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










Publication year range
1.
Neuropsychopharmacology ; 49(6): 915-923, 2024 May.
Article in English | MEDLINE | ID: mdl-38374364

ABSTRACT

Opioid use disorder is a chronic relapsing disorder encompassing misuse, dependence, and addiction to opioid drugs. Long term maintenance of associations between the reinforcing effects of the drug and the cues associated with its intake are a leading cause of relapse. Indeed, exposure to the salient drug-associated cues can lead to drug cravings and drug seeking behavior. The dorsal hippocampus (dHPC) and locus coeruleus (LC) have emerged as important structures for linking the subjective rewarding effects of opioids with environmental cues. However, their role in cue-induced reinstatement of opioid use remains to be further elucidated. In this study, we showed that chemogenetic inhibition of excitatory dHPC neurons during re-exposure to drug-associated cues significantly attenuates cue-induced reinstatement of morphine-seeking behavior. In addition, the same manipulation reduced reinstatement of sucrose-seeking behavior but failed to alter memory recall in the object location task. Finally, intact activity of tyrosine hydroxylase (TH) LC-dHPCTh afferents is necessary to drive cue induced reinstatement of morphine-seeking as inhibition of this pathway blunts cue-induced drug-seeking behavior. Altogether, these studies show an important role of the dHPC and LC-dHPCTh pathway in mediating cue-induced reinstatement of opioid seeking.


Subject(s)
Cues , Drug-Seeking Behavior , Hippocampus , Locus Coeruleus , Self Administration , Animals , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Female , Drug-Seeking Behavior/drug effects , Drug-Seeking Behavior/physiology , Morphine/pharmacology , Morphine/administration & dosage , Rats, Sprague-Dawley , Neural Pathways/drug effects , Neural Pathways/physiology , Analgesics, Opioid/pharmacology , Analgesics, Opioid/administration & dosage , Opioid-Related Disorders/physiopathology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology
2.
Nat Commun ; 15(1): 750, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38286800

ABSTRACT

The hippocampus is pivotal in integrating emotional processing, learning, memory, and reward-related behaviors. The dorsal hippocampus (dHPC) is particularly crucial for episodic, spatial, and associative memory, and has been shown to be necessary for context- and cue-associated reward behaviors. The nucleus accumbens (NAc), a central structure in the mesolimbic reward pathway, integrates the salience of aversive and rewarding stimuli. Despite extensive research on dHPC→NAc direct projections, their sufficiency in driving reinforcement and reward-related behavior remains to be determined. Our study establishes that activating excitatory neurons in the dHPC is sufficient to induce reinforcing behaviors through its direct projections to the dorso-medial subregion of the NAc shell (dmNAcSh). Notably, dynorphin-containing neurons specifically contribute to dHPC-driven reinforcing behavior, even though both dmNAcSh dynorphin- and enkephalin-containing neurons are activated with dHPC stimulation. Our findings unveil a pathway governing reinforcement, advancing our understanding of the hippocampal circuity's role in reward-seeking behaviors.


Subject(s)
Dynorphins , Nucleus Accumbens , Phospholipid Ethers , Nucleus Accumbens/physiology , Hippocampus/physiology , Reward , Neurons/physiology
4.
Front Syst Neurosci ; 16: 1014768, 2022.
Article in English | MEDLINE | ID: mdl-36341476

ABSTRACT

Decades of research advances have established a central role for endogenous opioid systems in regulating reward processing, mood, motivation, learning and memory, gastrointestinal function, and pain relief. Endogenous opioid systems are present ubiquitously throughout the central and peripheral nervous system. They are composed of four families, namely the µ (MOPR), κ (KOPR), δ (DOPR), and nociceptin/orphanin FQ (NOPR) opioid receptors systems. These receptors signal through the action of their endogenous opioid peptides ß-endorphins, dynorphins, enkephalins, and nociceptins, respectfully, to maintain homeostasis under normal physiological states. Due to their prominent role in pain regulation, exogenous opioids-primarily targeting the MOPR, have been historically used in medicine as analgesics, but their ability to produce euphoric effects also present high risks for abuse. The ability of pain and opioid use to perturb endogenous opioid system function, particularly within the central nervous system, may increase the likelihood of developing opioid use disorder (OUD). Today, the opioid crisis represents a major social, economic, and public health concern. In this review, we summarize the current state of the literature on the function, expression, pharmacology, and regulation of endogenous opioid systems in pain. Additionally, we discuss the adaptations in the endogenous opioid systems upon use of exogenous opioids which contribute to the development of OUD. Finally, we describe the intricate relationship between pain, endogenous opioid systems, and the proclivity for opioid misuse, as well as potential advances in generating safer and more efficient pain therapies.

5.
Pain ; 163(5): 809-819, 2022 05 01.
Article in English | MEDLINE | ID: mdl-34510137

ABSTRACT

ABSTRACT: Many analgesics inadequately address the psychiatric comorbidities of chronic and persistent pain, but there is no standard preclinical model of pain-altered behavior to support the development of new therapies. To explore this conflicting and inconclusive literature, we conducted a focused systematic review and meta-analysis on the effect of complete Freund adjuvant-induced (CFA) rodent hind paw inflammation on multiple classical indicators of exploratory behavior, stress coping, and naturalistic behavior. Our primary objective was to define CFA's effect on assays including, but not limited to, the elevated plus maze and forced swim test. Our secondary objective was to discover how variables such as species and strain may influence outcomes in such assays. We searched Ovid MEDLINE, Embase, Scopus, and Web of Science in April and October 2020 for studies with adult rodents injected with CFA into the hind paw and subsequently tested for aspects of "anxiety-like" or "depressive-like" behaviors. Forty-four studies evaluated performance in the elevated plus or zero maze, open field test, light-dark box, place escape and avoidance paradigm, forced swim test, tail suspension test, sucrose preference test, wheel running, and burrowing assay. Complete Freund adjuvant modestly but significantly decreased exploratory behavior, significantly increased passive stress coping in the tail suspension test but not the forced swim test, and significantly decreased preference for sucrose and naturally rewarding activity. Subgroup analyses revealed significant differences between species and animal sourcing. Based on the evidence provided here, we conclude future studies should focus on CFA's effect on natural rewards and naturalistic behaviors.


Subject(s)
Motor Activity , Rodentia , Animals , Behavior, Animal , Disease Models, Animal , Freund's Adjuvant/toxicity , Pain/chemically induced , Pain/psychology , Sucrose/pharmacology
6.
Nat Neurosci ; 24(11): 1601-1613, 2021 11.
Article in English | MEDLINE | ID: mdl-34663957

ABSTRACT

The persistence of negative affect in pain leads to co-morbid symptoms such as anhedonia and depression-major health issues in the United States. The neuronal circuitry and contribution of specific cellular populations underlying these behavioral adaptations remains unknown. A common characteristic of negative affect is a decrease in motivation to initiate and complete goal-directed behavior, known as anhedonia. We report that in rodents, inflammatory pain decreased the activity of ventral tegmental area (VTA) dopamine (DA) neurons, which are critical mediators of motivational states. Pain increased rostromedial tegmental nucleus inhibitory tone onto VTA DA neurons, making them less excitable. Furthermore, the decreased activity of DA neurons was associated with reduced motivation for natural rewards, consistent with anhedonia-like behavior. Selective activation of VTA DA neurons was sufficient to restore baseline motivation and hedonic responses to natural rewards. These findings reveal pain-induced adaptations within VTA DA neurons that underlie anhedonia-like behavior.


Subject(s)
Adaptation, Physiological/physiology , Anhedonia/physiology , Dopaminergic Neurons/metabolism , Pain/metabolism , Ventral Tegmental Area/metabolism , Animals , Conditioning, Operant/physiology , Dopaminergic Neurons/chemistry , Female , Male , Optogenetics/methods , Pain/genetics , Rats , Rats, Long-Evans , Rats, Transgenic , Ventral Tegmental Area/chemistry
7.
Int Rev Neurobiol ; 157: 31-68, 2021.
Article in English | MEDLINE | ID: mdl-33648672

ABSTRACT

Across centuries and civilizations opioids have been used to relieve pain. In our modern societies, opioid-based analgesics remain one of the most efficient treatments for acute pain. However, the long-term use of opioids can lead to the development of analgesic tolerance, opioid-induced hyperalgesia, opioid use disorders, and overdose, which can ultimately produce respiratory depressant effects with fatal consequences. In addition to the nociceptive sensory component of pain, negative affective states arising from persistent pain represent a risk factor for developing an opioid use disorder. Several studies have indicated that the increase in prescribed opioid analgesics since the 1990s represents the root of our current opioid epidemic. In this review, we will present our current knowledge on the endogenous opioid system within the pain neuroaxis and the plastic changes occurring in this system that may underlie the occurrence of pain-induced negative affect leading to misuse and abuse of opioid medications. Dissecting the allostatic neuronal changes occurring during pain is the most promising avenue to uncover novel targets for the development of safer pain medications. We will discuss this along with current and potential approaches to treat pain-induced negative affective states that lead to drug misuse. Moreover, this chapter will provide a discussion on potential avenues to reduce the abuse potential of new analgesic drugs and highlight a basis for future research and drug development based on recent advances in this field.


Subject(s)
Acute Pain , Analgesics, Opioid , Pain Management , Acute Pain/drug therapy , Affect , Analgesics, Opioid/adverse effects , Humans , Opioid-Related Disorders/prevention & control , Pain Management/adverse effects , Substance-Related Disorders/prevention & control
8.
Pain ; 162(6): 1705-1721, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33433146

ABSTRACT

ABSTRACT: Pain puts patients at risk for developing psychiatric conditions such as anxiety and depression. Preclinical mouse models of pain-induced affective behavior vary widely in methodology and results, impairing progress towards improved therapeutics. To systematically investigate the effect of long-term inflammatory pain on exploratory behavior and stress coping strategy, we assessed male C57BL/6J mice in the forced swim test (FST), elevated zero maze, and open field test at 4 and 6 weeks postinjection of Complete Freund's Adjuvant, while controlling for testing order and combination. Inflammatory pain did not induce a passive stress coping strategy in the FST and did not reduce exploratory behavior in the elevated zero maze or the open field test. Using systematic correlational analysis and composite behavioral scores, we found no consistent association among measures for mice with or without inflammatory pain. A meta-analysis of similar studies indicated a modest, significant effect of Complete Freund's Adjuvant on exploratory behavior, but not immobility in the FST, and high heterogeneity among effect sizes in all 3 paradigms. Given the urgency for understanding the mechanisms of pain comorbidities and identifying novel therapies, these findings support the reallocation of our limited resources away from such unreliable assays and toward motivated and naturalistic behaviors. Future studies in pain and psychiatric translational research may benefit by considering outcomes beyond binary categorization, quantifying the associations between multiple measured behaviors, and agnostically identifying subtle yet meaningful patterns in behaviors.


Subject(s)
Exploratory Behavior , Pain , Adaptation, Psychological , Animals , Anxiety , Behavior, Animal , Depression/etiology , Disease Models, Animal , Humans , Male , Maze Learning , Mice , Mice, Inbred C57BL , Pain/etiology
9.
J Med Chem ; 63(22): 13618-13637, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33170687

ABSTRACT

In this work, we studied a series of carfentanyl amide-based opioid derivatives targeting the mu opioid receptor (µOR) and the delta opioid receptor (δOR) heteromer as a credible novel target in pain management therapy. We identified a lead compound named MP135 that exhibits high G-protein activity at µ-δ heteromers compared to the homomeric δOR or µOR and low ß-arrestin2 recruitment activity at all three. Furthermore, MP135 exhibits distinct signaling profile, as compared to the previously identified agonist targeting µ-δ heteromers, CYM51010. Pharmacological characterization of MP135 supports the utility of this compound as a molecule that could be developed as an antinociceptive agent similar to morphine in rodents. In vivo characterization reveals that MP135 maintains untoward side effects such as respiratory depression and reward behavior; together, these results suggest that optimization of MP135 is necessary for the development of therapeutics that suppress the classical side effects associated with conventional clinical opioids.


Subject(s)
Fentanyl/analogs & derivatives , Receptors, Opioid, delta/agonists , Analgesics/chemical synthesis , Analgesics/pharmacology , Animals , Cell Line , Fentanyl/chemical synthesis , Fentanyl/pharmacology , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Long-Evans , Receptors, Opioid, delta/metabolism
10.
Pain ; 161(12): 2798-2804, 2020 12.
Article in English | MEDLINE | ID: mdl-32639370

ABSTRACT

Currently, opioids targeting mu-opioid receptors are the most potent drugs for acute and cancer pain. However, opioids produce adverse side effects such as constipation, respiratory depression, or addiction potential. We recently developed (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide (NFEPP), a compound that does not evoke central or intestinal side effects due to its selective activation of mu-opioid receptors at low pH in peripheral injured tissues. Although we demonstrated that NFEPP effectively abolishes injury-induced pain, hyperalgesia, and allodynia in rodents, the efficacy of NFEPP in nonevoked ongoing pain remains to be established. Here, we examined reward, locomotor activity, and defecation in rats with complete Freund's adjuvant-induced paw inflammation to compare fentanyl's and NFEPP's potentials to induce side effects and to inhibit spontaneous pain. We demonstrate that low, but not higher, doses of NFEPP produce conditioned place preference but not constipation or motor disturbance, in contrast to fentanyl. Using a peripherally restricted antagonist, we provide evidence that NFEPP-induced place preference is mediated by peripheral opioid receptors. Our results indicate that a low dose of NFEPP produces reward by abolishing spontaneous inflammatory pain.


Subject(s)
Analgesics, Opioid , Pain , Analgesics, Opioid/therapeutic use , Animals , Hydrogen-Ion Concentration , Hyperalgesia/drug therapy , Pain/drug therapy , Rats , Receptors, Opioid , Receptors, Opioid, mu/genetics
11.
Br J Pharmacol ; 176(23): 4510-4520, 2019 12.
Article in English | MEDLINE | ID: mdl-31355457

ABSTRACT

BACKGROUND AND PURPOSE: Adverse side effects of conventional opioids can be avoided if ligands selectively activate peripheral opioid receptors in injured tissue. Injury and inflammation are typically accompanied by acidification. In this study, we examined influences of low pH and mutation of the ionizable amino acid residue H2976.52 on µ-opioid receptor binding and signalling induced by the µ-opioid receptor ligands fentanyl, DAMGO, and naloxone. EXPERIMENTAL APPROACH: HEK 293 cells stably transfected with µ-opioid receptors were used to study opioid ligand binding, [35 S]-GTPγS binding, and cAMP reduction at physiological and acidic pH. We used µ-opioid receptors mutated at H2976.52 to A (MOR-H2976.52 A) to delineate ligand-specific interactions with H2976.52 . KEY RESULTS: Low pH and the mutant receptor MOR-H2976.52 A impaired naloxone binding and antagonism of cAMP reduction. In addition, DAMGO binding and G-protein activation were decreased under these conditions. Fentanyl-induced signalling was not influenced by pH and largely independent of H2976.52 . CONCLUSIONS AND IMPLICATIONS: Our investigations indicate that low pH selectively impairs µ-opioid receptor signalling modulated by ligands capable of forming hydrogen bonds with H2976.52 . We propose that protonation of H2976.52 at acidic pH reduces binding and subsequent signalling of such ligands. Novel agonists targeting opioid receptors in injured tissue might benefit from lack of hydrogen bond formation with H2976.52 .


Subject(s)
Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Fentanyl/pharmacology , Histamine/metabolism , Naloxone/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Cells, Cultured , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/chemistry , Fentanyl/chemistry , HEK293 Cells , Histamine/genetics , Humans , Hydrogen-Ion Concentration , Ligands , Molecular Structure , Mutation , Naloxone/chemistry , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
12.
Curr Opin Behav Sci ; 26: 69-74, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30984806

ABSTRACT

Pain has a useful protective role; through avoidance learning, it helps to decrease the probability of engaging in tissue-damaging, or otherwise dangerous experiences. In our modern society, the experience of acute post-surgical pain and the development of chronic pain states represent an unnecessary negative outcome. This has become an important health issue as more than 30% of the US population reports experiencing "unnecessary" pain at any given time. Opioid therapies are often efficacious treatments for severe and acute pain; however, in addition to their powerful analgesic properties, opioids produce potent reinforcing properties and their inappropriate use has led to the current opioid overdose epidemic in North America. Dissecting the allostatic changes occurring in nociceptors and neuronal pathways in response to pain are the first and most important steps in understanding the physiologic changes underlying the opioid epidemic. Full characterization of these adaptations will provide novel targets for the development of safer pharmacotherapies. In this review, we highlight the current efforts toward safer opioid treatments and describe our current knowledge of the interaction between pain and opioid systems.

13.
Neuron ; 102(3): 564-573.e6, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30878290

ABSTRACT

Negative affective states affect quality of life for patients suffering from pain. These maladaptive emotional states can lead to involuntary opioid overdose and many neuropsychiatric comorbidities. Uncovering the mechanisms responsible for pain-induced negative affect is critical in addressing these comorbid outcomes. The nucleus accumbens (NAc) shell, which integrates the aversive and rewarding valence of stimuli, exhibits plastic adaptations in the presence of pain. In discrete regions of the NAc, activation of the kappa opioid receptor (KOR) decreases the reinforcing properties of rewards and induces aversive behaviors. Using complementary techniques, we report that in vivo recruitment of NAc shell dynorphin neurons, acting through KOR, is necessary and sufficient to drive pain-induced negative affect. Taken together, our results provide evidence that pain-induced adaptations in the kappa opioid system within the NAc shell represent a functional target for therapeutic intervention that could circumvent pain-induced affective disorders. VIDEO ABSTRACT.


Subject(s)
Affect/physiology , Dynorphins/metabolism , Inflammation/metabolism , Mood Disorders/metabolism , Neurons/metabolism , Nucleus Accumbens/metabolism , Pain/metabolism , Receptors, Opioid, kappa/metabolism , Animals , Inflammation/complications , Inflammation/psychology , Mice , Mood Disorders/etiology , Mood Disorders/psychology , Neural Inhibition , Neuronal Plasticity , Nucleus Accumbens/cytology , Pain/complications , Pain/psychology , Rats
14.
Front Neurosci ; 10: 480, 2016.
Article in English | MEDLINE | ID: mdl-27872581

ABSTRACT

Pain and stress are protective mechanisms essential in avoiding harmful or threatening stimuli and ensuring survival. Despite these beneficial roles, chronic exposure to either pain or stress can lead to maladaptive hormonal and neuronal modulations that can result in chronic pain and a wide spectrum of stress-related disorders including anxiety and depression. By inducing allostatic changes in the mesolimbic dopaminergic pathway, both chronic pain and stress disorders affect the rewarding values of both natural reinforcers, such as food or social interaction, and drugs of abuse. Despite opioids representing the best therapeutic strategy in pain conditions, they are often misused as a result of these allostatic changes induced by chronic pain and stress. The kappa opioid receptor (KOR) system is critically involved in these neuronal adaptations in part through its control of dopamine release in the nucleus accumbens. Therefore, it is likely that changes in the kappa opioid system following chronic exposure to pain and stress play a key role in increasing the misuse liability observed in pain patients treated with opioids. In this review, we will discuss how chronic pain and stress-induced pathologies can affect mesolimbic dopaminergic transmission, leading to increased abuse liability. We will also assess how the kappa opioid system may underlie these pathological changes.

15.
Neuropsychopharmacology ; 41(11): 2668-78, 2016 10.
Article in English | MEDLINE | ID: mdl-27170097

ABSTRACT

In people with a prior history of opioid misuse, cues associated with previous drug intake can trigger relapse even after years of abstinence. Examining the processes that lead to the formation and maintenance of the memories between cues/context and the opioid may help to discover new therapeutic candidates to treat drug-seeking behavior. The hippocampus is a brain region essential for learning and memory, which has been involved in the mechanisms underlying opioid cravings. The formation of memories and associations are thought to be dependent on synaptic strengthening associated with structural plasticity of dendritic spines. Here, we assess how dendritic spines in the CA1 region of the hippocampus are affected by morphine-conditioning training. Our results show that morphine pairing with environmental cues (ie, the conditioned place preference (CPP) apparatus) triggers a significant decrease in the number of thin dendritic spines in the hippocampus. Interestingly, this effect was observed regardless of the expression of a conditioned response when mice were trained using an unpaired morphine CPP design and was absent when morphine was administered in the home cage. To investigate the mechanism underlying this structural plasticity, we examined the role of Rho GTPase in dendritic spine remodeling. We found that synaptic expression of RhoA increased with morphine conditioning and blocking RhoA signaling prevented the expression of morphine-induced CPP. Our findings uncover novel mechanisms in response to morphine-associated environmental cues and the underlying alterations in spine plasticity.


Subject(s)
CA1 Region, Hippocampal/cytology , Conditioning, Operant/drug effects , Cues , Morphine/pharmacology , Narcotics/pharmacology , Neuronal Plasticity/drug effects , Pyramidal Cells/drug effects , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Post-Synaptic Density/drug effects , Post-Synaptic Density/metabolism , Post-Synaptic Density/ultrastructure , Pyramidal Cells/ultrastructure , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/ultrastructure , Time Factors , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
16.
Front Mol Neurosci ; 7: 99, 2014.
Article in English | MEDLINE | ID: mdl-25610367

ABSTRACT

Because of its ability to regulate the abundance of selected proteins the ubiquitin proteasome system (UPS) plays an important role in neuronal and synaptic plasticity. As a result various stages of learning and memory depend on UPS activity. Drug addiction, another phenomenon that relies on neuroplasticity, shares molecular substrates with memory processes. However, the necessity of proteasome-dependent protein degradation for the development of addiction has been poorly studied. Here we first review evidences from the literature that drugs of abuse regulate the expression and activity of the UPS system in the brain. We then provide a list of proteins which have been shown to be targeted to the proteasome following drug treatment and could thus be involved in neuronal adaptations underlying behaviors associated with drug use and abuse. Finally we describe the few studies that addressed the need for UPS-dependent protein degradation in animal models of addiction-related behaviors.

17.
Neuropsychopharmacology ; 38(4): 596-604, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23169349

ABSTRACT

Plastic changes in the nucleus accumbens (NAcc), a structure occupying a key position in the neural circuitry related to motivation, are among the critical cellular processes responsible for drug addiction. During the last decade, it has been shown that memory formation and related neuronal plasticity may rely not only on protein synthesis but also on protein degradation by the ubiquitin proteasome system (UPS). In this study, we assess the role of protein degradation in the NAcc in opiate-related behaviors. For this purpose, we coupled behavioral experiments to intra-accumbens injections of lactacystin, an inhibitor of the UPS. We show that protein degradation in the NAcc is mandatory for a full range of animal models of opiate addiction including morphine locomotor sensitization, morphine conditioned place preference, intra-ventral tegmental area morphine self-administration and intra-venous heroin self-administration but not for discrimination learning rewarded by highly palatable food. This study provides the first evidence of a specific role of protein degradation by the UPS in addiction.


Subject(s)
Behavior, Addictive/metabolism , Morphine/administration & dosage , Opioid-Related Disorders/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism , Animals , Behavior, Addictive/psychology , Male , Mice , Mice, Inbred C57BL , Opioid-Related Disorders/psychology , Proteolysis/drug effects , Rats , Rats, Sprague-Dawley , Self Administration
18.
Antioxid Redox Signal ; 15(6): 1477-96, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21395477

ABSTRACT

AIMS: Apelin and its receptor have emerged as promising targets for the treatment of insulin resistance. Indeed, peripheral administration of apelin stimulates glucose utilization and insulin sensitivity via a nitric oxide (NO) pathway. In addition to being expressed on peripheral metabolically active adipose tissues, apelin is also found in the brain. However, no data are available on the role of central effects of apelin on metabolic control. We studied glucose metabolism in response to acute and chronic intracerebroventricular (i.c.v.) injection of apelin performed in normal and obese/diabetic mice. RESULTS: We demonstrate that i.c.v. injection of apelin into fed mice improves glucose control via NO-dependent mechanisms. These results have been strengthened by transgenic (eNOS-KO mice), pharmacological (L-NMMA i.c.v. treated mice), and real-time measurement of NO release with amperometric probes detection. High-fat diet-fed mice displayed a severely blunted response to i.c.v. apelin associated with a lack of NO response by the hypothalamus. Moreover, central administration of high dose apelin in fasted normal mice provoked hyperinsulinemia, hyperglycemia, glucose intolerance, and insulin resistance. CONCLUSION: These data provide compelling evidence that central apelin participates in the regulation of glucose homeostasis and suggest a novel pathophysiological mechanism involved in the transition from normal to diabetic state.


Subject(s)
Glucose/metabolism , Homeostasis , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nitric Oxide/metabolism , Adipokines , Animals , Apelin , Biosynthetic Pathways , Brain/metabolism , Circadian Rhythm , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans , Hypothalamus/cytology , Hypothalamus/drug effects , Infusions, Intraventricular , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/blood , Mice , Neurons/drug effects , Neurons/metabolism
19.
Antioxid Redox Signal ; 14(3): 415-23, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20879900

ABSTRACT

Intestinal detection of nutrients is a crucial step to inform the whole body of the nutritional status. In this paradigm, peripheral information generated by nutrients is transferred to the brain, which in turn controls physiological functions, including glucose metabolism. Here, we investigated the effect of enteric glucose sensors stimulation on hypothalamic nitric oxide (NO) release in lean or in obese/diabetic (db/db) mice. By using specific NO amperometric probes implanted directly in the hypothalamus of mice, we demonstrated that NO release is stimulated in response to enteric glucose sensors activation in lean but not in db/db mice. Alteration of gut to hypothalamic NO signaling in db/db mice is associated with a drastic increase in inflammatory, oxidative/nitric oxide (iNOS, IL-1ß), and endoplasmic reticulum stress (CHOP, ATF4) genes expression in the jejunum. Although we could not exclude the importance of the hypothalamic inflammatory state in obese and diabetic mice, our results provide compelling evidence that enteric glucose sensors could be considered as potential targets for metabolic diseases.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Inflammation/metabolism , Jejunum/pathology , Mice, Obese , Nitric Oxide/metabolism , Animals , Biomarkers/metabolism , Inflammation/pathology , Jejunum/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...