Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 229
Filter
1.
Sleep ; 47(5)2024 May 10.
Article in English | MEDLINE | ID: mdl-38447008

ABSTRACT

Dynorphin is an endogenous opiate localized in many brain regions and spinal cord, but the activity of dynorphin neurons during sleep is unknown. Dynorphin is an inhibitory neuropeptide that is coreleased with orexin, an excitatory neuropeptide. We used microendoscopy to test the hypothesis that, like orexin, the dynorphin neurons are wake-active. Dynorphin-cre mice (n = 3) were administered rAAV8-Ef1a-Con/Foff 2.0-GCaMP6M into the zona incerta-perifornical area, implanted with a GRIN lens (gradient reflective index), and electrodes to the skull that recorded sleep. One month later, a miniscope imaged calcium fluorescence in dynorphin neurons during multiple bouts of wake, non-rapid-eye movement (NREM), and rapid-eye movement (REM) sleep. Unbiased data analysis identified changes in calcium fluorescence in 64 dynorphin neurons. Most of the dynorphin neurons (72%) had the highest fluorescence during bouts of active and quiet waking compared to NREM or REM sleep; a subset (20%) were REM-max. Our results are consistent with the emerging evidence that the activity of orexin neurons can be classified as wake-max or REM-max. Since the two neuropeptides are coexpressed and coreleased, we suggest that dynorphin-cre-driven calcium sensors could increase understanding of the role of this endogenous opiate in pain and sleep.


Subject(s)
Dynorphins , Neurons , Sleep, REM , Wakefulness , Zona Incerta , Animals , Mice , Dynorphins/metabolism , Dynorphins/physiology , Neurons/physiology , Orexins/metabolism , Orexins/physiology , Sleep, REM/physiology , Wakefulness/physiology , Zona Incerta/physiology , Zona Incerta/physiopathology
2.
J Neuroendocrinol ; 33(3): e12945, 2021 03.
Article in English | MEDLINE | ID: mdl-33713519

ABSTRACT

Neurones in the arcuate nucleus co-expressing kisspeptin, neurokinin B (NKB) and dynorphin (KNDy) play a critical role in the control of gonadotrophin-releasing hormone (GnRH) and luteinising hormone (LH) secretion. In sheep, KNDy neurones mediate both steroid-negative- and -positive-feedback during pulsatile and preovulatory surge secretions of GnRH/LH, respectively. In addition, KNDy neurones receive glutamatergic inputs expressing vGlut2, a glutamate transporter that serves as a marker for those terminals, from both KNDy neurones and other populations of glutamatergic neurones. Previous work reported higher numbers of vGlut2-positive axonal inputs onto KNDy neurones during the LH surge than in luteal phase ewes. In the present study, we further examined the effects of the ovarian steroids progesterone (P) and oestradiol (E2 ) on glutamatergic inputs to KNDy neurones. Ovariectomised (OVX) ewes received either no further treatment (OVX) or steroid treatments that mimicked the luteal phase (low E2  + P), and early (low E2 ) or late follicular (high E2 ) phases of the oestrous cycle (n = 4 or 5 per group). Brain sections were processed for triple-label immunofluorescent detection of NKB/vGlut2/synaptophysin and analysed using confocal microscopy. We found higher numbers of vGlut2 inputs onto KNDy neurones in high E2 compared to the other three treatment groups. These results suggest that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase depend on increasing levels of E2 required for the preovulatory GnRH/surge. These synaptic changes likely contribute to the positive-feedback action of oestrogen on GnRH/LH secretion and thus the generation of the preovulatory surge in the sheep.


Subject(s)
Dynorphins/physiology , Estradiol/physiology , Follicular Phase/physiology , Glutamates/physiology , Kisspeptins/physiology , Neurokinin B/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Estradiol/metabolism , Female , Gonadotropin-Releasing Hormone/blood , Luteal Phase/drug effects , Luteinizing Hormone/blood , Ovariectomy , Sheep , Vesicular Glutamate Transport Protein 2/metabolism
3.
Nat Rev Endocrinol ; 16(8): 407-420, 2020 08.
Article in English | MEDLINE | ID: mdl-32427949

ABSTRACT

Hypothalamic kisspeptin neurons serve as the nodal regulatory centre of reproductive function. These neurons are subjected to a plethora of regulatory factors that ultimately affect the release of kisspeptin, which modulates gonadotropin-releasing hormone (GnRH) release from GnRH neurons to control the reproductive axis. The presence of sufficient energy reserves is critical to achieve successful reproduction. Consequently, metabolic factors impose a very tight control over kisspeptin synthesis and release. This Review offers a synoptic overview of the different steps in which kisspeptin neurons are subjected to metabolic regulation, from early developmental stages to adulthood. We cover an ample array of known mechanisms that underlie the metabolic regulation of KISS1 expression and kisspeptin release. Furthermore, the novel role of kisspeptin neurons as active players within the neuronal circuits that govern energy balance is discussed, offering evidence of a bidirectional role of these neurons as a nexus between metabolism and reproduction.


Subject(s)
Energy Metabolism/physiology , Kisspeptins/physiology , Reproduction/physiology , Animals , Dynorphins/physiology , Female , Gonadotropin-Releasing Hormone/physiology , Homeostasis , Humans , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Kisspeptins/genetics , Luteinizing Hormone/physiology , Neurokinin B/physiology , Neurons/physiology , Ovary/physiology , Puberty/physiology
4.
J Neurosci ; 39(49): 9738-9747, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31645462

ABSTRACT

Fertility critically depends on the gonadotropin-releasing hormone (GnRH) pulse generator, a neural construct comprised of hypothalamic neurons coexpressing kisspeptin, neurokoinin-B and dynorphin. Here, using mathematical modeling and in vivo optogenetics we reveal for the first time how this neural construct initiates and sustains the appropriate ultradian frequency essential for reproduction. Prompted by mathematical modeling, we show experimentally using female estrous mice that robust pulsatile release of luteinizing hormone, a proxy for GnRH, emerges abruptly as we increase the basal activity of the neuronal network using continuous low-frequency optogenetic stimulation. Further increase in basal activity markedly increases pulse frequency and eventually leads to pulse termination. Additional model predictions that pulsatile dynamics emerge from nonlinear positive and negative feedback interactions mediated through neurokinin-B and dynorphin signaling respectively are confirmed neuropharmacologically. Our results shed light on the long-elusive GnRH pulse generator offering new horizons for reproductive health and wellbeing.SIGNIFICANCE STATEMENT The gonadotropin-releasing hormone (GnRH) pulse generator controls the pulsatile secretion of the gonadotropic hormones LH and FSH and is critical for fertility. The hypothalamic arcuate kisspeptin neurons are thought to represent the GnRH pulse generator, since their oscillatory activity is coincident with LH pulses in the blood; a proxy for GnRH pulses. However, the mechanisms underlying GnRH pulse generation remain elusive. We developed a mathematical model of the kisspeptin neuronal network and confirmed its predictions experimentally, showing how LH secretion is frequency-modulated as we increase the basal activity of the arcuate kisspeptin neurons in vivo using continuous optogenetic stimulation. Our model provides a quantitative framework for understanding the reproductive neuroendocrine system and opens new horizons for fertility regulation.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Animals , Dynorphins/physiology , Estrous Cycle/physiology , Feedback, Physiological , Female , Kisspeptins/physiology , Luteinizing Hormone/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Theoretical , Neurokinin B/physiology , Neurons/physiology , Optogenetics , Pregnancy , Reproduction/physiology , Ultradian Rhythm/physiology
5.
Alcohol Res ; 40(1)2019 10 17.
Article in English | MEDLINE | ID: mdl-31649835

ABSTRACT

In this review, the effects of stress on alcohol drinking are discussed. The interactions between biological stress systems and alcohol drinking are examined, with a focus on the hypothalamic pituitary adrenal axis, corticotropin releasing factor, dynorphin, neuropeptide Y, and norepinephrine systems. Findings from animal models suggest that these biological stress systems may be useful targets for medications development for alcohol use disorder and co-occurring stress-related disorders in humans.


Subject(s)
Alcohol Drinking/epidemiology , Stress, Psychological/epidemiology , Alcohol Drinking/physiopathology , Animals , Comorbidity , Corticotropin-Releasing Hormone/physiology , Dynorphins/physiology , Humans , Hypothalamo-Hypophyseal System/physiopathology , Neuropeptide Y/physiology , Norepinephrine/physiology , Pituitary-Adrenal System/physiopathology , Stress, Psychological/physiopathology
6.
Vitam Horm ; 111: 17-47, 2019.
Article in English | MEDLINE | ID: mdl-31421700

ABSTRACT

Dynorphin is a neuropeptide involved in pain, addiction and mood regulation. It exerts its activity by binding to the kappa opioid receptor (KOP) which belongs to the large family of G protein-coupled receptors. The dynorphin peptide was discovered in 1975, while its receptor was cloned in 1993. This review will describe: (a) the activities and physiological functions of dynorphin and its receptor, (b) early structure-activity relationship studies performed before cloning of the receptor (mostly pharmacological and biophysical studies of peptide analogues), (c) structure-activity relationship studies performed after cloning of the receptor via receptor mutagenesis and the development of recombinant receptor expression systems, (d) structural biology of the opiate receptors culminating in X-ray structures of the four opioid receptors in their inactive state and structures of MOP and KOP receptors in their active state. X-ray and EM structures are combined with NMR data, which gives complementary insight into receptor and peptide dynamics. Molecular modeling greatly benefited from the availability of atomic resolution 3D structures of receptor-ligand complexes and an example of the strategy used to model a dynorphin-KOP receptor complex using NMR data will be described. These achievements have led to a better understanding of the complex dynamics of KOP receptor activation and to the development of new ligands and drugs.


Subject(s)
Dynorphins/chemistry , Dynorphins/physiology , Receptors, Opioid/chemistry , Receptors, Opioid/physiology , Amino Acid Sequence , Animals , Cloning, Molecular , Dynorphins/genetics , Humans , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Pain , Protein Binding , Receptors, Opioid/genetics , Structure-Activity Relationship , Substance-Related Disorders
7.
Vitam Horm ; 111: 227-246, 2019.
Article in English | MEDLINE | ID: mdl-31421702

ABSTRACT

The organization of estrogenic signaling in the CNS is exceedingly complex. It is comprised of peripherally and centrally synthesized estrogens, and a plethora of types of estrogen receptor that can localize to both the nucleus and the plasma membrane. Moreover, CNS estrogen receptors can exist independent of aromatase (aka estrogen synthase) as well as oligomerize with it, along with a host of other membrane signaling proteins. This ability of CNS estrogen receptors to either to physically pair or exist separately enables locally produced estrogens to act on multiple spatial levels, with a high degree of gradated regulation and plasticity, signaling either in-phase or out-of phase with circulating estrogens. This complexity explains the numerous contradictory findings regarding sex-dependent pain processing and sexually dimorphic opioid antinociception. This review highlights the increasing awareness that estrogens are major endogenous arbiters of both opioid analgesic actions and the mechanisms used to achieve them. This behooves us to understand, and possibly intercede at, the points of intersection of estrogenic signaling and opioid functionality. Factors that integrate estrogenic actions at subcellular, synaptic, and CNS regional levels are likely to be prime drug targets for novel pharmacotherapies designed to modulate CNS estrogen-dependent opioid functionalities and possibly circumvent the current opioid epidemic.


Subject(s)
Analgesics, Opioid/pharmacology , Estrogens/physiology , Reproduction/physiology , Sex Characteristics , Analgesia , Animals , Aromatase , Brain/physiology , Dynorphins/physiology , Female , Humans , Male , Neurosecretory Systems/physiology , Nociception/drug effects , Nociception/physiology , Receptors, Estrogen/physiology , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/physiology , Signal Transduction/physiology
8.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-31297186

ABSTRACT

This review recounts the origins and development of the concept of the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator. It starts in the late 1960s when striking rhythmic episodes of luteinizing hormone secretion, as reflected by circulating concentrations of this gonadotropin, were first observed in monkeys and ends in the present day. It is currently an exciting time witnessing the application, primarily to the mouse, of contemporary neurobiological approaches to delineate the mechanisms whereby Kiss1/NKB/Dyn (KNDy) neurons in the arcuate nucleus of the hypothalamus generate and time the pulsatile output of kisspeptin from their terminals in the median eminence that in turn dictates intermittent GnRH release and entry of this decapeptide into the primary plexus of the hypophysial portal circulation. The review concludes with an examination of questions that remain to be addressed.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Gonadotropin-Releasing Hormone/physiology , Kisspeptins/physiology , Animals , Dynorphins/physiology , Mice , Neurokinin B/physiology , Neurons/physiology
9.
Neurochem Int ; 129: 104504, 2019 10.
Article in English | MEDLINE | ID: mdl-31301327

ABSTRACT

Neural circuits that enable an organism to protect itself by promoting escape from immediate threat and avoidance of future injury are conceptualized to carry an "aversive" signal. One of the key molecular elements of these circuits is the kappa opioid receptor (KOR) and its endogenous peptide agonist, dynorphin. In many cases, the aversive response to an experimental manipulation can be eliminated by selective blockade of KOR function, indicating its necessity in transmitting this signal. The dopamine system, through its contributions to reinforcement learning, is also involved in processing of aversive stimuli, and KOR control of dopamine in the context of aversive behavioral states has been intensely studied. In this review, we have discussed the multiple ways in which the KORs regulate dopamine dynamics with a central focus on dopamine neurons and projections from the ventral tegmental area. At the neuronal level, KOR agonists inhibit dopamine neurons both in the somatodendritic region as well as at terminal release sites, through various signaling pathways and ion channels, and these effects are specific to different synaptic sites. While the dominant hypotheses are that aversive states are driven by decreases in dopamine and increases in dynorphin, reported exceptions to these patterns indicate these ideas require refinement. This is critical given that KOR is being considered as a target for development of new therapeutics for anxiety, depression, pain, and other psychiatric disorders.


Subject(s)
Avoidance Learning/physiology , Dopamine/physiology , Dopaminergic Neurons/physiology , Receptors, Opioid, kappa/physiology , Amygdala/metabolism , Animals , Avoidance Learning/drug effects , Dynorphins/physiology , Forecasting , Learning/physiology , Nucleus Accumbens/metabolism , Punishment , Reinforcement, Psychology , Signal Transduction/physiology , Ventral Tegmental Area/metabolism
10.
Brain Res ; 1713: 91-101, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30244022

ABSTRACT

Negative affective states are prevalent symptoms in a plethora of neuropsychiatric disorders, including depression and drug addiction. Dysfunction of mesocorticolimbic dopamine systems has been implicated in negative affective states in neuropsychiatric disorders. The dynorphin/kappa-opioid receptor system is a powerful effector of stress-related behavior and is highly enriched within the mesocorticolimbic dopamine system. Dysfunction of dynorphin/KOR signaling within the mesocorticolimbic dopamine system is implicated in promoting symptoms in neuropsychiatric disorders. As such, the kappa-opioid receptor system provides an important therapeutic target to treat negative affective states associated with psychiatric disorders. In this review, we provide a comprehensive overview of the dynorphin/kappa-opioid receptor system and its role in regulating the mesocorticolimbic dopamine system, motivation, and emotional behavior. Furthermore, we highlight unresolved issues in the field and offer some insights for future research.


Subject(s)
Affect/physiology , Dopamine/metabolism , Dynorphins/metabolism , Receptors, Opioid, kappa/metabolism , Affect/drug effects , Animals , Dopamine/physiology , Dynorphins/physiology , Emotions , Humans , Mental Disorders/metabolism , Mental Disorders/physiopathology , Psychotic Disorders/metabolism , Psychotic Disorders/physiopathology , Receptors, Opioid, kappa/physiology , Signal Transduction
11.
Reproduction ; 156(3): R83-R99, 2018 09.
Article in English | MEDLINE | ID: mdl-29880718

ABSTRACT

Early work in ewes provided a wealth of information on the physiological regulation of pulsatile gonadotropin-releasing hormone (GnRH) secretion by internal and external inputs. Identification of the neural systems involved, however, was limited by the lack of information on neural mechanisms underlying generation of GnRH pulses. Over the last decade, considerable evidence supported the hypothesis that a group of neurons in the arcuate nucleus that contain kisspeptin, neurokinin B and dynorphin (KNDy neurons) are responsible for synchronizing secretion of GnRH during each pulse in ewes. In this review, we describe our current understanding of the neural systems mediating the actions of ovarian steroids and three external inputs on GnRH pulsatility in light of the hypothesis that KNDy neurons play a key role in GnRH pulse generation. In breeding season adults, estradiol (E2) and progesterone decrease GnRH pulse amplitude and frequency, respectively, by actions on KNDy neurons, with E2 decreasing kisspeptin and progesterone increasing dynorphin release onto GnRH neurons. In pre-pubertal lambs, E2 inhibits GnRH pulse frequency by decreasing kisspeptin and increasing dynorphin release, actions that wane as the lamb matures to allow increased pulsatile GnRH secretion at puberty. Less is known about mediators of undernutrition and stress, although some evidence implicates kisspeptin and dynorphin, respectively, in the inhibition of GnRH pulse frequency by these factors. During the anoestrus, inhibitory photoperiod acting via melatonin activates A15 dopaminergic neurons that innervate KNDy neurons; E2 increases dopamine release from these neurons to inhibit KNDy neurons and suppress the frequency of kisspeptin and GnRH release.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Homeostasis/physiology , Sheep/physiology , Animals , Arcuate Nucleus of Hypothalamus/physiology , Breeding , Dynorphins/physiology , Estradiol/pharmacology , Estrous Cycle , Feedback, Physiological , Female , Kisspeptins/physiology , Luteinizing Hormone/metabolism , Neurokinin B/physiology , Neurons/physiology , Periodicity , Progesterone/pharmacology , Seasons , Sexual Maturation/physiology
12.
Gynecol Endocrinol ; 34(11): 913-919, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29902942

ABSTRACT

Women during perimenopausal period experience a range of symptoms, which interfere with physical, sexual, and social life. About 65-75% of symptoms connected with postmenopausal period are vasomotor symptoms (VMS), such as hot flushes and night sweats. Hot flushes are subjective sensation of heat associated with cutaneous vasodilatation and drop in core temperature. It is suspected that VMS are strongly correlated with pulsatile oversecretion of gonadotropin-releasing hormone (GnRH) and subsequently luteinizing hormone (LH). Evidence has accumulated in parallel showing that lack of negative feedback of steroid hormones synthesized in ovary causes overactivation of hypertrophied kisspeptin/neurokinin B/dynorphin (KNDy) neurons, located in infundibular nucleus. Oversecretion of both kisspeptin (KISS1) and neurokinin B (NKB), as well as downregulation of dynorphin, plays dominant role in creation of GnRH pulses. This in turn causes VMS. Administration of senktide, highly potent and selective NK3R agonist, resulted in increase of serum LH concentration, induction of VMS, increase in heart rate, and skin temperature in postmenopausal women. These finding suggest that modulation of KNDy neurons may become new therapeutic approach in the treatment of VMS.


Subject(s)
Hot Flashes/etiology , Hypothalamus/physiology , Neurons/physiology , Postmenopause/physiology , Vasomotor System/physiology , Dynorphins/physiology , Feedback, Physiological , Female , Hot Flashes/drug therapy , Humans , Kisspeptins/physiology , Neurokinin B/physiology
13.
Handb Exp Pharmacol ; 248: 473-503, 2018.
Article in English | MEDLINE | ID: mdl-29526023

ABSTRACT

Understanding the neural systems that drive alcohol motivation and are disrupted in alcohol use disorders is of critical importance in developing novel treatments. The dynorphin and orexin/hypocretin neuropeptide systems are particularly relevant with respect to alcohol use and misuse. Both systems are strongly associated with alcohol-seeking behaviors, particularly in cases of high levels of alcohol use as seen in dependence. Furthermore, both systems also play a role in stress and anxiety, indicating that disruption of these systems may underlie long-term homeostatic dysregulation seen in alcohol use disorders. These systems are also closely interrelated with one another - dynorphin/kappa opioid receptors and orexin/hypocretin receptors are found in similar regions and hypocretin/orexin neurons also express dynorphin - suggesting that these two systems may work together in the regulation of alcohol seeking and may be mutually disrupted in alcohol use disorders. This chapter reviews studies demonstrating a role for each of these systems in motivated behavior, with a focus on their roles in regulating alcohol-seeking and self-administration behaviors. Consideration is also given to evidence indicating that these neuropeptide systems may be viable targets for the development of potential treatments for alcohol use disorders.


Subject(s)
Alcoholism , Dynorphins/physiology , Ethanol/pharmacology , Motivation , Orexins/physiology , Humans
14.
J Neuroendocrinol ; 29(6)2017 06.
Article in English | MEDLINE | ID: mdl-28475285

ABSTRACT

Pulsatile secretion of gonadotrophin-releasing hormone (GnRH)/luteinising hormone is indispensable for the onset of puberty and reproductive activities at adulthood in mammalian species. A cohort of neurones expressing three neuropeptides, namely kisspeptin, encoded by the Kiss1 gene, neurokinin B (NKB) and dynorphin A, localised in the hypothalamic arcuate nucleus (ARC), so-called KNDy neurones, comprises a putative intrinsic source of the GnRH pulse generator. Synchronous activity among KNDy neurones is considered to be required for pulsatile GnRH secretion. It has been reported that gap junctions play a key role in synchronising electrical activity in the central nervous system. Thus, we hypothesised that gap junctions are involved in the synchronised activities of KNDy neurones, which is induced by NKB-NK3R signalling. We determined the role of NKB-NK3R signalling in Ca2+ oscillation (an indicator of neuronal activities) of KNDy neurones and its synchronisation mechanism among KNDy neurones. Senktide, a selective agonist for NK3R, increased the frequency of Ca2+ oscillations in cultured Kiss1-GFP cells collected from the mediobasal hypothalamus of the foetal Kiss1-green fluorescent protein (GFP) mice. The senktide-induced Ca2+ oscillations were synchronised in the Kiss1-GFP and neighbouring glial cells. Confocal microscopy analysis of these cells, which have shown synchronised Ca2+ oscillations, revealed close contacts between Kiss1-GFP cells, as well as between Kiss1-GFP cells and glial cells. Dye coupling experiments suggest cell-to-cell communication through gap junctions between Kiss1-GFP cells and neighbouring glial cells. Connexin-26 and -37 mRNA were found in isolated ARC Kiss1 cells taken from adult female Kiss1-GFP transgenic mice. Furthermore, 18ß-glycyrrhetinic acids and mefloquine, which are gap junction inhibitors, attenuated senktide-induced Ca2+ oscillations in Kiss1-GFP cells. Taken together, these results suggest that NKB-NK3R signalling enhances synchronised activities among neighbouring KNDy neurones, and that both neurone-neurone and neurone-glia communications via gap junctions possibly contribute to synchronised activities among KNDy neurones.


Subject(s)
Gap Junctions/physiology , Neuroglia/physiology , Neurons/physiology , Peptide Fragments/pharmacology , Substance P/analogs & derivatives , Animals , Cells, Cultured , Connexins/metabolism , Dynorphins/physiology , Gap Junctions/drug effects , Gap Junctions/metabolism , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Kisspeptins/genetics , Medulla Oblongata/metabolism , Mefloquine/pharmacology , Mice, Transgenic , Neuroglia/metabolism , Neurokinin B/physiology , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/antagonists & inhibitors , Substance P/antagonists & inhibitors , Substance P/pharmacology
15.
J Physiol ; 595(16): 5637-5652, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28449351

ABSTRACT

KEY POINTS: Both endogenous opioids and opiate drugs of abuse modulate learning of habitual and goal-directed actions, and can also modify long-term plasticity of corticostriatal synapses. Striatal projection neurons of the direct pathway co-release the opioid neuropeptide dynorphin which can inhibit dopamine release via κ-opioid receptors. Theta-burst stimulation of corticostriatal fibres produces long-term potentiation (LTP) in striatal projection neurons when measured using whole-cell patch recording. Optogenetic activation of direct pathway striatal projection neurons inhibits LTP while reducing dopamine release. Because the endogenous release of opioids is activity dependent, this modulation of synaptic plasticity represents a negative feedback mechanism that may limit runaway enhancement of striatal neuron activity in response to drugs of abuse. ABSTRACT: Synaptic plasticity in the striatum adjusts behaviour adaptively during skill learning, or maladaptively in the case of addiction. Just as dopamine plays a critical role in synaptic plasticity underlying normal skill learning and addiction, endogenous and exogenous opiates also modulate learning and addiction-related striatal plasticity. Though the role of opioid receptors in long-term depression in striatum has been characterized, their effect on long-term potentiation (LTP) remains unknown. In particular, direct pathway (dopamine D1 receptor-containing; D1R-) spiny projection neurons (SPNs) co-release the opioid neuropeptide dynorphin, which acts at presynaptic κ-opioid receptors (KORs) on dopaminergic afferents and can negatively regulate dopamine release. Therefore, we evaluated the interaction of co-released dynorphin and KOR on striatal LTP. We optogenetically facilitate the release of endogenous dynorphin from D1R-SPNs in brain slice while using whole-cell patch recording to measure changes in the synaptic response of SPNs following theta-burst stimulation (TBS) of cortical afferents. Our results demonstrate that TBS evokes corticostriatal LTP, and that optogenetic activation of D1R-SPNs during induction impairs LTP. Additional experiments demonstrate that optogenetic activation of D1R-SPNs reduces stimulation-evoked dopamine release and that bath application of a KOR antagonist provides full rescue of both LTP induction and dopamine release during optogenetic activation of D1R-SPNs. These results suggest that an increase in the opioid neuropeptide dynorphin is responsible for reduced TBS LTP and illustrate a physiological phenomenon whereby heightened D1R-SPN activity can regulate corticostriatal plasticity. Our findings have important implications for learning in addictive states marked by elevated direct pathway activation.


Subject(s)
Dynorphins/physiology , Neuronal Plasticity/physiology , Receptors, Opioid, kappa/physiology , Animals , Corpus Striatum/physiology , Dopamine/physiology , Female , Learning , Light , Long-Term Potentiation , Male , Mice, Transgenic , Neurons/physiology , Receptors, Dopamine D1/physiology , Synapses/physiology
16.
Neuron ; 93(1): 147-163, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-28056342

ABSTRACT

Endogenous dynorphin signaling via the kappa-opioid receptor (KOR) in the nucleus accumbens (NAcc) powerfully mediates negative affective states and stress reactivity. Excitatory inputs from the hippocampus and amygdala play a fundamental role in shaping the activity of both NAcc D1 and D2 MSNs, which encode positive and negative motivational valences, respectively. However, a circuit-based mechanism by which KOR modulation of excitation-inhibition balance modifies D1 and D2 MSN activity is lacking. Here, we provide a comprehensive synaptic framework wherein presynaptic KOR inhibition decreases the excitatory drive of D1 MSN activity by the amygdala, but not the hippocampus. Conversely, presynaptic inhibition by KORs of inhibitory synapses on D2 MSNs enhances integration of excitatory drive by the amygdala and hippocampus. In conclusion, we describe a circuit-based mechanism showing differential gating of afferent control of D1 and D2 MSN activity by KORs in a pathway-specific manner.


Subject(s)
Affect/physiology , Amygdala/metabolism , Dynorphins/metabolism , Hippocampus/metabolism , Neural Inhibition/physiology , Neurons/metabolism , Nucleus Accumbens/metabolism , Receptors, Opioid, kappa/metabolism , Amygdala/physiology , Animals , Dynorphins/physiology , Female , Gene Knockdown Techniques , Hippocampus/physiology , Male , Mice , Motivation , Neurons/physiology , Nucleus Accumbens/physiology , Patch-Clamp Techniques , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/physiology
17.
Neurobiol Aging ; 50: 30-38, 2017 02.
Article in English | MEDLINE | ID: mdl-27842268

ABSTRACT

Pulsatile secretion of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) decreases during aging. Kisspeptin (encoded by Kiss1) neurons in the arcuate nucleus coexpress neurokinin B (Tac3) and dynorphin (Pdyn) and are critical for regulating the GnRH/LH pulse. We therefore examined kisspeptin neurons by histochemistry and pulsatile LH release in rats aged 2-3 (Young), 12-13 (Young-Middle), 19-22 (Late-Middle), and 24-26 (Old) months. Total LH concentrations, sampled for 3 hours, decreased in both sexes with aging. In females, numbers of Tac3 and Pdyn neurons were significantly reduced in all aging rats, and numbers of Kiss1 neurons were significantly reduced in Late-Middle and Old rats. In males, numbers of all 3 neuron-types were significantly decreased in all aging rats. GnRH agonist induced LH release in all animals; however, the increased LH concentration in all aging rats was less than that in Young rats. These results suggest that expression of each gene in kisspeptin neurons may be controlled individually during aging, and that reduction of their expression or change in pituitary responsiveness may cause attenuated pulsatile LH secretion.


Subject(s)
Aging/pathology , Aging/physiology , Dynorphins/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Neurons/pathology , Animals , Dynorphins/physiology , Female , Histocytochemistry , Hypothalamus/cytology , Hypothalamus/pathology , Kisspeptins/physiology , Male , Menopause/metabolism , Menopause/physiology , Neurokinin B/physiology , Neurons/physiology , Pulsatile Flow , Rats, Wistar
18.
Depress Anxiety ; 33(10): 895-906, 2016 10.
Article in English | MEDLINE | ID: mdl-27699938

ABSTRACT

Kappa-opioid receptor (KOR) antagonists are currently being considered for the treatment of a variety of neuropsychiatric conditions, including depressive, anxiety, and substance abuse disorders. A general ability to mitigate the effects of stress, which can trigger or exacerbate these conditions, may explain their putative efficacy across such a broad array of conditions. The discovery of their potentially therapeutic effects evolved from preclinical research designed to characterize the molecular mechanisms by which experience causes neuroadaptations in the nucleus accumbens (NAc), a key element of brain reward circuitry. This research established that exposure to drugs of abuse or stress increases the activity of the transcription factor CREB (cAMP response element binding protein) in the NAc, which leads to elevated expression of the opioid peptide dynorphin that in turn causes core signs of depressive- and anxiety-related disorders. Disruption of KORs-the endogenous receptors for dynorphin-produces antidepressant- and anxiolytic-like actions in screening procedures that identify standard drugs of these classes, and reduces stress effects in tests used to study addiction and stress-related disorders. Although interest in this target is high, prototypical KOR antagonists have extraordinarily persistent pharmacodynamic effects that complicate clinical trials. The development of shorter acting KOR antagonists together with more rapid designs for clinical trials may soon provide insight on whether these drugs are efficacious as would be predicted by preclinical work. If successful, KOR antagonists would represent a unique example in psychiatry where the therapeutic mechanism of a drug class is understood before it is shown to be efficacious in humans.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Antidepressive Agents/therapeutic use , Anxiety Disorders/drug therapy , Dynorphins/physiology , Dynorphins/therapeutic use , Narcotic Antagonists/therapeutic use , Nucleus Accumbens/drug effects , Receptors, Opioid, kappa/antagonists & inhibitors , Animals , Anxiety Disorders/physiopathology , Brain/drug effects , Brain/physiopathology , CREB-Binding Protein/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Nucleus Accumbens/physiopathology , Receptors, Opioid, kappa/physiology , Reward , Stress, Psychological/complications , Stress, Psychological/physiopathology , Substance-Related Disorders/complications , Substance-Related Disorders/physiopathology , Translational Research, Biomedical
19.
Hum Mol Genet ; 25(13): 2728-2737, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27260403

ABSTRACT

Spinocerebellar ataxia type 23 (SCA23) is caused by missense mutations in prodynorphin, encoding the precursor protein for the opioid neuropeptides α-neoendorphin, Dynorphin (Dyn) A and Dyn B, leading to neurotoxic elevated mutant Dyn A levels. Dyn A acts on opioid receptors to reduce pain in the spinal cord, but its cerebellar function remains largely unknown. Increased concentration of or prolonged exposure to Dyn A is neurotoxic and these deleterious effects are very likely caused by an N-methyl-d-aspartate-mediated non-opioid mechanism as Dyn A peptides were shown to bind NMDA receptors and potentiate their glutamate-evoked currents. In the present study, we investigated the cellular mechanisms underlying SCA23-mutant Dyn A neurotoxicity. We show that SCA23 mutations in the Dyn A-coding region disrupted peptide secondary structure leading to a loss of the N-terminal α-helix associated with decreased κ-opioid receptor affinity. Additionally, the altered secondary structure led to increased peptide stability of R6W and R9C Dyn A, as these peptides showed marked degradation resistance, which coincided with decreased peptide solubility. Notably, L5S Dyn A displayed increased degradation and no aggregation. R6W and wt Dyn A peptides were most toxic to primary cerebellar neurons. For R6W Dyn A, this is likely because of a switch from opioid to NMDA- receptor signalling, while for wt Dyn A, this switch was not observed. We propose that the pathology of SCA23 results from converging mechanisms of loss of opioid-mediated neuroprotection and NMDA-mediated excitotoxicity.


Subject(s)
Dynorphins/metabolism , Spinocerebellar Degenerations/metabolism , Amino Acid Sequence , Animals , Cell Culture Techniques , Computer Simulation , Dynorphins/physiology , Endorphins/metabolism , Enkephalins/genetics , Enkephalins/metabolism , Mice , Mice, Inbred C57BL , N-Methylaspartate/metabolism , Neurons/metabolism , Neurotoxins , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Structure, Secondary , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Spinal Cord/metabolism , Spinocerebellar Degenerations/genetics
20.
Curr Neuropharmacol ; 14(2): 165-76, 2016.
Article in English | MEDLINE | ID: mdl-26903446

ABSTRACT

Opioids are traditionally associated with pain, analgesia and drug abuse. It is now clear, however, that the opioids are central players in mood. The implications for mood disorders, particularly clinical depression, suggest a paradigm shift from the monoamine neurotransmitters to the opioids either alone or in interaction with monoamine neurons. We have a special interest in dynorphin, the last of the major endogenous opioids to be isolated and identified. Dynorphin is derived from the Greek word for power, dynamis, which hints at the expectation that the neuropeptide held for its discoverers. Yet, dynorphin and its opioid receptor subtype, kappa, has always taken a backseat to the endogenous b-endorphin and the exogenous morphine that both bind the mu opioid receptor subtype. That may be changing as the dynorphin/ kappa system has been shown to have different, often opposite, neurophysiological and behavioral influences. This includes major depressive disorder (MDD). Here, we have undertaken a review of dynorphin/ kappa neurobiology as related to behaviors, especially MDD. Highlights include the unique features of dynorphin and kappa receptors and the special relation of a plant-based agonist of the kappa receptor salvinorin A. In addition to acting as a kappa opioid agonist, we conclude that salvinorin A has a complex pharmacologic profile, with potential additional mechanisms of action. Its unique neurophysiological effects make Salvinorina A an ideal candidate for MDD treatment research.


Subject(s)
Brain/physiopathology , Depressive Disorder, Major/physiopathology , Diterpenes, Clerodane/physiology , Dynorphins/physiology , Receptors, Opioid, kappa/physiology , Anhedonia/drug effects , Animals , Brain/drug effects , Depressive Disorder, Major/drug therapy , Diterpenes, Clerodane/therapeutic use , Humans , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...