Background:

Novel analgesics that do not suppress the respiratory drive are urgently needed. Glutamate signaling through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors plays important roles in central pain circuits. AMPAkines augment AMPA receptor function and have been shown to stimulate the respiratory drive to oppose opioid-induced hypoventilation. However, their role in chronic pain states remains unknown.

Methods:

The authors studied AMPAkines (CX546 and CX516) in rat spared nerve injury (SNI) model of neuropathic pain and Complete Freund’s Adjuvant (CFA) model of inflammatory pain. They measured the effect of AMPAkines on mechanical and cold allodynia. They also evaluated their effect on depressive symptoms of pain using the forced swim test, as time of immobility on this test has been used as a measure for behavioral despair, a feature of depression.

Results:

The authors found that CX546, compared with dimethyl sulfoxide (DMSO) control, reduced both mechanical and sensory allodynia in SNI (DMSO group, n = 9; CX546 group, n = 11) and CFA models (both DMSO and CX546 groups, n = 9). They found that CX546, compared with control, also reduced depressive symptoms of pain by decreasing immobility on the forced swim test in both SNI (both DMSO and CX546 groups, n = 8) and CFA models (both DMSO and CX546 groups, n = 10). Finally, they found that CX516, compared with control, also reduced mechanical and cold allodynia in the SNI model (both DMSO and CX516 groups, n = 10).

Conclusions:

AMPAkines alleviate pain hypersensitivity as well as depression-like behavior associated with long-lasting nerve injury and inflammatory insult.

What We Already Know about This Topic
  • AMPAkines are compounds that enhance AMPA receptor transmission in the central nervous system. Importantly, they stimulate respiration in the setting of opiate-induced hypoventilation.

  • AMPA receptors also play a significant role in nociceptive transmission. While AMPAkines might find utility in the treatment of opioid-induced respiratory depression, their effects on nociception are not fully understood.

  • In two experimental models of chronic pain, the effect of AMPAkines on nociception was determined.

What This Article Tells Us That Is New
  • Surprisingly, AMPAkines reduced mechanical and cold allodynia and attenuated symptoms of depression associated with chronic pain.

  • Given the combination of analgesia and reduction of opioid-induced hypoventilation, AMPAkines may find utility in the treatment of persistent postoperative and chronic pain.

DESPITE advances in pain research, pharmacologic options remain limited. Opioids remain the most potent postoperative analgesic. A feared complication of opioids is respiratory depression, which can happen in both acute postoperative and chronic pain settings.1–3  In recent years, there has been a sharp rise in opioid-related mortalities in chronic pain patients, mostly due to respiratory depression.1,4  Even as many of these opioid-related morbidities and mortalities involve inappropriate prescription and over- or mis-use, treatment or prevention of opioid-induced respiratory depression remains lacking. Similarly, the development of new analgesics, particularly agents that do not suppress the respiratory drive, is urgently needed.2 

Excitatory glutamate signaling in the central nervous system (CNS) plays an important role in the regulation of chronic pain. α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors are the primary glutamate receptors in the CNS.5  Increased transmission through AMPA receptors in the spinal dorsal horn has been shown to contribute to the induction and maintenance of chronic pain.6–10  AMPA receptor signaling in the anterior cingulate cortex (ACC) and amygdala can also facilitate pain transmission.11–13  On the other hand, transmission through AMPA receptors is critical for maintaining the integrity of the periaqueductal gray-rostral ventromedial medulla (PAG-RVM) descending inhibitory pathway.14–16  In addition, the nucleus accumbens (NAc), a key region for the regulation of mood and motivated behaviors, also utilizes AMPA receptor signaling to generate pain-induced analgesia.17,18  Thus, depending on the region in the CNS, AMPA receptor signaling may play either a pro-nociceptive or anti-nociceptive role.

AMPAkines are a class of compounds that enhance AMPA receptor transmission.19,20  AMPAkines bind to an allosteric site on the AMPA receptor to reduce the kinetics of channel deactivation and desensitization. By preventing AMPA receptors from closing, AMPAkines increase the inward synaptic current. AMPAkines have been found to have cognitive effects in animal and human studies,21  and have been studied in schizophrenia, depression, Huntington disease, and Alzheimer disease.19,22–25  Interestingly, recent animal and human studies have shown that AMPAkines stimulate the respiratory drive in the context of hypoventilation.26–28  For reasons not yet mechanistically understood, AMPAkines stimulate respiratory rhythmogenesis only when the respiratory drive has been suppressed, making these compounds ideal drugs for treating opioid-induced respiratory depression. A few limited studies found that AMPAkines do not alter the threshold of acute pain.26,28  However, the effect of AMPAkines on persistent or chronic pain has not been examined. Given the central role the AMPA receptors play in pain, we hypothesized that the systemic administration of AMPAkines can alter and affect the pain sensitivity.

Here we studied the effect of AMPAkines on chronic pain using the spared nerve injury (SNI) model for neuropathic pain and Complete Freund’s Adjuvant (CFA) model for inflammatory pain.29–31  We found that CX546, an established AMPAkine, which has been studied in hypoventilation, Rett syndrome, anxiety, and autism,27,32–35  has significant analgesic properties. We verified these findings using a second AMPAkine, CX516.36,37  Thus, our results suggest a novel analgesic role for AMPAkines in persistent pain states. This novel analgesic property, combined with the ability to stimulate the respiratory drive, makes AMPAkines promising drugs for postoperative and chronic pain.

Animals

All procedures in this study were approved by the New York University School of Medicine Institutional Animal Care and Use Committee (New York, NY) as consistent with the National Institute of Health Guide for the Care and Use of Laboratory Animals (publication number 85-23) to ensure minimal animal use and discomfort. Male Sprague-Dawley rats were purchased from Taconic Farms, Albany, NY, and kept in the Mispro Animal Facility in Alexandria Life Science Building (New York, NY), with controlled humidity, room temperature, and 12-h (6:30 AM–6:30 PM) light-dark cycle. Food and water were available ad libitum. Animals arrived to the animal facility at 250–300 g and were given approximately 7 days to adjust to the new environment before the onset of any experiments. For each experiment, the animals were randomized to either the control or experimental group.

SNI Surgery

The SNI surgery has been previously described in detail.29  Briefly, under isoflurane anesthesia (1.5–2%), the skin on the lateral surface of the right thigh of rat was incised and a section was made through the biceps femoris muscle to expose three branches of the sciatic nerve: sural, common peroneal, and tibial nerves. The common peroneal and tibial nerves were tied with non-absorbent 5.0 silk sutures at the point of trifurcation. The nerves were then cut distal to the knot, and about 3–5 mm of the distal ends were removed. In sham surgeries (control), above nerves were dissected, but not cut. Muscle and skin layers were then sutured closely in distinct layers.

CFA Administration

To produce inflammatory pain, CFA (Mycobacterium tuberculosis, Sigma–Aldrich [St. Louis, MO], 0.1 ml) was suspended in an oil–saline (1:1) emulsion and injected subcutaneously into the plantar aspect of the right hind paw. Control rats received equal volume of saline injection.

Drugs

CX546 and CX516 were injected intraperitoneally in rats to a volume of 0.5–1 ml, while a similar volume of dimethyl sulfoxide (DMSO) was injected intraperitoneally to the control group. Injections were given 14 days after SNI surgery and 7 days after CFA injection. Injections were followed by behavioral tests.

Animal Behavioral Tests

Mechanical Allodynia Testing.

A traditional Dixon up-down method with von Frey filaments was used to measure mechanical allodynia.38,39  In brief, rats were individually placed into plexiglass chambers over a mesh table and acclimated for 20 min before the onset of examination. Beginning with 2.55 g, von Frey filaments in a set with logarithmically incremental stiffness (0.45, 0.75, 1.20, 2.55, 4.40, 6.10, 10.50, 15.10 g) were applied to the lateral 1/3 of right paws (in the distribution of the sural nerve) of animals before and up to 14 days after SNI or sham surgery. Similar tests were done on CFA- or saline-treated rats. About 50% withdrawal threshold was calculated as described previously.39  Observers were blinded to the test conditions.

Cold Allodynia Testing

. Animals were individually placed into plexiglass chambers as above and acclimated for 20 min. A drop of acetone (Sigma–Aldrich) was applied to the lateral plantar surface of the paws (in the distribution of the sural nerve). As previously described,40,41  the following scoring system was applied. 0: no visible response or startle response lasting <0.5 s; 1: paw withdrawal lasting <5 s; 2: withdrawal lasting 5–10 s, ± licking of the paws; 3: prolonged repetitive withdrawal lasting > 10 s. Acetone was applied five times to each paw, and average score was calculated. Observers were blinded to the test conditions.

Forced Swim Test (FST).

As described previously,42  on the first session of the test, each animal was placed for 15 min into a standard clear Porsolt chamber (Lafayette Instrument Company, Lafayette, IN) with water at 25°C filled to 25 cm. Afterwards, the animal was taken out of the chamber, dried and put back in its home cage. Twenty-four hours later, the animal was placed into the Porsolt chamber again under the same condition for 5 min. Both sessions were videotaped, but only the second session was analyzed. Immobility was defined as a lack of movement of the hind paws lasting more than 1 s. An independent observer, blinded to the test conditions, examined and graded the total time of immobility for each rat, and the average grade was presented for each animal. FST was conducted 14 days after SNI/sham surgeries or 7 days after CFA/saline treatments.

Locomotion Activities.

As described previously,43  rats were acclimated in locomotor measurement chambers (Accuscan, Columbus, OH) before testing. On test days, locomotion activities were measured for a total of 30 min. The distance traveled was measured using the VersaMax System (Accuscan), which monitored the animal activity via a grid of 16 × 16 infrared light beams that traverse the animal cage (42 × 42 × 30 cm) front-to-back and left-to-right. Information about beam status, scanned at a rate of 100 times per second, was stored to disk. The activity was expressed as ambulatory distance measured during 10 different 3-min bins in a 30-min session.

Statistics

The results of behavioral experiments were given as mean ± SEM. For mechanical allodynia, a two-way ANOVA with repeated measures and post hoc multiple pair-wise comparison Bonferroni tests was used to compare the 50% withdrawal threshold of the SNI and sham animals and of the CFA and saline-treated groups. Cold allodynia was also analyzed using the two-way ANOVA test with repeated measures and post hoc multiple pair-wise comparison Bonferroni tests. Two-way ANOVA with post hoc multiple pair-wise Bonferroni comparison tests was also used for the dose–response experiments to compare the effect of AMPAkines at various doses on mechanical and cold allodynia. In the pharmacokinetic experiment, a two-way ANOVA with repeated measures and post hoc multiple pair-wise comparison Bonferroni tests was used to compare mechanical or cold allodynia in AMPAkine versus DMSO (control) treatments at various time points. For the FST, unpaired two-tailed Student t test was used to compare the performances of sham and SNI groups, CFA and saline groups, as well as AMPAkine and DMSO groups. A two-way ANOVA with repeated measures and post hoc multiple pair-wise comparison Bonferroni tests was also used to compare locomotion in SNI versus sham groups, CFA versus saline treatment groups, and in AMPAkine versus DMSO groups. For all tests, a P value <0.05 was considered statistically significant. All data were analyzed using GraphPad Prism version 5 software (GraphPad, La Jolla, CA).

SNI Produced Long-lasting Sensory Allodynia and Depression-like Behavior

The SNI model is commonly used to study chronic neuropathic pain or persistent postoperative pain. Consistent with previous results,29,42,44  we found that SNI surgery produced significant mechanical and cold allodynia in the (spared) sural nerve distribution (fig. 1, A and B). In contrast, rats that underwent sham operation did not demonstrate any sensory allodynia in the spared nerve distribution.

Fig. 1.

Spared nerve injury (SNI) induces long-lasting mechanical and cold allodynia and increases immobility during forced swim tests in rats. (A) Animals that underwent SNI surgery developed mechanical allodynia that lasted at least 14 days. Mechanical allodynia was tested using von Frey filaments. In comparison, sham-treated rats (control) did not demonstrate mechanical allodynia. A 50% withdrawal threshold was calculated (see Methods for details). ****P < 0.0001, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Sham group, n = 6. SNI group, n = 10 (B) SNI-treated animals, in contrast to control rats, developed cold allodynia that lasted at least 14 days. ****P < 0.0001, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Sham group, n = 6. SNI group, n = 10 (C) SNI-treated rats demonstrated increased immobility during the forced swim test compared to controls. *P < 0.05, unpaired two-tailed Student test. Sham group, n = 12. SNI group, n = 10. (D) Locomotion was unaffected by SNI surgery. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni tests. n = 6 for both Sham and SNI groups. Error bars represent standard error mean (SEM).

Fig. 1.

Spared nerve injury (SNI) induces long-lasting mechanical and cold allodynia and increases immobility during forced swim tests in rats. (A) Animals that underwent SNI surgery developed mechanical allodynia that lasted at least 14 days. Mechanical allodynia was tested using von Frey filaments. In comparison, sham-treated rats (control) did not demonstrate mechanical allodynia. A 50% withdrawal threshold was calculated (see Methods for details). ****P < 0.0001, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Sham group, n = 6. SNI group, n = 10 (B) SNI-treated animals, in contrast to control rats, developed cold allodynia that lasted at least 14 days. ****P < 0.0001, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Sham group, n = 6. SNI group, n = 10 (C) SNI-treated rats demonstrated increased immobility during the forced swim test compared to controls. *P < 0.05, unpaired two-tailed Student test. Sham group, n = 12. SNI group, n = 10. (D) Locomotion was unaffected by SNI surgery. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni tests. n = 6 for both Sham and SNI groups. Error bars represent standard error mean (SEM).

Close modal

We have previously shown that chronic neuropathic pain, in addition to producing sensory allodynia as tested by paw withdrawal, also leads to depression-like behavior in rats.42,44  Here, we applied the FST, a widely used test to assess depression-like behavior in rats.45  Numerous studies have demonstrated clinically relevant pharmacologic validity of this test. Increased time of immobility (instead of swimming) on the FST is considered a measure of behavioral despair, a salient feature of depression. We performed the FST 14 days after the surgery and found that compared with sham-operated rats, rats after SNI developed a significantly increased time of immobility (fig. 1C). To rule out the possibility that this immobility was caused by locomotor deficits secondary to peripheral neuropathy or by pain associated with movement, we performed locomotion tests on SNI-treated rats. SNI-treated rats, compared with sham-treated rats, did not display a change on the locomotion test for 30 min (fig. 1D), and thus these rats were unlikely to have overt locomotor deficits, which prevented them from swimming for 5 min. Increased immobility on FST, therefore, more likely reflected depression-like behavior exhibited by these animals in chronic pain.45 

CX546, an Established AMPAkine, Relieves Sensory Allodynia and Depression-like Behavior Associated with Chronic Neuropathic Pain

We tested the effect of CX546 on pain using the SNI model. CX546 is a well-established AMPAkine, which has been studied in a number of preclinical models of CNS diseases, including respiratory depression, Rett syndrome, anxiety, and autism.27,32–35  When we applied this compound systemically (via intraperitoneal injection) to SNI-treated rats, we were surprised to observe a drastic improvement in mechanical allodynia compared with control (DMSO) injection (fig. 2A). We began to observe anti-allodynic effects at a dose of 5 mg/kg, while a higher dose of 10 mg/kg produced an even greater effect. We further validated this anti-nociceptive effect in the cold allodynia test, where we similarly observed improvements in cold allodynic scores at 5 and 10 mg/kg doses (fig. 2B). Next, we evaluated the duration of this anti-nociceptive effect (fig. 3, A and B). We administered a 10 mg/kg dose of CX546 (fig. 2, A and B), and we found that the effect of CX546 lasted 6 h in the mechanical allodynia test. Its effect was shorter (2 h) in the cold allodynia test.

Fig. 2.

CX546 relieves mechanical and cold allodynia in spared nerve injury (SNI)-treated animals in a dose-dependent manner. (A) SNI-treated animals after intraperitoneal administration of CX546 demonstrated reduced mechanical allodynia compared to dimethyl sulfoxide (DMSO) control at doses of 10 mg/kg and 5 mg/kg, but not at 2.5 mg/kg. ***P < 0.001, two-way ANOVA with post hoc Bonferroni multiple comparison tests. DMSO group, n = 9. CX546 group, n = 11. (B) SNI-treated animals after intraperitoneal administration of CX546 demonstrated reduced cold allodynia compared to DMSO control at doses of 10 mg/kg and 5 mg/kg, but not at 2.5 mg/kg. **P < 0.01, *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. DMSO group, n = 9. CX546 group, n = 11. Error bars represent SEM.

Fig. 2.

CX546 relieves mechanical and cold allodynia in spared nerve injury (SNI)-treated animals in a dose-dependent manner. (A) SNI-treated animals after intraperitoneal administration of CX546 demonstrated reduced mechanical allodynia compared to dimethyl sulfoxide (DMSO) control at doses of 10 mg/kg and 5 mg/kg, but not at 2.5 mg/kg. ***P < 0.001, two-way ANOVA with post hoc Bonferroni multiple comparison tests. DMSO group, n = 9. CX546 group, n = 11. (B) SNI-treated animals after intraperitoneal administration of CX546 demonstrated reduced cold allodynia compared to DMSO control at doses of 10 mg/kg and 5 mg/kg, but not at 2.5 mg/kg. **P < 0.01, *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. DMSO group, n = 9. CX546 group, n = 11. Error bars represent SEM.

Close modal
Fig. 3.

A single administration of CX546 (10 mg/kg) improves mechanical and cold allodynia in spared nerve injury (SNI)-treated animals over several hours. (A) SNI-treated animals had reduced mechanical allodynia at 1, 2, 4, and 6 h after administration of CX546 (10 mg/kg). ****P < 0.0001, **P < 0.01, *P < 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Dimethyl sulfoxide (DMSO) group, n = 10. CX546 group, n = 9. (B) SNI-treated animals administered with CX546 (10 mg/kg) had reduced cold allodynia compared to DMSO control at 1 and 2 h. *P < 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 8 for both DMSO and CX546 groups. Error bars represent SEM.

Fig. 3.

A single administration of CX546 (10 mg/kg) improves mechanical and cold allodynia in spared nerve injury (SNI)-treated animals over several hours. (A) SNI-treated animals had reduced mechanical allodynia at 1, 2, 4, and 6 h after administration of CX546 (10 mg/kg). ****P < 0.0001, **P < 0.01, *P < 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Dimethyl sulfoxide (DMSO) group, n = 10. CX546 group, n = 9. (B) SNI-treated animals administered with CX546 (10 mg/kg) had reduced cold allodynia compared to DMSO control at 1 and 2 h. *P < 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 8 for both DMSO and CX546 groups. Error bars represent SEM.

Close modal

Sensory allodynia tests assay nociception, but amplifying AMPA receptor transmission can affect a number of brain regions that regulate both nociceptive and affective components of pain. Thus, we next used the FST to test whether CX546, in addition to its anti-nociceptive effects, can also treat depression-like behavior associated with pain. Here we found that CX546 also improved immobility on the FST in SNI-treated rats (fig. 4A). In contrast, CX546 did not affect locomotion (fig. 4B). Thus, the effect of CX546 on FST likely reflected an improvement in behavioral despair, rather than improved locomotion. Results from the FST augment and complement the results on allodynia tests.

Fig. 4.

Intraperitoneal injection of CX546 (10 mg/kg) in spared nerve injury (SNI)-treated animals reduces immobility time during forced swim tests without altering locomotor activities. (A) Animals with intraperitoneal injection of CX546 (10 mg/kg) exhibited reduced immobility compared to dimethyl sulfoxide (DMSO) control. *P < 0.05, unpaired two-tailed Student test. n = 8 for both DMSO and CX546 groups. (B) Locomotion in SNI-treated rats was not affected by CX546 injection (10 mg/kg) compared to DMSO injection. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 6 for both DMSO and CX546 groups. Error bars represent SEM.

Fig. 4.

Intraperitoneal injection of CX546 (10 mg/kg) in spared nerve injury (SNI)-treated animals reduces immobility time during forced swim tests without altering locomotor activities. (A) Animals with intraperitoneal injection of CX546 (10 mg/kg) exhibited reduced immobility compared to dimethyl sulfoxide (DMSO) control. *P < 0.05, unpaired two-tailed Student test. n = 8 for both DMSO and CX546 groups. (B) Locomotion in SNI-treated rats was not affected by CX546 injection (10 mg/kg) compared to DMSO injection. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 6 for both DMSO and CX546 groups. Error bars represent SEM.

Close modal

CX546 Relieves Sensory Allodynia and Depression-like Behavior Associated with Persistent Inflammatory Pain

Synaptic and circuit mechanisms for neuropathic pain are distinct in many aspects from other chronic or acute pain conditions.46  Thus, we applied the CFA model of inflammatory pain to test whether CX546 also confers analgesic effects in persistent inflammatory pain. As expected, CFA injection, compared with saline injection, resulted in consistent mechanical allodynia that lasted for at least 11 days (fig. 5A). In addition, we found that CFA injection also led to a drastic increase in immobility (fig. 5B). CFA did not alter locomotion (fig. 5C), suggesting that immobility on the FST indicates the development of behavioral despair rather than locomotor deficits. These results provide evidence that inflammatory pain, similar to neuropathic pain, leads to sensory allodynia as well as depression-like behavior.

Fig. 5.

Subcutaneous injection of Complete Freund’s Adjuvant (CFA) in the right hind paw of animals induces long-lasting mechanical allodynia and increased immobility during forced swim tests. (A) CFA-treated rats exhibited increased and long-lasting mechanical allodynia compared to saline-treated rats up to postoperative day 11. ****P < 0.0001, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 6 for both saline and CFA groups. (B) CFA-treated rats exhibited increased immobility during the forced swim test compared to saline-treated rats. *P < 0.05, unpaired two-tailed Student test. n = 6 for both saline and CFA groups. (C) CFA treatment did not alter the locomotor activity of animals. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Saline group, n = 6. CFA group, n = 8. Error bars represent SEM.

Fig. 5.

Subcutaneous injection of Complete Freund’s Adjuvant (CFA) in the right hind paw of animals induces long-lasting mechanical allodynia and increased immobility during forced swim tests. (A) CFA-treated rats exhibited increased and long-lasting mechanical allodynia compared to saline-treated rats up to postoperative day 11. ****P < 0.0001, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 6 for both saline and CFA groups. (B) CFA-treated rats exhibited increased immobility during the forced swim test compared to saline-treated rats. *P < 0.05, unpaired two-tailed Student test. n = 6 for both saline and CFA groups. (C) CFA treatment did not alter the locomotor activity of animals. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. Saline group, n = 6. CFA group, n = 8. Error bars represent SEM.

Close modal

Next, we tested whether the analgesic property of CX546 we found in the SNI model of neuropathic pain is preserved in the CFA model of inflammatory pain. After intraperitoneal administration of this AMPAkine, we observed a significant improvement in mechanical allodynia (fig. 6A) in a dose-dependent manner, which is similar to our findings in the SNI model. Furthermore, CX546, as expected, also reduced immobility in the FST, without altering locomotion (fig. 6, B and C). Thus, CX546 treated both sensory allodynia and depression-like behavior elicited by the CFA model. These results, therefore, demonstrate that the analgesic property of CX546 is preserved in both neuropathic and inflammatory pain conditions.

Fig. 6.

Intraperitoneal administration of CX546 in Complete Freund’s Adjuvant (CFA)-treated animals relieves mechanical allodynia and reduces immobility during forced swim tests without altering locomotor activities. (A) Intraperitoneal administration of CX546 in CFA-treated animals decreases mechanical allodynia at doses of 25 mg/kg and 12.5 mg/kg, but not at 6.25 mg/kg or 3.125 mg/kg dose. ***P < 0.001, *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. For the 25, 12.5, and 6.25 mg/kg dose, n = 9. For the 3.125 mg/kg dose, n = 6. (B) CFA-treated animals administered with CX546 (25 mg/kg) showed decreased immobility compared with control during the forced swim test. *P < 0.05, unpaired two-tailed Student test. n = 10 for both dimethyl sulfoxide (DMSO) and CX546 groups. (C) Administration of CX546 (25 mg/kg) did not affect locomotor activities of CFA-treated animals. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 7 for both DMSO and CX546 groups. Error bars represent SEM.

Fig. 6.

Intraperitoneal administration of CX546 in Complete Freund’s Adjuvant (CFA)-treated animals relieves mechanical allodynia and reduces immobility during forced swim tests without altering locomotor activities. (A) Intraperitoneal administration of CX546 in CFA-treated animals decreases mechanical allodynia at doses of 25 mg/kg and 12.5 mg/kg, but not at 6.25 mg/kg or 3.125 mg/kg dose. ***P < 0.001, *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. For the 25, 12.5, and 6.25 mg/kg dose, n = 9. For the 3.125 mg/kg dose, n = 6. (B) CFA-treated animals administered with CX546 (25 mg/kg) showed decreased immobility compared with control during the forced swim test. *P < 0.05, unpaired two-tailed Student test. n = 10 for both dimethyl sulfoxide (DMSO) and CX546 groups. (C) Administration of CX546 (25 mg/kg) did not affect locomotor activities of CFA-treated animals. P > 0.05, two-way ANOVA with repeated measures and post hoc Bonferroni multiple comparison tests. n = 7 for both DMSO and CX546 groups. Error bars represent SEM.

Close modal

CX516 Relieves Sensory Allodynia and Depression-like Behavior Associated with Chronic Pain

To further validate the analgesic effect of AMPAkines in chronic pain, we applied CX516, another compound with well-known AMPA-potentiation properties.36,37  Here, we found that CX516 also produced dose-dependent effects on sensory allodynia in SNI-treated rats (fig. 7, A and B). The dose required to produce maximal analgesic effects is higher in CX516 (20 mg/kg) than required for CX546 (<10 mg/kg). The maximal analgesic efficacy, however, is similar in both compounds. Thus, these results suggest that AMPAkines in general have important analgesic properties in chronic pain states.

Fig. 7.

Intraperitoneal administration of CX516 in spared nerve injury (SNI)-treated animals relieves mechanical and cold allodynia. (A) Intraperitoneal administration of CX516 in SNI-treated animals decreases their mechanical allodynia in a dose-dependent manner. Mechanical allodynia was decreased at CX516 doses of 40 mg/kg and 20 mg/kg, but not at 10 mg/kg. ***P < 0.001, *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. For 10 mg/kg, n = 7. For 20 mg/kg, n = 10. For 40mg/kg, n = 7. (B) Intraperitoneal administration of CX516 in SNI-treated animals decreases their cold allodynia in a dose-dependent manner. Cold allodynia was decreased at CX516 doses of 40 mg/kg, but not at 20 mg/kg or 10 mg/kg. *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. For 10 mg/kg, n = 7. For 20 mg/kg, n = 10. For 40mg/kg, n = 7. Error bars represent SEM. DMSO = dimethyl sulfoxide.

Fig. 7.

Intraperitoneal administration of CX516 in spared nerve injury (SNI)-treated animals relieves mechanical and cold allodynia. (A) Intraperitoneal administration of CX516 in SNI-treated animals decreases their mechanical allodynia in a dose-dependent manner. Mechanical allodynia was decreased at CX516 doses of 40 mg/kg and 20 mg/kg, but not at 10 mg/kg. ***P < 0.001, *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. For 10 mg/kg, n = 7. For 20 mg/kg, n = 10. For 40mg/kg, n = 7. (B) Intraperitoneal administration of CX516 in SNI-treated animals decreases their cold allodynia in a dose-dependent manner. Cold allodynia was decreased at CX516 doses of 40 mg/kg, but not at 20 mg/kg or 10 mg/kg. *P < 0.05, two-way ANOVA with post hoc Bonferroni multiple comparison tests. For 10 mg/kg, n = 7. For 20 mg/kg, n = 10. For 40mg/kg, n = 7. Error bars represent SEM. DMSO = dimethyl sulfoxide.

Close modal

In this study, we investigated the role of AMPAkines in chronic pain. We found that CX546 and CX516, two well-studied AMPAkines, reduce sensory allodynia and depression-like behavior in rodent neuropathic and inflammatory pain models.

New analgesics that do not suppress the respiratory drive are urgently needed. AMPAkines slow the kinetics of AMPA receptor deactivation to enhance the inward synaptic current.19,20  By increasing AMPA receptor-mediated currents in neurons of the pre-Botzinger complex in the medulla, AMPAkines directly stimulate the respiratory drive,26,27,47–49  and they have been shown to treat or prevent hypoventilation caused by opioids and propofol.26,28,50,51  The effect of AMPAkines in chronic pain, however, has not been previously studied. The analgesic dose of CX546 found in our study is comparable with the dose tested in rats to treat respiratory depression.27  Meanwhile, the time course of analgesia after a single administration (2–6 h) is slightly longer than the respiratory stimulatory effect of AMPAkines.26,27  Thus, the analgesic property of AMPAkines overlaps their respiratory stimulatory activity from a pharmacologic and pharmacokinetic standpoint. As a rare analgesic that can stimulate the respiratory drive, AMPAkines should be useful for postoperative and chronic pain.

The anti-nociceptive effect of AMPAkines can be explained by their action on AMPA receptors in two descending inhibitory circuits: PAG-RVM and NAc-RVM circuits. The PAG-RVM-spinal descending pathway is a well-known mechanism for pain regulation.52–54  In this pathway, neurons from the PAG form glutamatergic projections through AMPA receptors on GABAergic cells in the RVM to inhibit dorsal horn neurons.55  The administration of glutamate into the PAG is known to produce analgesia.56–58  Furthermore, there is evidence that in the RVM, AMPA receptor up-regulation mediates analgesia in inflammatory pain states,59,60  whereas their down-regulation in neuropathic pain causes hyperalgesia.61  Thus, transmission through AMPA receptors is required for the intact PAG-RVM-descending pathway.16,62  A second pain modulating center that depends on AMPA receptor signaling is the NAc. The NAc provides pain-induced analgesia, in part through its projection to the RVM.18  Intra-NAc administration of AMPA receptor antagonists, however, can disrupt this pain-induced analgesic mechanism.17 

At the same time, however, potentiation of AMPA receptors in neurons of the spinal dorsal horn, ACC and amygdala can have pro-nociceptive effects. In the dorsal horn, chronic inflammatory pain increases membrane targeting of GluA1 AMPA receptor subunits, but decreases GluA2 delivery,6,9,63  leading to the formation of GluA2-lacking receptors to augment pain transmission.8  Similarly, AMPA receptor signaling in the ACC and amygdala has also been suggested to mediate increased synaptic plasticity and confer hyperalgesia.11–13,64,65

Thus, in chronic pain conditions, AMPA receptor signaling plays both pro-nociceptive and anti-nociceptive roles, depending on the target CNS regions. In our study, we administered AMPAkines systemically, and we expected them to act peripherally or in the brain, with a smaller component of activity in the spinal cord. Thus, the behavioral phenotype we observed likely represents AMPAkine effects in the brain. Our data indicate that the net result favors analgesia rather than hyperalgesia. A possible reason for this net analgesic effect may lie in the distinct affinity of AMPAkines for neurons of different brain regions.66  AMPAkines are known to bind to neurons in the NAc and brain stem with high affinity,66  thereby facilitating the enhancement of AMPA receptor signaling in these descending inhibitory circuits. Future studies targeting specific regions in the brain and spinal cord are needed to further elucidate the precise roles of these drugs in pain states.

Symptoms of depression occur in many chronic pain patients,67,68  and our finding that AMPAkines can treat pain-induced depression is consistent with the role of central AMPA receptors in depression.69  For example, GluA1 subunits of AMPA receptors are reduced in the amygdala, prefrontal cortex, and hippocampus in rodent models of depression.70–72  Ketamine has been shown to increase GluA1 concentration in the prefrontal cortex to provide fast-acting relief from depression,73  and it is effective in treating pain-induced depression.42  Thus, increased AMPA receptor signaling (especially in the hippocampus and prefrontal cortex) has antidepressant effects. By directly amplifying postsynaptic currents through AMPA receptors, AMPAkines have been shown to treat depression in animal models.74  Our results show that this antidepressant effect of AMPAkines is preserved in pain models.

Distinct molecular mechanisms are found in the maintenance of neuropathic and inflammatory pain. The dose requirement for CX546 is higher in the CFA model than the SNI model, and the maximal anti-allodynia effect, is also quantitatively lower. Qualitatively, however, CX546 clearly has anti-nociceptive and antidepressant effects in both models. Our results, therefore, suggest a shared analgesic mechanism mediated by AMPA receptors in both neuropathic and inflammatory pain states.

In comparison to our finding here, previous studies on AMPAkines failed to demonstrate pro- or anti-nociceptive effects.26,28  However, these studies were performed in wild-type rodents and healthy human subjects with acute stimuli. In one study, nociceptive threshold was tested in rats by withdrawal to acute thermal stimuli.26  In another study, pain was tested by electric shock and thermal stimuli in healthy volunteers.28  The difference between these findings and ours may be due to the mechanistic differences between acute and chronic pain. Synaptic plasticity in the CNS is thought to mediate the transition from acute to chronic pain,46,75  and a general feature of synaptic plasticity is the amplification of excitatory postsynaptic currents conducted through AMPA receptors.76  Thus, it is not surprising that while potentiating these receptors in acute pain has an insignificant effect,26,28  it has a more profound effect in chronic pain states.

Both withdrawal tests and the FST can be confounded by locomotor deficits. Worse performance on the FST may reflect a decrease in locomotion due to movement-induced pain or neuropathy. Our locomotion tests, however, do not show any deficits, compatible with previous findings.44  If rats do not demonstrate locomotor deficits above 30 min, it is unlikely that they will have deficits while swimming for 5 min during the FST. Thus, results on the FST likely reflect the phenotype of depression, rather than deficiencies in locomotion. In addition, compatible with our findings, other studies on AMPAkines also demonstrate no effects on locomotion.21 

The timing of development of affective pain symptoms in rodents is not completely known. In our study, immobility on the FST manifested 2 weeks after SNI, compatible with two previous reports.42,44  In another study using this model, however, deficits on the FST were detected at 7 weeks, but data from previous time points were not shown.77  In the spinal nerve ligation model, Kontinen et al.78  reported no changes in immobility 14 days after surgery, whereas Suzuki et al.79  found increased immobility 15 days after surgery. In another study, rats developed immobility 7 days after chronic constriction injury.80  Sham surgery can induce transient reversible depressive pain symptoms, likely caused by acute pain from skin or muscle incision.42  Thus, depending on the rate of recovery from sham surgeries, the detectable differences on the FST between neuropathy and control groups at previous time points may be masked by acute pain-induced changes in the control group. In terms of inflammatory pain, we found the rats developed deficits on the FST 1 week after subcutaneous CFA injections. Similar to what we observe here, another study using CFA to induce peripheral inflammatory pain also demonstrated deficits on the FST within a week after pain induction.81  A study on monoarthritic pain, meanwhile, found deficits at 4 weeks after CFA injection into the tibiotarsal joint.82  In that study, however, injection was deep into the joint. Thus, the exact onset of affective pain symptoms may vary depending on the nature of pain models.

In our study, we used DMSO to deliver AMPAkines according to the manufacturer’s recommendation, as has been done in previous studies.27  In our study, there were small differences in the average allodynia measures for the SNI group without any treatment (fig. 1, A and B) and the group that received DMSO (fig. 2, A and B). These differences were small, however, compared with the changes in allodynia as the result of AMPAkine treatment (fig. 2, A and B). We did not observe any anti-allodynic effect for DMSO in the CFA model (fig. 5A and, 6A). To make sure that the use of DMSO as a solvent would not confound our findings, we used DMSO as control throughout our study. Thus, the minor anti-allodynic effect of DMSO should not have affected our results that the AMPAkines have significant pain-relieving properties.

A potential side effect of AMPAkines is CNS hyperexcitability owing to increased AMPA receptor activities.19,20  AMPAkines are classified into two classes: low- and high-impact AMPAkines. Low-impact AMPAkines (e.g. CX717 and CX1739) are less likely to cause hyperexcitability and have a broader therapeutic window. High-impact AMPAkines are more potent, but they have a narrower therapeutic window. We used low doses of high-impact AMPAkines and did not observe any overt behavioral deficits. Nevertheless, in the future, it will be important to investigate the pain-inhibiting properties of low-impact AMPAkines which have safer pharmacologic profiles.

In summary, we show that AMPAkines have novel analgesic properties in rat models of neuropathic and inflammatory pain. A combination of analgesic and respiratory stimulatory properties can make AMPAkines ideal drugs for the treatment of persistent postoperative and chronic pain.

This work was supported by the National Institute for General Medical Sciences (GM102691) (Bethesda, Maryland) and the Anesthesia Research Fund of the New York University Department of Anesthesiology (New York, New York).

Dr. Wang has filed a patent for the use of AMPA receptor potentiation in the treatment of pain and pain-induced depression. The other authors declare no competing financial interests.

1.
Dahan
A
,
Overdyk
F
,
Smith
T
,
Aarts
L
,
Niesters
M
:
Pharmacovigilance: A review of opioid-induced respiratory depression in chronic pain patients.
Pain Physician
2013
;
16
:
E85
94
2.
Dahan
A
,
Aarts
L
,
Smith
TW
:
Incidence, reversal, and prevention of opioid-induced respiratory depression.
Anesthesiology
2010
;
112
:
226
38
3.
Benyamin
R
,
Trescot
AM
,
Datta
S
,
Buenaventura
R
,
Adlaka
R
,
Sehgal
N
,
Glaser
SE
,
Vallejo
R
:
Opioid complications and side effects.
Pain Physician
2008
;
11
(
2 suppl
):
S105
20
4.
Paulozzi
LJ
,
Budnitz
DS
,
Xi
Y
:
Increasing deaths from opioid analgesics in the United States.
Pharmacoepidemiol Drug Saf
2006
;
15
:
618
27
5.
Song
I
,
Huganir
RL
:
Regulation of AMPA receptors during synaptic plasticity.
Trends Neurosci
2002
;
25
:
578
88
6.
Park
JS
,
Yaster
M
,
Guan
X
,
Xu
JT
,
Shih
MH
,
Guan
Y
,
Raja
SN
,
Tao
YX
:
Role of spinal cord alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors in complete Freund’s adjuvant-induced inflammatory pain.
Mol Pain
2008
;
4
:
67
7.
Stanfa
LC
,
Dickenson
AH
:
The role of non-N-methyl-.
d
-aspartate ionotropic glutamate receptors in the spinal transmission of nociception in normal animals and animals with carrageenan inflammation.
Neuroscience
1999
;
93
:
1391
8
8.
Hartmann
B
,
Ahmadi
S
,
Heppenstall
PA
,
Lewin
GR
,
Schott
C
,
Borchardt
T
,
Seeburg
PH
,
Zeilhofer
HU
,
Sprengel
R
,
Kuner
R
:
The AMPA receptor subunits GluR-A and GluR-B reciprocally modulate spinal synaptic plasticity and inflammatory pain.
Neuron
2004
;
44
:
637
50
9.
Katano
T
,
Furue
H
,
Okuda-Ashitaka
E
,
Tagaya
M
,
Watanabe
M
,
Yoshimura
M
,
Ito
S
:
N-ethylmaleimide-sensitive fusion protein (NSF) is involved in central sensitization in the spinal cord through GluR2 subunit composition switch after inflammation.
Eur J Neurosci
2008
;
27
:
3161
70
10.
Lu
Y
,
Sun
YN
,
Wu
X
,
Sun
Q
,
Liu
FY
,
Xing
GG
,
Wan
Y
:
Role of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptor subunit GluR1 in spinal dorsal horn in inflammatory nociception and neuropathic nociception in rat.
Brain Res
2008
;
1200
:
19
26
11.
Chen
J
,
Song
Y
,
Yang
J
,
Zhang
Y
,
Zhao
P
,
Zhu
XJ
,
Su
HC
:
The contribution of TNF-α in the amygdala to anxiety in mice with persistent inflammatory pain.
Neurosci Lett
2013
;
541
:
275
80
12.
Li
W
,
Neugebauer
V
:
Block of NMDA and non-NMDA receptor activation results in reduced background and evoked activity of central amygdala neurons in a model of arthritic pain.
Pain
2004
;
110
:
112
22
13.
Xu
H
,
Wu
LJ
,
Wang
H
,
Zhang
X
,
Vadakkan
KI
,
Kim
SS
,
Steenland
HW
,
Zhuo
M
:
Presynaptic and postsynaptic amplifications of neuropathic pain in the anterior cingulate cortex.
J Neurosci
2008
;
28
:
7445
53
14.
van Praag
H
,
Frenk
H
:
The role of glutamate in opiate descending inhibition of nociceptive spinal reflexes.
Brain Res
1990
;
524
:
101
5
15.
Spinella
M
,
Cooper
ML
,
Bodnar
RJ
:
Excitatory amino acid antagonists in the rostral ventromedial medulla inhibit mesencephalic morphine analgesia in rats.
Pain
1996
;
64
:
545
52
16.
Urban
MO
,
Coutinho
SV
,
Gebhart
GF
:
Involvement of excitatory amino acid receptors and nitric oxide in the rostral ventromedial medulla in modulating secondary hyperalgesia produced by mustard oil.
Pain
1999
;
81
:
45
55
17.
Ghalandari-Shamami
M
,
Hassanpour-Ezatti
M
,
Haghparast
A
:
Intra-accumbal NMDA but not AMPA/kainate receptor antagonist attenuates WIN55,212-2 cannabinoid receptor agonist-induced antinociception in the basolateral amygdala in a rat model of acute pain.
Pharmacol Biochem Behav
2011
;
100
:
213
9
18.
Gear
RW
,
Aley
KO
,
Levine
JD
:
Pain-induced analgesia mediated by mesolimbic reward circuits.
J Neurosci
1999
;
19
:
7175
81
19.
Arai
AC
,
Kessler
M
:
Pharmacology of ampakine modulators: From AMPA receptors to synapses and behavior.
Curr Drug Targets
2007
;
8
:
583
602
20.
Lynch
G
:
Glutamate-based therapeutic approaches: Ampakines.
Curr Opin Pharmacol
2006
;
6
:
82
8
21.
Lynch
G
,
Gall
CM
:
Ampakines and the threefold path to cognitive enhancement.
Trends Neurosci
2006
;
29
:
554
62
22.
Tuominen
HJ
,
Tiihonen
J
,
Wahlbeck
K
:
Glutamatergic drugs for schizophrenia: A systematic review and meta-analysis.
Schizophr Res
2005
;
72
:
225
34
23.
Skolnick
P
,
Popik
P
,
Trullas
R
:
Glutamate-based antidepressants: 20 years on.
Trends Pharmacol Sci
2009
;
30
:
563
9
24.
Zheng
YW
,
Balabhadrapatruni
S
,
Masumura
C
,
Darlington
CL
,
Smith
PF
:
Effects of the putative cognitive-enhancing ampakine, CX717, on attention and object recognition memory.
Curr Alzheimer Res
2011
;
8
:
876
82
25.
Simmons
DA
,
Rex
CS
,
Palmer
L
,
Pandyarajan
V
,
Fedulov
V
,
Gall
CM
,
Lynch
G
:
Up-regulating BDNF with an ampakine rescues synaptic plasticity and memory in Huntington’s disease knockin mice.
Proc Natl Acad Sci U S A
2009
;
106
:
4906
11
26.
Ren
J
,
Ding
X
,
Funk
GD
,
Greer
JJ
:
Ampakine CX717 protects against fentanyl-induced respiratory depression and lethal apnea in rats.
Anesthesiology
2009
;
110
:
1364
70
27.
Ren
J
,
Poon
BY
,
Tang
Y
,
Funk
GD
,
Greer
JJ
:
Ampakines alleviate respiratory depression in rats.
Am J Respir Crit Care Med
2006
;
174
:
1384
91
28.
Oertel
BG
,
Felden
L
,
Tran
PV
,
Bradshaw
MH
,
Angst
MS
,
Schmidt
H
,
Johnson
S
,
Greer
JJ
,
Geisslinger
G
,
Varney
MA
,
Lötsch
J
:
Selective antagonism of opioid-induced ventilatory depression by an ampakine molecule in humans without loss of opioid analgesia.
Clin Pharmacol Ther
2010
;
87
:
204
11
29.
Decosterd
I
,
Woolf
CJ
:
Spared nerve injury: An animal model of persistent peripheral neuropathic pain.
Pain
2000
;
87
:
149
58
30.
Ruda
MA
,
Iadarola
MJ
,
Cohen
LV
,
Young
WS
3rd
:
In situ hybridization histochemistry and immunocytochemistry reveal an increase in spinal dynorphin biosynthesis in a rat model of peripheral inflammation and hyperalgesia.
Proc Natl Acad Sci U S A
1988
;
85
:
622
6
31.
Iadarola
MJ
,
Brady
LS
,
Draisci
G
,
Dubner
R
:
Enhancement of dynorphin gene expression in spinal cord following experimental inflammation: Stimulus specificity, behavioral parameters and opioid receptor binding.
Pain
1988
;
35
:
313
26
32.
Nagarajan
N
,
Quast
C
,
Boxall
AR
,
Shahid
M
,
Rosenmund
C
:
Mechanism and impact of allosteric AMPA receptor modulation by the ampakine CX546.
Neuropharmacology
2001
;
41
:
650
63
33.
Ogier
M
,
Wang
H
,
Hong
E
,
Wang
Q
,
Greenberg
ME
,
Katz
DM
:
Brain-derived neurotrophic factor expression and respiratory function improve after ampakine treatment in a mouse model of Rett syndrome.
J Neurosci
2007
;
27
:
10912
7
34.
Silverman
JL
,
Oliver
CF
,
Karras
MN
,
Gastrell
PT
,
Crawley
JN
:
AMPAKINE enhancement of social interaction in the BTBR mouse model of autism.
Neuropharmacology
2013
;
64
:
268
82
35.
Procaccini
C
,
Aitta-aho
T
,
Jaako-Movits
K
,
Zharkovsky
A
,
Panhelainen
A
,
Sprengel
R
,
Linden
AM
,
Korpi
ER
:
Excessive novelty-induced c-Fos expression and altered neurogenesis in the hippocampus of GluA1 knockout mice.
Eur J Neurosci
2011
;
33
:
161
74
36.
Arai
A
,
Kessler
M
,
Rogers
G
,
Lynch
G
:
Effects of a memory-enhancing drug on.
dl
-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor currents and synaptic transmission in hippocampus.
J Pharmacol Exp Ther
1996
;
278
:
627
38
37.
Lynch
G
,
Kessler
M
,
Rogers
G
,
Ambros-Ingerson
J
,
Granger
R
,
Schehr
RS
:
Psychological effects of a drug that facilitates brain AMPA receptors.
Int Clin Psychopharmacol
1996
;
11
:
13
9
38.
Bourquin
AF
,
Süveges
M
,
Pertin
M
,
Gilliard
N
,
Sardy
S
,
Davison
AC
,
Spahn
DR
,
Decosterd
I
:
Assessment and analysis of mechanical allodynia-like behavior induced by spared nerve injury (SNI) in the mouse.
Pain
2006
;
122
:
14.e1
14
39.
Chaplan
SR
,
Bach
FW
,
Pogrel
JW
,
Chung
JM
,
Yaksh
TL
:
Quantitative assessment of tactile allodynia in the rat paw.
J Neurosci Methods
1994
;
53
:
55
63
40.
Hao
JX
,
Shi
TJ
,
Xu
IS
,
Kaupilla
T
,
Xu
XJ
,
Hökfelt
T
,
Bartfai
T
,
Wiesenfeld-Hallin
Z
:
Intrathecal galanin alleviates allodynia-like behaviour in rats after partial peripheral nerve injury.
Eur J Neurosci
1999
;
11
:
427
32
41.
Jørum
E
,
Warncke
T
,
Stubhaug
A
:
Cold allodynia and hyperalgesia in neuropathic pain: The effect of N-methyl-.
d
-aspartate (NMDA) receptor antagonist ketamine–a double-blind, cross-over comparison with alfentanil and placebo.
Pain
2003
;
101
:
229
35
42.
Wang
J
,
Goffer
Y
,
Xu
D
,
Tukey
DS
,
Shamir
DB
,
Eberle
SE
,
Zou
AH
,
Blanck
TJ
,
Ziff
EB
:
A single subanesthetic dose of ketamine relieves depression-like behaviors induced by neuropathic pain in rats.
Anesthesiology
2011
;
115
:
812
21
43.
Cabeza de Vaca
S
,
Carr
KD
:
Food restriction enhances the central rewarding effect of abused drugs.
J Neurosci
1998
;
18
:
7502
10
44.
Goffer
Y
,
Xu
D
,
Eberle
SE
,
D’amour
J
,
Lee
M
,
Tukey
D
,
Froemke
RC
,
Ziff
EB
,
Wang
J
:
Calcium-permeable AMPA receptors in the nucleus accumbens regulate depression-like behaviors in the chronic neuropathic pain state.
J Neurosci
2013
;
33
:
19034
44
45.
Nestler
EJ
,
Hyman
SE
:
Animal models of neuropsychiatric disorders.
Nat Neurosci
2010
;
13
:
1161
9
46.
von Hehn
CA
,
Baron
R
,
Woolf
CJ
:
Deconstructing the neuropathic pain phenotype to reveal neural mechanisms.
Neuron
2012
;
73
:
638
52
47.
Greer
JJ
,
Smith
JC
,
Feldman
JL
:
Role of excitatory amino acids in the generation and transmission of respiratory drive in neonatal rat.
J Physiol
1991
;
437
:
727
49
48.
Funk
GD
,
Smith
JC
,
Feldman
JL
:
Generation and transmission of respiratory oscillations in medullary slices: Role of excitatory amino acids.
J Neurophysiol
1993
;
70
:
1497
515
49.
Pace
RW
,
Mackay
DD
,
Feldman
JL
,
Del Negro
CA
:
Inspiratory bursts in the preBötzinger complex depend on a calcium-activated non-specific cation current linked to glutamate receptors in neonatal mice.
J Physiol
2007
;
582
(
pt 1
):
113
25
50.
Greer
JJ
,
Ren
J
:
Ampakine therapy to counter fentanyl-induced respiratory depression.
Respir Physiol Neurobiol
2009
;
168
:
153
7
51.
Ren
J
,
Lenal
F
,
Yang
M
,
Ding
X
,
Greer
JJ
:
Coadministration of the AMPAKINE CX717 with propofol reduces respiratory depression and fatal apneas.
Anesthesiology
2013
;
118
:
1437
45
52.
Fields
HL
,
Anderson
SD
,
Clanton
CH
,
Basbaum
AI
:
Nucleus raphe magnus: A common mediator of opiate- and stimulus-produced analgesia.
Trans Am Neurol Assoc
1976
;
101
:
208
10
53.
Basbaum
AI
,
Fields
HL
:
Endogenous pain control systems: Brainstem spinal pathways and endorphin circuitry.
Annu Rev Neurosci
1984
;
7
:
309
38
54.
Heinricher
MM
,
Tavares
I
,
Leith
JL
,
Lumb
BM
:
Descending control of nociception: Specificity, recruitment and plasticity.
Brain Res Rev
2009
;
60
:
214
25
55.
Morgan
MM
,
Whittier
KL
,
Hegarty
DM
,
Aicher
SA
:
Periaqueductal gray neurons project to spinally projecting GABAergic neurons in the rostral ventromedial medulla.
Pain
2008
;
140
:
376
86
56.
Behbehani
MM
,
Fields
HL
:
Evidence that an excitatory connection between the periaqueductal gray and nucleus raphe magnus mediates stimulation produced analgesia.
Brain Res
1979
;
170
:
85
93
57.
Jensen
TS
,
Yaksh
TL
:
Spinal monoamine and opiate systems partly mediate the antinociceptive effects produced by glutamate at brainstem sites.
Brain Res
1984
;
321
:
287
97
58.
Urca
G
,
Nahin
RL
,
Liebeskind
JC
:
Glutamate-induced analgesia: Blockade and potentiation by naloxone.
Brain Res
1980
;
192
:
523
30
59.
Guan
Y
,
Guo
W
,
Zou
SP
,
Dubner
R
,
Ren
K
:
Inflammation-induced upregulation of AMPA receptor subunit expression in brain stem pain modulatory circuitry.
Pain
2003
;
104
:
401
13
60.
Guan
Y
,
Guo
W
,
Robbins
MT
,
Dubner
R
,
Ren
K
:
Changes in AMPA receptor phosphorylation in the rostral ventromedial medulla after inflammatory hyperalgesia in rats.
Neurosci Lett
2004
;
366
:
201
5
61.
Ho
YC
,
Cheng
JK
,
Chiou
LC
:
Hypofunction of glutamatergic neurotransmission in the periaqueductal gray contributes to nerve-injury-induced neuropathic pain.
J Neurosci
2013
;
33
:
7825
36
62.
Guan
Y
,
Terayama
R
,
Dubner
R
,
Ren
K
:
Plasticity in excitatory amino acid receptor-mediated descending pain modulation after inflammation.
J Pharmacol Exp Ther
2002
;
300
:
513
20
63.
Park
JS
,
Voitenko
N
,
Petralia
RS
,
Guan
X
,
Xu
JT
,
Steinberg
JP
,
Takamiya
K
,
Sotnik
A
,
Kopach
O
,
Huganir
RL
,
Tao
YX
:
Persistent inflammation induces GluR2 internalization via NMDA receptor-triggered PKC activation in dorsal horn neurons.
J Neurosci
2009
;
29
:
3206
19
64.
Li
XY
,
Ko
HG
,
Chen
T
,
Descalzi
G
,
Koga
K
,
Wang
H
,
Kim
SS
,
Shang
Y
,
Kwak
C
,
Park
SW
,
Shim
J
,
Lee
K
,
Collingridge
GL
,
Kaang
BK
,
Zhuo
M
:
Alleviating neuropathic pain hypersensitivity by inhibiting PKMzeta in the anterior cingulate cortex.
Science
2010
;
330
:
1400
4
65.
Ji
G
,
Sun
H
,
Fu
Y
,
Li
Z
,
Pais-Vieira
M
,
Galhardo
V
,
Neugebauer
V
:
Cognitive impairment in pain through amygdala-driven prefrontal cortical deactivation.
J Neurosci
2010
;
30
:
5451
64
66.
Montgomery
KE
,
Kessler
M
,
Arai
AC
:
Modulation of agonist binding to AMPA receptors by 1-(1,4-benzodioxan-6-ylcarbonyl)piperidine (CX546): Differential effects across brain regions and GluA1-4/transmembrane AMPA receptor regulatory protein combinations.
J Pharmacol Exp Ther
2009
;
331
:
965
74
67.
Dworkin
RH
,
Gitlin
MJ
:
Clinical aspects of depression in chronic pain patients.
Clin J Pain
1991
;
7
:
79
94
68.
Miller
LR
,
Cano
A
:
Comorbid chronic pain and depression: Who is at risk?
J Pain
2009
;
10
:
619
27
69.
Sanacora
G
,
Treccani
G
,
Popoli
M
:
Towards a glutamate hypothesis of depression: An emerging frontier of neuropsychopharmacology for mood disorders.
Neuropharmacology
2012
;
62
:
63
77
70.
Chandran
A
,
Iyo
AH
,
Jernigan
CS
,
Legutko
B
,
Austin
MC
,
Karolewicz
B
:
Reduced phosphorylation of the mTOR signaling pathway components in the amygdala of rats exposed to chronic stress.
Prog Neuropsychopharmacol Biol Psychiatry
2013
;
40
:
240
5
71.
Duric
V
,
Banasr
M
,
Stockmeier
CA
,
Simen
AA
,
Newton
SS
,
Overholser
JC
,
Jurjus
GJ
,
Dieter
L
,
Duman
RS
:
Altered expression of synapse and glutamate related genes in post-mortem hippocampus of depressed subjects.
Int J Neuropsychopharmacol
2013
;
16
:
69
82
72.
Yuen
EY
,
Wei
J
,
Liu
W
,
Zhong
P
,
Li
X
,
Yan
Z
:
Repeated stress causes cognitive impairment by suppressing glutamate receptor expression and function in prefrontal cortex.
Neuron
2012
;
73
:
962
77
73.
Li
N
,
Lee
B
,
Liu
RJ
,
Banasr
M
,
Dwyer
JM
,
Iwata
M
,
Li
XY
,
Aghajanian
G
,
Duman
RS
:
mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists.
Science
2010
;
329
:
959
64
74.
Skolnick
P
:
AMPA receptors: A target for novel antidepressants?
Biol Psychiatry
2008
;
63
:
347
8
75.
Latremoliere
A
,
Woolf
CJ
:
Central sensitization: A generator of pain hypersensitivity by central neural plasticity.
J Pain
2009
;
10
:
895
926
76.
Kessels
HW
,
Malinow
R
:
Synaptic AMPA receptor plasticity and behavior.
Neuron
2009
;
61
:
340
50
77.
Gonçalves
L
,
Silva
R
,
Pinto-Ribeiro
F
,
Pêgo
JM
,
Bessa
JM
,
Pertovaara
A
,
Sousa
N
,
Almeida
A
:
Neuropathic pain is associated with depressive behaviour and induces neuroplasticity in the amygdala of the rat.
Exp Neurol
2008
;
213
:
48
56
78.
Kontinen
VK
,
Kauppila
T
,
Paananen
S
,
Pertovaara
A
,
Kalso
E
:
Behavioural measures of depression and anxiety in rats with spinal nerve ligation-induced neuropathy.
Pain
1999
;
80
:
341
6
79.
Suzuki
T
,
Amata
M
,
Sakaue
G
,
Nishimura
S
,
Inoue
T
,
Shibata
M
,
Mashimo
T
:
Experimental neuropathy in mice is associated with delayed behavioral changes related to anxiety and depression.
Anesth Analg
2007
;
104
:
1570
7
80.
Zeng
Q
,
Wang
S
,
Lim
G
,
Yang
L
,
Mao
J
,
Sung
B
,
Chang
Y
,
Lim
JA
,
Guo
G
,
Mao
J
:
Exacerbated mechanical allodynia in rats with depression-like behavior.
Brain Res
2008
;
1200
:
27
38
81.
Kim
H
,
Chen
L
,
Lim
G
,
Sung
B
,
Wang
S
,
McCabe
MF
,
Rusanescu
G
,
Yang
L
,
Tian
Y
,
Mao
J
:
Brain indoleamine 2,3-dioxygenase contributes to the comorbidity of pain and depression.
J Clin Invest
2012
;
122
:
2940
54
82.
Borges
G
,
Neto
F
,
Mico
JA
,
Berrocoso
E
:
Reversal of monoarthritis-induced affective disorders by diclofenac in rats.
Anesthesiology
2014
;
120
:
1476
90