Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 43(10): 1668-1681, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36781220

RESUMO

While the physical signs of opioid withdrawal are most readily observable, withdrawal insidiously drives relapse and contributes to compulsive drug use, by disrupting emotional learning circuits. How these circuits become disrupted during withdrawal is poorly understood. Because amygdala neurons mediate relapse, and are highly opioid sensitive, we hypothesized that opioid withdrawal would induce adaptations in these neurons, opening a window of disrupted emotional learning circuit function. Under normal physiological conditions, synaptic transmission between the basolateral amygdala (BLA) and the neighboring main island (Im) of GABAergic intercalated cells (ITCs) is strongly inhibited by endogenous opioids. Using patch-clamp electrophysiology in brain slices prepared from male rats, we reveal that opioid withdrawal abruptly reduces the ability of these peptides to inhibit neurotransmission, a direct consequence of a protein kinase A (PKA)-driven increase in the synaptic activity of peptidases. Reduced peptide control of neurotransmission in the amygdala shifts the excitatory/inhibitory balance of inputs onto accumbens-projecting amygdala cells involved in relapse. These findings provide novel insights into how peptidases control synaptic activity within the amygdala and presents restoration of endogenous peptide activity during withdrawal as a viable option to mitigate withdrawal-induced disruptions in emotional learning circuits and rescue the relapse behaviors exhibited during opioid withdrawal and beyond into abstinence.SIGNIFICANCE STATEMENT We find that opioid withdrawal dials down inhibitory neuropeptide activity in the amygdala. This disrupts both GABAergic and glutamatergic transmission through amygdala circuits, including reward-related outputs to the nucleus accumbens. This likely disrupts peptide-dependent emotional learning processes in the amygdala during withdrawal and may direct behavior toward compulsive drug use.


Assuntos
Analgésicos Opioides , Síndrome de Abstinência a Substâncias , Ratos , Masculino , Animais , Analgésicos Opioides/farmacologia , Tonsila do Cerebelo/fisiologia , Transmissão Sináptica/fisiologia , Peptídeos/farmacologia , Síndrome de Abstinência a Substâncias/metabolismo , Peptídeo Hidrolases/metabolismo
2.
Neuropharmacology ; 173: 108131, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32422213

RESUMO

The opioid epidemic has led to a serious examination of the use of opioids for the treatment of pain. Opioid drugs are effective due to the expression of opioid receptors throughout the body. These receptors respond to endogenous opioid peptides that are expressed as polypeptide hormones that are processed by proteolytic cleavage. Endogenous opioids are expressed throughout the peripheral and central nervous system and regulate many different neuronal circuits and functions. One of the key functions of endogenous opioid peptides is to modulate our responses to pain. This review will focus on the descending pain modulatory circuit which consists of the ventrolateral periaqueductal gray (PAG) projections to the rostral ventromedial medulla (RVM). RVM projections modulate incoming nociceptive afferents at the level of the spinal cord. Stimulation within either the PAG or RVM results in analgesia and this circuit has been studied in detail in terms of the actions of exogenous opioids, such as morphine and fentanyl. Further emphasis on understanding the complex regulation of endogenous opioids will help to make rational decisions with regard to the use of opioids for pain. We also include a discussion of the actions of endogenous opioids in the amygdala, an upstream brain structure that has reciprocal connections to the PAG that contribute to the brain's response to pain.


Assuntos
Peptídeos Opioides/farmacologia , Dor/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Humanos , Manejo da Dor , Percepção da Dor/fisiologia , Substância Cinzenta Periaquedutal/metabolismo , Receptores Opioides/metabolismo
3.
Br J Pharmacol ; 177(2): 420-431, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31596498

RESUMO

BACKGROUND AND PURPOSE: Pain is a subjective experience involving sensory discriminative and emotionally aversive components. Consistent with its role in pain processing and emotions, the amygdala modulates the aversive component of pain. The laterocapsular region of the central nucleus of the amygdala (CeLC) receives nociceptive information from the parabrachial nucleus (PB) and polymodal, including nociceptive, inputs from the basolateral nucleus of the amygdala (BLA). Opioids are strong analgesics and reduce both the sensory discriminative and the affective component of pain. However, it is unknown whether opioids regulate activity at the two nociceptive inputs to the amygdala. EXPERIMENTAL APPROACH: Using whole-cell electrophysiology, optogenetics, and immunohistochemistry, we investigated whether opioids inhibit the rat PB-CeLC and BLA-CeLC synapses. KEY RESULTS: Opioids inhibited glutamate release at the PB-CeLC and BLA-CeLC synapses. Opioid inhibition is via the µ-receptor at the PB-CeLC synapse, while at the BLA-CeLC synapse, inhibition is via µ-receptors in all neurons and via δ-receptors and κ-receptors in a subset of neurons. CONCLUSIONS AND IMPLICATIONS: Agonists of µ-receptors inhibited two of the synaptic inputs carrying nociceptive information into the laterocapsular amygdala. Therefore, µ-receptor agonists, such as morphine, will inhibit glutamate release from PB and BLA in the CeLC, and this could serve as a mechanism through which opioids reduce the affective component of pain and pain-induced associative learning. The lower than expected regulation of BLA synaptic outputs by δ-receptors does not support the proposal that opioid receptor subtypes segregate into subnuclei of brain regions.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/prevenção & controle , Percepção da Dor/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/fisiopatologia , Animais , Ácido Glutâmico/metabolismo , Masculino , Inibição Neural/efeitos dos fármacos , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Optogenética , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Sinapses/metabolismo
4.
Eur J Neurosci ; 50(3): 2065-2074, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30099803

RESUMO

Neural circuits in the amygdala are important for associating the positive experience of drug taking with the coincident environmental cues. During abstinence, cue re-exposure activates the amygdala, increases dopamine release in the amygdala and stimulates relapse to drug use in an opioid dependent manner. Neural circuits in the amygdala and the learning that underlies these behaviours are inhibited by GABAergic synaptic inhibition. A specialised subtype of GABAergic neurons in the amygdala are the clusters of intercalated cells. We focussed on the main-island of intercalated cells because these neurons, located ventromedial to the basolateral amygdala, express very high levels of dopamine D1-receptor and µ-opioid receptor, release enkephalin and are densely innervated by the ventral tegmental area. However, where these neurons project to was not fully described and their regulation by opioids and dopamine was incomplete. To address this issue we electrically stimulated in the main-island of the intercalated cells in rat brain slices and made patch-clamp recordings of GABAergic synaptics from amygdala neurons. We found that main-island neurons had a strong GABAergic inhibitory output to pyramidal neurons of the basolateral nucleus and the medial central nucleus, the major output zones of the amygdala. Opioids inhibited both these synaptic outputs of the intercalated neurons and thus would disinhibit these target zones. Additionally, dopamine acting at D1-receptors inhibited main-island neuron synapses onto other main-island neurons. This data indicates that the inhibitory projections from the main-island neurons could influence multiple aspects of addiction and emotional processing in an opioid and dopamine dependent manner.


Assuntos
Analgésicos Opioides/farmacologia , Dopamina/farmacologia , Sinapses/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Masculino , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Área Tegmentar Ventral/metabolismo
5.
J Physiol ; 2018 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-30004124

RESUMO

KEY POINTS: Chronic pain is disabling because sufferers form negative associations between pain and activities, such as work, leading to the sufferer limiting these activities. Pain information arriving in the amygdala is responsible for forming these associations and contributes to us feeling bad when we are in pain. Ongoing injuries enhance the delivery of pain information to the amygdala. If we want to understand why chronic pain can continue without ongoing injury, it is important to know whether this facilitation continues once the injury has healed. In the present study, we show that a 2 min noxious heat stimulus, without ongoing injury, is able to enhance delivery of pain information to the amygdala for 3 days. If the noxious heat stimulus is repeated, this enhancement persists even longer. These changes may prime this information pathway so that subsequent injuries may feel even worse and the associative learning that results in pain-related avoidance may be promoted. ABSTRACT: Pain is an important defence against dangers in our environment; however, some clinical conditions produce pain that outlasts this useful role and persists even after the injury has healed. The experience of pain consists of somatosensory elements of intensity and location, negative emotional/aversive feelings and subsequent restrictions on lifestyle as a result of a learned association between certain activities and pain. The amygdala contributes negative emotional value to nociceptive sensory information and forms the association between an aversive response and the environment in which it occurs. It is able to form this association because it receives nociceptive information via the spino-parabrachio-amygdaloid pathway and polymodal sensory information via cortical and thalamic inputs. Synaptic plasticity occurs at the parabrachial-amygdala synapse and other brain regions in chronic pain conditions with ongoing injury; however, very little is known about how plasticity occurs in conditions with no ongoing injury. Using immunohistochemistry, electrophysiology and behavioural assays, we show that a brief nociceptive stimulus with no ongoing injury is able to produce long-lasting synaptic plasticity at the rat parabrachial-amygdala synapse. We show that this plasticity is caused by an increase in postsynaptic AMPA receptors with a transient change in the AMPA receptor subunit, similar to long-term potentiation. Furthermore, this synaptic potentiation primes the synapse so that a subsequent noxious stimulus causes prolonged potentiation of the nociceptive information flow into the amygdala. As a result, a second injury could have an increased negative emotional value and promote associative learning that results in pain-related avoidance.

6.
Behav Neurosci ; 131(6): 454-459, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29083203

RESUMO

The midbrain periaqueductal gray (PAG) coordinates the expression and topography of defensive behaviors to threat and also plays an important role in Pavlovian fear learning itself. Whereas the role of PAG in the expression of defensive behavior is well understood, the relationship between the activity of PAG neurons and fear learning, the exact timing of PAG contributions to learning during the conditioning trial, and the contributions of different PAG columns to fear learning are poorly understood. We assessed the effects of optogenetic inhibition of lateral (LPAG) and ventrolateral PAG (VLPAG) neurons on fear learning. Using adenoassociated viral vectors expressing halorhodopsin, we show that brief optogenetic inhibition of LPAG or VLPAG during delivery of the shock unconditioned stimulus (US) augments acquisition of contextual or cued fear conditioning, and we also show that this inhibition augments postencounter defensive responses to a nonnoxious threat. Taken together, these results show that LPAG and VLPAG serve a key role in the regulation of Pavlovian fear learning at the time of US delivery. These findings provide strong support for existing models that state that LPAG and VLPAG contribute to a fear prediction error signal determining variations in the effectiveness of the aversive US in supporting learning. (PsycINFO Database Record


Assuntos
Comportamento Animal/fisiologia , Medo , Optogenética , Substância Cinzenta Periaquedutal/fisiopatologia , Animais , Condicionamento Clássico/fisiologia , Condicionamento Operante/fisiologia , Medo/fisiologia , Reação de Congelamento Cataléptica/fisiologia , Masculino , Neurônios/fisiologia , Optogenética/métodos , Ratos Sprague-Dawley
7.
Neurobiol Learn Mem ; 144: 186-197, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28716712

RESUMO

The midbrain periaqueductal gray (PAG) has been implicated in the generation and transmission of a prediction error signal that instructs amygdala-based fear and extinction learning. However, the PAG also plays a key role in the expression of conditioned fear responses. The evidence for a role of the PAG in fear learning and extinction learning has been obtained almost exclusively using PAG-dependent fear responses. It is less clear whether the PAG regulates fear learning when other measures of learned fear are used. Here we combined a chemogenetic approach, permitting excitation or inhibition of neurons in the ventrolateral PAG (VLPAG), with conditioned suppression as the measure of learned fear to assess the role of VLPAG in the acquisition and extinction of fear learning. We show that chemogenetic excitation of VLPAG (with some encroachment on lateral PAG [LPAG]) impairs acquisition of fear and, conversely, chemogenetic inhibition impairs extinction of fear. These effects on fear and extinction learning were specific to the combination of DREADD expression and injection of CNO because they were observed relative to both eYFP controls injected with CNO as well as DREADD expressing controls injected with vehicle. Taken together, these results show that activity of L/VLPAG neurons regulates both the acquisition and extinction of Pavlovian fear learning.


Assuntos
Condicionamento Psicológico/fisiologia , Extinção Psicológica/fisiologia , Medo , Neurônios/fisiologia , Substância Cinzenta Periaquedutal/fisiologia , Potenciais de Ação , Animais , Masculino , Inibição Neural , Ratos Sprague-Dawley
8.
Nat Commun ; 8: 14611, 2017 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-28327612

RESUMO

Fear and emotional learning are modulated by endogenous opioids but the cellular basis for this is unknown. The intercalated cells (ITCs) gate amygdala output and thus regulate the fear response. Here we find endogenous opioids are released by synaptic stimulation to act via two distinct mechanisms within the main ITC cluster. Endogenously released opioids inhibit glutamate release through the δ-opioid receptor (DOR), an effect potentiated by a DOR-positive allosteric modulator. Postsynaptically, the opioids activate a potassium conductance through the µ-opioid receptor (MOR), suggesting for the first time that endogenously released opioids directly regulate neuronal excitability. Ultrastructural localization of endogenous ligands support these functional findings. This study demonstrates a new role for endogenously released opioids as neuromodulators engaged by synaptic activity to regulate moment-to-moment neuronal communication and excitability. These distinct actions through MOR and DOR may underlie the opposing effect of these receptor systems on anxiety and fear.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/metabolismo , Medo/fisiologia , Interneurônios/metabolismo , Peptídeos Opioides/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Ácido Glutâmico/metabolismo , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp , Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides/metabolismo , Sinapses/metabolismo
9.
J Neurosci ; 36(2): 385-95, 2016 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-26758831

RESUMO

Basolateral amygdala (BLA) is critical for fear learning, and its heightened activation is widely thought to underpin a variety of anxiety disorders. Here we used chemogenetic techniques in rats to study the consequences of heightened BLA activation for fear learning and memory, and to specifically identify a mechanism linking increased activity of BLA glutamatergic neurons to aberrant fear. We expressed the excitatory hM3Dq DREADD in rat BLA glutamatergic neurons and showed that CNO acted selectively to increase their activity, depolarizing these neurons and increasing their firing rates. This chemogenetic excitation of BLA glutamatergic neurons had no effect on the acquisition of simple fear learning, regardless of whether this learning led to a weak or strong fear memory. However, in an associative blocking task, chemogenetic excitation of BLA glutamatergic neurons yielded significant learning to a blocked conditioned stimulus, which otherwise should not have been learned about. Moreover, in an overexpectation task, chemogenetic manipulation of BLA glutamatergic neurons prevented use of negative prediction error to reduce fear learning, leading to significant impairments in fear inhibition. These effects were not attributable to the chemogenetic manipulation enhancing arousal, increasing asymptotic levels of fear learning or fear memory consolidation. Instead, chemogenetic excitation of BLA glutamatergic neurons disrupted use of prediction error to regulate fear learning. SIGNIFICANCE STATEMENT: Several neuropsychiatric disorders are characterized by heightened activation of the amygdala. This heightened activation has been hypothesized to underlie increased emotional reactivity, fear over generalization, and deficits in fear inhibition. Yet the mechanisms linking heightened amygdala activation to heightened emotional learning are elusive. Here we combined chemogenetic excitation of rat basolateral amygdala glutamatergic neurons with a variety of behavioral approaches to show that, although simple fear learning is unaffected, the use of prediction error to regulate this learning is profoundly disrupted, leading to formation of inappropriate fear associations and impaired fear inhibition.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Condicionamento Psicológico/fisiologia , Medo , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Clozapina/análogos & derivados , Clozapina/farmacologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Condicionamento Psicológico/efeitos dos fármacos , Dependovirus/genética , Eletrochoque/efeitos adversos , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Medo/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Muscarínico M3/genética , Receptores de Droga/genética , Receptores de Droga/metabolismo
10.
Front Pharmacol ; 5: 148, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25009497

RESUMO

Opioids are intensely addictive, and cessation of their chronic use is associated with a highly aversive withdrawal syndrome. A cellular hallmark of withdrawal is an opioid sensitive protein kinase A-dependent increase in GABA transporter-1 (GAT-1) currents in periaqueductal gray (PAG) neurons. Elevated GAT-1 activity directly increases GABAergic neuronal excitability and synaptic GABA release, which will enhance GABAergic inhibition of PAG output neurons. This reduced activity of PAG output neurons to several brain regions, including the hypothalamus and medulla, contributes to many of the PAG-mediated signs of opioid withdrawal. The GABAB receptor agonist baclofen reduces some of the PAG mediated signs of opioid withdrawal. Like the opioid receptors the GABAB receptor is a Gi/Go coupled G-protein coupled receptor. This suggests it could be modulating GAT-1 activity in PAG neurons through its inhibition of the adenylyl cyclase/protein kinase A pathway. Opioid modulation of the GAT-1 activity can be detected by changes in the reversal potential of opioid membrane currents. We found that when opioids are reducing the GAT-1 cation conductance and increasing the GIRK conductance the opioid agonist reversal potential is much more negative than E k . Using this approach for GABAB receptors we show that the GABAB receptor agonist, baclofen, does not couple to inhibition of GAT-1 currents during opioid withdrawal. It is possible this differential signaling of the two Gi/Go coupled G-protein coupled receptors is due to the strong compartmentalization of the GABAB receptor that does not favor signaling to the adenylyl cyclase/protein kinase A/GAT-1 pathway. This highlights the importance of studying the effects of G-protein coupled receptors in native tissue with endogenous G-protein coupled receptors and the full complement of relevant proteins and signaling molecules. This study suggests that baclofen reduces opioid withdrawal symptoms through a non-GAT-1 effector.

11.
J Physiol ; 590(7): 1641-54, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22351628

RESUMO

Many aspects of synaptic transmission are modified during development, reflecting not only the consequence of developmental programmes of gene expression, but also the effects of ongoing neural activity. We investigated the role of synaptic activity in the maturation of Schaffer collateral (SC)-CA1 synapses using sustained low frequency field stimulation of acute brain slices. Between postnatal days 4-6 and 14-16, mouse SC-CA1 synapses in naïve slices showed a developmental decrease in the probability of transmitter release (P(r)) and an increase in the contribution of GluN2A (NR2A) subunits to the NMDA receptor-mediated excitatory postsynaptic current (EPSC). Surprisingly, these developmental changes could be mimicked by short term (4 h) in vitro synaptic activity in slices taken from postnatal days (PND) 4-6 mice. However, different activity levels were required to alter release probability compared to the NMDA receptor subunit composition. Spontaneous synaptic activity was sufficient to alter the NMDA receptor subunit composition, but sustained low-frequency field stimulation of the brain slice (0.1 Hz, 4 h) was necessary to reduce release probability, as assessed 1 h following the cessation of stimulation. The protein synthesis inhibitor anisomycin blocked the effect of field stimulation on release probability. These results indicate that features of mature excitatory synapses can be rapidly induced in immature neurons. The activity dependence of the P(r) and NMDA receptor subunit composition serves as a sensitive indicator of prior neural activity, and provides dual mechanisms for homeostatic control of excitatory synaptic efficacy.


Assuntos
Envelhecimento/fisiologia , Região CA1 Hipocampal/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Sinapses/fisiologia , Animais , Animais Recém-Nascidos , Maleato de Dizocilpina/farmacologia , Estimulação Elétrica , Potenciais Evocados/fisiologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
12.
Nat Neurosci ; 14(12): 1548-54, 2011 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-22037500

RESUMO

Neurotransmitter transporters can affect neuronal excitability indirectly via modulation of neurotransmitter concentrations or directly via transporter currents. A physiological or pathophysiological role for transporter currents has not been described. We found that GABA transporter 1 (GAT-1) cation currents directly increased GABAergic neuronal excitability and synaptic GABA release in the periaqueductal gray (PAG) during opioid withdrawal in rodents. In contrast, GAT-1 did not indirectly alter GABA receptor responses via modulation of extracellular GABA concentrations. Notably, we found that GAT-1-induced increases in GABAergic activity contributed to many PAG-mediated signs of opioid withdrawal. Together, these data support the hypothesis that GAT-1 activity directly produces opioid withdrawal signs through direct hyperexcitation of GABAergic PAG neurons and nerve terminals, which presumably enhances GABAergic inhibition of PAG output neurons. These data provide, to the best of our knowledge, the first evidence that dysregulation of a neurotransmitter transporter current is important for the maladaptive plasticity that underlies opiate withdrawal.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Morfina/efeitos adversos , Substância Cinzenta Periaquedutal/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Ácido gama-Aminobutírico/metabolismo , Análise de Variância , Animais , Modelos Animais de Doenças , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Glutamato Descarboxilase/genética , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microdiálise/métodos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ácidos Nipecóticos/farmacologia , Oximas/farmacologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Síndrome de Abstinência a Substâncias/patologia , Fatores de Tempo
13.
Mol Pharmacol ; 69(4): 1296-303, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16377766

RESUMO

Several forms of macroscopic N-methyl-D-aspartate (NMDA) receptor desensitization affect the amplitude and duration of postsynaptic responses. In addition to its functional significance, desensitization provides one means to examine the conformational coupling of ligand binding to channel gating. Segments flanking the ligand binding domain in the extracellular N terminus of the NMDA receptor NR2 subunit influence the glycine-independent form of desensitization. The NR2A pre-M1 region, the linker between the glutamate binding domain and the channel pore, plays a critical role in desensitization. Thus, we used the substituted-cysteine accessibility method to scan the accessibility of residues in the pre-M1 region and the first transmembrane domain (M1) of NR2A. Cysteine mutants were expressed with NR1 in human embryonic kidney 293 cells and were assayed by whole-cell recording. With activation of the receptor by glutamate and glycine, only a single mutant, V557C, which is located at the beginning of M1, led to irreversible inhibition by the methanethiosulfonate derivative methanethiosulfonate ethyltrimethylammonium (MTSET). The NR2 ligand glutamate was insufficient on its own to induce modification of V557C by MTSET, suggesting that the change in accessibility required channel gating. The rate of MTSET modification of the homologous residue on NR1 (NR1-1a(L562C)/NR2A) was much slower than V557C. We also substituted cysteine in the V557 site of mutant subunits that exhibit either enhanced or reduced desensitization. Modification by MTSET correlated with the degree of desensitization for these subunits, suggesting that V557C is a sensitive detector of desensitization gating.


Assuntos
Cisteína/química , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Linhagem Celular , Humanos , Ativação do Canal Iônico , Ligantes , Ligação Proteica , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo
14.
Br J Pharmacol ; 146(1): 68-76, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15980868

RESUMO

The midbrain periaqueductal gray (PAG) is a major site of opioid analgesic action, and a significant site of cellular adaptations to chronic morphine treatment (CMT). We examined mu-opioid receptor (MOP) regulation of voltage-gated calcium channel currents (I(Ca)) and G-protein-activated K channel currents (GIRK) in PAG neurons from CMT mice. Mice were injected s.c. with 300 mg kg(-1) of morphine base in a slow release emulsion three times over 5 days, or with emulsion alone (vehicles). This protocol produced significant tolerance to the antinociceptive effects of morphine in a test of thermal nociception. Voltage clamp recordings were made of I(Ca) in acutely isolated PAG neurons and GIRK in PAG slices. The MOP agonist DAMGO (Tyr-D-Ala-Gly-N-Me-Phe-Gly-ol enkephalin) inhibited I(Ca) in neurons from CMT mice (230 nM) with a similar potency to vehicle (150 nM), but with a reduced maximal effectiveness (37% inhibition in vehicle neurons, 27% in CMT neurons). Inhibition of I(Ca) by the GABA(B) agonist baclofen was not altered by CMT. Met-enkephalin-activated GIRK currents recorded in PAG slices were significantly smaller in neurons from CMT mice than vehicles, while GIRK currents activated by baclofen were unaltered. These data demonstrate that CMT-induced antinociceptive tolerance is accompanied by homologous reduction in the effectiveness of MOP agonists to inhibit I(Ca) and activate GIRK. Thus, a reduction in MOP number and/or functional coupling to G proteins accompanies the characteristic cellular adaptations to CMT previously described in PAG neurons.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos/fisiologia , Morfina/farmacologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Receptores Opioides mu/agonistas , Animais , Baclofeno/farmacologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/fisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Agonistas GABAérgicos/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Medição da Dor , Substância Cinzenta Periaquedutal/metabolismo , Substância Cinzenta Periaquedutal/fisiologia , Receptores Opioides mu/metabolismo , Receptores Opioides mu/fisiologia
15.
J Neurosci ; 25(12): 3192-8, 2005 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-15788776

RESUMO

Delta-opioid receptor (DOPr) activation fails to produce cellular physiological responses in many brain regions, including the periaqueductal gray (PAG), despite neural expression of high densities of the receptor. Previous histochemical studies have demonstrated that a variety of stimuli, including chronic morphine treatment, induce the translocation of DOPr from intracellular pools to the surface membrane of CNS neurons. PAG neurons in slices taken from untreated mice exhibited mu-opioid receptor (MOPr) but not DOPr-mediated presynaptic inhibition of GABAergic synaptic currents. In contrast, after 5-6 d of chronic morphine treatment, DOPr stimulation inhibited synaptic GABA release onto most neurons. Shorter exposure to morphine in vitro (upto 4 h) or in vivo (18 h) did not induce functional DOPr responses. DOPr-mediated presynaptic inhibition could not be induced in slices from untreated animals by increasing synaptic activity in vitro using high extracellular potassium concentrations or activation of protein kinase A. Induction of functional DOPr signaling by chronic morphine required MOPr expression, because no DOPr receptor responses were observed in MOPr knock-out mice. DOPr agonists also had no effect on miniature IPSCs in beta-arrestin-2 knock-out mice after chronic morphine. These results suggest that induction of DOPr-mediated actions in PAG by chronic morphine requires prolonged MOPr stimulation and expression of beta-arrestin-2.


Assuntos
Morfina/administração & dosagem , Entorpecentes/administração & dosagem , Neurônios/efeitos dos fármacos , Substância Cinzenta Periaquedutal/citologia , Receptores Opioides delta/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Arrestinas/deficiência , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Estimulação Elétrica/métodos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Encefalina Leucina/análogos & derivados , Encefalina Leucina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Glicinérgicos/farmacologia , Técnicas In Vitro , Isoquinolinas/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Potenciais da Membrana/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Técnicas de Patch-Clamp/métodos , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptores Opioides mu/deficiência , Estricnina/farmacologia , Sulfonamidas/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Fatores de Tempo , Xantinas/farmacologia , beta-Arrestina 2 , beta-Arrestinas , Ácido gama-Aminobutírico/metabolismo
16.
Neuron ; 45(3): 433-45, 2005 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-15694329

RESUMO

Adaptations in neurons of the midbrain periaqueductal gray (PAG) induced by chronic morphine treatment mediate expression of many signs of opioid withdrawal. The abnormally elevated action potential rate of opioid-sensitive PAG neurons is a likely cellular mechanism for withdrawal expression. We report here that opioid withdrawal in vitro induced an opioid-sensitive cation current that was mediated by the GABA transporter-1 (GAT-1) and required activation of protein kinase A (PKA) for its expression. Inhibition of GAT-1 or PKA also prevented withdrawal-induced hyperexcitation of PAG neurons. Our findings indicate that GAT-1 currents can directly increase the action potential rates of neurons and that GAT-1 may be a target for therapy to alleviate opioid-withdrawal symptoms.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Neurônios/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/fisiologia , Animais , Canais de Cloreto/efeitos dos fármacos , Canais de Cloreto/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Proteínas da Membrana Plasmática de Transporte de GABA , Masculino , Moduladores de Transporte de Membrana , Proteínas de Membrana Transportadoras/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neurônios/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Técnicas de Cultura de Órgãos , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/fisiopatologia , Ácido gama-Aminobutírico/metabolismo
17.
Br J Pharmacol ; 142(8): 1273-80, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15265812

RESUMO

Functional studies indicate that the midbrain periaqueductal grey (PAG) is involved in the analgesic actions of somatostatin; however, the cellular actions of somatostatin in this brain region are unknown. In the present study, whole-cell patch clamp recordings were made from rat PAG neurons in vitro. In 93% of acutely isolated neurons, somatostatin inhibited Ca(2+)-channel currents. This effect was mimicked by the sst-2 selective agonist BIM-23027, but not by the sst-1 and sst-5 selective agonists CH-275 and L-362855. In brain slices, 81% of neurons responded to somatostatin (300 nm) with an increase in K(+) conductance that reversed polarity at -114 mV. A greater proportion of somatostatin-sensitive neurons (93%) than somatostatin-insensitive neurons (53%) responded to the opioid agonist met-enkephalin (10 microm). Somatostatin also reduced the amplitude of evoked GABA(A)-mediated inhibitory postsynaptic currents (IPSCs). The actions of somatostatin in brain slices were mimicked by BIM-23027, but not by CH-275. Somatostatin had a variable effect on the rate of spontaneous miniature IPSCs in normal external potassium solutions. In high external potassium solutions, somatostatin reduced the rate of miniature IPSCs in all neurons, and this inhibition was abolished by addition of Cd(2+) (30 microm). Somatostatin had no effect on the amplitude of miniature IPSCs. These results indicate that somatostatin acts via sst-2 receptors to directly inhibit a subpopulation of PAG neurons by activating a potassium conductance and inhibits GABA release within PAG via a presynaptic Ca(2+)-dependent mechanism. Thus, like opioids, somatostatin has the potential to exert pre- and postsynaptic disinhibitory effects within the PAG.


Assuntos
Neurônios/citologia , Neurônios/efeitos dos fármacos , Substância Cinzenta Periaquedutal/citologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Somatostatina/farmacologia , Animais , Feminino , Técnicas In Vitro , Masculino , Neurônios/fisiologia , Peptídeos Cíclicos/farmacologia , Substância Cinzenta Periaquedutal/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo
18.
Br J Pharmacol ; 139(2): 362-7, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12770941

RESUMO

1 Patch clamp recordings were made from periaqueductal grey (PAG) neurons in vitro to investigate the cellular actions of opioids in wild-type C57B16/J mice and mutant mice lacking the first exon of the micro -opioid (MOP) receptor. 2 In wild-type mice, the kappa-(KOP) agonist U-69593 (300 nM) and the mixed micro /delta-opioid agonist met-enkephalin (10 micro M), but not the delta-(DOP) agonist deltorphin (300 nM), reduced the amplitude of evoked GABA(A)-mediated inhibitory postsynaptic currents (IPSCs). Met-enkephalin and U-69593 also reduced the rate of spontaneous miniature IPSCs, but had no effect on their amplitude and kinetics. In micro -receptor-deleted mice, only U-69593 (300 nM) reduced the amplitude of evoked IPSCs. 3 In wild-type mice, the MOP agonist DAMGO (3 micro M) produced an outward current in 76% of the neurons. Deltorphin and U-69593 produced outward currents in 24 and 32% of the neurons, respectively. In micro -receptor-deleted mice, deltorphin and U-69593 produced similar outward currents in 32 and 27% of the neurons, respectively, while DAMGO was without effect. All neurons in both the wild-type and micro -receptor-deleted mice responded with similar outward currents to either the GABA(B) receptor agonist baclofen (10 micro M), or the opioid-like receptor ORL1 (NOP) agonist nociceptin (300 nM). 4 The DAMGO-, deltorphin-, U-69593-, baclofen- and nociceptin-induced currents displayed inward rectification and reversed polarity at -109 to -116 mV. 5 These findings indicate that micro -, delta- and kappa-opioid receptor activation has complex pre- and postsynaptic actions within the mouse PAG. This differs to the rat PAG where only micro -opioid receptor actions have been observed.


Assuntos
Neurônios/fisiologia , Substância Cinzenta Periaquedutal/fisiologia , Receptores de GABA-A/fisiologia , Receptores Opioides mu/agonistas , Analgésicos Opioides/farmacologia , Animais , Benzenoacetamidas/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Encefalina Metionina/farmacologia , Feminino , Antagonistas de Receptores de GABA-A , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Oligopeptídeos/farmacologia , Técnicas de Patch-Clamp , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Pirrolidinas/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides delta/fisiologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/genética , Receptores Opioides mu/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA