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










Publication year range
1.
J Alzheimers Dis ; 91(4): 1495-1514, 2023.
Article in English | MEDLINE | ID: mdl-36641678

ABSTRACT

BACKGROUND: Amylin, a pancreatic amyloid peptide involved in energy homeostasis, is increasingly studied in the context of Alzheimer's disease (AD) etiology. To date, conflicting pathogenic and neuroprotective roles for this peptide and its analogs for AD pathogenesis have been described. OBJECTIVE: Whether the benefits of amylin are associated with peripheral improvement of metabolic tone/function or directly through the activation of central amylin receptors is also unknown and downstream signaling mechanisms of amylin receptors are major objectives of this study. METHODS: To address these questions more directly we delivered the amylin analog pramlintide systemically (IP), at previously identified therapeutic doses, while centrally (ICV) inhibiting the receptor using an amylin receptor antagonist (AC187), at doses known to impact CNS function. RESULTS: Here we show that pramlintide improved cognitive function independently of CNS receptor activation and provide transcriptomic data that highlights potential mechanisms. Furthermore, we show than inhibition of the amylin receptor increased amyloid-beta pathology in female APP/PS1 mice, an effect than was mitigated by peripheral delivery of pramlintide. Through transcriptomic analysis of pramlintide therapy in AD-modeled mice we found sexual dimorphic modulation of neuroprotective mechanisms: oxidative stress protection in females and membrane stability and reduced neuronal excitability markers in males. CONCLUSION: These data suggest an uncoupling of functional and pathology-related events and highlighting a more complex receptor system and pharmacological relationship that must be carefully studied to clarify the role of amylin in CNS function and AD.


Subject(s)
Alzheimer Disease , Amylin Receptor Agonists , Receptors, Islet Amyloid Polypeptide , Animals , Female , Male , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Disease Models, Animal , Islet Amyloid Polypeptide/metabolism , Mice, Transgenic , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/metabolism , Amylin Receptor Agonists/pharmacology
2.
Sci Rep ; 9(1): 10942, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358858

ABSTRACT

Recent evidence supports involvement of amylin and the amylin receptor in the pathogenesis of Alzheimer's disease (AD). We have previously shown that amylin receptor antagonist, AC253, improves spatial memory in AD mouse models. Herein, we generated and screened a peptide library and identified two short sequence amylin peptides (12-14 aa) that are proteolytically stable, brain penetrant when administered intraperitoneally, neuroprotective against Aß toxicity and restore diminished levels of hippocampal long term potentiation in AD mice. Systemic administration of the peptides for five weeks in aged 5XFAD mice improved spatial memory, reduced amyloid plaque burden, and neuroinflammation. The common residue SQELHRLQTY within the peptides is an essential sequence for preservation of the beneficial effects of the fragments that we report here and constitutes a new pharmacological target. These findings suggest that the amylin receptor antagonism may represent a novel therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Animals , Female , Hippocampus/drug effects , Islet Amyloid Polypeptide/chemistry , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Receptors, Islet Amyloid Polypeptide/metabolism , Spatial Memory
3.
Neuropsychopharmacology ; 44(6): 1093-1102, 2019 05.
Article in English | MEDLINE | ID: mdl-30710109

ABSTRACT

Recent findings have identified salmon calcitonin (sCT), an amylin receptor agonist and analogue of endogenous amylin, as a potential regulator of alcohol-induced activation of the mesolimbic dopamine system and alcohol consumption. Providing that the role of amylin signalling in alcohol-related behaviours remains unknown, the present experiments investigate the effect of sCT on these behaviours and the mechanisms involved. We showed that repeated sCT administration decreased alcohol and food intake in outbred rats. Moreover, single administration of the potent amylin receptor antagonist, AC187, increased short-term alcohol intake in outbred alcohol-consuming rats, but did not affect food intake. Acute administration of sCT prevented relapse-like drinking in the "alcohol deprivation effect" model in outbred alcohol-experienced rats. Additionally, acute sCT administration reduced operant oral alcohol self-administration (under the fixed ratio 4 schedule of reinforcement) in selectively bred Sardinian alcohol-preferring rats, while it did not alter operant self-administration (under the progressive ratio schedule of reinforcement) of a highly palatable chocolate-flavoured beverage in outbred rats. Lastly, we identified differential amylin receptor expression in high compared to low alcohol-consuming rats, as reflected by decreased calcitonin receptor and increased receptor activity modifying protein 1 expression in the nucleus accumbens (NAc) of high consumers. Collectively, our data suggest that amylin signalling, especially in the NAc, may contribute to reduction of various alcohol-related behaviours.


Subject(s)
Alcoholism , Amylin Receptor Agonists/pharmacology , Behavior, Animal/drug effects , Calcitonin/pharmacology , Drinking Behavior/drug effects , Nucleus Accumbens/metabolism , Peptide Fragments/pharmacology , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/metabolism , Alcohol Drinking , Amylin Receptor Agonists/administration & dosage , Animals , Calcitonin/administration & dosage , Disease Models, Animal , Eating/drug effects , Male , Peptide Fragments/administration & dosage , Rats , Rats, Wistar , Self Administration
4.
Addict Biol ; 24(3): 388-402, 2019 05.
Article in English | MEDLINE | ID: mdl-29405517

ABSTRACT

Alcohol expresses its reinforcing properties by activating areas of the mesolimbic dopamine system, which consists of dopaminergic neurons projecting from the ventral tegmental area to the nucleus accumbens. The findings that reward induced by food and addictive drugs involve common mechanisms raise the possibility that gut-brain hormones, which control appetite, such as amylin, could be involved in reward regulation. Amylin decreases food intake, and despite its implication in the regulation of natural rewards, tenuous evidence support amylinergic mediation of artificial rewards, such as alcohol. Therefore, the present experiments were designed to investigate the effect of salmon calcitonin (sCT), an amylin receptor agonist and analogue of endogenous amylin, on various alcohol-related behaviours in rodents. We showed that acute sCT administration attenuated the established effects of alcohol on the mesolimbic dopamine system, particularly alcohol-induced locomotor stimulation and accumbal dopamine release. Using the conditioned place preference model, we demonstrated that repeated sCT administration prevented the expression of alcohol's rewarding properties and that acute sCT administration blocked the reward-dependent memory consolidation. In addition, sCT pre-treatment attenuated alcohol intake in low alcohol-consuming rats, with a more evident decrease in high alcohol consumers in the intermittent alcohol access model. Lastly, sCT did not alter peanut butter intake, blood alcohol concentration and plasma corticosterone levels in mice. Taken together, the present data support that amylin signalling is involved in the expression of alcohol reinforcement and that amylin receptor agonists could be considered for the treatment of alcohol use disorder in humans.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptors, Islet Amyloid Polypeptide/physiology , Animals , Brain/physiology , Calcitonin/pharmacology , Conditioning, Psychological/drug effects , Corticosterone/metabolism , Dopaminergic Neurons/drug effects , Feeding Behavior/drug effects , Intestines/physiology , Limbic System/drug effects , Male , Mice , Motor Activity/drug effects , Rats, Wistar , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Reward
5.
Eur J Pain ; 23(4): 784-799, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30506955

ABSTRACT

BACKGROUND: Amylin is a calcitonin gene-related peptide family member expressed by nociceptors. Amylin's expression is down-regulated following nerve damage, and studies suggested it affects nociception. We aimed at clarifying amylin's effects on chronic neuropathic pain and investigating its site of action. METHODS: Chronic neuropathic pain was induced in rats by spared nerve injury (SNI) surgery. Mechanical allodynia/hyperalgesia and cold allodynia/hyperalgesia were assessed by the von Frey, pinprick, acetone and cold plate behavioural tests, respectively. Amylin, amylin-receptor antagonist (AC187) or vehicle solutions were delivered chronically, by a subcutaneous (SC) mini-osmotic pump, or acutely, by SC or intrathecal (IT) injections. Cellular and fibre markers were used to detect spinal cord alterations in SNI rats after chronic amylin administration. RESULTS: Continuous subcutaneous amylin administration aggravated cold allodynia in SNI animals, possibly via amylin-receptors (AmyR) in supraspinal areas. Acute intrathecal administration of amylin attenuated mechanical hyperalgesia, whereas AC187 reduced mechanical allodynia, suggesting distinct roles of endogenous amylin and of pharmacological amylin doses when targeting spinal cord amylin receptors. Chronic amylin administration promoted c-Fos activation only in the dorsal horn neurons of SHAM animals, suggesting a distinctive role of amylin in the activation of the spinal neuronal circuitry under neuropathic and physiological conditions. ERK1/2 phosphorylation increased in the dorsal horn neurons of SNI rats chronically treated with amylin. This ERK1/2 cascade activation may be related to amylin's effect on the aggravation of cold allodynia in SNI rats. CONCLUSIONS: Amylin's nociceptive effects seem to depend on the treatment duration and route of administration by acting at different levels of the nervous system. SIGNIFICANCE: Amylin modulated neuropathic pain by acting at different levels of the nervous system. Whereas supraspinal areas may be involved in amylin's induced pronociception, modulation of spinal cord amylin receptors by endogenous or pharmacological amylin doses triggers both pro- and antinociceptive effects.


Subject(s)
Amylin Receptor Agonists/pharmacology , Islet Amyloid Polypeptide/pharmacology , Neuralgia/metabolism , Pain Perception/drug effects , Peptide Fragments/pharmacology , Posterior Horn Cells/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Spinal Cord/drug effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Hyperalgesia/metabolism , Injections, Spinal , Male , Nociceptors/metabolism , Posterior Horn Cells/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Spinal Cord/metabolism , Spinal Cord/pathology
6.
Cephalalgia ; 38(3): 437-451, 2018 03.
Article in English | MEDLINE | ID: mdl-28165287

ABSTRACT

Background Calcitonin gene-related peptide (CGRP) is a neuropeptide that acts in the trigeminovascular system and is believed to play an important role in migraine. CGRP activates two receptors that are both present in the trigeminovascular system; the CGRP receptor and the amylin 1 (AMY1) receptor. CGRP receptor antagonists, including olcegepant (BIBN4096BS) and telcagepant (MK-0974), can treat migraine. This study aimed to determine the effectiveness of these antagonists at blocking CGRP receptor signalling in trigeminal ganglia (TG) neurons and transfected CGRP and AMY1 receptors in Cos7 cells, to better understand their mechanism of action. Methods CGRP stimulation of four intracellular signalling molecules relevant to pain (cAMP, CREB, p38 and ERK) were examined in rat TG neurons and compared to transfected CGRP and AMY1 receptors in Cos7 cells. Results In TG neurons, olcegepant displayed signal-specific differences in antagonism of CGRP responses. This effect was also evident in transfected Cos7 cells, where olcegepant blocked CREB phosphorylation more potently than expected at the AMY1 receptor, suggesting that the affinity of this antagonist can be dependent on the signalling pathway activated. Conclusions CGRP receptor antagonist activity appears to be assay-dependent. Thus, these molecules may not be as selective for the CGRP receptor as commonly reported.


Subject(s)
Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Dipeptides/pharmacology , Neurons/drug effects , Quinazolines/pharmacology , Signal Transduction/drug effects , Animals , COS Cells , Chlorocebus aethiops , Humans , Piperazines , Rats , Rats, Wistar , Receptors, Calcitonin Gene-Related Peptide/drug effects , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Signal Transduction/physiology , Transfection , Trigeminal Ganglion/drug effects
7.
Trends Mol Med ; 23(8): 709-720, 2017 08.
Article in English | MEDLINE | ID: mdl-28694141

ABSTRACT

Alzheimer'sdisease (AD) is a progressive neurodegenerative disorder, characterized by senile plaques constituting extracellular deposits of ß-amyloid (Aß) fibrils. Since Aß accumulation in the brain is considered an early event preceding, by decades, cognitive dysfunction, disease-modifying treatments are aimed at facilitating clearance of this protein from the brain or ameliorating its toxic effects. Recent studies have identified the amylin receptor as a capable mediator of the deleterious actions of Aß and furthermore, administration of amylin receptor-based peptides has been shown to improve spatial memory and learning in transgenic mouse models of AD. Here, by discussing available evidence, we posit that the amylin receptor could be considered a potential therapeutic target for AD, and present the rationale for using amylin receptor antagonists to treat this debilitating condition.


Subject(s)
Alzheimer Disease/drug therapy , Peptides/therapeutic use , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Humans , Mice , Mice, Transgenic , Receptors, Islet Amyloid Polypeptide/genetics
8.
Neuropharmacology ; 119: 170-181, 2017 06.
Article in English | MEDLINE | ID: mdl-28363773

ABSTRACT

Amylin is an important gut-brain axis hormone. Since amylin and amyloid-ß peptide (Aß) share similar ß sheet secondary structure despite not having the same primary sequences, we hypothesized that the accumulation of Aß in the brains of subjects with Alzheimer's disease (AD) might compete with amylin for binding to the amylin receptor (AmR). If true, adding exogenous amylin type peptides would compete with Aß and reduce the AD pathological cascade, improving cognition. Here we report that a 10-week course of peripheral treatment with human amylin significantly reduced multiple different markers associated with AD pathology, including reducing levels of phospho-tau, insoluble tau, two inflammatory markers (Iba1 and CD68), as well as cerebral Aß. Amylin treatment also led to improvements in learning and memory in two AD mouse models. Mechanistic studies showed that an amylin receptor antagonist successfully antagonized some protective effects of amylin in vivo, suggesting that the protective effects of amylin require interaction with its cognate receptor. Comparison of signaling cascades emanating from AmR suggest that amylin electively suppresses activation of the CDK5 pathway by Aß. Treatment with amylin significantly reduced CDK5 signaling in a receptor dependent manner, dramatically decreasing the levels of p25, the active form of CDK5 with a corresponding reduction in tau phosphorylation. This is the first report documenting the ability of amylin treatment to reduce tauopathy and inflammation in animal models of AD. The data suggest that the clinical analog of amylin, pramlintide, might exhibit utility as a therapeutic agent for AD and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Receptors, Islet Amyloid Polypeptide/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/pharmacology , Ligands , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Mutation/genetics , Peptide Fragments/therapeutic use , Presenilin-1/genetics , Presenilin-1/metabolism , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Spatial Memory/drug effects , tau Proteins/genetics , tau Proteins/metabolism
9.
Biol Psychiatry ; 82(11): 828-838, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28237459

ABSTRACT

BACKGROUND: The pancreatic- and brain-derived hormone amylin promotes negative energy balance and is receiving increasing attention as a promising obesity therapeutic. However, the neurobiological substrates mediating amylin's effects are not fully characterized. We postulated that amylin acts in the lateral dorsal tegmental nucleus (LDTg), an understudied neural processing hub for reward and homeostatic feeding signals. METHODS: We used immunohistochemical and quantitative polymerase chain reaction analyses to examine expression of the amylin receptor complex in rat LDTg tissue. Behavioral experiments were performed to examine the mechanisms underlying the hypophagic effects of amylin receptor activation in the LDTg. RESULTS: Immunohistochemical and quantitative polymerase chain reaction analyses show expression of the amylin receptor complex in the LDTg. Activation of LDTg amylin receptors by the agonist salmon calcitonin dose-dependently reduces body weight, food intake, and motivated feeding behaviors. Acute pharmacological studies and longer-term adeno-associated viral knockdown experiments indicate that LDTg amylin receptor signaling is physiologically and potentially preclinically relevant for energy balance control. Finally, immunohistochemical data indicate that LDTg amylin receptors are expressed on gamma-aminobutyric acidergic neurons, and behavioral results suggest that local gamma-aminobutyric acid receptor signaling mediates the hypophagia after LDTg amylin receptor activation. CONCLUSIONS: These findings identify the LDTg as a novel nucleus with therapeutic potential in mediating amylin's effects on energy balance through gamma-aminobutyric acid receptor signaling.


Subject(s)
Amylin Receptor Agonists/therapeutic use , Gene Expression Regulation/drug effects , Islet Amyloid Polypeptide/pharmacology , Signal Transduction/physiology , Ventral Tegmental Area/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Body Weight/drug effects , Body Weight/physiology , Calcitonin/pharmacology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Eating/drug effects , Food Preferences/drug effects , GABA Agents/pharmacology , Male , Motivation/drug effects , Peptide Fragments/pharmacology , Phosphopyruvate Hydratase/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor Activity-Modifying Proteins/genetics , Receptor Activity-Modifying Proteins/metabolism , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/genetics , Receptors, Islet Amyloid Polypeptide/metabolism , Signal Transduction/drug effects
10.
J Alzheimers Dis ; 56(3): 1087-1099, 2017.
Article in English | MEDLINE | ID: mdl-28059785

ABSTRACT

Findings from Alzheimer's disease (AD) mouse models showed that amylin treatment improved AD pathology and enhanced amyloid-ß (Aß) brain to blood clearance; however, the mechanism was not investigated. Using the Tg2576 AD mouse model, a single intraperitoneal injection of amylin significantly increased Aß serum levels, and the effect was abolished by AC253, an amylin receptor antagonist, suggesting that amylin effect could be mediated by its receptor. Subsequent mechanistic studies showed amylin enhanced Aß transport across a cell-based model of the blood-brain barrier (BBB), an effect that was abolished when the amylin receptor was inhibited by two amylin antagonists and by siRNA knockdown of amylin receptor Ramp3. To explain this finding, amylin effect on Aß transport proteins expressed at the BBB was evaluated. Findings indicated that cells treated with amylin induced LRP1 expression, a major receptor involved in brain Aß efflux, in plasma membrane fraction, suggesting intracellular translocation of LRP1 from the cytoplasmic pool. Increased LRP1 in membrane fraction could explain, at least in part, the enhanced uptake and transport of Aß across the BBB. Collectively, our findings indicated that amylin induced Aß brain to blood clearance through amylin receptor by inducing LRP1 subcellular translocation to the plasma membrane of the BBB endothelium.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Capillary Permeability/physiology , Islet Amyloid Polypeptide/metabolism , Animals , Blood-Brain Barrier/drug effects , Cell Culture Techniques , Cells, Cultured , Central Nervous System Agents/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Islet Amyloid Polypeptide/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1 , Mice, Transgenic , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/genetics , Receptors, Islet Amyloid Polypeptide/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism
11.
Neuropeptides ; 57: 95-100, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26778650

ABSTRACT

Amylin (AMY) is a member of calcitonin family of peptides. In this study, the effects of intrathecal (i.t) injection of AMY on the inflammatory pain and on the cAMP accumulation in the rat spinal cells were investigated. By using AMY receptor antagonists, we also studied the pharmacology of AMY receptors in the spinal cells. Formalin model of inflammatory pain was induced by intraplantar injection of formalin. AMY (0.06250-2500pmol/rat) was administrated i.t 15min before the injection of formalin. Antagonists were injected i.t 10min before the injection of AMY and/or morphine. AMY reduced formalin-induced pain in a dose dependent mode. This effect was inhibited by the potent AMY antagonist, AC187 but not CGRP8-37. rAMY8-37, most commonly reported as a weak AMY antagonist, showed to be equally or more potent than AC187 in antagonizing the above effects. The opioid antagonist, naloxone, had no significant effects on AMY antinociceptive effects. Primary dissociated cell culture was used to investigate the effect of AMY on cAMP production and to characterize AMY receptors in the spinal cells. AMY moderately increases cAMP accumulation in the spinal cells with an EC50 value of 74.62nM. This effect was not affected by CGRP8-37 but was inhibited by AC187 and rAMY8-37 with pA2 values of 7.94 and 7.87 respectively. In conclusion, effects of AMY in reducing formalin induced pain and on the cAMP accumulation by spinal cells are mediated through undefined receptors.


Subject(s)
Analgesics/administration & dosage , Cyclic AMP/metabolism , Inflammation/metabolism , Islet Amyloid Polypeptide/administration & dosage , Nociception/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Amylin Receptor Agonists/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Calcitonin Gene-Related Peptide/administration & dosage , Formaldehyde , Inflammation/chemically induced , Injections, Spinal , Islet Amyloid Polypeptide/antagonists & inhibitors , Male , Morphine/administration & dosage , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Pain/chemically induced , Pain Threshold/drug effects , Peptide Fragments/administration & dosage , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Spinal Cord/embryology
12.
FASEB J ; 29(7): 2970-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808537

ABSTRACT

The toxic effects of human islet amyloid polypeptide (IAPP) on pancreatic islets have been widely studied. However, much less attention has been paid to the physiologic actions of IAPP on pancreatic ß cells, which secrete this peptide together with insulin upon glucose stimulation. Here, we aimed to explore the signaling pathways and mitogenic actions of IAPP on ß cells. We show that IAPP activated Erk1/2 and v-akt murine thymoma viral oncogene homolog 1 (Akt) at the picomolar range (10-100 pM) in mouse pancreatic islets and MIN6 ß cells cultured at low glucose concentrations. In contrast, IAPP decreased the induction of these pathways by high glucose levels. Consistently, IAPP induced a 1.7-fold increase of ß-cell proliferation at low-glucose conditions, whereas it reduced ß-cell proliferation at high glucose levels. Strikingly, the specific antagonist of the IAPP receptor AC187 (100 nM) decreased the activation of Erk1/2 and Akt and reduced ß-cell proliferation by 24% in glucose-stimulated ß cells, uncovering a key role of endogenously released IAPP in ß-cell responses to glucose. We conclude that exogenously added IAPP exerts a dual effect on ß-cell mitogenic signaling and proliferation, depending on the glucose concentration. Importantly, secreted IAPP contributes to the signaling and mitogenic response of ß cells to glucose through an autocrine mechanism.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/metabolism , Animals , Autocrine Communication/drug effects , Cell Line , Cell Proliferation/drug effects , Glucose/metabolism , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Islet Amyloid Polypeptide/pharmacology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/metabolism
13.
Am J Physiol Endocrinol Metab ; 307(1): E24-33, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24801386

ABSTRACT

The present study investigated a novel oral dual amylin and calcitonin receptor agonist (DACRA), KBP-042, in head-to-head comparison with salmon calcitonin (sCT) with regard to in vitro receptor pharmacology, ex vivo pancreatic islet studies, and in vivo proof of concept studies in diet-induced obese (DIO) and Zucker diabetic fatty (ZDF) rats. In vitro, KBP-042 demonstrated superior binding affinity and activation of amylin and calcitonin receptors, and ex vivo, KBP-042 exerted inhibitory action on stimulated insulin and glucagon release from isolated islets. In vivo, KBP-042 induced a superior and pronounced reduction in food intake in conjunction with a sustained pair-fed corrected weight loss in DIO rats. Concomitantly, KBP-042 improved glucose homeostasis and reduced hyperinsulinemia and hyperleptinemia in conjunction with enhanced insulin sensitivity. In ZDF rats, KBP-042 induced a superior attenuation of diabetic hyperglycemia and alleviated impaired glucose and insulin tolerance. Concomitantly, KBP-042 preserved insulinotropic and induced glucagonostatic action, ultimately preserving pancreatic insulin and glucagon content. In conclusion, oral KBP-042 is a novel DACRA, which exerts antiobesity and antidiabetic efficacy by dual modulation of insulin sensitivity and directly decelerating stress on the pancreatic α- and ß-cells. These results could provide the basis for oral KBP-042 as a novel therapeutic agent in type 2 diabetes.


Subject(s)
Anti-Obesity Agents/administration & dosage , Calcitonin/analogs & derivatives , Hypoglycemic Agents/administration & dosage , Receptors, Calcitonin/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Administration, Oral , Animals , Blood Glucose/drug effects , Calcitonin/administration & dosage , Insulin Resistance , Male , Rats , Rats, Sprague-Dawley , Rats, Zucker , Treatment Outcome , Weight Loss/drug effects
14.
Neuropsychopharmacology ; 38(9): 1685-97, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23474592

ABSTRACT

The ability of amylin, a pancreatic ß-cell-derived neuropeptide, to promote negative energy balance has been ascribed to neural activation at the area postrema. However, despite amylin binding throughout the brain, the possible role of amylin signaling at other nuclei in the control of food intake has been largely neglected. We show that mRNA for all components of the amylin receptor complex is expressed in the ventral tegmental area (VTA), a mesolimbic structure mediating food intake and reward. Direct activation of VTA amylin receptors reduces the intake of chow and palatable sucrose solution in rats. This effect is mediated by reductions in meal size and is not due to nausea/malaise or prolonged suppression of locomotor activity. VTA amylin receptor activation also reduces sucrose self-administration on a progressive ratio schedule. Finally, antagonist studies provide novel evidence that VTA amylin receptor blockade increases food intake and attenuates the intake-suppressive effects of a peripherally administered amylin analog, suggesting that amylin receptor signaling in the VTA is physiologically relevant for food intake control and potentially clinically relevant for the treatment of obesity.


Subject(s)
Eating/physiology , Receptors, Islet Amyloid Polypeptide/physiology , Ventral Tegmental Area/physiology , Amylin Receptor Agonists , Animals , Calcitonin/administration & dosage , Calcitonin/antagonists & inhibitors , Calcitonin/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Eating/drug effects , Male , Microinjections , Motor Activity/drug effects , Nausea/chemically induced , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , Rats , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/metabolism , Reinforcement Schedule , Reward , Self Administration , Sucrose/administration & dosage , Sucrose/pharmacology , Ventral Tegmental Area/drug effects
15.
J Neurosci ; 32(48): 17401-6, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197731

ABSTRACT

Alzheimer's disease (AD) is characterized by accumulation of amyloid-ß peptide (Aß) in the brain regions that subserve memory and cognition. The amylin receptor is a potential target receptor for expression of the deleterious actions of soluble oligomeric Aß species. We investigated whether the amylin receptor antagonist, AC253, neutralizes the depressant effects of Aß(1-42) and human amylin on hippocampal long-term potentiation (LTP). Furthermore, we examined whether depressed levels of LTP observed in transgenic mice, which overexpress amyloid precursor protein (TgCRND8), could be restored with AC253. In mouse hippocampal brain slices, field EPSPs were recorded from the stratum radiatum layer of the CA1 area (cornu ammonis 1 region of the hippocampus) in response to electrical stimulation of Schaeffer collateral afferents. LTP was induced by 3-theta burst stimulation protocols. Aß(1-42) (50 nM) and human amylin (50 nM), but not Aß(42-1) (50 nM), depressed LTP evoked using both stimulation protocols. Preapplication of AC253 (250 nM) blocked Aß- and human amylin-induced reduction of LTP without affecting baseline transmission or LTP on its own. In contrast to wild-type controls, where robust LTP is observed, 6- to 12-month-old TgCRND8 mice show blunted LTP that is significantly enhanced by application of AC253. Our data demonstrate that the effects of Aß(1-42) and human amylin on LTP are expressed via the amylin receptor, and moreover, blockade of this receptor increases LTP in transgenic mice that show increased brain amyloid burden. Amylin receptor antagonists could serve as potentially useful therapeutic agents in AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Hippocampus/drug effects , Islet Amyloid Polypeptide/pharmacology , Long-Term Potentiation/drug effects , Peptide Fragments/pharmacology , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Animals , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Mice , Mice, Transgenic
16.
Apoptosis ; 17(1): 37-47, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21947943

ABSTRACT

Deposition of amyloid-beta (Aß) protein, a 39-43 amino acid peptide, in the brain is a major pathological feature of Alzheimer's disease (AD). We have previously provided evidence that in primary cultures of rat basal forebrain and human fetal neurons (HFNs), neurotoxic effects of oligomeric Aß are expressed through the amylin receptor. In this study, we utilized RT-PCR arrays to compare RNA expression levels of 84 markers for pro and anti- apoptotic signalling pathways following exposure of HFNs to either Aß(1-42) (20 µM) or human amylin (2 µM). Oligomeric Aß(1-42) or human amylin was applied to HFNs alone or after pre-treatment of cultures with the amylin receptor antagonist, AC253. Changes in RNA levels were then quantified and compared to each other in order to identify increases or decreases in gene expression of apoptotic markers. Applications of Aß(1-42) or human amylin, but not the inactive inverse sequence Aß(42-1) or rat amylin, resulted in a time-dependent marked increase in mediators of apoptosis including a 10- to 30-fold elevations in caspases 3, 6, 9, BID and XIAP levels. Amylin receptor antagonists, AC253 (10 µM) or AC187 (10 µM), significantly attenuated the induction of several pro-apoptotic mediators up-regulated following exposure to Aß(1-42) or human amylin and increased the expression of several anti-apoptotic markers. These data allow us to identify key elements in the Aß-induced apoptosis that are blocked by antagonism of the amylin receptor and further support the potential for amylin receptor blockade as a potential therapeutic avenue in AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis , Islet Amyloid Polypeptide/metabolism , Peptide Fragments/metabolism , Receptors, Islet Amyloid Polypeptide/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Neurons/cytology , Neurons/metabolism , Rats , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/genetics
17.
Br J Pharmacol ; 166(1): 151-67, 2012 May.
Article in English | MEDLINE | ID: mdl-22014233

ABSTRACT

BACKGROUND AND PURPOSE: Amylin (Amy) is an important glucoregulatory peptide and AMY receptors are clinical targets for diabetes and obesity. Human (h) AMY receptor subtypes are complexes of the calcitonin (CT) receptor with receptor activity-modifying proteins (RAMPs); their rodent counterparts have not been characterized. To allow identification of the most clinically relevant receptor subtype, the elucidation of rat (r) AMY receptor pharmacology is necessary. EXPERIMENTAL APPROACH: Receptors were transiently transfected into COS-7 cells and cAMP responses measured in response to different agonists, with or without antagonists. Competition binding experiments were performed to determine rAmy affinity. KEY RESULTS: rCT was the most potent agonist of rCT((a)) receptors, whereas rAmy was most potent at rAMY(1(a)) and rAMY(3(a)) receptors. rAmy bound to these receptors with high affinity. Rat α-calcitonin gene-related peptide (CGRP) was equipotent to rAmy at both AMY receptors. Rat adrenomedullin (AM) and rAM2/intermedin activated all three receptors but were most effective at rAMY(3(a)) . AC187, AC413 and sCT(8-32) were potent antagonists at all three receptors. rαCGRP(8-37) displayed selectivity for rAMY receptors over rCT((a)) receptors. rAMY(8-37) was a weak antagonist but was more effective at rAMY(1(a)) than rAMY(3(a)) . CONCLUSIONS AND IMPLICATIONS: AMY receptors were generated by co-expression of rCT((a)) with rRAMP1 or 3, forming rAMY(1(a)) and rAMY(3(a)) receptors, respectively. CGRP was more potent at rAMY than at hAMY receptors. No antagonist tested was able to differentiate the rAMY receptor subtypes. The data emphasize the need for and provide a useful resource for developing new CT or AMY receptor ligands as pharmacological tools or potential clinical candidates.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Cyclic AMP/metabolism , Islet Amyloid Polypeptide/metabolism , Receptors, Islet Amyloid Polypeptide/metabolism , Adrenomedullin/metabolism , Amylin Receptor Agonists , Animals , Binding, Competitive , COS Cells , Chlorocebus aethiops , Male , Mice , Neuropeptides/metabolism , Protein Binding , Rats , Rats, Wistar , Receptor Activity-Modifying Protein 1/metabolism , Receptor Activity-Modifying Protein 3/metabolism , Receptors, Calcitonin/agonists , Receptors, Calcitonin/antagonists & inhibitors , Receptors, Calcitonin/metabolism , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors
18.
Assay Drug Dev Technol ; 9(5): 522-31, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21561374

ABSTRACT

Activation of G(i)-coupled G protein-coupled receptor (GPCRs) by their ligands leads to inhibition of adenylyl cyclase (AC) and reduction of cyclic adenosine monophosphate (cAMP) levels in cells. The traditional cAMP assay for G(i)-coupled GPCRs commonly uses forskolin, a nonspecific AC activator, to increase the basal cAMP level in cells to create an assay window for ligand detection. However, there is still a need to develop a nonforskolin-based cAMP assay because of the challenges inherent in titrating the concentration of forskolin to achieve a reliable assay window, along with issues related to the cAMP-independent effects of forskolin. Herein, we describe such an assay by utilizing the endogenous activity of the calcitonin receptor in Chinese hamster ovary (CHO) cells. The calcitonin receptor is a G(s)-coupled GPCR that, when activated by calcitonin, leads to the stimulation of AC and increases cAMP in cells. Thus, we use calcitonin, instead of forskolin, to increase the basal cAMP level in CHO cells to achieve an assay window. We demonstrated that calcitonin peptides robustly increased cAMP accumulation in several CHO cell lines stably expressing well-known G(i)-coupled GPCRs, such as the Dopamine D2 receptor, the Opioid µ receptor, or the Cannabinoid receptor-1. Agonists of these G(i)-coupled GPCRs attenuated calcitonin-induced cAMP production in their receptor stable cell lines. On the other hand, antagonists and/or inverse agonists blocked the effects of their agonists on calcitonin-induced cAMP production. This calcitonin-based cAMP assay has been demonstrated to be sensitive and robust and exhibited acceptable assay windows (signal/noise ratio) and, thus, can be applied to screen for agonists and antagonists/inverse agonists of G(i)-coupled GPCRs in high-throughput screening formats.


Subject(s)
Calcitonin/physiology , Cyclic AMP/analysis , Receptors, Calcitonin/physiology , Receptors, G-Protein-Coupled/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adenylyl Cyclase Inhibitors , Animals , CHO Cells , Cell Culture Techniques , Colforsin/metabolism , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Drug Evaluation, Preclinical , Female , Humans , Inhibitory Concentration 50 , Ligands , Molecular Targeted Therapy , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Radioligand Assay , Rats , Receptors, Calcitonin/agonists , Receptors, Calcitonin/analysis , Receptors, Calcitonin/antagonists & inhibitors , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Opioid, mu/analysis , Receptors, Opioid, mu/antagonists & inhibitors , Salmon
19.
Am J Pathol ; 178(1): 140-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224052

ABSTRACT

Disruption of neurotoxic effects of amyloid ß protein (Aß) is one of the major, but as yet elusive, goals in the treatment of Alzheimer's disease (AD). The amylin receptor, activated by a pancreatic polypeptide isolated from diabetic patients, is a putative target for the actions of Aß in the brain. Here we show that in primary cultures of human fetal neurons (HFNs), AC253, an amylin receptor antagonist, blocks electrophysiological effects of Aß. Pharmacological blockade of the amylin receptor or its down-regulation using siRNA in HFNs confers neuroprotection against oligomeric Aß-induced caspase-dependent and caspase-independent apoptotic cell death. In transgenic mice (TgCRND8) that overexpress amyloid precursor protein, amylin receptor is up-regulated in specific brain regions that also demonstrate an elevated amyloid burden. The expression of Aß actions through the amylin receptor in human neurons and temporospatial interrelationship of Aß and the amylin receptor in an in vivo model of AD together provide a persuasive rationale for this receptor as a novel therapeutic target in the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Apoptosis , Neurons/physiology , Receptors, Islet Amyloid Polypeptide/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Animals , Brain/metabolism , Cytoprotection , Gene Knockdown Techniques , Humans , Mice , Mice, Transgenic , Neurons/drug effects , Patch-Clamp Techniques , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Receptors, Islet Amyloid Polypeptide/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...