Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 920
Filter
1.
J Neurosci ; 44(39)2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39187377

ABSTRACT

Dopamine (DA) neurons in the ventral tegmental area (VTA) respond to motivationally relevant cues, and circuit-specific signaling drives different aspects of motivated behavior. Orexin (ox; also known as hypocretin) and dynorphin (dyn) are coexpressed lateral hypothalamic (LH) neuropeptides that project to the VTA. These peptides have opposing effects on the firing activity of VTADA neurons via orexin 1 (Ox1R) or kappa opioid (KOR) receptors. Given that Ox1R activation increases VTADA firing, and KOR decreases firing, it is unclear how the coreleased peptides contribute to the net activity of DA neurons. We tested if optical stimulation of LHox/dyn neuromodulates VTADA neuronal activity via peptide release and if the effects of optically driven LHox/dyn release segregate based on VTADA projection targets including the basolateral amygdala (BLA) or the lateral or medial shell of the nucleus accumbens (lAcbSh, mAchSh). Using a combination of circuit tracing, optogenetics, and patch-clamp electrophysiology in male and female orexincre mice, we showed a diverse response of LHox/dyn optical stimulation on VTADA neuronal firing, which is not mediated by fast transmitter release and is blocked by antagonists to KOR and Ox1R signaling. Additionally, where optical stimulation of LHox/dyn inputs in the VTA inhibited firing of the majority of BLA-projecting VTADA neurons, optical stimulation of LHox/dyn inputs in the VTA bidirectionally affects firing of either lAcbSh- or mAchSh-projecting VTADA neurons. These findings indicate that LHox/dyn corelease may influence the output of the VTA by balancing ensembles of neurons within each population which contribute to different aspects of reward seeking.


Subject(s)
Dopaminergic Neurons , Dynorphins , Orexins , Ventral Tegmental Area , Animals , Orexins/metabolism , Orexins/pharmacology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology , Dynorphins/metabolism , Dynorphins/pharmacology , Mice , Dopaminergic Neurons/physiology , Dopaminergic Neurons/drug effects , Male , Mice, Inbred C57BL , Female , Neural Pathways/physiology , Neural Pathways/drug effects , Hypothalamic Area, Lateral/physiology , Hypothalamic Area, Lateral/drug effects , Mice, Transgenic , Optogenetics , Orexin Receptors/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology
2.
Biol Reprod ; 110(2): 275-287, 2024 Feb 10.
Article in English | MEDLINE | ID: mdl-37930247

ABSTRACT

The timing of puberty onset is reliant on increased gonadotropin-releasing hormone (GnRH). This elicits a corresponding increase in luteinizing hormone (LH) due to a lessening of sensitivity to the inhibitory actions of estradiol (E2). The mechanisms underlying the increase in GnRH release likely involve a subset of neurons within the arcuate (ARC) nucleus of the hypothalamus that contain kisspeptin, neurokinin B (NKB), and dynorphin (KNDy neurons). We aimed to determine if KNDy neurons in female sheep are critical for: timely puberty onset; the LH surge; and the response to an intravenous injection of the neurokinin-3 receptor (NK3R) agonist, senktide. Prepubertal ewes received injections aimed at the ARC containing blank-saporin (control, n = 5) or NK3-saporin (NK3-SAP, n = 6) to ablate neurons expressing NK3R. Blood samples taken 3/week for 65 days following surgery were assessed for progesterone to determine onset of puberty. Control ewes exhibited onset of puberty at 33.2 ± 3.9 days post sampling initiation, whereas 5/6 NK3-SAP treated ewes didn't display an increase in progesterone. After an artificial LH surge protocol, surge amplitude was lower in NK3-SAP ewes. Finally, ewes were treated with senktide to determine if an LH response was elicited. LH pulses were evident in both groups in the absence of injections, but the response to senktide vs saline was similar between groups. These results show that KNDy cells are necessary for timely puberty onset and for full expresson of the LH surge. The occurrence of LH pulses in NK3-SAP treated ewes may indicate a recovery from an apulsatile state.


Subject(s)
Arcuate Nucleus of Hypothalamus , Luteinizing Hormone , Peptide Fragments , Substance P/analogs & derivatives , Female , Animals , Sheep , Luteinizing Hormone/pharmacology , Arcuate Nucleus of Hypothalamus/metabolism , Saporins/pharmacology , Progesterone/pharmacology , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Neurokinin B/metabolism , Dynorphins/pharmacology , Dynorphins/metabolism , Kisspeptins/metabolism
3.
Appetite ; 200: 107504, 2024 09 01.
Article in English | MEDLINE | ID: mdl-38768926

ABSTRACT

The dynorphin peptides are the endogenous ligands for the kappa opioid receptor (KOR) and regulate food intake. Administration of dynorphin-A1-13 (DYN) in the paraventricular hypothalamic nucleus (PVN) increases palatable food intake, and this effect is blocked by co-administration of the orexin-A neuropeptide, which is co-released with DYN in PVN from neurons located in the lateral hypothalamus. While PVN administration of DYN increases palatable food intake, whether it increases food-seeking behaviors has yet to be examined. We tested the effects of DYN and norBNI (a KOR antagonist) on the seeking and consumption of sucrose using a progressive ratio (PR) and demand curve (DC) tasks. In PVN, DYN did not alter the sucrose breaking point (BP) in the PR task nor the elasticity or intensity of demand for sucrose in the DC task. Still, DYN reduced the delay in obtaining sucrose and increased licks during sucrose intake in the PR task, irrespective of the co-administration of orexin-A. In PVN, norBNI increased the delay in obtaining sucrose and reduced licks during sucrose intake in the PR task while increasing elasticity without altering intensity of demand in the DC task. However, subcutaneous norBNI reduced the BP for sucrose and increased the delay in obtaining sucrose in the PR task while reducing the elasticity of demand. Together, these data show different effects of systemic and PVN blockade of KOR on food-seeking, consummatory behaviors, and incentive motivation for sucrose and suggest that KOR activity in PVN is necessary but not sufficient to drive seeking behaviors for palatable food.


Subject(s)
Dynorphins , Motivation , Paraventricular Hypothalamic Nucleus , Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , Dynorphins/pharmacology , Dynorphins/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Male , Motivation/drug effects , Orexins , Rats , Rats, Sprague-Dawley , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Eating/drug effects , Eating/physiology , Eating/psychology , Sucrose , Feeding Behavior/drug effects , Feeding Behavior/psychology , Narcotic Antagonists/pharmacology
4.
Molecules ; 29(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38999061

ABSTRACT

Kappa opioid receptor (KOR) antagonists have potential therapeutic applications in the treatment of stress-induced relapse to substance abuse and mood disorders. The dynorphin A analog arodyn (Ac[Phe1,2,3,Arg4,D-Ala8]dynorphin A-(1-11)-NH2) exhibits potent and selective kappa opioid receptor antagonism. Multiple cyclizations in longer peptides, such as dynorphin and its analogs, can extend the conformational constraint to additional regions of the peptide beyond what is typically constrained by a single cyclization. Here, we report the design, synthesis, and pharmacological evaluation of a bicyclic arodyn analog with two constraints in the opioid peptide sequence. The peptide, designed based on structure-activity relationships of monocyclic arodyn analogs, was synthesized by solid-phase peptide synthesis and cyclized by sequential ring-closing metathesis (RCM) in the C- and N-terminal sequences. Molecular modeling studies suggest similar interactions of key aromatic and basic residues in the bicyclic peptide with KOR as found in the cryoEM structure of KOR-bound dynorphin, despite substantial differences in the backbone conformations of the two peptides. The bicyclic peptide's affinities at KOR and mu opioid receptors (MOR) were determined in radioligand binding assays, and its KOR antagonism was determined in the [35S]GTPγS assay in KOR-expressing cells. The bicyclic analog retains KOR affinity and selectivity (Ki = 26 nM, 97-fold selectivity over MOR) similar to arodyn and exhibits potent KOR antagonism in the dynorphin-stimulated [35S]GTPγS assay. This bicyclic peptide represents a promising advance in preparing cyclic opioid peptide ligands and opens avenues for the rational design of additional bicyclic opioid peptide analogs.


Subject(s)
Dynorphins , Receptors, Opioid, kappa , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/metabolism , Dynorphins/chemistry , Dynorphins/pharmacology , Humans , Animals , Structure-Activity Relationship , Models, Molecular , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemical synthesis , Amino Acid Sequence
5.
J Physiol ; 600(22): 4897-4916, 2022 11.
Article in English | MEDLINE | ID: mdl-36156249

ABSTRACT

Excitatory inputs drive burst firing of locus coeruleus (LC) noradrenaline (NA) neurons in response to a variety of stimuli. Though a small number of glutamatergic LC afferents have been investigated, the overall landscape of these excitatory inputs is largely unknown. The current study used an optogenetic approach to isolate three glutamatergic afferents: the prefrontal cortex (PFC), lateral hypothalamus (LH) and periaqueductal grey (PAG). AAV5-DIO-ChR2 was injected into each region in male and female CaMKII-Cre mice and the properties of excitatory inputs on LC-NA cells were measured. Notably we found differences among these inputs. First, the pattern of axonal innervation differed between inputs such that LH afferents were concentrated in the posterior portion of the LC-NA somatic region while PFC afferents were denser in the medial dendritic region. Second, basal intrinsic properties varied for afferents, with LH inputs having the highest connectivity and the largest amplitude excitatory postsynaptic currents while PAG inputs had the lowest initial release probability. Third, while orexin and oxytocin had minimal effects on any input, dynorphin strongly inhibited excitatory inputs originating from the LH and PAG, and corticotrophin releasing factor (CRF) selectively inhibited inputs from the PAG. Overall, these results demonstrate that individual afferents to the LC have differing properties, which may contribute to the modularity of the LC and its ability to mediate various behavioural outcomes. KEY POINTS: Excitatory inputs to the locus coeruleus (LC) are important for driving noradrenaline neuron activity and downstream behaviours in response to salient stimuli, but little is known about the functional properties of different glutamate inputs that innervate these neurons We used a virus-mediated optogenetic approach to compare glutamate afferents from the prefrontal cortex (PFC), the lateral hypothalamus (LH) and the periaqueductal grey (PAG). While PFC was predicted to make synaptic inputs, we found that the LH and PAG also drove robust excitatory events in LC noradrenaline neurons. The strength, kinetics, and short-term plasticity of each input differed as did the extent of neuromodulation by both dynorphin and corticotrophin releasing factor. Thus each input displayed a unique set of basal properties and modulation by peptides. This characterization is an important step in deciphering the heterogeneity of the LC.


Subject(s)
Dynorphins , Locus Coeruleus , Male , Female , Mice , Animals , Locus Coeruleus/metabolism , Dynorphins/pharmacology , Glutamic Acid/pharmacology , Corticotropin-Releasing Hormone/metabolism , Norepinephrine/pharmacology , Adrenocorticotropic Hormone
6.
BMC Neurosci ; 23(1): 58, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36217122

ABSTRACT

BACKGROUND: Opioids are among the most effective and commonly prescribed analgesics for the treatment of acute pain after spinal cord injury (SCI). However, morphine administration in the early phase of SCI undermines locomotor recovery, increases cell death, and decreases overall health in a rodent contusion model. Based on our previous studies we hypothesize that morphine acts on classic opioid receptors to alter the immune response. Indeed, we found that a single dose of intrathecal morphine increases the expression of activated microglia and macrophages at the injury site. Whether similar effects of morphine would be seen with repeated intravenous administration, more closely simulating clinical treatment, is not known. METHODS: To address this, we used flow cytometry to examine changes in the temporal expression of microglia and macrophages after SCI and intravenous morphine. Next, we explored whether morphine changed the function of these cells through the engagement of cell-signaling pathways linked to neurotoxicity using Western blot analysis. RESULTS: Our flow cytometry studies showed that 3 consecutive days of morphine administration after an SCI significantly increased the number of microglia and macrophages around the lesion. Using Western blot analysis, we also found that repeated administration of morphine increases ß-arrestin, ERK-1 and dynorphin (an endogenous kappa opioid receptor agonist) production by microglia and macrophages. CONCLUSIONS: These results suggest that morphine administered immediately after an SCI changes the innate immune response by increasing the number of immune cells and altering neuropeptide synthesis by these cells.


Subject(s)
Morphine , Spinal Cord Injuries , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Dynorphins/metabolism , Dynorphins/pharmacology , Dynorphins/therapeutic use , Macrophages , Microglia/pathology , Morphine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/therapeutic use , Recovery of Function , Spinal Cord/metabolism , Spinal Cord Injuries/pathology , beta-Arrestins/metabolism , beta-Arrestins/pharmacology , beta-Arrestins/therapeutic use
7.
Stem Cells ; 39(5): 600-616, 2021 05.
Article in English | MEDLINE | ID: mdl-33452745

ABSTRACT

Although the roles of opioid receptors in neurogenesis have been implicated in previous studies, the mechanism by which κ-opioid receptor (OPRK1) regulates adult neurogenesis remains elusive. We now demonstrate that two agonists of OPRK1, U50,488H and dynorphin A, inhibit adult neurogenesis by hindering neuronal differentiation of mouse hippocampal neural stem cells (NSCs), both in vitro and in vivo. This effect was blocked by nor-binaltorphimine (nor-BNI), a specific antagonist of OPRK1. By examining neurogenesis-related genes, we found that OPRK1 agonists were able to downregulate the expression of Pax6, Neurog2, and NeuroD1 in mouse hippocampal NSCs, in a way that Pax6 regulates the transcription of Neurog2 and Neurod1 by directly interacting with their promoters. Moreover, this effect of OPRK1 was accomplished by inducing expression of miR-7a, a miRNA that specifically targeted Pax6 by direct interaction with its 3'-UTR sequence, and thereby decreased the levels of Pax6, Neurog2, and NeuroD1, thus resulted in hindrance of neuronal differentiation of NSCs. Thus, by modulating Pax6/Neurog2/NeuroD1 activities via upregulation of miR-7a expression, OPRK1 agonists hinder the neuronal differentiation of NSCs and hence inhibit adult neurogenesis in mouse hippocampus.


Subject(s)
MicroRNAs/genetics , Neural Stem Cells/cytology , Neurogenesis/genetics , PAX6 Transcription Factor/genetics , Receptors, Opioid, kappa/genetics , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Dynorphins/pharmacology , Gene Expression Regulation, Developmental/drug effects , Hippocampus/drug effects , Humans , Mice , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Nerve Tissue Proteins/genetics , Neurogenesis/drug effects , Receptors, Opioid, kappa/agonists , Signal Transduction/drug effects
8.
Reprod Biol Endocrinol ; 20(1): 91, 2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35729637

ABSTRACT

BACKGROUND: Kisspeptin released from Kiss-1 neurons in the hypothalamus plays an essential role in the control of the hypothalamic-pituitary-gonadal axis by regulating the release of gonadotropin-releasing hormone (GnRH). In this study, we examined how androgen supplementation affects the characteristics of Kiss-1 neurons. METHODS: We used a Kiss-1-expressing mHypoA-55 cell model that originated from the arcuate nucleus (ARC) of the mouse hypothalamus. These cells are KNDy neurons that co-express neurokinin B (NKB) and dynorphin A (DynA). We stimulated these cells with androgens and examined them. We also examined the ARC region of the hypothalamus in ovary-intact female rats after supplementation with androgens. RESULTS: Stimulation of mHypoA-55 cells with 100 nM testosterone significantly increased Kiss-1 gene expression by 3.20 ± 0.44-fold; testosterone also increased kisspeptin protein expression. The expression of Tac3, the gene encoding NKB, was also increased by 2.69 ± 0.64-fold following stimulation of mHypoA-55 cells with 100 nM testosterone. DynA gene expression in these cells was unchanged by testosterone stimulation, but it was significantly reduced at the protein level. Dihydrotestosterone (DHT) had a similar effect to testosterone in mHypoA-55 cells; kisspeptin and NKB protein expression was significantly increased by DHT, whereas it significantly reduced DynA expression. In ovary-intact female rats, DTH administration significantly increased the gene expression of Kiss-1 and Tac3, but not DynA, in the arcuate nucleus. Exogenous NKB and DynA stimulation failed to modulate Kiss-1 gene expression in mHypoA-55 cells. Unlike androgen stimulation, prolactin stimulation did not modulate kisspeptin, NKB, or DynA protein expression in these cells. CONCLUSIONS: Our observations imply that hyperandrogenemia affects KNDy neurons and changes their neuronal characteristics by increasing kisspeptin and NKB levels and decreasing DynA levels. These changes might cause dysfunction of the hypothalamic-pituitary-gonadal axis.


Subject(s)
Dynorphins , Hyperandrogenism , Androgens/metabolism , Animals , Dynorphins/genetics , Dynorphins/metabolism , Dynorphins/pharmacology , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Hyperandrogenism/metabolism , Hypothalamus/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Mice , Neurokinin B/genetics , Neurokinin B/metabolism , Neurokinin B/pharmacology , Neurons/metabolism , Rats , Tachykinins , Testosterone/metabolism , Testosterone/pharmacology
9.
Nutr Neurosci ; 25(5): 1105-1114, 2022 May.
Article in English | MEDLINE | ID: mdl-33151127

ABSTRACT

The orexin peptides promote hedonic intake and other reward behaviors through different brain sites. The opioid dynorphin peptides are co-released with orexin peptides but block their effects on reward in the ventral tegmental area (VTA). We previously showed that in the paraventricular hypothalamic nucleus (PVN), dynorphin and not orexin peptides enhance hedonic intake, suggesting they have brain-site-specific effects. Obesity alters the expression of orexin and dynorphin receptors, but whether their expression across different brain sites is important to hedonic intake is unclear. We hypothesized that hedonic intake is regulated by orexin and dynorphin peptides in PVN and that hedonic intake in obesity correlates with expression of their receptors. Here we show that in mice, injection of DYN-A1-13 (an opioid dynorphin peptide) in the PVN enhanced hedonic intake, whereas in the VTA, injection of OXA (orexin-A, an orexin peptide) enhanced hedonic intake. In PVN, OXA blunted the increase in hedonic intake caused by DYN-A1-13. In PVN, injection of norBNI (opioid receptor antagonist) reduced hedonic intake but a subsequent OXA injection failed to increase hedonic intake, suggesting that OXA activity in PVN is not influenced by endogenous opioid activity. In the PVN, DYN-A1-13 increased the intake of the less-preferred food in a two-food choice task. In obese mice fed a cafeteria diet, orexin 1 receptor mRNA across brain sites involved in hedonic intake correlated with fat preference but not caloric intake. Together, these data support that orexin and dynorphin peptides regulate hedonic intake in an opposing manner with brain-site-specific effects.


Subject(s)
Dynorphins , Paraventricular Hypothalamic Nucleus , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain/metabolism , Dynorphins/metabolism , Dynorphins/pharmacology , Mice , Obesity/metabolism , Orexins/metabolism
10.
Int J Mol Sci ; 22(15)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34360762

ABSTRACT

Peptide therapeutics offer numerous advantages in the treatment of diseases and disorders of the central nervous system (CNS). However, they are not without limitations, especially in terms of their pharmacokinetics where their metabolic lability and low blood-brain barrier penetration hinder their application. Targeted nanoparticle delivery systems are being tapped for their ability to improve the delivery of therapeutics into the brain non-invasively. We have developed a family of mannosylated glycoliposome delivery systems for targeted drug delivery applications. Herein, we demonstrate via in vivo distribution studies the potential of these glycoliposomes to improve the utility of CNS active therapeutics using dynantin, a potent and selective dynorphin peptide analogue antagonist of the kappa opioid receptor (KOR). Glycoliposomal entrapment protected dynantin against known rapid metabolic degradation and ultimately improved brain levels of the peptide by approximately 3-3.5-fold. Moreover, we linked this improved brain delivery with improved KOR antagonist activity by way of an approximately 30-40% positive modulation of striatal dopamine levels 20 min after intranasal administration. Overall, the results clearly highlight the potential of our glycoliposomes as a targeted delivery system for therapeutic agents of the CNS.


Subject(s)
Dynorphins , Peptides , Receptors, Opioid, kappa/antagonists & inhibitors , Corpus Striatum/metabolism , Dopamine , Dynorphins/chemistry , Dynorphins/pharmacokinetics , Dynorphins/pharmacology , Humans , Liposomes , Peptides/chemistry , Peptides/pharmacokinetics , Peptides/pharmacology
11.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33671048

ABSTRACT

Previous studies have shown that genetically selected Marchigian Sardinian alcohol-preferring (msP) rats consume excessive amounts of ethanol to self-medicate from negative moods and to relieve innate hypersensitivity to stress. This phenotype resembling a subset of alcohol use disorder (AUD) patients, appears to be linked to a dysregulation of the equilibrium between stress and antistress mechanisms in the extended amygdala. Here, comparing water and alcohol exposed msP and Wistar rats we evaluate the transcript expression of the anti-stress opioid-like peptide nociceptin/orphanin FQ (N/OFQ) and its receptor NOP as well as of dynorphin (DYN) and its cognate κ-opioid receptor (KOP). In addition, we measured the transcript levels of corticotropin-releasing factor (CRF), CRF receptor 1 (CRF1R), brain-derived neurotrophic factor (BDNF) and of the tropomyosin receptor kinase B receptor (Trk-B). Results showed an innately up-regulation of the CRFergic system, mediating negative mood and stress responses, as well as an inherent up-regulation of the anti-stress N/OFQ system, both in the amygdala (AMY) and bed nucleus of the stria terminalis (BNST) of msP rats. The up-regulation of this latter system may reflect an attempt to buffer the negative condition elicited by the hyperactivity of pro-stress mechanisms since results showed that voluntary alcohol consumption dampened N/OFQ. Alcohol exposure also reduced the expression of dynorphin and CRF transmissions in the AMY of msP rats. In the BNST, alcohol intake led to a more complex reorganization of these systems increasing receptor transcripts in msP rats, along with an increase of CRF and a decrease of N/OFQ transcripts, respectively. Moreover, mimicking the effects of alcohol in the AMY we observed that the activation of NOP receptor by intracerebroventricular administration of N/OFQ in msP rats caused an increase of BDNF and a decrease of CRF transcripts. Our study indicates that both stress and anti-stress mechanisms are dysregulated in the extended AMY of msP rats. The voluntary alcohol drinking, as well as NOP agonism, have a significant impact on neuropeptidergic systems arrangement, bringing the systems back to normalization.


Subject(s)
Alcohol Drinking/physiopathology , Alcoholism/pathology , Amygdala/pathology , Dynorphins/pharmacology , Ethanol/toxicity , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Receptors, Opioid/metabolism , Alcoholism/etiology , Amygdala/drug effects , Amygdala/metabolism , Animals , Behavior, Animal , Male , Neurotransmitter Agents/pharmacology , Rats , Rats, Wistar , Receptors, Opioid/genetics
12.
Mol Pharmacol ; 98(4): 462-474, 2020 10.
Article in English | MEDLINE | ID: mdl-32958572

ABSTRACT

Opioid receptors (ORs) convert extracellular messages to signaling events by coupling to the heterotrimeric G proteins, Gα•ßγ Classic pharmacological methods, such as [35S]GTPγS binding and inhibition of cyclic AMP production, allow for general opioid characterization, but they are subject to the varying endogenous Gα proteins in a given cell type. Bioluminescence resonance energy transfer (BRET) technology offers new insight by allowing the direct observation of Gα subunit-specific effects on opioid pharmacology. Using a Venus-tagged Gßγ and nanoluciferase-tagged truncated G protein receptor kinase 3, an increase in BRET signal correlated with OR activation mediated by a specific Gα protein. The magnitude of the BRET signal was normalized to the maximum response obtained with 10 µM 2-(3,4-dichlorophenyl)-N-methyl-N-[(1R,2R)-2-pyrrolidin-1-ylcyclohexyl]acetamide (U50,488) for the kappa OR (KOR). Opioids reached equilibrium with the KOR, and concentration-response curves were generated. Although the full agonists U50,488, salvinorin A, nalfurafine, and dynorphin peptides were equally efficacious regardless of the Gα subunit present, the concentration-response curves were leftward shifted when the KOR was signaling through Gαz compared with other Gαi/o subunits. In contrast, the Gα subunit distinctly affected both the efficacy and potency of partial kappa agonists, such as the benzomorphans, and the classic mu opioid antagonists, naloxone, naltrexone, and nalmefene. For example, (-)pentazocine had EC50 values of 7.3 and 110 nM and maximal stimulation values of 79% and 35% when the KOR signaled through Gαz and Gαi1, respectively. Together, these observations suggest KOR pharmacology varies based on the specific Gα subunit coupled to the KOR. SIGNIFICANCE STATEMENT: Opioid receptors couple to various heterotrimeric Gαßγ proteins to convert extracellular cues to precise intracellular events. This paper focuses on how the various inhibitory Gα subunits influence the pharmacology of full and partial agonists at the kappa opioid receptor. Using a bioluminescent assay, the efficacy and potency of kappa opioids was determined. Opioid signaling was more potent through Gαz compared with other Gα proteins. These observations suggest that Gαz may impact opioid pharmacology and cellular physiology more than previously thought.


Subject(s)
Analgesics, Opioid/pharmacology , Bioluminescence Resonance Energy Transfer Techniques/methods , GTP-Binding Protein alpha Subunits/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Diterpenes, Clerodane/pharmacology , Dynorphins/pharmacology , HEK293 Cells , Humans , Morphinans/pharmacology , Signal Transduction/drug effects , Spiro Compounds/pharmacology
13.
J Fish Biol ; 96(3): 747-754, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32003470

ABSTRACT

The opioid peptides are involved in the regulation of neuroendocrine functions in vertebrates. Nonetheless, the influence of an opioid peptide, dynorphin A (DYN), on reproduction in fish is understudied. The aim of this work was to study the influence of DYN on the pituitary-ovary axis in Oreochromis mossambicus. Daily injections (ip) of 250 µg DYN kg-1 body weight for 22 days during the ovarian cycle caused a reduction in the intensity and the per cent area of luteinizing hormone (LH) immunoreactive content in the proximal pars distalis region of the pituitary gland compared with an intense immunostaining in time-matched controls. In the ovary, DYN treatment caused a decrease in the number of stage I (previtellogenic) follicles compared with time-matched controls. No difference was observed in the number of stage IV (vitellogenic) follicles among different experimental groups, whereas the numbers of stage II and stage III follicles (previtellogenic) were higher in DYN-treated fish than in time-matched controls. Nonetheless, there was a reduction in the number of stage V (preovulatory) follicles in DYN-treated fish compared with time-matched controls. Taken together, these results indicate that DYN exerts an inhibitory effect on follicular recruitment at the late vitellogenic stage, through the suppression of LH secretion in fish.


Subject(s)
Dynorphins/pharmacology , Ovary/drug effects , Pituitary Gland/drug effects , Tilapia/physiology , Animals , Female , Luteinizing Hormone/metabolism , Reproduction/drug effects , Vitellogenesis/drug effects
14.
J Neurosci ; 38(5): 1061-1072, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29114074

ABSTRACT

Estradiol feedback regulates gonadotropin-releasing hormone (GnRH) neurons and subsequent luteinizing hormone (LH) release. Estradiol acts via estrogen receptor α (ERα)-expressing afferents of GnRH neurons, including kisspeptin neurons in the anteroventral periventricular (AVPV) and arcuate nuclei, providing homeostatic feedback on episodic GnRH/LH release as well as positive feedback to control ovulation. Ionotropic glutamate receptors are important for estradiol feedback, but it is not known where they fit in the circuitry. Estradiol-negative feedback decreased glutamatergic transmission to AVPV and increased it to arcuate kisspeptin neurons; positive feedback had the opposite effect. Deletion of ERα in kisspeptin cells decreased glutamate transmission to AVPV neurons and markedly increased it to arcuate kisspeptin neurons, which also exhibited increased spontaneous firing rate. KERKO mice had increased LH pulse frequency, indicating loss of negative feedback. These observations indicate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and neuroendocrine output by estradiol.SIGNIFICANCE STATEMENT The brain regulates fertility through gonadotropin-releasing hormone (GnRH) neurons. Ovarian estradiol regulates the pattern of GnRH (negative feedback) and initiates a surge of release that triggers ovulation (positive feedback). GnRH neurons do not express the estrogen receptor needed for feedback (estrogen receptor α [ERα]); kisspeptin neurons in the arcuate and anteroventral periventricular nuclei are postulated to mediate negative and positive feedback, respectively. Here we extend the network through which feedback is mediated by demonstrating that glutamatergic transmission to these kisspeptin populations is differentially regulated during the reproductive cycle and by estradiol. Electrophysiological and in vivo hormone profile experiments on kisspeptin-specific ERα knock-out mice demonstrate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and for neuroendocrine output.


Subject(s)
Estradiol/pharmacology , Glutamates/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Kisspeptins/physiology , Neurons/physiology , Receptors, Estrogen/drug effects , Synaptic Transmission/physiology , Animals , Arcuate Nucleus of Hypothalamus/physiology , Dynorphins/pharmacology , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Hypothalamus/drug effects , Luteinizing Hormone/physiology , Mice , Midline Thalamic Nuclei/physiology , Neurons/drug effects , Pituitary Gland/drug effects , Pituitary Gland/physiology , Proestrus/physiology , Receptors, Ionotropic Glutamate/drug effects , Receptors, Ionotropic Glutamate/physiology , Synaptic Transmission/drug effects , ERRalpha Estrogen-Related Receptor
15.
J Neurosci ; 38(6): 1588-1599, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29311142

ABSTRACT

Orexin (also known as hypocretin) neurons are considered a key component of the ascending arousal system. They are active during wakefulness, at which time they drive and maintain arousal, and are silent during sleep. Their activity is controlled by long-range inputs from many sources, as well as by more short-range inputs, including from presumptive GABAergic neurons in the lateral hypothalamus/perifornical region (LH/PF). To characterize local GABAergic input to orexin neurons, we used channelrhodopsin-2-assisted circuit mapping in brain slices. We expressed channelrhodopsin-2 in GABAergic neurons (Vgat+) in the LH/PF and recorded from genetically identified surrounding orexin neurons (LH/PFVgat → Orx). We performed all experiments in mice of either sex. Photostimulation of LH/PF GABAergic neurons inhibited the firing of orexin neurons through the release of GABA, evoking GABAA-mediated IPSCs in orexin neurons. These photo-evoked IPSCs were maintained in the presence of TTX, indicating direct connectivity. Carbachol inhibited LH/PFVgat → Orx input through muscarinic receptors. By contrast, application of orexin was without effect on LH/PFVgat → Orx input, whereas dynorphin, another peptide produced by orexin neurons, inhibited LH/PFVgat → Orx input through κ-opioid receptors. Our results demonstrate that orexin neurons are under inhibitory control by local GABAergic neurons and that this input is depressed by cholinergic signaling, unaffected by orexin and inhibited by dynorphin. We propose that local release of dynorphin may, via collaterals, provides a positive feedback to orexin neurons and that, during wakefulness, orexin neurons may be disinhibited by acetylcholine and by their own release of dynorphin.SIGNIFICANCE STATEMENT The lateral hypothalamus contains important wake-promoting cell populations, including orexin-producing neurons. Intermingled with the orexin neurons, there are other cell populations that selectively discharge during nonrapid eye movement or rapid eye movement sleep. Some of these sleep-active neurons release GABA and are thought to inhibit wake-active neurons during rapid eye movement and nonrapid eye movement sleep. However, this hypothesis had not been tested. Here we show that orexin neurons are inhibited by a local GABAergic input. We propose that this local GABAergic input inhibits orexin neurons during sleep but that, during wakefulness, this input is depressed, possibly through cholinergically mediated disinhibition and/or by release of dynorphin from orexin neurons themselves.


Subject(s)
GABAergic Neurons/physiology , Hypothalamic Area, Lateral/metabolism , Hypothalamic Area, Lateral/physiology , Orexins/physiology , Animals , Brain Mapping , Carbachol/pharmacology , Channelrhodopsins/physiology , Dynorphins/pharmacology , Excitatory Postsynaptic Potentials/physiology , Female , GABAergic Neurons/drug effects , Hypothalamic Area, Lateral/drug effects , Male , Mice , Muscarinic Agonists/pharmacology , Orexins/pharmacology , Parasympathetic Nervous System/physiology , Photic Stimulation , Prefrontal Cortex/metabolism , Receptors, GABA-A/metabolism , Tetrodotoxin/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins/physiology
16.
J Physiol ; 597(6): 1605-1625, 2019 03.
Article in English | MEDLINE | ID: mdl-30618146

ABSTRACT

KEY POINTS: Excitatory glutamate neurons are sparse in the rostral hypothalamic arcuate nucleus (ARC), the subregion that has received the most attention in the past. In striking contrast, excitatory neurons are far more common (by a factor of 10) in the caudal ARC, an area which has received relatively little attention. These glutamate cells may play a negative role in energy balance and food intake. They can show an increase in phosphorylated Stat-3 in the presence of leptin, are electrically excited by the anorectic neuromodulator cholecystokinin, and inhibited by orexigenic neuromodulators neuropeptide Y, met-enkephalin, dynorphin and the catecholamine dopamine. The neurons project local axonal connections that excite other ARC neurons including proopiomelanocortin neurons that can play an important role in obesity. These data are consistent with models suggesting that the ARC glutamatergic neurons may play both a rapid and a slower role in acting as anorectic neurons in CNS control of food intake and energy homeostasis. ABSTRACT: Here we interrogate a unique class of excitatory neurons in the hypothalamic arcuate nucleus (ARC) that utilizes glutamate as a fast neurotransmitter using mice expressing GFP under control of the vesicular glutamate transporter 2 (vGluT2) promoter. These neurons show a unique distribution, synaptic characterization, cellular physiology and response to neuropeptides involved in energy homeostasis. Although apparently not previously appreciated, the caudal ARC showed a far greater density of vGluT2 cells than the rostral ARC, as seen in transgenic vGluT2-GFP mice and mRNA analysis. After food deprivation, leptin induced an increase in phosphorylated Stat-3 in vGluT2-positive neurons, indicating a response to hormonal cues of energy state. Based on whole-cell recording electrophysiology in brain slices, vGluT2 neurons were spontaneously active with a spike frequency around 2 Hz. vGluT2 cells were responsive to a number of neuropeptides related to energy homeostasis; they were excited by the anorectic peptide cholecystokinin, but inhibited by orexigenic neuropeptide Y, dynorphin and met-enkephalin, consistent with an anorexic role in energy homeostasis. Dopamine, associated with the hedonic aspect of enhancing food intake, inhibited vGluT2 neurons. Optogenetic excitation of vGluT2 cells evoked EPSCs in neighbouring neurons, indicating local synaptic excitation of other ARC neurons. Microdrop excitation of ARC glutamate cells in brain slices rapidly increased excitatory synaptic activity in anorexigenic proopiomelanocortin neurons. Together these data support the perspective that vGluT2 cells may be more prevalent in the ARC than previously appreciated, and play predominantly an anorectic role in energy metabolism.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Eating , Energy Metabolism , Excitatory Postsynaptic Potentials , Neurons/metabolism , Action Potentials , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Cholecystokinin/pharmacology , Dopamine/pharmacology , Dynorphins/pharmacology , Enkephalin, Methionine/pharmacology , Glutamic Acid/metabolism , Homeostasis , Leptin/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Neuropeptide Y/pharmacology , Pro-Opiomelanocortin/metabolism , STAT3 Transcription Factor/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
17.
Am J Physiol Regul Integr Comp Physiol ; 314(5): R716-R723, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29341829

ABSTRACT

Amphibian neuromuscular junctions (NMJs) become relatively more silent during the dry winter season in Australia. During the dry, calcium sensitivity is reduced, whereas calcium dependence remains unchanged. Endogenous opioid peptides play an important role in the regulation of the physiological functions of active and dormant vertebrates. Previous findings suggest that dynorphin-A is more potent than other opiates in decreasing evoked neurotransmission in amphibian NMJs. Dynorphin-A has been shown not to alter the amplitude or the frequency of miniature quantal neurotransmitter release. In the present study, we report that dynorphin-A exerted a more pronounced inhibitory effect on evoked neurotransmitter release during the dry (hibernating period) when compared with the wet (active period) season. Dynorphin-A increased the frequency and decreased the amplitude of miniature neurotransmitter release only at relatively high concentration during the dry season. In the present study, we propose that dynorphin-A suppresses evoked neurotransmitter release and thus contraction of skeletal muscles, while allowing subthreshold activation of the NMJ by miniature neurotransmission, thus preventing any significant neuromuscular remodeling. The inhibitory effect of dynorphin-A on evoked transmitter release is reduced by increasing the extracellular calcium concentration.


Subject(s)
Amphibian Proteins/metabolism , Bufo marinus/metabolism , Climate , Dynorphins/metabolism , Hibernation , Muscle Contraction , Muscle, Skeletal/innervation , Neuromuscular Junction/metabolism , Synaptic Transmission , Amphibian Proteins/pharmacology , Animals , Calcium/metabolism , Calcium Signaling , Dynorphins/pharmacology , Electric Stimulation , Miniature Postsynaptic Potentials , Muscle Contraction/drug effects , Neuromuscular Junction/drug effects , Synaptic Transmission/drug effects
18.
Biol Pharm Bull ; 41(6): 957-960, 2018.
Article in English | MEDLINE | ID: mdl-29863085

ABSTRACT

Olfactory bulbectomized (OBX) mice are characterized by impaired performance in the passive avoidance test and decreased number of cholinergic neurons in the hippocampus. Several studies have reported that κ-opioid receptor agonists improve cognitive function in mice. However, their influence on OBX-induced cognitive dysfunction remains unclear. To address this question, we evaluated the effects of the endogenous κ-opioid receptor agonist dynorphin A (Dyn A) and the selective agonist trans-(-)-U-50488 on the behavior of OBX mice in the passive avoidance test. The cognitive dysfunction of OBX mice was significantly recovered by the intracerebroventricular administration of Dyn A or trans-(-)-U-50488. The effects of these two agonists were counteracted by the selective κ-opioid receptor antagonist nor-binaltorphimine or the inhibitor of acetylcholine release ß-bungarotoxin. These findings suggest that κ-opioid receptor agonists produce anti-dementia effects through activation of cholinergic neurons in OBX mice.


Subject(s)
Cognitive Dysfunction/drug therapy , Memory Disorders/drug therapy , Receptors, Opioid, kappa/agonists , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/therapeutic use , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Cognitive Dysfunction/physiopathology , Dynorphins/pharmacology , Dynorphins/therapeutic use , Hippocampus/physiology , Male , Memory Disorders/physiopathology , Mice , Olfactory Bulb/surgery , Receptors, Opioid, kappa/physiology
19.
Biochim Biophys Acta ; 1863(11): 2719-2738, 2016 11.
Article in English | MEDLINE | ID: mdl-27523794

ABSTRACT

Together with its endogenous ligands (dynorphin), the kappa opioid receptor (KOR) plays an important role in modulating various physiological and pharmacological responses, with a classical G protein-coupled pathway mediating analgesia and non-G protein-dependent pathway, especially the ß-arrestin-dependent pathway, eliciting side effects of dysphoria, aversion, drug-seeking in addicts, or even relapse to addiction. Although mounting evidence has verified a functional overlap between dynorphin/KOR and neurotensin/neurotensin receptor 1 (NTSR1) systems, little is known about direct interaction between the two receptors. Here, we showed that KOR and NTSR1 form a heterodimer that functions as a novel pharmacological entity, and this heterodimer, in turn, brings about a switch in KOR-mediated signal transduction, from G protein-dependent to ß-arrestin-2-dependent. This was simultaneously verified by analyzing a KOR mutant (196th residue) that lost the ability to dimerize with NTSR1. We also found that dual occupancy of the heterodimer forced the ß-arrestin-2-dependent pathway back into Gi protein-dependent signaling, according to KOR activation. These data provide new insights into the interaction between KOR and NTSR1, and the newly discovered role of NTSR1 acting as a switch between G protein- and ß-arrestin-dependent pathways of KOR also suggests a new approach for utilizing pathologically elevated dynorphin/KOR system into full play for its analgesic effect with limited side effects.


Subject(s)
Basal Ganglia/metabolism , Neurons/metabolism , Receptors, Neurotensin/metabolism , Receptors, Opioid, kappa/metabolism , Signal Transduction , beta-Arrestin 2/metabolism , Animals , Animals, Newborn , Basal Ganglia/cytology , Basal Ganglia/drug effects , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Dynorphins/pharmacology , Female , HEK293 Cells , Humans , Kinetics , Male , Mutation , Neurons/drug effects , Peptide Fragments/pharmacology , Primary Cell Culture , Protein Binding , RNA Interference , Rats, Sprague-Dawley , Receptors, Neurotensin/genetics , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/genetics , Signal Transduction/drug effects , Transfection , beta-Arrestin 2/genetics
20.
J Am Chem Soc ; 139(17): 6152-6159, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28350441

ABSTRACT

Host-microbe communication via small molecule signals is important for both symbiotic and pathogenic relationships, but is often poorly understood at the molecular level. Under conditions of host stress, levels of the human opioid peptide dynorphin are elevated, triggering virulence in the opportunistic pathogenic bacterium Pseudomonas aeruginosa via an unknown pathway. Here we apply a multilayered chemical biology strategy to unravel the mode of action of this putative interkingdom signal. We designed and applied dynorphin-inspired photoaffinity probes to reveal the protein targets of the peptide in live bacteria via chemical proteomics. ParS, a largely uncharacterized membrane sensor of a two-component system, was identified as the most promising hit. Subsequent full proteome studies revealed that dynorphin(1-13) induces an antimicrobial peptide-like response in Pseudomonas, with specific upregulation of membrane defense mechanisms. No such response was observed in a parS mutant, which was more susceptible to dynorphin-induced toxicity. Thus, P. aeruginosa exploits the ParS sensing machinery to defend itself against the host in response to dynorphin as a signal. This study highlights interkingdom communication as a potential essential strategy not only for induction of P. aeruginosa virulence but also for maintaining viability in the hostile environment of the host.


Subject(s)
Anti-Bacterial Agents/chemistry , Dynorphins/chemistry , Molecular Probes/chemistry , Protein Kinases/chemistry , Pseudomonas aeruginosa/enzymology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Binding Sites , Dose-Response Relationship, Drug , Dynorphins/metabolism , Dynorphins/pharmacology , Humans , Molecular Structure , Protein Kinases/metabolism , Proteomics , Pseudomonas aeruginosa/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL