Author Response: Morphine Disinhibits Glutamatergic Input to VTA Dopamine Neurons and Promotes Dopamine Neuron Excitation
Ming Chen,Ying Zhao,Hualan Yang,Wenjie Luan,Jiaojiao Song,Dongyang Cui,Yi Dong,Bin Lai,Lan Ma,Ping Zheng
DOI: https://doi.org/10.7554/elife.09275.016
2015-01-01
Abstract:Article Figures and data Abstract eLife digest Introduction Results Discussion Materials and methods References Decision letter Author response Article and author information Metrics Abstract One reported mechanism for morphine activation of dopamine (DA) neurons of the ventral tegmental area (VTA) is the disinhibition model of VTA-DA neurons. Morphine inhibits GABA inhibitory neurons, which shifts the balance between inhibitory and excitatory input to VTA-DA neurons in favor of excitation and then leads to VTA-DA neuron excitation. However, it is not known whether morphine has an additional strengthening effect on excitatory input. Our results suggest that glutamatergic input to VTA-DA neurons is inhibited by GABAergic interneurons via GABAB receptors and that morphine promotes presynaptic glutamate release by removing this inhibition. We also studied the contribution of the morphine-induced disinhibitory effect on the presynaptic glutamate release to the overall excitatory effect of morphine on VTA-DA neurons and related behavior. Our results suggest that the disinhibitory action of morphine on presynaptic glutamate release might be the main mechanism for morphine-induced increase in VTA-DA neuron firing and related behaviors. https://doi.org/10.7554/eLife.09275.001 eLife digest Morphine is one of the most commonly used drugs for the treatment of severe pain. It is derived from opium, which is extracted from poppies, and binds to the same receptors in the brain as the body's own naturally produced painkillers. As well as providing pain relief, morphine can act directly on the brain's reward system to trigger a state of euphoria, and can therefore be highly addictive. One of the key components of the brain's reward circuit that morphine affects is called the ventral tegmental area (VTA). The activity of the VTA is regulated by the combined efforts of two groups of cells: excitatory glutamatergic neurons that increase VTA activity and inhibitory interneuronsthat reduce the activity of the VTA. Morphine inhibits the interneurons, thereby allowing the glutamatergic neurons to activate the VTA. But does morphine also strengthen this excitatory input directly? By examining the effects of morphine on individual VTA neurons, Chen et al. show that the drug does indeed enhance the activity of the glutamatergic neurons. However, it does so indirectly by inhibiting another group of interneurons that would otherwise silence the glutamatergic neurons. This effect of morphine is dependent on the drug acting on a specific receptor type on the interneurons. Chen et al. show that injecting a drug that blocks these receptors straight into the VTA of rats prevents morphine from increasing the animals' activity levels. It also prevents the animals from developing a preference for being in locations where they have previously received morphine. This suggests that morphine could primarily exert its pleasurable effects by preventing the glutamatergic neurons from being inhibited, and thus allowing them to activate the VTA neurons. https://doi.org/10.7554/eLife.09275.002 Introduction Morphine is a potent analgesic with high addictive potential. Morphine-induced addictive behaviors are strongly dependent on the activation of dopamine (DA) neurons of the ventral tegmental area (VTA) (Wise, 1989; Gardner, 2011; Luscher and Malenka, 2011). The hedonic response produced by the activation of VTA-DA neurons by morphine is a primary factor behind drug dependence. One previously reported mechanism for morphine activation of VTA-DA neurons is the VTA-DA neuron disinhibition model (Johnson and North, 1992; Kalivas, 1993; White, 1996). Morphine first inhibits neighboring GABA inhibitory neurons, which shifts the balance between inhibitory and excitatory input to VTA-DA neurons in favor of excitation and then leads to the promotion of VTA-DA neuron excitation. It was also reported that the release of glutamate from least some of the glutamate terminals that synapsed on VTA-DA neurons was inhibited by opioids (Manzoni and Williams, 1999; Margolis et al., 2005). This inhibitory effect of opioids on glutamate release is puzzling (Chartoff and Connery, 2014) because one would expect morphine to produce rapid activation of VTA-DA neurons through both the inhibition of GABAergic input and the excitation of glutamatergic input. In addition, postsynaptic inhibition (Ford et al., 2006) or excitation (Margolis et al., 2014) in response to μ opioid receptor activation has also been reported in some VTA-DA neurons. However, the amplitude of the inhibitory outward currents produced by opioids in postsynaptic VTA-DA neurons in the study of Ford et al. was small (2.1 ± 1.5 pA in VTA-DA neurons projecting to the nucleus accumbens and 14 ± 4 pA in those projecting to the basolateral amygdaloid nucleus) (Ford et al., 2006), while only a small population of VTA-DA neurons (19%) showed depolarization or an increase in firing rate in response to opioids in the study of Margolis et al. (2014). This low postsynaptic responsiveness to opioids was consistent with physiological and anatomical evidence that only a few VTA-DA neurons had µ receptors (Ford et al., 2006). Here, we propose that morphine might have a strengthening, rather than an inhibitory, effect on glutamatergic input to VTA-DA neurons because it was reported that blockade of excitatory glutamatergic signaling in the VTA suppressed morphine-induced VTA-DA neuron activation (Jalabert et al., 2011) and related behaviors (Kalivas and Alesdatter, 1993; Harris and Aston-Jones, 2003; Harris et al., 2004). Moreover, the strengthening influence of morphine on glutamatergic input to VTA-DA neurons is more relevant to its promoting effect on VTA-DA neuron excitation. In this article, we report our observations of the effect of morphine on glutamatergic input to VTA-DA neurons using the whole-cell patch-clamp method, and the results of our study of its mechanism and functional consequences using electrophysiological, biochemical, optogenetic, pharmacological, and behavioral approaches. Results Morphine-induced increase in spontaneous firing frequency of VTA-DA neurons is dependent on glutamatergic input We observed the effect of morphine (10 µM) on the spontaneous firing frequency of VTA-DA neurons in rats. We select 10 µM of morphine because this concentration is commonly used in in vitro experiments as it elicits a significant effect useful for further analyses (Akaishi et al., 2000), and the effect of morphine at this concentration can be essentially abolished by the opioid receptor antagonist naloxone (Valentino and Dingledine, 1982). VTA-DA neurons were identified based on Ih currents (Figure 1A) and tyrosine hydroxylase (TH) staining (Figure 1B). Details of the identification method are given in the ‘Materials and methods’ section. Using original recordings of spontaneous firing (left panel of Figure 2A) and the time course of spontaneous firing (middle panel of Figure 2A) in VTA-DA neurons for comparison, we could see that morphine (10 μM) increased the frequency of spontaneous firing in VTA-DA neurons. The average frequency of spontaneous firing increased from 1.0 ± 0.3 Hz before to 1.4 ± 0.4 Hz for 10–15 min after morphine application (n = 6 cells from five rats, paired t test, p < 0.05, compared to control before morphine, right panel of Figure 2A). In order to determine the role of glutamatergic input in the morphine-induced increase in the spontaneous firing frequency of VTA-DA neurons, we observed the influence of the N-methyl-D-aspartic acid (NMDA) receptor antagonist DL-2-amino-5-phosphonovaleric acid (APV) (50 μM) and the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist 6, 7-Dinitroquinoxalie-2, 3-dione (DNQX) (10 μM) on the effect of morphine. In the presence of APV and DNQX, morphine no longer increased spontaneous firing frequency (Figure 2B). The average frequency of spontaneous firing was 0.7 ± 0.1 Hz before and 0.8 ± 0.1 Hz for 10–15 min after morphine application in the presence of APV and DNQX (n = 6 cells from five rats, paired t test, p > 0.05, compared to control with APV and DNQX before morphine, right panel of Figure 2B). These results suggest that the morphine-induced increase in the spontaneous firing frequency of VTA-DA neurons requires AMPA and NMDA receptor-mediated glutamatergic input, consistent with a recent report using the NMDA antagonist APV and the AMPA receptor antagonist 6-Cyano-7-nitroquinoxaline-2, 3-dione (CNQX) in in vivo experiments (Jalabert et al., 2011). Figure 1 Download asset Open asset Identification of VTA-DA neurons in rats. (A) Electrophysiological properties of VTA-DA neurons. Left panel: representative traces showing a large hyperpolarization-activated current (Ih) in whole-cell voltage-clamp recording. Holding potential: −70 mV. Right panel: representative traces showing a large voltage ‘sag’ when hyperpolarized in whole-cell current-clamp recording. Holding current: 0 pA. (B) Immunohistochemical labeling of identified VTA-DA neurons. Panel 1: images of a Lucifer yellow-labeled neuron from the ventral tegmental area (VTA) after whole-cell patch-clamp recording under infrared differential interference contrast and fluorescent microscopy. Panel 2: the same neuron labeled with Lucifer yellow (green color) under confocal microscopy. Panel 3: VTA images showing tyrosine hydroxylase (TH)-positive neurons after immunostaining. Panel 4: Lucifer yellow-filled neuron co-labeled with TH. Scale bar: 20 μm. https://doi.org/10.7554/eLife.09275.003 Figure 2 Download asset Open asset Effect of morphine on spontaneous firing of VTA-DA neurons in rats and the influence of the NMDA receptor antagonist APV and the AMPA receptor antagonist DNQX on the effect of morphine on spontaneous firing of VTA-DA neurons in rats. (A) Effect of morphine on spontaneous firing of VTA-DA neurons. Left panel: representative spontaneous firing traces before and after morphine (10 μM). Middle panel: time course of spontaneous firing before and after morphine (10 μM) (n = 6 cells from five rats). Right panel: average frequency of spontaneous firing before and after morphine (n = 6 cells from five rats, p < 0.05, compared to control before morphine). (B) Influence of the NMDA receptor antagonist APV and the AMPA receptor antagonist DNQX on the effect of morphine on spontaneous firing in VTA-DA neurons. Left panel: representative spontaneous firing traces before and after morphine (10 μM) in the presence of APV (50 μM) and DNQX (10 μM). Middle panel: time course of spontaneous firing before and after morphine in the presence of APV (50 μM) and DNQX (10 μM) (n = 6 cells from five rats). Right panel: average frequency of spontaneous firing before and after morphine in the presence of APV (50 μM) and DNQX (10 μM) (n = 6 cells from five rats, p = 0.34). Data are shown as the mean ±s.e.m. *p < 0.05. https://doi.org/10.7554/eLife.09275.004 Morphine has an additional promoting effect on presynaptic glutamate release in VTA-DA neurons In order to study the effect of morphine on glutamatergic input to VTA-DA neurons, we examined the effect of morphine on the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in VTA-DA neurons in rats. First, we observed the effect of morphine on the frequency of sEPSCs when the GABAA receptor antagonist picrotoxin (PTX) was added to a bath solution to remove spontaneous inhibitory postsynaptic currents (sIPSCs). Consistent with earlier reports (Manzoni and Williams, 1999; Margolis et al., 2005), in the presence of extracellularly applied PTX (100 μM), morphine (10 μM) decreased the frequency of sEPSCs (Figure 3A). The average frequency of sEPSCs decreased from 4.2 ± 0.7 Hz before to 3.5 ± 0.7 Hz for 10–15 min after morphine application (n = 6 cells from four rats, paired t test, p < 0.05, compared to control before morphine, right panel of Figure 3A). However, bath application of GABAA receptor antagonists can lead to a wide blocking effect on GABAA receptors present on different types of neurons, including DA-neurons and GABA neurons, in VTA slices. To circumvent this wide influence and target the inhibition of GABAA receptors to VTA-DA neurons, we added the GABAA receptor antagonist PTX (100 µM) to the internal solution of microelectrodes as described by Akaike et al. (1985) when recording sEPSCs. To demonstrate the effectiveness of the blockade of GABAA receptors by intracellularly applied PTX, we first examined the effect of intracellularly applied PTX (100 μM) on inhibitory postsynaptic currents (IPSC) in VTA-DA neurons. The results showed that under this experimental paradigm, IPSCs disappeared (top of panel 1 in Figure 3B, n = 5 cells from two rats), demonstrating that GABAA receptors in the recorded VTA-DA neurons were blocked by intracellularly applied PTX. In addition, to further confirm that the spontaneous events we measured in the presence of intracellularly applied PTX were in fact sEPSCs, we observed the effect of the AMPA receptor antagonist DNQX on spontaneous events in the presence of intracellularly applied PTX. The results showed that the spontaneous events were completely blocked by DNQX (10 μM) (bottom of panel 1 in Figure 3B). We repeated the experiment in three cells from different slices and obtained similar results. On this basis, we observed the effect of morphine on the sEPSCs in the presence of intracellularly applied PTX. Raw current traces (panel 2 of Figure 3B) and the time course of sEPSCs (panel 3 of Figure 3B) before and after morphine application in the presence of intracellularly applied PTX showed that morphine (10 μM) increased the frequency of sEPSCs. The average frequency of sEPSCs increased from 4.8 ± 0.6 Hz before to 5.5 ± 0.6 Hz for 10–15 min after morphine application (n = 8 cells from five rats, paired t test, p < 0.05, compared to control before morphine, panel 4 of Figure 3B). However, morphine (10 μM) had no significant effect on the amplitude of sEPSCs. The average amplitude of sEPSCs was 16.6 ± 1.3 pA before and 15.3 ± 1.3 pA for 10–15 min after morphine application (n = 8 cells from five rats, paired t test, p > 0.05, compared to control before morphine, panel 5 of Figure 3B). To confirm that the increased spontaneous events we measured in the presence of intracellularly applied PTX after morphine were also in fact sEPSCs, we observed the effect of the AMPA receptor antagonist DNQX on spontaneous events after morphine application in the presence of intracellularly applied PTX. The results showed that the spontaneous events after morphine application were completely blocked by adding DNQX (10 μM) (panel 6 of Figure 3B). We repeated the experiment in three cells from different slices and obtained similar results. We also used the first excitatory postsynaptic current (EPSC) of paired pulse facilitation (PPF) as an index of EPSC (Maejima et al., 2001), and the PPF of paired EPSC as an indicator of presynaptic glutamate release (Zucker and Regehr, 2002) to confirm the effect of morphine on presynaptic glutamate release in VTA-DA neurons in the presence of intracellularly applied PTX. As shown in the left panel of Figure 3C, morphine (10 μM) increased the amplitude of the first EPSC, which was accompanied by a clear change in the presynaptic parameter PPF. The average amplitude of the first EPSCs was 124.1 ± 9.0 pA before and 161.9 ± 10.8 pA for 10–15 min after morphine application (n = 6 cells from four rats, paired t test, p < 0.05, compared to control before morphine, middle panel of Figure 3C). The average PPF was decreased from 1.5 ± 0.2 before to 1.2 ± 0.1 for 10–15 min after morphine application (n = 6 cells from four rats, paired t test, p < 0.05, compared to control before morphine, right panel of Figure 3C). These results supported the suggestion that morphine increased presynaptic glutamate release in VTA-DA neurons in the presence of intracellularly applied PTX. More importantly, when we clamped the membrane potential of VTA-DA neurons at the reversal potential of Cl− channels to remove sIPSCs, as an alternative to the application of a GABAA antagonist, either by the intracellular or bath approach, morphine still exerted a promoting effect on presynaptic glutamate release in VTA-DA neurons (Figure 3D). The average frequency of sEPSCs in this condition increased from 4.4 ± 0.3 Hz before to 5.5 ± 0.4 Hz for 10–15 min after morphine application (n = 6 cells from four rats, paired t test, p < 0.05, compared to control before morphine, right panel of Figure 3D). Figure 3 Download asset Open asset Effects of morphine on the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) and paired pulse facilitation (PPF) of VTA-DA neurons in rats. (A) Effects of morphine on the frequency of sEPSCs in the presence of extracellularly applied picrotoxin (PTX) in VTA-DA neurons. Left panel: typical current traces of sEPSCs before and after morphine (10 μM) in the presence of extracellularly applied PTX. Middle panel: typical time course of the frequency of sEPSCs before and after morphine (10 μM) in the presence of extracellularly applied PTX. Right panel: average frequency of sEPSCs before and after morphine (10 μM) in the presence of extracellularly applied PTX (n = 6 cells from four rats, p < 0.05, compared to control before morphine). (B) Effects of morphine on sEPSCs in the presence of intracellularly applied PTX in VTA-DA neurons. Top of panel 1: inhibitory postsynaptic currents (IPSCs) in the normal intracellular recording solution and the presence of intracellularly applied PTX. Bottom of panel 1: typical current traces of sEPSCs before and after DNQX (10 μM) in the presence of intracellularly applied PTX. Panel 2: typical current traces of sEPSCs before and after morphine (10 μM) in the presence of intracellularly applied PTX. Panel 3: typical time course of the frequency of sEPSCs before and after morphine (10 μM) in the presence of intracellularly applied PTX. Panel 4: average frequency of sEPSCs before and after morphine (10 μM) in the presence of intracellularly applied PTX (n = 8 cells from five rats, p < 0.05, compared to control before morphine). Panel 5: average amplitude of sEPSCs before and after morphine (10 μM) in the presence of intracellularly applied PTX (n = 8 cells from five rats, p = 0.24, compared to control before morphine). Panel 6: typical current traces of sEPSCs before and after DNQX (10 μM) in the presence of intracellularly applied PTX and morphine. (C) Effects of morphine on the PPF in VTA-DA neurons. Left panel: representative traces of the PPF before and after morphine (10 μM), and superimposition of the two traces normalized to the first excitatory postsynaptic current (EPSC) before and after morphine (10 μM) in the presence of intracellularly applied PTX. Middle panel: average amplitude of the first EPSC in control and morphine (10 μM) in the presence of intracellularly applied PTX (n = 6 cells from four rats, p < 0.05, compared to control before morphine). Right panel: average PPF before and after morphine (10 μM) in the presence of intracellularly applied PTX (n = 6 cells from four rats, p < 0.05, compared to control before morphine). (D) Effects of morphine on the frequency of sEPSCs when VTA-DA neurons were clamped the membrane potential at the reversal potential of Cl− channels in VTA-DA neurons. Left panel: typical current traces of sEPSCs before and after morphine (10 μM) when DA neurons was clamped the membrane potential at the reversal potential of Cl− channels. Middle panel: typical time course of the frequency of sEPSCs before and after morphine (10 μM) when DA neurons was clamped the membrane potential at the reversal potential of Cl− channels. Right panel: average frequency of sEPSCs before and after morphine (10 μM) when DA neurons was clamped the membrane potential at the reversal potential of Cl− channels (n = 6 cells from four rats, p < 0.05, compared to control before morphine). Data are shown as the mean ±s.e.m. *p < 0.05. https://doi.org/10.7554/eLife.09275.005 Morphine promotes presynaptic glutamate release in VTA-DA neurons via presynaptic disinhibition The mechanism underlying the promoting effect of morphine on presynaptic glutamate release in VTA-DA neurons may involve different processes. One suggestion is that morphine may directly act at glutamatergic terminals to promote glutamate release. To test this hypothesis, we studied the effect of morphine on glutamate release from the VTA synaptosomes of rats, which are sealed particles containing vesicles, viable mitochondria, and all components necessary to store, release, and retain neurotransmitters (Breukel et al., 1997), using on-line fluorometry. However, we did not find that morphine (10 μM) had a direct effect at glutamatergic terminals to promote glutamate release. The average concentration of glutamate was 8.1 ± 0.1 nmol/mg before and 8.5 ± 0.7 nmol/mg after morphine application (n = 6 samples from eight rats, paired t test, p > 0.05, Figure 4A). We also studied the effect of morphine on the frequency of sEPSCs from mechanically dissociated single VTA-DA neurons in rats, which retained functional synaptic terminals (Akaike and Moorhouse, 2003; Ye et al., 2004; Deng et al., 2009). The left top image of panel 1 of Figure 4B shows mechanically dissociated neurons from the VTA. These VTA-DA neurons were identified by only using Ih currents (the graph on the right of panel 1 of Figure 4B) without TH staining because in dissociated DA neurons, it was difficult to fix the cell after recording for TH staining. The results showed that morphine (10 µM) had no significant effect on the frequency of sEPSCs (panels 2 and 3 of Figure 4B). The average frequency of sEPSCs was 2.1 ± 0.3 Hz before and 2.0 ± 0.2 Hz for 10–15 min after morphine application (10 µM) (n = 6 cells from six rats, paired t test, p > 0.05, compared to control before morphine, panel 4 of Figure 4B). These results suggest that morphine may promote presynaptic glutamate release in VTA-DA neurons in an indirect way. Figure 4 Download asset Open asset Effects of morphine on glutamate release from the ventral tegmental area (VTA) synaptosomes of rats and the frequency of sEPSCs in mechanically dissociated VTA-DA neurons from rats. (A) Effects of morphine on glutamate release from VTA synaptosomes. Average concentration of glutamate release before and after application of morphine (10 μM) from VTA synaptosomes (n = 6 samples from eight rats, p = 0.46). (B) Effects of morphine on the frequency of sEPSCs in mechanically dissociated VTA-DA neurons. Panel 1, left: images of an acutely dissociated single neuron from the VTA under phase contrast microscopy. Scale bar: 10 μm. Panel 1, right: representative current traces showing a large hyperpolarization-activated current (Ih) in whole-cell voltage-clamp recording. Holding potential: −70 mV. Panel 2: typical current traces of sEPSC before and after morphine (10 μM) in the presence of intracellularly applied PTX. Panel 3: typical time course of the frequency of sEPSCs before and after morphine (10 μM) in the presence of intracellularly applied PTX. Panel 4: Average frequency of sEPSCs before and after morphine (10 μM) in the presence of intracellularly applied PTX (n = 6 cells from six rats, p = 0.65). Data are shown as the mean ±s.e.m. *p < 0.05. https://doi.org/10.7554/eLife.09275.006 To explore how morphine promoted presynaptic glutamate release in VTA-DA neurons, we hypothesized that glutamatergic input to VTA-DA neurons was inhibited by GABAergic interneurons and morphine disinhibited glutamatergic input by removing this inhibition, thus promoting glutamate release. To test this hypothesis, we first studied whether GABA could inhibit presynaptic glutamate release in VTA-DA neurons by examining the effect of exogenous applied of GABA on the frequency of sEPSCs of VTA-DA neurons in rats. From raw current traces (left panel of Figure 5A) and the time course of sEPSCs (middle panel of Figure 5A), we could see that GABA (10 μM) apparently decreased the frequency of sEPSCs. The average frequency of sEPSCs decreased from 6.7 ± 0.7 Hz before to 4.7 ± 0.6 Hz for 10–15 min after GABA application (n = 6 cells from four rats, paired t test, p < 0.05, compared to control before GABA, right panel of Figure 5A). Then, we observed whether the activation of intrinsic GABAergic neurons could inhibit presynaptic glutamate release of VTA-DA neurons in mice. To do this, AAV virus expressing a double floxed-stopped channelrhodopsin-2 (ChR2)-mCherry was stereotaxically injected into the VTA of mice expressing Cre recombinase in GABA neurons. 2 weeks after infection, expression of ChR2–mCherry was observed in the VTA (panel 1 of Figure 5B). We then performed whole-cell patch-clamp recording in GABA neurons of the VTA and observed the light-induced action potentials in GABA neurons in order to confirm that ChR2 was indeed expressed in the GABA neurons of the VTA in mice. The results showed that 470 nm light stimulation (20 Hz) elicited action potentials in GABA neurons (panel 2 of Figure 5B). On this basis, whole-cell patch-clamp recording was performed in VTA-DA neurons to observe the effect of 470 nm light stimulation on the frequency of sEPSCs in the presence of intracellularly applied PTX (100 µM). Following 5–10 min of baseline recording of sEPSCs, twenty 470 nm light pulses of 5 ms at 20 Hz were delivered every 4 s for 15 min. Raw current traces (panel 3 of Figure 5B) and the time course of sEPSCs (panel 4 of Figure 5B) before and after light stimulation showed that the light stimulation apparently decreased the frequency of sEPSCs. The average frequency of sEPSCs decreased from 4.1 ± 0.7 Hz before to 2.7 ± 0.3 Hz for 10–15 min after light stimulation (n = 6 cells from five mice, paired t test, p < 0.05, compared to control before light stimulation, panel 5 of Figure 5B). Figure 5 Download asset Open asset Effect of exogenous application of GABA and 470 nm light stimulation on the frequency of sEPSCs in the presence of intracellularly applied PTX in VTA-DA neurons. (A) Effect of exogenous application of GABA on the frequency of sEPSCs of VTA-DA neurons in rats. Left panel: typical current traces of sEPSCs before and after GABA (10 μM) in the presence of intracellularly applied PTX. Middle panel: typical time course of the frequency of sEPSCs before and after GABA (10 μM) in the presence of intracellularly applied PTX. Right panel: average frequency of sEPSCs before and after GABA (10 μM) in the presence of intracellularly applied PTX (n = 6 cells from four rats, p < 0.05, compared to control before GABA). (B) Effect of 470 nm light stimulation on the frequency of sEPSCs of VTA-DA neurons in mice. Panel 1: coronal image showing the expression of ChR2-mCherry (red) following injection of the viral construct bilaterally into the ventral tegmental area (VTA) of GADcre+ mice. Scale bar: 500 µm. Panel 2: 470 nm light (20 Hz)-induced firing of VTA GABA neurons in current-clamp mode. Panel 3: typical current traces of sEPSCs before and after blue light (470 nm) stimulation in the presence of intracellularly applied PTX. Panel 4: typical time course of the frequency of sEPSCs before and after blue light (470 nm) stimulation in the presence of intracellularly applied PTX. Panel 5: average frequency of sEPSCs before and after blue light (470 nm) stimulation in the presence of intracellularly applied PTX (n = 6 cells from five mice, p < 0.05, compared to control before light stimulation). Data are shown as the mean ±s.e.m. *p < 0.05. https://doi.org/10.7554/eLife.09275.007 We also studied which kinds of GABA receptors (GABAA or GABAB receptors) mediated the decreasing effect of GABA on presynaptic glutamate release in VTA-DA neurons by examining the influence of the GABAA or GABAB receptor antagonist on the effect of GABA on the frequency of sEPSCs in rats. The results showed that the GABAA receptor antagonist PTX (100 µM) had no significant influence on the effect of GABA (Figure 6A). The average frequency of sEPSCs still decreased from 4.0 ± 0.3 Hz before to 3.0 ± 0.2 Hz for 10–15 min after GABA application in the presence of PTX (n = 6 cells from four rats, paired t test, p < 0.05, compared to PTX before GABA, right panel of Figure 6A). The percentage of GABA-produced response in the presence of PTX (−25.8 ± 3.8%) was not statistically significant (n = 6, independent t test, p > 0.05) compared to that without PTX (−29.2 ± 6.0%). However, in the presence of the GABAB receptor antagonist CGP54626, the effect of GABA on the frequency of sEPSCs disappeared (Figure 6B). The average frequency of sEPSCs was 3.8 ± 0.4 Hz before and 3.8 ± 0.4 Hz for 10–15 min after GABA application in the presence of CGP54626 (2 µM) (n = 6 cells from four rats, paired t test, p > 0.05, compared to CGP54626 before GABA, right panel of Figure 6B). These results suggest that it is GABAB receptors, rather than GABAA receptors, that mediate the decreasing effect of GABA on presynaptic glutamate release in VTA-DA neurons. Figure 6 Download asset Open asset Influence of the GABAA receptor antagonist PTX and the GABAB receptor antagonist CGP54626 on the effect of exogenous application of GABA as well as the influence of the GABAB receptor antagonist CGP54626 on 470 nm light-induced inhibition of the frequency of sEPSCs in VTA-DA neurons. (A) Influence of the GABAA receptor antagon
What problem does this paper attempt to address?
-
Morphine Disinhibits Glutamatergic Input to VTA Dopamine Neurons and Promotes Dopamine Neuron Excitation
Ming Chen,Yanfang Zhao,Hualan Yang,Wenjie Luan,Jiaojiao Song,Dongyang Cui,Yi Dong,Bin Lai,Lan Ma,Ping Zheng
DOI: https://doi.org/10.7554/elife.09275
IF: 7.7
2015-01-01
eLife
Abstract:One reported mechanism for morphine activation of dopamine (DA) neurons of the ventral tegmental area (VTA) is the disinhibition model of VTA-DA neurons. Morphine inhibits GABA inhibitory neurons, which shifts the balance between inhibitory and excitatory input to VTA-DA neurons in favor of excitation and then leads to VTA-DA neuron excitation. However, it is not known whether morphine has an additional strengthening effect on excitatory input. Our results suggest that glutamatergic input to VTA-DA neurons is inhibited by GABAergic interneurons via GABAB receptors and that morphine promotes presynaptic glutamate release by removing this inhibition. We also studied the contribution of the morphine-induced disinhibitory effect on the presynaptic glutamate release to the overall excitatory effect of morphine on VTA-DA neurons and related behavior. Our results suggest that the disinhibitory action of morphine on presynaptic glutamate release might be the main mechanism for morphine-induced increase in VTA-DA neuron firing and related behaviors.
-
Morphine selectively disinhibits glutamatergic input from mPFC onto dopamine neurons of VTA, inducing reward.
Li Yang,Ming Chen,Qianqian Ma,Huan Sheng,Dongyang Cui,Da Shao,Bin Lai,Ping Zheng
DOI: https://doi.org/10.1016/j.neuropharm.2020.108217
IF: 5.273
2020-01-01
Neuropharmacology
Abstract:Ventral tegmental area (VTA) dopamine (DA) neurons presynaptic glutamate release plays a very important role in the mechanism of morphine. Previously, a study from our lab found that morphine disinhibited glutamatergic input onto the VTA-DA neurons, which was an important mechanism for the morphine-induced increase in the VTA-DA neuron firing and related behaviors (Chen et al., 2015). However, what source of glutamatergic inputs is disinhibited by morphine remains to be elucidated. Using optogenetic strategy combined with whole-cell patch-clamp, qRT-PCR, immunofluorescence and chemical genetic approach combined with behavioral methods, our results show that: 1) morphine promotes glutamate release from glutamatergic terminals of medial prefrontal cortex (mPFC) neurons projecting to VTA-DA neurons but does not on those from glutamatergic terminals of the lateral hypothalamus (LH) neurons projecting to VTA-DA neurons; 2) different response of glutamatergic neurons projecting to VTA-DA neurons from the mPFC or the LH to morphine is related to the expression of GABAB receptors at terminals of these neurons; 3) inhibition of projection neurons from the mPFC to the VTA significantly reduces morphine-induced locomotor activity increase and conditioned place preference but inhibition of projection neurons from the LH to the VTA does not. These results suggest that morphine selectively promotes glutamate release of the glutamatergic input from mPFC onto VTA-DA neurons by removing the inhibition of the GABAB receptors in this glutamatergic input from mPFC.
-
Morphine selectively promotes glutamate release from glutamatergic terminals of projection neurons from medial prefrontal cortex to dopamine neurons of ventral tegmental area
Ping Zheng,Li Yang,Ming Chen
DOI: https://doi.org/10.1101/133090
2017-01-01
bioRxiv
Abstract:Recently, we found that morphine promoted presynaptic glutamate release of dopamine (DA) neurons in the ventral tegmental area (VTA), which constituted the main mechanism for morphine-induced increase in VTA-DA neuron firing and related behaviors (Chen et al., 2015). However, what source of presynaptic glutamate release of DA neurons in the VTA is promoted by morphine remains unknown. To address this question, we used optogenetic strategy to selectively activate glutamatergic inputs from different projection neurons and then observed the effect of morphine on them. The result shows that morphine promotes glutamate release from glutamatergic terminals of projection neurons from the medial prefrontal cortex (mPFC) to VTA DA neurons, but has no effect on that from the basolateral amygdala (BLA) or the lateral hypothalamus (LH) to VTA DA neurons, and the inhibition of glutamatergic projection neurons from the mPFC to the VTA significantly reduces morphine-induced increase in locomotor activity of mice.
-
Morphine Epigenomically Regulates Behavior Through Alterations in Histone H3 Lysine 9 Dimethylation in the Nucleus Accumbens
HaoSheng Sun,Ian Maze,David M. Dietz,Kimberly N. Scobie,Pamela J. Kennedy,Diane Damez-Werno,Rachael L. Neve,Venetia Zachariou,Li Shen,Eric J. Nestler
DOI: https://doi.org/10.1523/jneurosci.1357-12.2012
2012-01-01
Journal of Neuroscience
Abstract:Dysregulation of histone modifying enzymes has been associated with numerous psychiatric disorders. Alterations in G9a (Ehmt2), a histone methyltransferase that catalyzes the euchromatic dimethylation of histone H3 at lysine 9 (H3K9me2), has been implicated recently in mediating neural and behavioral plasticity in response to chronic cocaine administration. Here, we show that chronic morphine, like cocaine, decreases G9a expression, and global levels of H3K9me2, in mouse nucleus accumbens (NAc), a key brain reward region. In contrast, levels of other histone methyltransferases or demethylases, or of other methylated histone marks, were not affected in NAc by chronic morphine. Through viral-mediated gene transfer and conditional mutagenesis, we found that overexpression of G9a in NAc opposes morphine reward and locomotor sensitization and concomitantly promotes analgesic tolerance and naloxone-precipitated withdrawal, whereas downregulation of G9a in NAc enhances locomotor sensitization and delays the development of analgesic tolerance. We identified downstream targets of G9a by providing a comprehensive chromatin immunoprecipitation followed by massively parallel sequencing analysis of H3K9me2 distribution in NAc in the absence and presence of chronic morphine. These data provide novel insight into the epigenomic regulation of H3K9me2 by chronic morphine and suggest novel chromatin-based mechanisms through which morphine-induced addictive-like behaviors arise.
-
Morphine treatment enhances glutamatergic input onto neurons of the nucleus accumbens via both disinhibitory and stimulating effect.
Kejing Yuan,Huan Sheng,Jiaojiao Song,Li Yang,Dongyang Cui,Qianqian Ma,Wen Zhang,Bin Lai,Ming Chen,Ping Zheng
DOI: https://doi.org/10.1111/adb.12438
2017-01-01
Addiction Biology
Abstract:Drug addiction is a chronic brain disorder characterized by the compulsive repeated use of drugs. The reinforcing effect of repeated use of drugs on reward plays an important role in morphine-induced addictive behaviors. The nucleus accumbens (NAc) is an important site where morphine treatment produces its reinforcing effect on reward. However, how morphine treatment produces its reinforcing effect on reward in the NAc remains to be clarified. In the present study, we studied the influence of morphine treatment on the effects of DA and observed whether morphine treatment could directly change glutamatergic synaptic transmission in the NAc. We also explored the functional significance of morphine-induced potentiation of glutamatergic synaptic transmission in the NAc at behavioral level. Our results show that (1) morphine treatment removes the inhibitory effect of DA on glutamatergic input onto NAc neurons; (2) morphine treatment potentiates glutamatergic input onto NAc neurons, especially the one from the basolateral amygdala (BLA) to the NAc; (3) blockade of glutamatergic synaptic transmission in the NAc or ablation of projection neurons from BLA to NAc significantly decreases morphine treatment-induced increase in locomotor activity. These results suggest that morphine treatment enhances glutamatergic input onto neurons of the NAc via both disinhibitory and stimulating effect and therefore increases locomotor activity.
-
Increased Glutamate Synaptic Transmission in the Nucleus Raphe Magnus Neurons from Morphine-Tolerant Rats
Bihua Bie,Zhizhong Z Pan
DOI: https://doi.org/10.1186/1744-8069-1-7
IF: 3.37
2005-01-01
Molecular Pain
Abstract:Currently, opioid-based drugs are the most effective pain relievers that are widely used in the treatment of pain. However, the analgesic efficacy of opioids is significantly limited by the development of tolerance after repeated opioid administration. Glutamate receptors have been reported to critically participate in the development and maintenance of opioid tolerance, but the underlying mechanisms remain unclear. Using whole-cell voltage-clamp recordings in brainstem slices, the present study investigated chronic morphine-induced adaptations in glutamatergic synaptic transmission in neurons of the nucleus raphe magnus (NRM), a key supraspinal relay for pain modulation and opioid analgesia. Chronic morphine significantly increased glutamate synaptic transmission exclusively in one class of NRM cells that contains μ-opioid receptors in a morphine-tolerant state. The adenylyl cyclase activator forskolin and the cAMP analog 8-bromo-cAMP mimicked the chronic morphine effect in control neurons and their potency in enhancing the glutamate synaptic current was significantly increased in neurons from morphine-tolerant rats. MDL12330a, an adenylyl cyclase inhibitor, and H89, a protein kinase A (PKA) inhibitor, reversed the increase in glutamate synaptic transmission induced by chronic morphine. In addition, PMA, a phorbol ester activator of protein kinase C (PKC), also showed an increased potency in enhancing the glutamate synaptic current in these morphine-tolerant cells. The PKC inhibitor GF109203X attenuated the chronic morphine effect. Taken together, these results suggest that chronic morphine increases presynaptic glutamate release in μ receptor-containing NRM neurons in a morphine-tolerant state, and that the increased glutamate synaptic transmission appears to involve an upregulation of both the cAMP/PKA pathway and the PKC pathway. This glutamate-mediated activation of these NRM neurons that are thought to facilitate spinal pain transmission may contribute to the reduced opioid analgesia during opioid tolerance.
-
Α2δ-1–Boundn-methyl-d-aspartate Receptors Mediate Morphine-induced Hyperalgesia and Analgesic Tolerance by Potentiating Glutamatergic Input in Rodents
Meichun Deng,Shao-Rui Chen,Hong Chen,Hui-Lin Pan
DOI: https://doi.org/10.1097/aln.0000000000002648
IF: 8.986
2019-01-01
Anesthesiology
Abstract:Background: Chronic use of mu-opioid receptor agonists paradoxically causes both hyperalgesia and the loss of analgesic efficacy. Opioid treatment increases presynaptic N-methyl-d-aspartate receptor activity to potentiate nociceptive input to spinal dorsal horn neurons. However, the mechanism responsible for this opioid- induced activation of presynaptic N-methyl-d-aspartate receptors remains unclear. alpha 2 delta-1, formerly known as a calcium channel subunit, interacts with N-methyl-d-aspartate receptors and is primarily expressed at presynaptic terminals. This study tested the hypothesis that alpha 2 delta-1-bound N-methyl-d-aspartate receptors contribute to presynaptic N-methyl-d-aspartate receptor hyperactivity associated with opioid-induced hyperalgesia and analgesic tolerance. Methods: Rats (5 mg/kg) and wild-type and alpha 2 delta-1-knockout mice (10 mg/kg) were treated intraperitoneally with morphine twice/day for 8 consecutive days, and nociceptive thresholds were examined. Presynaptic N-methyl-d-aspartate receptor activity was recorded in spinal cord slices. Coimmunoprecipitation was performed to examine protein-protein interactions. Results: Chronic morphine treatment in rats increased alpha 2 delta-1 protein amounts in the dorsal root ganglion and spinal cord. Chronic morphine exposure also increased the physical interaction between alpha 2 delta-1 and N-methyl-d-aspartate receptors by 1.5 +/- 0.3 fold (means +/- SD, P = 0.009, n = 6) and the prevalence of alpha 2 delta-1-bound N-methyl-d-aspartate receptors at spinal cord synapses. Inhibiting alpha 2 delta-1 with gabapentin or genetic knockout of alpha 2 delta-1 abolished the increase in presynaptic N-methyl-d-aspartate receptor activity in the spinal dorsal horn induced by morphine treatment. Furthermore, uncoupling the alpha 2 delta-1-Nmethyl- d-aspartate receptor interaction with an alpha 2 delta-1 C terminus-interfering peptide fully reversed morphine-induced tonic activation of N-methyl-d-aspartate receptors at the central terminal of primary afferents. Finally, intraperitoneal injection of gabapentin or intrathecal injection of an alpha 2 delta-1 C terminus-interfering peptide or alpha 2 delta-1 genetic knockout abolished the mechanical and thermal hyperalgesia induced by chronic morphine exposure and largely preserved morphine's analgesic effect during 8 days of morphine treatment. Conclusions: alpha 2 delta-1-Bound N-methyl-d-aspartate receptors contribute to opioid-induced hyperalgesia and tolerance by augmenting presynaptic N-methyl-d-aspartate receptor expression and activity at the spinal cord level.
-
GABAA Receptors in VTA Mediate the Morphine-Induced Release of Ascorbic Acid in Rat Nucleus Accumbens
Ji-Ye Sun,Jing-Yu Yang,Fang Wang,Yue Hou,Ying-Xu Dong,Chun-Fu Wu
DOI: https://doi.org/10.1016/j.brainres.2010.10.029
IF: 3.61
2010-01-01
Brain Research
Abstract:Local perfusion of morphine produces increased levels of extracellular ascorbic acid (AA) in the nucleus accumbens (NAc) of freely moving rats. However, the pathways that regulate morphine-induced AA release in the NAc are unclear. In the present study, we used high performance liquid chromatography with electrochemical detection (HPLC-ECD) to examine the effects of intra-ventral tegmental area (VTA) administration of a GABA(A) agonist and antagonist on morphine-induced increases in AA of the NAc. Also, using high performance liquid chromatography with fluorescent detection (HPLC-FD) and HPLC-ECD, the releases of γ-aminobutyric acid (GABA) and dopamine (DA) in the NAc induced by intra-VTA administration of a GABA(A) agonist and antagonist were also investigated. The results obtained showed that morphine (1 mM), locally perfused into the NAc, significantly increased AA release in the NAc and also GABA release. Intra-VTA infusion of bicuculline (150 ng/rat), a GABA receptor antagonist, not only abolished the enhanced extracellular AA and GABA levels produced by local perfusion of morphine but also decreased the basal release of extracellular GABA and increased the basal release of extracellular DA in the NAc. Muscimol (100 ng/rat), a GABA receptor agonist, affected the basal release of GABA and DA, but not the basal AA levels, or the morphine-induced changes in AA and GABA levels. These findings suggest that the GABA(A) receptors in the VTA play an important role in the modulation of morphine-induced AA release in the NAc, and the effect of morphine on AA release in the NAc is partially regulated by the GABA(A) receptor-mediated action of DA afferents from the VTA.
-
Morphine Differentially Alters the Synaptic and Intrinsic Properties of D1R- and D2R-Expressing Medium Spiny Neurons in the Nucleus Accumbens
Dillon S. McDevitt,Benjamin Jonik,Nicholas M. Graziane
DOI: https://doi.org/10.3389/fnsyn.2019.00035
2019-12-20
Frontiers in Synaptic Neuroscience
Abstract:Exposure to opioids reshapes future reward and motivated behaviors partially by altering the functional output of medium spiny neurons (MSNs) in the nucleus accumbens shell. Here, we investigated how morphine, a highly addictive opioid, alters synaptic transmission and intrinsic excitability on dopamine D1-receptor (D1R) expressing and dopamine D2-receptor (D2R) expressing MSNs, the two main output neurons in the nucleus accumbens shell. Using whole-cell electrophysiology recordings, we show, that 24 h abstinence following repeated non-contingent administration of morphine (10 mg/kg, i.p.) in mice reduces the miniature excitatory postsynaptic current (mEPSC) frequency and miniature inhibitory postsynaptic current (mIPSC) frequency on D2R-MSNs, with concomitant increases in D2R-MSN intrinsic membrane excitability. We did not observe any changes in synaptic or intrinsic changes on D1R-MSNs. Last, in an attempt to determine the integrated effect of the synaptic and intrinsic alterations on the overall functional output of D2R-MSNs, we measured the input-output efficacy by measuring synaptically-driven action potential firing. We found that both D1R-MSN and D2R-MSN output was unchanged following morphine treatment.
neurosciences
-
Projection-specific dopamine neurons in the ventral tegmental area participated in morphine-induced hyperalgesia and anti-nociceptive tolerance in male mice
Guo-Lin Sun,Zhi-Jing Song,Xiao-Han Peng,Pan-Pan Chen,Ying Song,Xia Qin,Rong Hua,Yong-Mei Zhang
DOI: https://doi.org/10.1177/0269881120985183
2021-03-21
Journal of Psychopharmacology
Abstract:Background: Long-term morphine use is associated with serious side effects, such as morphine-induced hyperalgesia and analgesic tolerance. Previous investigations have documented the association between dopamine (DA) neurons in the ventral tegmental area (VTA) and pain. However, whether VTA DA neurons are implicated in morphine-induced hyperalgesia and analgesic tolerance remains elusive. Methods: Initially, we observed behavioural effects of lidocaine administration into VTA or ablation of VTA DA neurons on morphine-induced hyperalgesia and anti-nociceptive tolerance. Subsequently, c-Fos expression in nucleus accumbens (NAc) shell-projecting and medial prefrontal cortex (mPFC)-projecting VTA DA neurons after chronic morphine treatment was respectively investigated. Afterwards, the effects of chemogenetic manipulation of NAc shell-projecting or mPFC-projecting DA neurons on morphine-induced hyperalgesia and anti-nociceptive tolerance were observed. Additionally, effects of chemogenetic manipulation of VTA GABA neurons on c-Fos expression in VTA DA neurons were investigated. Results: Lidocaine injection into VTA relieved established hyperalgesia and anti-nociceptive tolerance whereas ablation of VTA DA neurons prevented the development of morphine-induced hyperalgesia and anti-nociceptive tolerance. Chronic morphine treatment increased c-Fos expression in NAc shell-projecting DA neurons, rather than in mPFC-projecting DA neurons. Chemogenetic manipulation of NAc shell-projecting DA neurons had influence on morphine-induced hyperalgesia and tolerance. However, chemogenetic manipulation of mPFC-projecting DA neurons had no significant effects on morphine-induced hyperalgesia and anti-nociceptive tolerance. Chemogenetic manipulation of VTA GABA neurons affected the c-Fos expression in VTA DA neurons. Conclusions: These findings revealed the involvement of NAc shell-projecting VTA DA neurons in morphine-induced hyperalgesia and anti-nociceptive tolerance, and may shed new light on the clinical management of morphine-induced hyperalgesia and analgesic tolerance. Perspective: This study demonstrated that NAc shell-projecting DA neurons rather than mPFC-projecting DA neurons in the VTA were implicated in morphine-induced hyperalgesia and anti-nociceptive tolerance. Our findings may pave the way for the discovery of novel therapies for morphine-induced hyperalgesia and analgesic tolerance.
pharmacology & pharmacy,neurosciences,psychiatry,clinical neurology
-
Influences of morphine on the spontaneous and evoked excitatory postsynaptic currents in lateral amygdala of rats.
J-J Zhang,X-D Liu,L-C Yu
DOI: https://doi.org/10.33549/physiolres.933027
2016-01-01
Physiological Research
Abstract:Acute morphine exposure induces antinociceptive activity, but the underlying mechanisms in the central nervous system are unclear. Using whole-cell patch clamp recordings, we explore the role of morphine in the modulation of excitatory synaptic transmission in lateral amygdala neurons of rats. The results demonstrate that perfusion of 10 mu M of morphine to the lateral amygdala inhibits the discharge frequency significantly. We further find that there are no significant influences of morphine on the amplitude of spontaneous excitatory postsynaptic currents (sEPSCs). Interestingly, morphine shows no marked influence on the evoked excitatory postsynaptic currents (eEPSCs) in the lateral amygdala neurons. These results indicate that acute morphine treatment plays an important role in the modulation on the excitatory synaptic transmission in lateral amygdala neurons of rats.
-
Nucleus accumbens sub-regions experience distinct dopamine release responses following acute and chronic morphine exposure
Sarah Warren Gooding,Elinor Lewis,Christine Chau,Suhail Sandhu,Julianna Glienke,Jennifer Whistler
DOI: https://doi.org/10.1101/2024.06.28.601282
2024-07-02
Abstract:It is well established that dopamine neurons of the ventral tegmental area (VTA) play a critical role in reward and aversion as well as pathologies including drug dependence and addiction. The distinct effects of acute and chronic opioid exposure have been previously characterized at VTA synapses. Recent work suggests that distinct VTA projections that target the medial and lateral shell of the nucleus accumbens (NAc), may play opposing roles in modulating behavior. It is possible that these two anatomically and functionally distinct pathways also have disparate roles in opioid reward, tolerance, and withdrawal in the brain. In this study we monitored dopamine release in the medial or lateral shell of the NAc of male mice during a week-long morphine treatment paradigm. We measured dopamine release in response to an intravenous morphine injection both acutely and following a week of repeated morphine. We also measured dopamine in response to a naloxone injection both prior to and following repeated morphine treatment. Morphine induced a transient increase in dopamine in the medial NAc shell that was much larger than the slower rise observed in the lateral shell. Surprisingly, chronic morphine treatment induced a sensitization of the medial dopamine response to morphine that opposed a diminished response observed in the saline-treated control group. This study expands on our current understanding of the medial NAc shell as hub of opioid-induced dopamine fluctuation. It also highlights the need for future opioid studies to appreciate the heterogeneity of dopamine neurons.
Neuroscience
-
Morphine Coordinates SST and PV Interneurons in the Prelimbic Cortex to Disinhibit Pyramidal Neurons and Enhance Reward
Changyou Jiang,Xueying Wang,Qiumin Le,Peipei Liu,Cao Liu,Zhilin Wang,Guanhong He,Ping Zheng,Feifei Wang,Lan Ma
DOI: https://doi.org/10.1038/s41380-019-0480-7
IF: 11
2019-01-01
Molecular Psychiatry
Abstract:Opioids, such as morphine, are clinic analgesics which induce euphoria. Morphine exposure modifies the excitability and functional interactions between neurons, while the underlying cellular and molecular mechanisms, especially how morphine assembles heterogeneous interneurons (INs) in prelimbic cortex (PrL) to mediate disinhibition and reward, are not clear. Using approaches of optogenetics, electrophysiology, and cell type-specific RNA-seq, we show that morphine attenuates the inhibitory synaptic transmission from parvalbumin+ (PV)-INs onto pyramidal neurons in PrL via μ-opioid receptor (MOR) in PV-INs. Meanwhile, morphine enhances the inhibitory inputs from somatostatin+ (SST)-INs onto PV-INs, and thus disinhibits pyramidal neurons via δ-opioid receptor (DOR)-dependent Rac1 upregulation in SST-INs. We show that MOR in PV-INs is required for morphine-induced behavioral sensitization, while DOR as well as Rac1 activity in SST-INs is required for morphine-induced conditioned place preference and hyper-locomotion. These results reveal that SST- and PV-INs, functioning in PrL as a disinhibitory architecture, are coordinated by morphine via different opioid receptors to disinhibit pyramidal neurons and enhance reward.
-
Morphine activation of mu opioid receptors causes disinhibition of neurons in the ventral tegmental area mediated by β-arrestin2 and c-Src
Fiona A Bull,Daniel T Baptista-Hon,Jeremy J Lambert,Wendy Walwyn,Tim G Hales
DOI: https://doi.org/10.1038/s41598-017-10360-8
2017-08-30
Abstract:The tyrosine kinase, c-Src, participates in mu opioid receptor (MOP) mediated inhibition in sensory neurons in which β-arrestin2 (β-arr2) is implicated in its recruitment. Mice lacking β-arr2 exhibit increased sensitivity to morphine reinforcement; however, whether β-arr2 and/or c-Src participate in the actions of opioids in neurons within the reward pathway is unknown. It is also unclear whether morphine acts exclusively through MOPs, or involves delta opioid receptors (DOPs). We examined the involvement of MOPs, DOPs, β-arr2 and c-Src in the inhibition by morphine of GABAergic inhibitory postsynaptic currents (IPSCs) recorded from neurons in the mouse ventral tegmental area. Morphine inhibited spontaneous IPSC frequency, mainly through MOPs, with only a negligible effect remaining in MOP-/- neurons. However, a reduction in the inhibition by morphine for DOP-/- c.f. WT neurons and a DPDPE-induced decrease of IPSC frequency revealed a role for DOPs. The application of the c-Src inhibitor, PP2, to WT neurons also reduced inhibition by morphine, while the inactive PP3, and the MEK inhibitor, SL327, had no effect. Inhibition of IPSC frequency by morphine was also reduced in β-arr2-/- neurons in which PP2 caused no further reduction. These data suggest that inhibition of IPSCs by morphine involves a β-arr2/c-Src mediated mechanism.
-
Morphine modulates glutamate release in the hippocampal CA1 area in mice.
Ming Guo,Nan-Jie Xu,Yu-Ting Li,Jing-Yu Yang,Chun-Fu Wu,Gang Pei
DOI: https://doi.org/10.1016/j.neulet.2005.01.071
IF: 3.197
2005-01-01
Neuroscience Letters
Abstract:Opiate abuse is associated with long-lasting neural adaptative changes in the brain. Increasing evidence demonstrates that opiates significantly alter the function of the glutamatergic system, while how the system is regulated still remains elusive. In the present study, we studied the effect of morphine on extracellular glutamate concentration in the hippocampus, a nucleus rich of the glutamatergic neurons. The results showed that glutamate concentration in the extracellular fluid of the hippocampus was decreased following either acute or chronic treatment of morphine. However, naloxone-induced withdrawal increased glutamate concentration significantly. These results suggest an adaptation of the glutamatergic neuronal transmission in the hippocampus after morphine treatment.
-
Chronic Morphine Treatment Switches The Effect Of Dopamine On Excitatory Synaptic Transmission From Inhibition To Excitation In Pyramidal Cells Of The Basolateral Amygdala
Zicheng Li,Wenjie Luan,Yang Chen,Ming Chen,Yi Dong,Bin Lai,Lan Ma,Ping Zheng
DOI: https://doi.org/10.1523/JNEUROSCI.3806-11.2011
2011-01-01
Journal of Neuroscience
Abstract:Dopaminergic signaling in the basolateral amygdala (BLA) is important for drug-stimulus learning that triggers relapse to drug-seeking behavior. However, little is known about adaptive changes in this signaling pathway upon chronic morphine treatment. In this paper, we observed the influence of chronic morphine treatment on the effect of dopamine (DA) on the excitatory transmission in the pyramidal cells of BLA in slices with the whole-cell patch-clamp method. We also studied its mechanism and significance with pharmacological approaches combined with biochemical and behavioral techniques. The results showed that chronic morphine exposure switched the effect of DA on the excitatory synaptic transmission from inhibition to excitation; the chronic morphine-induced switching action on the effect of DA was due to its influence on D1 receptors; the site of the effect of chronic morphine treatment on D1 receptors was at presynaptic locus; chronic morphine treatment induced a significant increase in the amount of D1 receptor expression in the synaptosomes and synaptosomal membrane fraction from BLA; the enhancement of presynaptic glutamate release by D1 receptor agonist upon chronic morphine treatment was dependent on the activation of cAMP-dependent protein kinase; and the intra-BLA injection of D1 receptor antagonist canceled the conditioned place aversion (CPA) in morphine-dependent rats. In conclusion, chronic morphine treatment switches the effect of DA on the excitatory synaptic transmission from inhibition to excitation by the presynaptic D1 receptor amount increase-mediated glutamate release in the pyramidal cells of BLA and the blockade of D1 receptors in BLA cancels CPA in morphine-dependent rats.
-
The effect of morphine administration on GluN3B NMDA receptor subunit mRNA expression in rat brain
Shahrzad Nazari,Mitra-Sadat Sadat-Shirazi,Ali Shahbazi,Kamran Ghaffari,Nasim Vousooghi,Mitra‑Sadat Sadat‑Shirazi
DOI: https://doi.org/10.55782/ane-2024-2545
2024-03-26
Acta Neurobiologiae Experimentalis
Abstract:Opioid addiction is critically dependent on the activation of N‐methyl‐D‐aspartate (NMDA) receptors, which are widely found in the mesocorticolimbic system. Meanwhile, opioid addiction may affect the expression level of NMDA receptor subunits. The existence of GluN3 subunits in the NMDA receptor’s tetramer structure reduces the excitatory current of the receptor channel. We evaluated the changes in the mRNA expression pattern of the GluN3B subunit of the NMDA receptor in rat brains following acute and chronic exposure to morphine. Chronic, escalating intraperitoneal doses of morphine or saline were administered twice daily to male Wistar rats for six days. Two other groups were injected with a single acute dose of morphine or saline. The mRNA level of the GluN3B subunit of the NMDA receptor in the striatum, hippocampus, and nucleus accumbens (NAc) was measured by real‐time PCR. mRNA expression of the GluN3B subunit was considerably augmented (3.15 fold) in the NAc of animals chronically treated with morphine compared to the control group. The difference between rats that were chronically administered morphine and control rats was not statistically significant for other evaluated brain areas. In rats acutely treated with morphine, no significant differences were found for GluN3B subunit expression in the examined brain regions compared to the control group. It was concluded that chronic exposure to morphine notably increased the GluN3B subunit of the NMDA receptor in NAc. The extent of the impact of this finding on opioid addiction and its features requires further evaluation in future studies.
neurosciences
-
Morphine inhibits acid-sensing ion channel currents in rat dorsal root ganglion neurons
Qi Cai,Chun-Yu Qiu,Fang Qiu,Ting-Ting Liu,Zu-Wei Qu,Yu-Min Liu,Wang-Ping Hu
DOI: https://doi.org/10.1016/j.brainres.2014.01.042
IF: 3.61
2014-01-01
Brain Research
Abstract:Extracellular acidosis is a common feature in pain-generating pathological conditions. Acid-sensing ion channels (ASICs), pH sensors, are distributed in peripheral sensory neurons and participate in nociception. Morphine exerts potent analgesic effects through the activation of opioid receptors for various pain conditions. A cross-talk between ASICs and opioid receptors in peripheral sensory neurons has not been shown so far. Here, we have found that morphine inhibits the activity of native ASICs in rat dorsal root ganglion (DRG) neurons. Morphine dose-dependently inhibited proton-gated currents mediated by ASICs in the presence of the TRPV1 inhibitor capsazepine. Morphine shifted the proton concentration–response curve downwards, with a decrease of 51.4±3.8% in the maximum current response but with no significant change in the pH0.5 value. Another μ-opioid receptor agonist DAMGO induced a similar decrease in ASIC currents compared with morphine. The morphine inhibition of ASIC currents was blocked by naloxone, a specific opioid receptor antagonist. Pretreatment of forskolin, an adenylyl cyclase activator, or the addition of cAMP reversed the inhibitory effect of morphine. Moreover, morphine altered acid-evoked excitability of rat DRG neurons and decreased the number of action potentials induced by acid stimuli. Finally, peripheral applied morphine relieved pain evoked by intraplantar of acetic acid in rats. Our results indicate that morphine can inhibit the activity of ASICs via μ-opioid receptor and cAMP dependent signal pathway. These observations demonstrate a cross-talk between ASICs and opioid receptors in peripheral sensory neurons, which was a novel analgesic mechanism of morphine.
-
Contribution of adrenomedullin to the switch of G protein-coupled μ-opioid receptors from Gi to Gs in the spinal dorsal horn following chronic morphine exposure in rats
Dongmei Wang,Juan Zeng,Qi Li,Jianzhong Huang,Réjean Couture,Yanguo Hong
DOI: https://doi.org/10.1111/bph.13419
Abstract:Background and purpose: Chronic exposure to morphine increases spinal adrenomedullin (AM) bioactivity resulting in the development and maintenance of morphine tolerance. This study investigated the possible involvement of AM in morphine-evoked alteration in μ-opioid receptor-coupled G proteins. Experimental approach: Agents were administered intrathecally (i.t.) in rats. Nociceptive behaviours and cumulative dose-response of morphine analgesia were assessed. Neurochemicals in the spinal dorsal horn were assayed by immunoprecipitation, Western blot analysis and ELISA. Key results: Intrathecal injection of AM (8 μg) for 9 days decreased and increased the levels of μ receptor-coupled Gi and Gs proteins respectively. Morphine stimulation (5 μg) after chronic treatment with AM also induced an increase in cAMP production in the spinal dorsal horn. Co-administration of the selective AM receptor antagonist AM22-52 inhibited chronic morphine-evoked switch of G protein-coupled μ receptor from Gi to Gs. Chronic exposure to AM increased the phosphorylation of cAMP-responsive element-binding protein (CREB) and ERK. Co-administration of the PKA inhibitor H-89 (5 μg) or MEK1 inhibitor PD98059 (1 μg) reversed the AM-induced thermal/mechanical hypersensitivity, decline in morphine analgesic potency, switch of G protein-coupled μ receptor and increase in cAMP. Conclusions and implications: The present study supports the hypothesis that an increase in AM activity in the spinal dorsal horn contributes to the switch of the μ receptor-coupled G protein from Gi to Gs protein via the activation of cAMP/PKA/CREB and ERK signalling pathways in chronic morphine use.
-
Nmda Receptors in the Midbrain Play A Critical Role in Dopamine-Mediated Hippocampal Synaptic Potentiation Caused by Morphine
Ling Hu,Xiang-Hong Jing,Cai-Lian Cui,Guo-Gang Xing,Bing Zhu
DOI: https://doi.org/10.1111/adb.12010
2012-01-01
Addiction Biology
Abstract:A single exposure to drugs of abuse produces an NMDAR (N-methyl-D-aspartate receptor)-dependent synaptic potentiation at excitatory synapses of dopamine (DA) neurons in the ventral tegmental area (VTA) of the midbrain. All addictive drugs can increase DA concentrations in projection areas of the midbrain, including the hippocampus. Hippocampal DA release subsequently modulates hippocampal plasticity and drug-associated memories. Using in vivo electrophysiological recording techniques in anesthetized rats, we show that systemic injection of morphine induced hippocampal synaptic potentiation in a dose-dependent manner. Intra-VTA but not intra-hippocampus injection of morphine evoked this potentiation. Local hippocampal dopamine D1 receptors (D1R) are required in the morphine-induced synaptic potentiation and conditioned place preference (CPP). Moreover, both NMDAR activation in the VTA and VTA/hippocampus dopaminergic connections are essential for the morphine-evoked potentiation and CPP. These findings suggest that NMDAR signalings in the midbrain play a key role in regulating dopamine-mediated hippocampal synaptic plasticity underlying drug-induced associative memory.