a moderately reduced tyrosine phosphorylation of the insulin receptor. No reduction of the phosphorylation of the downstream effectors of insulin signaling Akt and AS160 was however observed. Similarly only a modest reduction of the activating phosphorylation of the AMP-activated kinase (AMPK) was observed in response to oligomycin. Similar to our previous observations with free fatty acids, inhibition of fatty acid oxidation restored oligomycin-stimulated glucose uptake. In conclusions, VLDL-derived fatty acids impair stimulated glucose transport in cardiac myocytes by a mechanism that seems to be mediated by a fatty acid oxidation intermediate. Thus, in the clinical context of the metabolic syndrome high VLDL may contribute to enhancement of ischemic injury by reduction of metabolic stress-stimulated glucose uptake.


Subject(s)
Deoxyglucose/metabolism , Lipoproteins, VLDL/pharmacology , Myocytes, Cardiac/drug effects , Stress, Physiological/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Dose-Response Relationship, Drug , Fatty Acids, Nonesterified/metabolism , GTPase-Activating Proteins/metabolism , Humans , Insulin/pharmacology , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Oligomycins/pharmacology , Oxidation-Reduction , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Tyrosine , Uncoupling Agents/pharmacology
16.
Chin J Physiol ; 59(2): 109-18, 2016 Apr 30.
Article in English | MEDLINE | ID: mdl-27080466

ABSTRACT

Risk factors for prostate cancer (PCa) include age, hormones, race, family history and diet. Recently, epidemiologic evidence has indicated that history of diabetes mellitus (DM) is inversely associated with risk of PCa. However, epidemiological investigations have yielded inconsistent results. Hence, the exact mechanism of DM-induced reduction in the incidence of PCa has yet to be fully elucidated. The aim of this study was to investigate the effects of DM factors, including glucose, insulin and insulin-like growth factor-1 (IGF-1), on the proliferation of PCa cell lines in vitro. Cell proliferation and expression of hormone receptors was examined in MTT assay and Western blot analysis, respectively. The results showed that DM factors did not affect the viability of androgen receptor (AR)-expressing PCa cell lines. However, cell proliferation increased after treatment with DM factors in androgen-independent PCa cell lines. On PCa tissue arrays, intensities of total AR and nuclear IGF-1R were higher in malignant tissues than in normal prostate glands. In terms of hormonal receptors, androgen-dependent LNCaP cells treated with insulin and IGF-1 in a low-serum medium showed decreased expression of insulin receptor beta (IRß) and elevated expression of IGF-1 receptor beta (IGF-1Rß). Moreover, expression of AR was upregulated after insulin and IGF-1 treatment in LNCaP cells, but not in the other PCa cell lines. Most of the studied antidiabetic drugs promoted the viability of PCa cells. However, metformin decreased the viability of AR-expressing PCa cells. These results suggest that diabetic factors modify the expression of AR, IR and IGF-1R to increase cancer cell proliferation. Moreover, the growth suppressing effects of metformin on PCa may be via the regulation of the AR signaling pathway.


Subject(s)
Diabetes Mellitus/physiopathology , Hypoglycemic Agents/pharmacology , Prostatic Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Glucose/pharmacology , Humans , Immunohistochemistry , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Male , Prostatic Neoplasms/physiopathology , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/drug effects , Receptor, Insulin/biosynthesis , Receptor, Insulin/drug effects , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Signal Transduction/drug effects
17.
J Neurochem ; 135(2): 332-46, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26173704

ABSTRACT

Hypoxia has been reported to cause hippocampal neurodegeneration resulting in learning and memory deficits. In the present study, we investigated the potential of salidroside, a glucoside derivative of tyrosol, in ameliorating hypoxia-induced neurodegeneration and memory impairment. Morris water maze test showed improvement in learning and spatial memory of salidroside-treated hypoxic rats correlating with increased dendritic intersections and arborization. Salidroside administration increased phosphorylation of insulin receptor subunit A (IRA) at Y972, Y1162/63, and Y1146 sites and subsequent activation of AMP-activated protein kinase (AMPK) α subunit isoforms pAMPKα1 and pAMPKα2 resulting in mitochondrial biogenesis. Contrarily, silencing of IRA in salidroside-supplemented hypoxic hippocampal cells could not improve cell viability or alter pAMPKα1 and pAMPKα2 expression. Rats administered with salidroside showed elevated expression of phosphorylated cAMP response element-binding protein in the hippocampus. Salidroside administration also resulted in increased sirtuin 1 (SIRT1) activity through a cytochrome P4502E1 (CYP2E1)-regulated mechanism that was independent of pIRA. Taken together, these findings suggest a synergistic role of pIRA and SIRT1 in salidroside-mediated neuroprotection, mitochondrial biogenesis, and cognitive improvement during hypoxia. We propose a novel mechanism for salidroside-mediated neuroprotection in hypoxia.


Subject(s)
Glucosides/pharmacology , Hypoxia/psychology , Maze Learning/drug effects , Phenols/pharmacology , Receptor, Insulin/drug effects , Sirtuin 1/pharmacology , Spatial Memory/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Blood-Brain Barrier/drug effects , Cell Survival , Cyclic AMP Response Element-Binding Protein , DNA, Mitochondrial/genetics , Glucosides/pharmacokinetics , Hippocampus/drug effects , Hippocampus/pathology , Male , Mitochondria/drug effects , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Phenols/pharmacokinetics , Phosphorylation , Rats , Rats, Sprague-Dawley
18.
Am J Physiol Endocrinol Metab ; 307(11): E955-68, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25249503

ABSTRACT

Connecting Peptide, or C-peptide, is a product of the insulin prohormone, and is released with and in amounts equimolar to those of insulin. While it was once thought that C-peptide was biologically inert and had little biological significance beyond its role in the proper folding of insulin, it is now known that C-peptide binds specifically to the cell membranes of a variety of tissues and initiates specific intracellular signaling cascades that are pertussis toxin sensitive. Although it is now clear that C-peptide is a biologically active molecule, controversy still remains as to the physiological significance of the peptide. Interestingly, C-peptide appears to reverse the deleterious effects of high glucose in some tissues, including the kidney, the peripheral nerves, and the vasculature. C-peptide is thus a potential therapeutic agent for the treatment of diabetes-associated long-term complications. This review addresses the possible physiologically relevant roles of C-peptide in both normal and disease states and discusses the effects of the peptide on sensory nerve, renal, and vascular function. Furthermore, we highlight the intracellular effects of the peptide and present novel strategies for the determination of the C-peptide receptor(s). Finally, a hypothesis is offered concerning the relationship between C-peptide and the development of microvascular complications of diabetes.


Subject(s)
C-Peptide/pharmacology , C-Peptide/therapeutic use , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Animals , Diabetes Complications/prevention & control , Humans , Receptor, Insulin/drug effects
19.
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
20.
Int J Obes (Lond) ; 37(4): 584-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22641065

ABSTRACT

OBJECTIVE: To investigate the inhibitory effect of cocoa polyphenol extract (CPE) on adipogenesis and obesity along with its mechanism of action. METHODS AND RESULTS: 3T3-L1 preadipocytes were cultured with isobutylmethylxanthine, dexamethasone and insulin (MDI), and male C57BL/6N mice (N=44) were fed a high-fat diet (HFD) for 5 weeks with or without CPE. CPE at 100 or 200 µg ml(-1) inhibited MDI-induced lipid accumulation without diminishing cell viability. In particular, CPE reduced the protein expression levels of PPARγ and CEBPα, and blocked mitotic clonal expansion (MCE) of preadipocytes by reducing proliferating signaling pathways. This in turn attenuates lipid accumulation during the differentiation of 3T3-L1 preadipocytes. CPE effectively suppressed MDI-induced phosphorylation of extracellular signal-regulated kinase (ERK) and Akt, and their downstream signals. We then examined whether CPE regulates insulin receptor (IR), a common upstream regulator of ERK and Akt. We found that although CPE does not affect the protein expression level of IR, it significantly inhibits the activity of IR kinase via direct binding. Collectively, the results suggested that CPE, a direct inhibitor of IR kinase activity, inhibits cellular differentiation and lipid accumulation in 3T3-L1 preadipocytes. Consistently, CPE attenuated HFD-induced body weight gain and fat accumulation in obese mice fed with a HFD. We also found that HFD-induced increased fasting glucose levels remained unaffected by CPE. CONCLUSION: This study demonstrates that CPE inhibits IR kinase activity and its proliferative downstream signaling markers, such as ERK and Akt, in 3T3-L1 preadipocytes, and also prevents the development of obesity in mice fed with a HFD.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Cacao/chemistry , Obesity/drug therapy , Obesity/metabolism , Polyphenols/pharmacology , Receptor, Insulin/drug effects , 3T3-L1 Cells/drug effects , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Diet, High-Fat , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Obesity/prevention & control , Phosphorylation , Polyphenols/chemistry , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL