Abstract
Opioid-induced hyperalgesia and analgesic tolerance can lead to dose escalation and inadequate pain treatment with μ-opioid receptor agonists. Opioids cause tonic activation of glutamate NMDA receptors (NMDARs) at primary afferent terminals, increasing nociceptive input. However, the signaling mechanisms responsible for opioid-induced activation of presynaptic NMDARs in the spinal dorsal horn remain unclear. In the present study, we determined the role of mitogen-activated protein kinase (MAPK) signaling in opioid-induced presynaptic NMDAR activation caused by chronic morphine administration. Whole-cell recordings of excitatory postsynaptic currents (EPSCs) were performed on dorsal horn neurons in rat spinal cord slices. Chronic morphine administration markedly increased the frequency of miniature EPSCs, increased the amplitude of monosynaptic EPSCs evoked from the dorsal root, and reduced the paired-pulse ratio of evoked EPSCs. These changes were fully reversed by an NMDAR antagonist and normalized by inhibiting extracellular signal-regulated kinase 1/2 (ERK1/2), p38, or c-Jun N-terminal kinase (JNK). Furthermore, intrathecal injection of a selective ERK1/2, p38, or JNK inhibitor blocked pain hypersensitivity induced by chronic morphine treatment. These inhibitors also similarly attenuated a reduction in morphine’s analgesic effect in rats. In addition, co-immunoprecipitation assays revealed that NMDARs formed a protein complex with ERK1/2, p38, and JNK in the spinal cord and that chronic morphine treatment increased physical interactions of NMDARs with these three MAPKs. Our findings suggest that opioid-induced hyperalgesia and analgesic tolerance are mediated by tonic activation of presynaptic NMDARs via three functionally interrelated MAPKs at the spinal cord level.
Keywords: opioid tolerance, NMDA receptor, synaptic plasticity, spinal cord, μ-opioid receptor, G protein-coupled receptor, primary afferent nerves
Graphical Abstract
Under the normal condition, NMDA receptors at the presynaptic terminals in the spinal cord are not phosphorylated or functionally active. However, presynaptic NMDA receptors in the spinal dorsal horn become tonically activated during chronic morphine treatment. Here we found that chronic treatment with morphine increases the association of NMDA receptors with three MAPKs (ERK1/2, p38, and JNK) in the spinal cord. This increased interaction leads to NMDA receptor phosphorylation at presynaptic terminals. As a result, presynaptic NMDARs are endogenously activated to augment synaptic glutamate release to postsynaptic neurons in the spinal dorsal horn to cause opioid-induced hyperalgesia and analgesic tolerance.
Introduction
Opioid analgesics are the mainstay of treatment of severe pain caused by cancer or tissue and nerve injury. However, repeated use of μ-opioid receptor (MOR) agonists causes loss of analgesic efficacy and opioid dose escalation, which contributes to the current epidemic of opioid abuse in the United States. Also, acute and chronic opioid use can paradoxically increase pain sensitivity, termed opioid-induced hyperalgesia, in both animals and humans (Celerier et al. 2000, Chia et al. 1999, Chen et al. 2007, Mao et al. 1994). Repeated morphine injections can progressively reduce the nociceptive threshold, which occurs in parallel with development of analgesic tolerance (Chen et al. 2007, Mao et al. 1994, Zhao et al. 2012). Concurrent development of hyperalgesia associated with opioid use can be perceived as the diminished opioid analgesic effect (i.e., analgesic tolerance). Furthermore, intrathecal administration of glutamate N-methyl-D-aspartate receptor (NMDAR) antagonists blocks both hyperalgesia and analgesic tolerance induced by chronic morphine use (Mao et al. 1994, Zhao et al. 2012), suggesting that increased NMDAR activity at the spinal cord level is essential for opioid-induced hyperalgesia and tolerance. Nevertheless, chronic morphine treatment reduces the activity of postsynaptic NMDARs in spinal dorsal horn neurons (Zhao et al. 2012).
NMDARs are also expressed at primary afferent terminals in the spinal dorsal horn (Liu et al. 1994). However, these NMDARs are not functionally active during the normal condition. After acute or chronic opioid exposure, presynaptic NMDARs become tonically activated, increasing glutamatergic input to spinal dorsal horn neurons (Zhao et al. 2012, Zhou et al. 2010, Zeng et al. 2006). Hence, tonic activation of presynaptic NMDARs can increase nociceptive input from primary afferent nerves to spinal dorsal horn neurons, resulting in hyperalgesia and opioid tolerance (Zhou et al. 2010, Zhao et al. 2012). However, how MOR stimulation leads to activation of presynaptic NMDARs in the spinal dorsal horn remains unclear.
Following MOR activation, MORs become phosphorylated by G protein receptor kinase, which initiates a β-arrestin—dependent desensitization process to terminate the MOR signaling (Bohn et al. 2004, Keith et al. 1996). Furthermore, β-arrestin may act as a scaffolding protein and promote the stable association of signaling proteins with MORs to activate the mitogen-activated protein kinase (MAPK) pathways (McDonald et al. 2000, Luttrell et al. 2001). Mice lacking β-arrestin 2 exhibit prolonged opioid analgesic effects and reduced tolerance to morphine (Bohn et al. 1999), suggesting that arrestin/MAPK signaling contributes to opioid tolerance development. The three major MAPKs, extracellular signal-regulated kinases 1/2 (ERK1/2), p38, and c-Jun N-terminal kinase (JNK), are activated in primary sensory neurons by nociceptive stimuli (Mizushima et al. 2005, Modol et al. 2015, Obata & Noguchi 2004). Although opioid-induced MAPK signaling is well documented (Belcheva et al. 2001, Fukuda et al. 1996), there is currently no evidence showing whether MAPK signaling contributes to opioid-induced activation of presynaptic NMDARs at the spinal cord level.
In the present study, we used a spinal cord slice preparation that has native MORs and preserves the intrinsic connection between primary afferents and dorsal horn neurons to define the role of ERK1/2, p38, and JNK in chronic morphine treatment-induced tonic activation of presynaptic NMDARs. We provide the first in vivo evidence that chronic morphine treatment potentiates the physical interaction between the NMDAR and ERK1/2, p38, and JNK in the spinal cord, leading to increased activity of presynaptic NMDARs at primary afferent terminals. This new information extends our understanding of signaling mechanisms involved in opioid-induced hyperalgesia and analgesic tolerance.
Materials and Methods
Animal Model and Morphine Treatment —
Adult male Sprague-Dawley rats (280–320 g; Harlan, Indianapolis, IN; RRID: RGD_5508397) were used in our experiments. All procedures and protocols were approved by The University of Texas MD Anderson Cancer Center (approval #882-RN01) and were performed in accordance with the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The timeline of experimental procedures and # of animals used for each protocol was specified in Fig. 1. To induce opioid hyperalgesia and analgesic tolerance, morphine sulfate (West Ward Pharmaceuticals, Eatontown, NJ) was intraperitoneally injected into the rats at 5 mg/kg twice a day for 8 consecutive days (Zhao et al. 2012, Chen et al. 2007).
Intrathecal Catheterization and Drug Delivery —
A polyethylene PE10 catheter was implanted in the lumbar subarachnoid space in each rat under isoflurane-induced anesthesia according to the method described previously (Chen & Pan 2006a). Briefly, each animal was placed prone on a stereotaxic frame, and a small incision was made at the back of the neck. The catheter was inserted after a small puncture was made in the atlanto-occipital membrane of the cisterna magna. The caudal tip of the catheter was then advanced and reached the lumbar enlargement of the spinal cord. To minimize animal’s suffering, 1% lidocaine was injected around the incision site during the brief surgery. The animals were allowed to recover for 7 days before intrathecal injection, and no other medications were administered. Rats exhibiting neurological deficits (e.g., paralysis) or poor grooming were promptly killed via CO2 asphyxiation. In 43 rats instrumented with intrathecal catheters, 4 rats were killed due to motor weakness and impairment. U0126 (5 μg; EMD-Calbiochem, La Jolla, CA), 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)-1H-imidazole (SB203580, 5 μg; EMD-Calbiochem), and SP600125 (30 μg; Cayman Chemical, Ann Arbor, MI) were dissolved in DMSO and intrathecally injected 20 min before morphine administration on each testing day (days 1–8).
Spinal Cord Slice Preparation and Electrophysiological Recordings of Dorsal Horn Neurons —
The lumbar spinal cords were rapidly removed from the rats via laminectomy under isoflurane-induced anesthesia. Each spinal cord was immediately placed in ice-cold sucrose artificial cerebrospinal fluid containing (in mM) 3.6 KCl, 1.2 MgCl2, 2.5 CaCl2, 1.2 NaH2PO4, 25.0 NaHCO3, 234 sucrose, and 12.0 glucose presaturated with 95% O2 and 5% CO2. The spinal cord was then placed in a shallow groove formed in an agar block and glued onto the stage of a vibratome (Leica, Wetzlar, Germany). Transverse slices of the spinal cords (400 μm thick) were cut in ice-cold sucrose artificial cerebrospinal fluid and preincubated in Krebs solution containing (in mM) 3.6 KCl, 1.2 MgCl2, 117.0 NaCl, 2.5 CaCl2, 1.2 NaH2PO4, 11.0 glucose, and 25.0 NaHCO3 oxygenated with 95% O2 and 5% CO2 at 34 °C for at least 1 h before being transferred to a recording chamber. The spinal cord slice was placed in an incubation chamber and continuously perfused with Krebs solution at 5 ml/min and 34 °C maintained by an inline solution heater and a temperature controller.
The spinal cord lamina II, a translucent region in the superficial dorsal horn, was identified in each slice under an upright fixed-stage microscope (BX50WI; Olympus, Tokyo, Japan) with differential interference contrast/infrared illumination. Neurons in the lamina II outer zone were visualized and selected for whole-cell recordings because they are important for nociceptive transmission (Pan & Pan 2004, Wang et al. 2018). A previous study shows that most lamina II neurons are glutamate-releasing excitatory interneurons (Santos et al. 2007), and these neurons form networks that play a critical role in transmitting nociceptive information from the primary afferents. The impedance of the glass electrode was 4–7 MΩ when a pipette was filled with an internal solution containing (in mM) 135 potassium gluconate, 0.5 CaCl2, 5 KCl, 2.0 MgCl2, 5.0 HEPES, 5.0 ATP-Mg, 0.5 Na-GTP, 5.0 EGTA, and 10 QX314 (adjusted to pH 7.25 with 1.0 M KOH, 280–300 mOsm).
Excitatory postsynaptic currents (EPSCs) were recorded at a holding potential of −60 mV using whole-cell voltage-clamp techniques. The input resistance was monitored, and the recording was abandoned if it changed by more than 15%. EPSCs were recorded using an amplifier (MultiClamp 700A, Axon Instruments, Foster City, CA), filtered at 1–2 kHz, and digitized at 10 kHz. Miniature EPSCs (mEPSCs) were recorded in the presence of 1 μM tetrodotoxin (Chen et al. 2014, Zhou et al. 2010). Because our whole-cell recording was done at a holding potential of −60 mV and in the presence of 1.2 mM Mg2+ (in the Krebs solution), the postsynaptic NMDARs were largely closed. To evoke glutamate release from primary afferent nerves, a bipolar tungsten electrode connected to a stimulator (0.2 ms, 0.3–0.6 mA, 0.1 Hz; Grass Instruments, Quincy, MA) was used to electrically stimulate the dorsal root, and monosynaptic EPSCs were identified on the basis of the constant latency and absence of conduction failure of evoked EPSCs in response to 20-Hz electrical stimulation (Zhou et al. 2010, Xie et al. 2017b). To calculate the paired-pulse ratio (PPR), two EPSCs were evoked by a pair of stimuli at 50-ms intervals. The PPR was expressed as the ratio of the amplitude of the second synaptic response to the amplitude of the first synaptic response (Xie et al. 2017b, Zhao et al. 2012). All the drugs were prepared in artificial cerebrospinal fluid immediately before the experiments and delivered via syringe pumps to reach their final concentrations. AP5 and tetrodotoxin were purchased from Hello Bio (Princeton, NJ).
Behavioral Assessment of Nociception —
To quantify the mechanical nociceptive threshold in the rats, a paw pressure test on a hindpaw was conducted using an analgesiometer (Ugo Basile, Varese, Italy). The device was activated by pressing a foot pedal to generate a constantly increasing force. When the animal displayed pain by either withdrawing its paw or vocalizing, the device was immediately inactivated, and the animal’s withdrawal threshold was read on the scale (Chen et al. 2007, Zhao et al. 2012). A maximum of 400 g of pressure was used as a cutoff to avoid tissue injury to the rats.
To assess thermal sensitivity, rats were placed on the glass surface of a thermal testing apparatus (IITC Life Science, Woodland Hills, CA) and allowed to acclimate for 30 min before testing. The temperature of the glass surface was maintained constant at 30 °C. A mobile radiant heat source located under the glass was focused onto a hindpaw of each rat. The paw withdrawal latency was recorded using a timer, and the hindpaw was tested twice to obtain the average. A cut-off of 30 s was set to prevent tissue damage (Li et al. 2016, Chen & Pan 2006b).
Co-immunoprecipitation —
After treatment with morphine and vehicle for 8 days, the dorsal quadrants of lumbar spinal cord tissues were quickly removed from rats anesthetized with 3% isoflurane. The spinal tissues were dissected and homogenized in ice-cold immunoprecipitation buffer (50 mM Tris [pH7.4], 250 mM NaCl, 20 mM NaF, 1 mM Na3VO4, 10% glycerol, 0.5% NP-40, 10 mM N-ethylmaleimide, 1 mM phenylmethylsulfonyl fluoride, and 2 mM benzamide) containing cocktails of protease and phosphatase inhibitors (Sigma-Aldrich, St. Louis, MO). Samples were then put on ice for 30 min with constant shaking. Lysates were centrifuged at 13,000 × g for 30 min at 4 °C. The supernatant was carefully collected, and the protein concentration was measured using a DC Protein Assay Kit (Bio-Rad, Hercules, CA). The soluble fraction was incubated at 4 °C overnight with Protein A/G beads (#16–266, Millipore, Darmstadt, Germany) prebound to a mouse anti-GluN1 antibody (#75–272, 1:1,000, NeuroMab, Davis, CA; RRID: AB_11000180). Protein A/G beads prebound to mouse IgG were used as controls. Samples were washed 3 times with an IP buffer and then immunoblotted. The following antibodies were selected for immunoblotting: rabbit anti-GluN1 (#G-8913, 1:1,000, Sigma-Aldrich; RRID: AB_259978), rabbit anti-ERK1/2 (#4695, 1:1,000, Cell Signaling Technology, Danvers, MA; RRID: AB_390779), rabbit anti-p38 (#8690, 1:1,000, Cell Signaling Technology; RRID: AB_10999090), and rabbit anti-SAPK/JNK (#9252, 1:1,000, Cell Signaling Technology; RRID:AB_2250373). The specificity of these antibodies was validated in previous studies (Rudhard et al. 2003, Brady et al. 2017, Li et al. 2017, Sun et al. 2017). For immunoblots, lysates were separated on a 4–12% gradient SDS-polyacrylamide gel and transferred to a polyvinylidene fluoride membrane. The membrane was blocked by 5% blotting-grade buffer (#170–6404, Bio-Rad, Hercules, CA). After the antibody incubation, the membrane was washed 3 times with Tris-buffered saline with Tween 20 (#9997S, Cell Signaling Technology). An ECL kit (#PI34580, Fisher Scientific, Lenexa, KS) was used to detect the protein band, which was visualized and quantified using an Odyssey Fc Imager (LI-COR Biotechnology, Lincoln, NE).
Study Design and Data Analysis —
Data were expressed as means ± SEM. No statistical methods were used to predetermine sample sizes for the studies, but our sample sizes were similar to those generally employed in the field. The animals were assigned (1:1 allocation) to the control and treatment groups, and no randomization methods were used. We did not exclude any animals from data analysis and did not use any test for outliers on the data. The investigators performing electrophysiological and behavioral experiments were blinded to drug treatment. In electrophysiological experiments, only one neuron was recorded in each tissue slice, and at least three rats were used in each group. The amplitude of 10 consecutive evoked EPSCs was analyzed and averaged using the Clampfit 10.0 software program (Axon Instruments). mEPSCs were analyzed off-line using the MiniAnalysis peak detection program (Synaptosoft, Leonia, NJ). D’Agostino-Pearson normality test was used to assess the normality of data. The Student’s t test was used to compare two groups, and one-way analysis of variance followed by Dunnett’s post hoc test was used to compare more than two groups. Two-way analysis of variance followed by Bonferroni’s post hoc test was used to determine the difference in development of hyperalgesia and opioid tolerance between the vehicle- and morphine-treated rats. Statistical analyses were performed using Prism software (version 7, GraphPad Software Inc., La Jolla, CA). P <0.05 was considered to be statistically significant.
Results
ERK1/2 Mediates Chronic Morphine-induced Increases in Presynaptic NMDAR Activity in the Spinal Cord —
We first determined whether ERK1/2 plays a role in increased presynaptic NMDAR activity in the spinal cord induced by chronic morphine treatment. Presynaptic ERK signaling plays a role in synaptic plasticity in the hippocampus (Vara et al. 2009), and presynaptic ERK1/2 is important for the control of central glutamatergic synapses (Sindreu et al. 2011). We recorded miniature excitatory postsynaptic currents (mEPSCs) in spinal cord lamina II neurons, which reflect quantal glutamate release from presynaptic terminals (Xie et al. 2017a, Chen et al. 2014). Bath application of 50 μM AP5, a specific NMDAR antagonist, rapidly normalized the increased frequency of mEPSCs in lamina II neurons obtained from morphine-treated rats (baseline, 6.45 ± 0.60 Hz; AP5, 4.17 ± 0.45 Hz; n = 11 neurons, Fig. 2, A and B). Bath application of AP5 does not affect the frequency or amplitude of mEPSCs in lamina II neurons from control rats (Zhao et al. 2012, Zhou et al. 2010). These results suggest that chronic morphine potentiates presynaptic NMDAR activity of spinal dorsal horn neurons, a finding similar to that in our previous study (Zhao et al. 2012). Also, U0126 is a highly selective ERK1/2 inhibitor used to study ERK1/2-mediated signaling (Favata et al. 1998, Arana-Argaez et al. 2010). Pretreatment with U0126 (10 μM for 1 h) significantly reduced the baseline frequency (3.00 ± 0.26 Hz vs. 6.45 ± 0.60 Hz), but not the amplitude, of mEPSCs in lamina II neurons in spinal cord slices obtained from morphine treated rats (n = 10 neurons, 6.45 ± 0.60 Hz vs. 3.00 ± 0.26 Hz, P < 0.05; Fig. 2, B and D). In these 10 neurons, subsequent application of 50 μM AP5 had no effect on the frequency or amplitude of mEPSCs (Fig. 2, C and D).
The synapse formed by central terminals of primary afferent neurons and spinal dorsal horn neurons is essential for nociceptive transmission and modulation by opioids (Chen & Pan 2006a, Zhou et al. 2010, Zhou et al. 2008). We thus determined whether ERK1/2 is involved in morphine-induced increases in the activity of NMDARs expressed at primary afferent nerve terminals in the spinal dorsal horn. Monosynaptic EPSCs of spinal dorsal horn neurons were evoked from the dorsal root, which represents elicited glutamate released from primary afferent terminals (Xie et al. 2017b, Zhou et al. 2010). Bath application of 50 μM AP5 considerably reduced the amplitude of monosynaptic EPSCs (363.7 ± 21.6 pA vs. 441.4 ± 23.9 pA) and increased the paired-pulse ratio (PPR, 0.76 ± 0.06 vs. 0.65 ± 0.05) of monosynaptically evoked EPSCs of lamina II neurons obtained from morphine-treated rats (n = 10 neurons, Fig. 3, A – C). These data are consistent with those in our previous study demonstrating opioid-induced tonic activation of NMDARs at primary afferent terminals (Zhao et al. 2012). After pretreatment of lamina II neurons from morphine-treated rats with U0126 (10 μM for 1 h), bath application of 50 μM AP5 no longer had an effect on the amplitude (n = 10 neurons) or PPR of monosynaptic EPSCs (n = 9 neurons, Fig. 3, D and F). Taken together, these data indicate that ERK1/2 plays a critical role in opioid-induced tonic activation of presynaptic NMDARs in the spinal cord.
P38 Is Involved in Morphine-induced Presynaptic NMDAR Activation in the Spinal Cord —
We next determined whether the p38 MAPK is involved in chronic morphine-induced increases in presynaptic NMDAR activity in the spinal cord. p38 is expressed presynaptically (Nakata et al. 2005), and SB203580 is a pyridinyl imidazole inhibitor commonly used to study p38 activity in vitro and in vivo (Arana-Argaez et al. 2010, Jin et al. 2013). Pretreatment with SB203580 (5 μM for 1 h) caused a large reduction in the baseline frequency of mEPSCs in spinal lamina II neurons obtained from morphine-treated rats (3.63 ± 0.41 Hz vs. 6.45 ± 0.60 Hz; n = 10 neurons, P < 0.05; Fig. 2B and Fig. 4B). In these neurons, further treatment with AP5 failed to affect the frequency or amplitude of mEPSCs (n = 10 neurons, Fig. 4, A and B).
We also examined whether p38 inhibition affects morphine-induced activation of NMDARs at primary afferent terminals. After treatment of spinal lamina II neurons obtained from morphine-treated rats with SB203580 (5 μM for 1 h), subsequent bath application of 50 μM AP5 had no effect on the amplitude of evoked monosynaptic EPSCs (n = 10 neurons, Fig. 4, C and E) or the PPR of monosynaptic EPSCs (n = 9 neurons, Fig. 4, D and E). These results suggest that p38 mediates opioid-induced tonic activation of NMDARs at primary afferent terminals.
JNK Participates in Morphine-induced Increases in Presynaptic NMDAR Activity in the Spinal Cord —
We then examined whether JNK is involved in increased presynaptic NMDAR activity in the spinal dorsal horn in morphine-treated rats. JNK2 is expressed presynaptically and plays a role in NMDAR-mediated glutamate release in the cortex (Nistico et al. 2015). SP600125 is an anthrapyrazolone capable of inhibiting JNK1, JNK2, and JNK3 with high affinity (Kocab et al. 2015, Bennett et al. 2001). In lamina II neurons obtained from morphine-treated rats, pretreatment with SP600125 (40 μM for 1 h) significantly reduced the frequency (4.40 ± 0.44 Hz vs .6.45 ± 0.60 Hz), but not the amplitude, of mEPSCs (n = 10 neurons, Fig. 5). In the same neurons, subsequent bath application of AP5 did not significantly affect the frequency or amplitude of mEPSCs (Fig. 5, A and B).
In addition, after treatment of lamina II neurons from morphine-treated rats with SP600125, bath application of AP5 (50 μM) did not have a significant effect on the amplitude of monosynaptically evoked EPSCs (n = 10 neurons, Fig. 5, C and E) or the PPR of monosynaptic EPSCs (n = 9 neurons, Fig. 5, D and E). These findings suggest that increased JNK activity critically contributes to opioid-induced tonic activation of presynaptic NMDARs in the spinal dorsal horn.
MAPK Signaling at the Spinal Cord Level Mediates the Development of Opioid-induced Hyperalgesia and Tolerance —
The electrophysiological results in this study suggest that ERK1/2, p38, and JNK are all involved in opioid-induced tonic activation of presynaptic NMDARs, which can increase nociceptive input to spinal dorsal horn neurons. To determine whether these three MAPKs are similarly involved in opioid-induced hyperalgesia and tolerance, we determined the effects of intrathecal administration of individual MAPK inhibitors on hyperalgesia and tolerance caused by chronic morphine administration. We induced morphine analgesic tolerance in the rats via daily intraperitoneal injection of morphine (5 mg/kg twice a day) for 8 consecutive days (Chen et al. 2007, Zhao et al. 2012). We also intrathecally injected 5 μg of U0126, 5 μg of SB203580, 30 μg of SP600125, or a vehicle control (in a volume of 10 μl) 20 min before each morphine administration in separate groups of rats. The intrathecal doses of MAPK inhibitors were derived from previous studies (Chen et al. 2008, Daulhac et al. 2006, Tsai et al. 2009). We measured their withdrawal thresholds in response to noxious pressure and thermal stimuli 30 min before (baseline) and 30 min after morphine injection daily. In vehicle-treated rats, daily morphine administration caused a gradual reduction in the baseline pressure and thermal withdrawal thresholds, indicating the presence of mechanical and thermal hyperalgesia (n = 9 rats, Fig. 6, A and B). In contrast, treatment with U0126 (n = 7 rats), SB203580 (n = 7 rats), or SP600125 (n = 8 rats) blocked the decrease in the baseline withdrawal thresholds induced by chronic morphine administration (Fig. 6, A and B). Furthermore, when 5 μg of U0126, 5 μg of SB203580, and 30 μg of SP600125 were combined for intrathecal injection, no additive effect on morphine-induced mechanical and thermal hyperalgesia was observed compared to a single MAPK inhibitor (n = 8 rats, Fig. 6, A and B).
Also, in vehicle-treated rats, intraperitoneal injection of 5 mg/kg morphine on day 1 produced robust increases in the pressure and thermal withdrawal thresholds. Daily morphine administration caused gradual attenuation of the analgesic effect of morphine. By day 6, morphine produced no analgesic effect, indicating the development of analgesic tolerance (n = 9 rats, Fig. 6, C and D). In comparison, treatment with U0126 (n = 7 rats), SB203580 (n = 7 rats), or SP600125 (n = 8 rats) largely attenuated the reduction in the analgesic effect of morphine (Fig. 6, C and D). Even by day 8, intraperitoneal injection of 5 mg/kg morphine still markedly increased the mechanical and thermal withdrawal thresholds in rats given any one of the three MAPK inhibitors. Additionally, when U0126, SB203580, and SP600125 were combined for intrathecal injection, no additive on the development of morphine-induced analgesic tolerance effect was detected compared to a single MAPK inhibitor (n = 8 rats, Fig. 6, C and D). These data suggest that increased activity of ERK1/2, p38, and JNK at the spinal level is required for the development of opioid-induced hyperalgesia and analgesic tolerance.
Chronic Morphine Treatment Increases the Physical Interaction between MAPKs and NMDARs in the Spinal Cord —
Because our electrophysiological and behavioral data unexpectedly showed that ERK1/2, p38, and JNK are similarly involved in opioid-induced presynaptic NMDAR activation, hyperalgesia, and analgesic tolerance, we reasoned whether chronic morphine treatment promotes the association of these three functionally redundant MAPKs with NMDARs at the spinal cord level. To determine the interaction between the NMDAR and ERK1/2, p38, and JNK in vivo, we conducted co-immunoprecipitation (co-IP) assays using total protein extracts of rat dorsal spinal cords obtained from vehicle- and chronic morphine-treated rats. We used an anti-GluN1 antibody for co-IP assays because GluN1 is an obligatory subunit of NMDARs. Co-IP analysis showed that the anti-GluN1 antibody, but not an irrelevant anti-immunoglobulin G (anti-IgG) antibody, precipitated ERK1/2, p38, and JNK proteins in spinal cord protein extracts (Fig. 7, A and B). Furthermore, co-IP analysis demonstrated that the anti-GluN1 antibody precipitated considerably more ERK1/2, p38, and JNK proteins in the spinal protein extracts from chronic morphine-treated than from vehicle-treated rats (P < 0.05, n = 6 rats per group; Fig. 7, A and B). However, the total protein amounts (i.e., input controls) for GluN1, ERK1/2, p38, and JNK in the spinal cord did not differ significantly between the vehicle-treated and morphine-treated groups (Fig. 7, A and B). These results indicate that NMDARs form a large protein complex with ERK1/2, p38, and JNK at the spinal cord level and that chronic opioid treatment promotes the association of NMDARs with the three MAPKs.
Discussion
Our current understanding of the dynamic opioid signaling mechanism remains fragmented, and the in vitro opioid signaling is often not directly linked with the in vivo physiological or pharmacological effects of opioids. MORs at the spinal cord level are essential for the analgesic effect of systemically administered opioids (Chen & Pan 2006a). Activation of MORs typically causes G protein-mediated inhibition of voltage-activated calcium channels, resulting in attenuation of nociceptive glutamatergic transmission in the spinal dorsal horn, producing an analgesic effect (Wu et al. 2004, Zhang et al. 2016). On the other hand, β-arrestin-mediated signaling is involved in rapid attenuation of opioid analgesic actions and opioid tolerance (Pierce & Lefkowitz 2001, Bohn et al. 1999). Arrestin may function as MAPK scaffolds and facilitate activation of MAPKs (Lefkowitz & Shenoy 2005, Song et al. 2009). ERK1/2, p38, and JNK are the most extensively studied MAPKs. In general, ERK1/2 is preferentially stimulated in response to exposure to growth factors and phorbol esters, whereas JNK and p38α/β are more responsive to stress stimuli, including osmotic shock and cytokine stimulation (Pearson et al. 2001). It has been recognized that phosphorylated arrestin-bound MOR complexes not only inactivate MOR’s effects but also recruit MAPK signaling (Lefkowitz & Shenoy 2005). In the present study, we showed that inhibiting the activity of ERK1/2, p38, or JNK at the spinal cord level not only preserved the analgesic effect of morphine but also abolished morphine-induced hyperalgesia. Consistent with our results, morphine tolerance is blocked in JNK2-knockout mice (Melief et al. 2010). Our findings suggest that ERK1/2, p38, and JNK play a critical role in the development of opioid-induced hyperalgesia and analgesic tolerance.
A striking finding of our study is that increased activity of ERK1/2, p38, and JNK is required for morphine-induced tonic activation of presynaptic NMDARs in the spinal cord. NMDARs at the spinal cord level play an essential role in both the development and maintenance of opioid-induced hyperalgesia and tolerance (Mao et al. 1994, Trujillo & Akil 1991, Zhao et al. 2012). We showed that inhibiting ERK1/2, p38, or JNK activity completely blocked opioid-induced activation of presynaptic NMDARs in the spinal dorsal horn. We previously reported that even brief stimulation of MORs can trigger long-lasting NMDAR-mediated glutamate release from primary afferent terminals in the spinal cord (Zhou et al. 2010). Chronic opioid treatment can sustain activation of presynaptic NMDARs to increase nociceptive input to the spinal dorsal horn, thereby producing prolonged hyperalgesia and opposing the opioid’s analgesic effects (i.e., causing tolerance). Notably, chronic morphine treatment increases the phosphorylation of ERK1/2, p38, and JNK in primary sensory neurons (Ma et al. 2001, Chen et al. 2008). Also, ERK activation can stimulate NMDAR activity in the brain (Nateri et al. 2007), and presynaptic JNK2 regulates NMDAR-dependent glutamate release in the brain (Nistico et al. 2015). Both GluN2A- or GluN2B-containing NMDARs are involved in the increased presynaptic NMDAR activity by chronic morphine treatment (Zhao et al. 2012). The increased presynaptic NMDAR activity after chronic morphine exposure is abolished by inhibition of PKC and MAPKs, suggesting that increased NMDAR phosphorylation may enhance presynaptic NMDAR function by promoting NMDAR trafficking and surface expression and by reducing the Mg2+ block (Zhao et al. 2012, Lan et al. 2001, Chen & Huang 1992, Chen et al. 2018). The sources of endogenous glutamate and glycine that cause tonic activation of presynaptic NMDARs during chronic morphine treatment are not clear. Glutamate may originate from the same primary afferent nerve terminals on which the NMDARs are expressed or from excitatory interneurons in the superficial dorsal horn (Wang et al. 2018). Also, glutamate and glycine may diffuse from an adjacent active terminal or reduced glutamate/glycine reuptake due to reduced glutamate/glycine transporter activity (Mao et al. 2002). Our present findings reveal a critical role of presynaptic MAPK signaling in opioid-induced stimulation of NMDARs in the spinal cord.
Another salient finding of our study is that NMDARs and ERK1/2, p38, and JNK can form a protein complex in the spinal cord and that chronic morphine treatment substantially increases the interaction of presynaptic NMDARs with the three MAPKs. This increased association may lead to presynaptic NMDAR phosphorylation to potentiate NMDAR activity in the spinal dorsal horn. It is uncertain how the three major groups of distinctly regulated MAPK cascades converge to NMDARs in the spinal cord after MOR stimulation, although the increased presynaptic NMDAR phosphorylation seems to be the common downstream signaling mechanism for the three MAPKs. The signaling cascade and/or crosstalk among the three MAPKs may be necessary for potentiation of presynaptic NMDAR phosphorylation and activity in the spinal dorsal horn during the development of opioid-induced hyperalgesia and tolerance. In cell lines, phosphorylated p38 is capable of forming a complex with ERK1/2 (Zhang et al. 2001). In addition, PKC inhibition can block opioid-induced presynaptic NMDAR activation, hyperalgesia, and tolerance (Zhao et al. 2012). PKC is considered an early signaling component in the opioid-induced MAPK signaling pathway (Belcheva et al. 2001, Fukuda et al. 1996). In cell lines, PKC activity is required for both MOR-induced JNK activation and ERK1/2 phosphorylation (Kuhar et al. 2015, Zheng et al. 2008). Although all MOR agonists activate ERK signaling, in vitro experiments suggest that morphine uses the PKC pathway to induce ERK phosphorylation, whereas etorphine and fentanyl use the β-arrestin pathway (Zheng et al. 2008). Further studies are warranted to determine whether β-arrestin and PKC are required for the recruitment of MAPKs to NMDARs in the spinal cord during chronic opioid treatment.
In summary, our study provides important new evidence that chronic opioid treatment enhances the physical association of NMDARs with MAPKs, including ERK1/2, p38, and JNK, in the spinal cord. These three functionally interrelated MAPKs critically contribute to tonic activation of presynaptic NMDARs in the spinal cord, which is responsible for opioid-induced hyperalgesia and analgesic tolerance. This new information is important to understanding the underlying signaling mechanisms of opioid-induced hyperalgesia and analgesic tolerance. Clinically used NMDAR antagonists are effective in reducing opioid tolerance in patients (Mercadante et al. 2000, Xuerong et al. 2008). However, these drugs often produce intolerable adverse effects. MAPKs may be targeted for inhibiting opioid-induce abnormal presynaptic NMDAR activation, thereby improving opioid analgesic efficacy and minimizing opioid side effects.
Open Science Badges.
This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Acknowledgements
This work was supported by grants from the National Institutes of Health (R01 DA041711) and by the N.G. and Helen T. Hawkins Endowment (to H.-L.P.).
Abbreviations:
- AP5
2-amino-5-phosphonopentanoic acid
- DRG
dorsal root ganglion
- EPSC
excitatory postsynaptic current
- mEPSC
miniature excitatory postsynaptic current
- NMDAR
N-methyl-D-aspartate receptor
- PPR
paired-pulse ratio
- ERK1/2
extracellular signal-regulated kinase 1/2
- JNK
c-Jun N-terminal kinase
- MOR
μ-opioid receptor
- MAPK
mitogen-activated protein kinase
- IP
immunoprecipitation
- RRID
Research Resource Identifiers
Footnotes
Conflict of Interest
The authors declare that they have no conflicts of interest with the contents of this study.
References
- Arana-Argaez VE, Delgado-Rizo V, Pizano-Martinez OE et al. (2010) Inhibitors of MAPK pathway ERK1/2 or p38 prevent the IL-1{beta}-induced up-regulation of SRP72 autoantigen in Jurkat cells. J Biol Chem, 285, 32824–32833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belcheva MM, Szucs M, Wang D, Sadee W and Coscia CJ (2001) mu-Opioid receptor-mediated ERK activation involves calmodulin-dependent epidermal growth factor receptor transactivation. J Biol Chem, 276, 33847–33853. [DOI] [PubMed] [Google Scholar]
- Bennett BL, Sasaki DT, Murray BW et al. (2001) SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc Natl Acad Sci U S A, 98, 13681–13686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bohn LM, Dykstra LA, Lefkowitz RJ, Caron MG and Barak LS (2004) Relative opioid efficacy is determined by the complements of the G protein-coupled receptor desensitization machinery. Molecular pharmacology, 66, 106–112. [DOI] [PubMed] [Google Scholar]
- Bohn LM, Lefkowitz RJ, Gainetdinov RR, Peppel K, Caron MG and Lin FT (1999) Enhanced morphine analgesia in mice lacking beta-arrestin 2. Science, 286, 2495–2498. [DOI] [PubMed] [Google Scholar]
- Brady SW, McQuerry JA, Qiao Y et al. (2017) Combating subclonal evolution of resistant cancer phenotypes. Nat Commun, 8, 1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Celerier E, Rivat C, Jun Y, Laulin JP, Larcher A, Reynier P and Simonnet G (2000) Long-lasting hyperalgesia induced by fentanyl in rats: preventive effect of ketamine. Anesthesiology, 92, 465–472. [DOI] [PubMed] [Google Scholar]
- Chen J, Li L, Chen SR et al. (2018) The alpha2delta-1-NMDA Receptor Complex Is Critically Involved in Neuropathic Pain Development and Gabapentin Therapeutic Actions. Cell reports, 22, 2307–2321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen L and Huang LY (1992) Protein kinase C reduces Mg2+ block of NMDA-receptor channels as a mechanism of modulation. Nature, 356, 521–523. [DOI] [PubMed] [Google Scholar]
- Chen SR, Hu YM, Chen H and Pan HL (2014) Calcineurin inhibitor induces pain hypersensitivity by potentiating pre- and postsynaptic NMDA receptor activity in spinal cords. J Physiol, 592, 215–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen SR and Pan HL (2006a) Blocking mu opioid receptors in the spinal cord prevents the analgesic action by subsequent systemic opioids. Brain Res, 1081, 119–125. [DOI] [PubMed] [Google Scholar]
- Chen SR and Pan HL (2006b) Loss of TRPV1-expressing sensory neurons reduces spinal mu opioid receptors but paradoxically potentiates opioid analgesia. J Neurophysiol, 95, 3086–3096. [DOI] [PubMed] [Google Scholar]
- Chen SR, Prunean A, Pan HM, Welker KL and Pan HL (2007) Resistance to morphine analgesic tolerance in rats with deleted transient receptor potential vanilloid type 1-expressing sensory neurons. Neuroscience, 145, 676–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Geis C and Sommer C (2008) Activation of TRPV1 contributes to morphine tolerance: involvement of the mitogen-activated protein kinase signaling pathway. The Journal of neuroscience : the official journal of the Society for Neuroscience, 28, 5836–5845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chia YY, Liu K, Wang JJ, Kuo MC and Ho ST (1999) Intraoperative high dose fentanyl induces postoperative fentanyl tolerance. Can J Anaesth, 46, 872–877. [DOI] [PubMed] [Google Scholar]
- Daulhac L, Mallet C, Courteix C, Etienne M, Duroux E, Privat AM, Eschalier A and Fialip J (2006) Diabetes-induced mechanical hyperalgesia involves spinal mitogen-activated protein kinase activation in neurons and microglia via N-methyl-D-aspartate-dependent mechanisms. Molecular pharmacology, 70, 1246–1254. [DOI] [PubMed] [Google Scholar]
- Favata MF, Horiuchi KY, Manos EJ et al. (1998) Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem, 273, 18623–18632. [DOI] [PubMed] [Google Scholar]
- Fukuda K, Kato S, Morikawa H, Shoda T and Mori K (1996) Functional coupling of the delta-, mu-, and kappa-opioid receptors to mitogen-activated protein kinase and arachidonate release in Chinese hamster ovary cells. Journal of neurochemistry, 67, 1309–1316. [DOI] [PubMed] [Google Scholar]
- Jin SX, Arai J, Tian X, Kumar-Singh R and Feig LA (2013) Acquisition of contextual discrimination involves the appearance of a RAS-GRF1/p38 mitogen-activated protein (MAP) kinase-mediated signaling pathway that promotes long term potentiation (LTP). J Biol Chem, 288, 21703–21713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keith DE, Murray SR, Zaki PA, Chu PC, Lissin DV, Kang L, Evans CJ and von Zastrow M (1996) Morphine activates opioid receptors without causing their rapid internalization. J Biol Chem, 271, 19021–19024. [DOI] [PubMed] [Google Scholar]
- Kocab AJ, Veloso A, Paulsen MT, Ljungman M and Duckett CS (2015) Effects of physiological and synthetic IAP antagonism on c-IAP-dependent signaling. Oncogene, 34, 5472–5481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuhar JR, Bedini A, Melief EJ, Chiu YC, Striegel HN and Chavkin C (2015) Mu opioid receptor stimulation activates c-Jun N-terminal kinase 2 by distinct arrestin-dependent and independent mechanisms. Cell Signal, 27, 1799–1806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lan JY, Skeberdis VA, Jover T, Grooms SY, Lin Y, Araneda RC, Zheng X, Bennett MV and Zukin RS (2001) Protein kinase C modulates NMDA receptor trafficking and gating. Nat Neurosci, 4, 382–390. [DOI] [PubMed] [Google Scholar]
- Lefkowitz RJ and Shenoy SK (2005) Transduction of receptor signals by beta-arrestins. Science, 308, 512–517. [DOI] [PubMed] [Google Scholar]
- Li L, Chen SR, Chen H, Wen L, Hittelman WN, Xie JD and Pan HL (2016) Chloride Homeostasis Critically Regulates Synaptic NMDA Receptor Activity in Neuropathic Pain. Cell reports, 15, 1376–1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, Zhu J, Dou J, She H, Tao K, Xu H, Yang Q and Mao Z (2017) Phosphorylation of LAMP2A by p38 MAPK couples ER stress to chaperone-mediated autophagy. Nat Commun, 8, 1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H, Wang H, Sheng M, Jan LY, Jan YN and Basbaum AI (1994) Evidence for presynaptic N-methyl-D-aspartate autoreceptors in the spinal cord dorsal horn. Proc Natl Acad Sci U S A, 91, 8383–8387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luttrell LM, Roudabush FL, Choy EW, Miller WE, Field ME, Pierce KL and Lefkowitz RJ (2001) Activation and targeting of extracellular signal-regulated kinases by beta-arrestin scaffolds. Proc Natl Acad Sci U S A, 98, 2449–2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma W, Zheng WH, Powell K, Jhamandas K and Quirion R (2001) Chronic morphine exposure increases the phosphorylation of MAP kinases and the transcription factor CREB in dorsal root ganglion neurons: an in vitro and in vivo study. Eur J Neurosci, 14, 1091–1104. [DOI] [PubMed] [Google Scholar]
- Mao J, Price DD and Mayer DJ (1994) Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C. The Journal of neuroscience : the official journal of the Society for Neuroscience, 14, 2301–2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mao J, Sung B, Ji RR and Lim G (2002) Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity. The Journal of neuroscience : the official journal of the Society for Neuroscience, 22, 8312–8323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonald PH, Chow CW, Miller WE, Laporte SA, Field ME, Lin FT, Davis RJ and Lefkowitz RJ (2000) Beta-arrestin 2: a receptor-regulated MAPK scaffold for the activation of JNK3. Science, 290, 1574–1577. [DOI] [PubMed] [Google Scholar]
- Melief EJ, Miyatake M, Bruchas MR and Chavkin C (2010) Ligand-directed c-Jun N-terminal kinase activation disrupts opioid receptor signaling. Proc Natl Acad Sci U S A, 107, 11608–11613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mercadante S, Arcuri E, Tirelli W and Casuccio A (2000) Analgesic effect of intravenous ketamine in cancer patients on morphine therapy: a randomized, controlled, double-blind, crossover, double-dose study. J Pain Symptom Manage, 20, 246–252. [DOI] [PubMed] [Google Scholar]
- Mizushima T, Obata K, Yamanaka H, Dai Y, Fukuoka T, Tokunaga A, Mashimo T and Noguchi K (2005) Activation of p38 MAPK in primary afferent neurons by noxious stimulation and its involvement in the development of thermal hyperalgesia. Pain, 113, 51–60. [DOI] [PubMed] [Google Scholar]
- Modol L, Santos D, Cobianchi S, Gonzalez-Perez F, Lopez-Alvarez V and Navarro X (2015) NKCC1 Activation Is Required for Myelinated Sensory Neurons Regeneration through JNK-Dependent Pathway. J Neurosci, 35, 7414–7427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakata K, Abrams B, Grill B, Goncharov A, Huang X, Chisholm AD and Jin Y (2005) Regulation of a DLK-1 and p38 MAP kinase pathway by the ubiquitin ligase RPM-1 is required for presynaptic development. Cell, 120, 407–420. [DOI] [PubMed] [Google Scholar]
- Nateri AS, Raivich G, Gebhardt C et al. (2007) ERK activation causes epilepsy by stimulating NMDA receptor activity. Embo j, 26, 4891–4901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nistico R, Florenzano F, Mango D et al. (2015) Presynaptic c-Jun N-terminal Kinase 2 regulates NMDA receptor-dependent glutamate release. Sci Rep, 5, 9035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obata K and Noguchi K (2004) MAPK activation in nociceptive neurons and pain hypersensitivity. Life Sci, 74, 2643–2653. [DOI] [PubMed] [Google Scholar]
- Pan YZ and Pan HL (2004) Primary afferent stimulation differentially potentiates excitatory and inhibitory inputs to spinal lamina II outer and inner neurons. J Neurophysiol, 91, 2413–2421. [DOI] [PubMed] [Google Scholar]
- Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K and Cobb MH (2001) Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev, 22, 153–183. [DOI] [PubMed] [Google Scholar]
- Pierce KL and Lefkowitz RJ (2001) Classical and new roles of beta-arrestins in the regulation of G-protein-coupled receptors. Nat Rev Neurosci, 2, 727–733. [DOI] [PubMed] [Google Scholar]
- Rudhard Y, Kneussel M, Nassar MA et al. (2003) Absence of Whisker-related pattern formation in mice with NMDA receptors lacking coincidence detection properties and calcium signaling. The Journal of neuroscience : the official journal of the Society for Neuroscience, 23, 2323–2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santos SF, Rebelo S, Derkach VA and Safronov BV (2007) Excitatory interneurons dominate sensory processing in the spinal substantia gelatinosa of rat. J Physiol, 581, 241–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sindreu C, Palmiter RD and Storm DR (2011) Zinc transporter ZnT-3 regulates presynaptic Erk1/2 signaling and hippocampus-dependent memory. Proc Natl Acad Sci U S A, 108, 3366–3370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song X, Coffa S, Fu H and Gurevich VV (2009) How does arrestin assemble MAPKs into a signaling complex? J Biol Chem, 284, 685–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun L, Huang T, Xu W, Sun J, Lv Y and Wang Y (2017) Advanced glycation end products promote VEGF expression and thus choroidal neovascularization via Cyr61-PI3K/AKT signaling pathway. Sci Rep, 7, 14925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trujillo KA and Akil H (1991) Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science, 251, 85–87. [DOI] [PubMed] [Google Scholar]
- Tsai RY, Tai YH, Tzeng JI, Lin SL, Shen CH, Yang CP, Hsin ST, Wang CB and Wong CS (2009) Ultra-low dose naloxone restores the antinociceptive effect of morphine in pertussis toxin-treated rats and prevents glutamate transporter downregulation by suppressing the p38 mitogen-activated protein kinase signaling pathway. Neuroscience, 159, 1244–1256. [DOI] [PubMed] [Google Scholar]
- Vara H, Onofri F, Benfenati F, Sassoe-Pognetto M and Giustetto M (2009) ERK activation in axonal varicosities modulates presynaptic plasticity in the CA3 region of the hippocampus through synapsin I. Proc Natl Acad Sci U S A, 106, 9872–9877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L, Chen SR, Ma H, Chen H, Hittelman WN and Pan HL (2018) Regulating nociceptive transmission by VGluT2-expressing spinal dorsal horn neurons. Journal of neurochemistry, doi: 10.1111/jnc.14588.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu ZZ, Chen SR and Pan HL (2004) Differential sensitivity of N- and P/Q-type Ca2+ channel currents to a mu opioid in isolectin B4-positive and -negative dorsal root ganglion neurons. J Pharmacol Exp Ther, 311, 939–947. [DOI] [PubMed] [Google Scholar]
- Xie JD, Chen SR, Chen H and Pan HL (2017a) Bortezomib induces neuropathic pain through protein kinase C-mediated activation of presynaptic NMDA receptors in the spinal cord. Neuropharmacology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie JD, Chen SR and Pan HL (2017b) Presynaptic mGluR5 receptor controls glutamatergic input through protein kinase C-NMDA receptors in paclitaxel-induced neuropathic pain. J Biol Chem, 292, 20644–20654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xuerong Y, Yuguang H, Xia J and Hailan W (2008) Ketamine and lornoxicam for preventing a fentanyl-induced increase in postoperative morphine requirement. Anesth Analg, 107, 2032–2037. [DOI] [PubMed] [Google Scholar]
- Zeng J, Thomson LM, Aicher SA and Terman GW (2006) Primary afferent NMDA receptors increase dorsal horn excitation and mediate opiate tolerance in neonatal rats. J Neurosci, 26, 12033–12042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H, Shi X, Hampong M, Blanis L and Pelech S (2001) Stress-induced inhibition of ERK1 and ERK2 by direct interaction with p38 MAP kinase. J Biol Chem, 276, 6905–6908. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Chen SR, Laumet G, Chen H and Pan HL (2016) Nerve Injury Diminishes Opioid Analgesia through Lysine Methyltransferase-mediated Transcriptional Repression of mu-Opioid Receptors in Primary Sensory Neurons. The Journal of biological chemistry, 291, 8475–8485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao YL, Chen SR, Chen H and Pan HL (2012) Chronic opioid potentiates presynaptic but impairs postsynaptic N-methyl-D-aspartic acid receptor activity in spinal cords: implications for opioid hyperalgesia and tolerance. J. Biol. Chem, 287, 25073–25085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng H, Loh HH and Law PY (2008) Beta-arrestin-dependent mu-opioid receptor-activated extracellular signal-regulated kinases (ERKs) Translocate to Nucleus in Contrast to G protein-dependent ERK activation. Mol Pharmacol, 73, 178–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou HY, Chen SR, Chen H and Pan HL (2008) Sustained inhibition of neurotransmitter release from nontransient receptor potential vanilloid type 1-expressing primary afferents by mu-opioid receptor activation-enkephalin in the spinal cord. J Pharmacol Exp Ther, 327, 375–382. [DOI] [PubMed] [Google Scholar]
- Zhou HY, Chen SR, Chen H and Pan HL (2010) Opioid-induced long-term potentiation in the spinal cord is a presynaptic event. The Journal of neuroscience : the official journal of the Society for Neuroscience, 30, 4460–4466. [DOI] [PMC free article] [PubMed] [Google Scholar]