Inhibition of AAK1 Kinase as a Novel Therapeutic Approach to Treat Neuropathic Pain

Walter Kostich, Brian D Hamman, Yu-Wen Li, Sreenivasulu Naidu, Kumaran Dandapani, Jianlin Feng, Amy Easton, Clotilde Bourin, Kevin Baker, Jason Allen, Katerina Savelieva, Justin V Louis, Manoj Dokania, Saravanan Elavazhagan, Pradeep Vattikundala, Vivek Sharma, Manish Lal Das, Ganesh Shankar, Anoop Kumar, Vinay K Holenarsipur, Michael Gulianello, Ted Molski, Jeffrey M Brown, Martin Lewis, Yanling Huang, Yifeng Lu, Rick Pieschl, Kevin O'Malley, Jonathan Lippy, Amr Nouraldeen, Thomas H Lanthorn, Guilan Ye, Alan Wilson, Anand Balakrishnan, Rex Denton, James E Grace, Kimberley A Lentz, Kenneth S Santone, Yingzhi Bi, Alan Main, Jon Swaffield, Ken Carson, Sandhya Mandlekar, Reeba K Vikramadithyan, Susheel J Nara, Carolyn Dzierba, Joanne Bronson, John E Macor, Robert Zaczek, Ryan Westphal, Laszlo Kiss, Linda Bristow, Charles M Conway, Brian Zambrowicz, Charles F Albright, Walter Kostich, Brian D Hamman, Yu-Wen Li, Sreenivasulu Naidu, Kumaran Dandapani, Jianlin Feng, Amy Easton, Clotilde Bourin, Kevin Baker, Jason Allen, Katerina Savelieva, Justin V Louis, Manoj Dokania, Saravanan Elavazhagan, Pradeep Vattikundala, Vivek Sharma, Manish Lal Das, Ganesh Shankar, Anoop Kumar, Vinay K Holenarsipur, Michael Gulianello, Ted Molski, Jeffrey M Brown, Martin Lewis, Yanling Huang, Yifeng Lu, Rick Pieschl, Kevin O'Malley, Jonathan Lippy, Amr Nouraldeen, Thomas H Lanthorn, Guilan Ye, Alan Wilson, Anand Balakrishnan, Rex Denton, James E Grace, Kimberley A Lentz, Kenneth S Santone, Yingzhi Bi, Alan Main, Jon Swaffield, Ken Carson, Sandhya Mandlekar, Reeba K Vikramadithyan, Susheel J Nara, Carolyn Dzierba, Joanne Bronson, John E Macor, Robert Zaczek, Ryan Westphal, Laszlo Kiss, Linda Bristow, Charles M Conway, Brian Zambrowicz, Charles F Albright

Abstract

To identify novel targets for neuropathic pain, 3097 mouse knockout lines were tested in acute and persistent pain behavior assays. One of the lines from this screen, which contained a null allele of the adapter protein-2 associated kinase 1 (AAK1) gene, had a normal response in acute pain assays (hot plate, phase I formalin), but a markedly reduced response to persistent pain in phase II formalin. AAK1 knockout mice also failed to develop tactile allodynia following the Chung procedure of spinal nerve ligation (SNL). Based on these findings, potent, small-molecule inhibitors of AAK1 were identified. Studies in mice showed that one such inhibitor, LP-935509, caused a reduced pain response in phase II formalin and reversed fully established pain behavior following the SNL procedure. Further studies showed that the inhibitor also reduced evoked pain responses in the rat chronic constriction injury (CCI) model and the rat streptozotocin model of diabetic peripheral neuropathy. Using a nonbrain-penetrant AAK1 inhibitor and local administration of an AAK1 inhibitor, the relevant pool of AAK1 for antineuropathic action was found to be in the spinal cord. Consistent with these results, AAK1 inhibitors dose-dependently reduced the increased spontaneous neural activity in the spinal cord caused by CCI and blocked the development of windup induced by repeated electrical stimulation of the paw. The mechanism of AAK1 antinociception was further investigated with inhibitors of α2 adrenergic and opioid receptors. These studies showed that α2 adrenergic receptor inhibitors, but not opioid receptor inhibitors, not only prevented AAK1 inhibitor antineuropathic action in behavioral assays, but also blocked the AAK1 inhibitor-induced reduction in spinal neural activity in the rat CCI model. Hence, AAK1 inhibitors are a novel therapeutic approach to neuropathic pain with activity in animal models that is mechanistically linked (behaviorally and electrophysiologically) to α2 adrenergic signaling, a pathway known to be antinociceptive in humans.

Copyright © 2016 The Author(s).

Figures

Fig. 1.
Fig. 1.
AAK1 knockout mice have an antinociceptive phenotype in the formalin assay and SNL model. (A) Hind paw flinches in formalin phase I and phase II from wild-type (WT) and AAK1 knockout (AAK1 KO) mice (n = 25–56 per group, male and female mice distributed equally between groups). ***P < 0.001 versus WT by two-way repeated measures analysis of variance, followed by Bonferroni’s post hoc test. (B) Distance traveled in open field testing of WT (n = 15) and AAK1 KO mice (n = 20). Male and female mice were used and distributed equally between groups. P = 0.27 unpaired t test. (C) Mechanical allodynia measured using manual von Frey (mVF) fibers after SNL surgery for WT (n = 15) and AAK1 KO mice (n = 22). Male and female mice were used and distributed equally between groups. ***P < 0.001 versus WT by two-way repeated measures analysis of variance, followed by Bonferroni’s post hoc test.
Fig. 2.
Fig. 2.
Chemical structure of AAK1 inhibitors. (A) LP-935509. (B) BMT-090605. (C) LP-922761. (D) BMT-124110 (E) LP-927443. (F) BMS-901715.
Fig. 3.
Fig. 3.
AAK1 inhibitor LP-935509 recapitulates the knockout phenotype in mice. (A) Mouse phase II formalin responses after oral vehicle, gabapentin (GBP) (200 mg/kg), or LP-935509 (10, 30, or 60 mg/kg) (n = 8–10 male mice per group). **P < 0.01 versus Veh by one-way analysis of variance, followed by Dunnett’s t test. (B) Mechanical allodynia over time postdose [doses as in (A)] in mice with SNL injury after oral vehicle, GBP, or LP-935509 (n = 9–13 male mice per group). ***P < 0.001 versus vehicle by repeated measures analysis of variance, followed by Dunnett’s t test. (C) Open field locomotor activity of mice at 30 minutes after oral [doses as in (A)] vehicle, GBP, or LP-935509 (n = 7 male mice per group). P = 0.27 by one-way analysis of variance.
Fig. 4.
Fig. 4.
AAK1 inhibitor LP-935509 efficacy in SNL model follows plasma exposure. Mouse SNL mechanical allodynia responses after oral vehicle, gabapentin (GBP) (200 mg/kg), or LP-935509 (3, 10, or 30 mg/kg) (n = 4–7 male mice per group). *P < 0.05 and ***P < 0.001 versus vehicle by repeated measures analysis of variance, followed by Dunnett’s t test.
Fig. 5.
Fig. 5.
AAK1 inhibitor LP-935509 efficacy in SNL model does not change with repeat dosing. Mouse SNL mechanical allodynia responses with twice-daily dosing over 5 consecutive days with oral vehicle, gabapentin (200 mg/kg), or LP-935509 (3, 10, or 30 mg/kg) (n = 4–7 male mice per group). *P < 0.05, **P < 0.01, and ***P < 0.001 versus vehicle by repeated measures analysis of variance, followed by Dunnett’s t test.
Fig. 6.
Fig. 6.
AAK1 inhibitor LP-935509 is antinociceptive in multiple rat models of neuropathic pain, but not acute pain (A–D). CCI-operated rats were tested at 3 hours after oral vehicle (0), LP-935509 (at indicated doses), or gabapentin (GBP; 100 mg/kg) in assays of (A) thermal hyperalgesia, (B) cold allodynia, (C) mechanical allodynia using Manual von Frey, or (D) mechanical hyperalgesia (n = 7–8 male rats per group). For comparison, sham-operated animals (Sham) are included. (E) Naive rats were testing in the accelerating rotarod assay, dosed as in Fig. 4, (n = 5–10 male rats per group). (F) STZ-injured rats were tested for mechanical allodynia, dosed as in Fig. 4, (n = 7–8 male rats per group). For comparison, sham-injected animals (Sham) are included. (G and H) Naive rats were tested in assays of acute pain, including the following: (G) hot plate and (H) tail flick at 1.5 hours after LP-935509 (30 mg/kg, Per Os po) or 0.5 hour after morphine (5 mg/kg, s.c.) (n = 7–8 male rats per group). ***P < 0.001 versus vehicle, or †††P < 0.001 versus sham controls by one-way analysis of variance, followed by Bonferroni’s post hoc test; ###P < 0.001 versus vehicle by paired t test.
Fig. 7.
Fig. 7.
AAK1 inhibitors cause antinociception by inhibiting AAK1 in the spinal cord. (A) Mice with SNL injury were tested for thermal hyperalgesia 2 hours after oral vehicle, LP-922761 (poorly brain-penetrant AAK1 inhibitor), or gabapentin (GBP) at indicated doses (n = 7–8 male rats per group). (B) Rats with CCI surgery were tested for thermal hyperalgesia 15 minutes after intrathecal vehicle (0), BMT-090605 (0.3–3 µg/rat), or clonidine (Clon, 3 µg) (n = 7–8 male rats per group). For comparison, sham-operated animals (Sham) are included. **P < 0.01 ***P < 0.001 versus vehicle/0, †††P < 0.001 versus Sham by one-way analysis of variance, followed by Bonferroni’s post hoc test.
Fig. 8.
Fig. 8.
AAK1 inhibitors decrease neural activity in pain-related circuits. (A) CCI rat spontaneous activity over time of a single spinal dorsal horn neuron showing inhibition by LP-935509 (1 mg/kg, i.v.). Arrow indicates time of LP-935509 injection (inset shows response to vehicle). (B) Dose-dependent inhibition by LP-935509 (0.3–3 mg/kg, i.v.) on spontaneous activity of spinal dorsal horn neurons in CCI male rats (responders, n = 3/4 at 1 mg/kg, n = 4/6 at 3 mg/kg). *P < 0.05 versus vehicle (0) by one-way analysis of variance, followed by Dunnett’s t test. (C) Absence of effect of thoracic T8–T10 transection on LP-935509 (1 mg/kg, i.v.) inhibition of spontaneous activity over time in a single spinal dorsal horn neuron of a CCI male rat (arrow indicates time of injection). (D) Effect of LP-935509 (1 mg/kg, i.v.) on normalized spontaneous activity of CCI male rats (n = 4) with thoracic T8–10 transection. *P < 0.05 versus baseline by unpaired t test. (E) Naive wide-dynamic range neuron spikes evoked by hind limb repetitive electrical stimulation (16 pulses, 2 ms, 2.0 mA) with effect of systemic vehicle or LP-935509 (1–3 mg/kg, i.v.) across pulses (n = 4–8 male rats per group). (F) Systemic inhibition by LP-935509 (0.3–3 mg/kg, i.v.) of electrically evoked windup in spinal wide-dynamic range neurons from naive male rats (responders, n = 5/8 at 1 mg/kg, n = 4/4 at 3 mg/kg). *P < 0.05 versus vehicle (0) by one-way analysis of variance, followed by Dunnett's t test. (G) Naive rat spinal wide-dynamic range neuron spikes across pulses (as in Fig. 6E) with effect of microiontophoretically-applied BMT-124110 (30 mM, pH 4, +10–30 nA, 3 minutes) (n = 10 male rats per group). (H) Spinal inhibition by BMT-124110 (as in Fig. 6G) of electrically evoked windup in dorsal horn wide-dynamic range neurons from naive male rats (responders, n = 10/14). *P < 0.05 by unpaired t test.
Fig. 9.
Fig. 9.
α2 adrenergic antagonist blocks AAK1 inhibition–induced antinociception in CCI and STZ rats. (A) STZ-treated rats were tested for mechanical allodynia using electronic von Frey 1.5 hours after vehicle (per os), LP-935509 (30 mg/kg, per os), or morphine (3 mg/kg, s.c.) delivered in the presence (+) or absence (−) of naloxone (1 mg/kg, s.c.) given 30 minutes prior to the other agents (n = 8 male rats per group). For comparison, sham-treated (CTL) animals are included. ***P < 0.001 versus vehicle by one-way analysis of variance, followed by Bonferroni’s post hoc test. (B) CCI-operated rats were tested for thermal hyperalgesia 30 minutes after tizanidine (1 mg/kg, i.p.), or 1.5 hours after either vehicle (per os) or LP-935509 (30 mg/kg, po) all delivered in the presence (+) or absence (−) of yohimbine (1 mg/kg, i.p.) given 100 minutes before testing (n = 6–11 male rats per group). CTL as in (A). ***P < 0.001 versus (−) yohimbine pair by one-way analysis of variance, followed by Bonferroni’s post hoc test. (C) STZ-treated rats were tested for mechanical allodynia with CTL and dosing as in (B) (n = 8 male rats per group). ***P < 0.001 versus (−) yohimbine as in (B).
Fig. 10.
Fig. 10.
α2 adrenergic antagonist blocks AAK1 inhibition–induced reduction in spontaneous neuronal activity in CCI rats. (A) CCI rat spontaneous activity time course of two individual spinal dorsal horn neurons treated with LP-935509 (1 mg/kg, i.v.; filled arrow) without (top trace) or with (bottom trace) pretreatment of yohimbine (0.3 mg/kg, i.v.; open arrow). (B) CCI rat group data for spontaneous activity with design and dosing as in (A) plus baseline and yohimbine controls (n = 6–8 male rats per group). *P < 0.05 versus baseline by one-way analysis of variance, followed by Dunnett's t test.

References

    1. Arendt-Nielsen L, Mansikka H, Staahl C, Rees H, Tan K, Smart TS, Monhemius R, Suzuki R, Drewes AM. (2011) A translational study of the effects of ketamine and pregabalin on temporal summation of experimental pain. Reg Anesth Pain Med 36:585–591.
    1. Beltran del Rio H, Kern F, Lanthorn T, Oravecz T, Piggott J, Powell D, Ramirez-Solis R, Sands AT, and Zambrowicz B (2003) Saturation screening of the druggable mammalian genome, in Model Organisms in Drug Discovery (Carroll P and Fitzgerald K eds) pp 251-278, Wiley & Sons, Chichester, UK.
    1. Benarroch EE. (2008) Descending monoaminergic pain modulation: bidirectional control and clinical relevance. Neurology 71:217–221.
    1. Bennett GJ, Xie YK. (1988) A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 33:87–107.
    1. Brommage R, Desai U, Revelli JP, Donoviel DB, Fontenot GK, Dacosta CM, Smith DD, Kirkpatrick LL, Coker KJ, Donoviel MS, et al. (2008) High-throughput screening of mouse knockout lines identifies true lean and obese phenotypes. Obesity 16:2362–2367.
    1. Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR, Vogel P. (2014) High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res 2:14034.
    1. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. (1994) Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 53:55–63.
    1. Chaumont S, André C, Perrais D, Boué-Grabot E, Taly A, Garret M. (2013) Agonist-dependent endocytosis of γ-aminobutyric acid type A (GABAA) receptors revealed by a γ2(R43Q) epilepsy mutation. J Biol Chem 288:28254–28265.
    1. Conner SD, Schmid SL. (2002) Identification of an adaptor-associated kinase, AAK1, as a regulator of clathrin-mediated endocytosis. J Cell Biol 156:921–929.
    1. Costigan M, Scholz J, Woolf CJ. (2009) Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci 32:1–32.
    1. Courteix C, Eschalier A, Lavarenne J. (1993) Streptozocin-induced diabetic rats: behavioural evidence for a model of chronic pain. Pain 53:81–88.
    1. D’Amour FE, Smith DL. (1941) A method for determining loss of pain sensation. J Pharmacol Exp Ther 72:74–79.
    1. Dolphin AC. (2012) Calcium channel auxiliary α2δ and β subunits: trafficking and one step beyond. Nat Rev Neurosci 13:542–555.
    1. Dubuisson D, Dennis SG. (1977) The formalin test: a quantitative study of the analgesic effects of morphine, meperidine, and brain stem stimulation in rats and cats. Pain 4:161–174.
    1. Dunham NW, Miya TS. (1957) A note on a simple apparatus for detecting neurological deficit in rats and mice. J Am Pharm Assoc Am Pharm Assoc 46:208–209.
    1. Engelman E, Marsala C. (2013) Efficacy of adding clonidine to intrathecal morphine in acute postoperative pain: meta-analysis. Br J Anaesth 110:21–27.
    1. Fairbanks CA. (2003) Spinal delivery of analgesics in experimental models of pain and analgesia. Adv Drug Deliv Rev 55:1007–1041.
    1. Fairbanks CA, Stone LS, Wilcox GL. (2009) Pharmacological profiles of alpha 2 adrenergic receptor agonists identified using genetically altered mice and isobolographic analysis. Pharmacol Ther 123:224–238.
    1. Fields H. (2004) State-dependent opioid control of pain. Nat Rev Neurosci 5:565–575.
    1. Finnerup NB, Sindrup SH, Jensen TS. (2010) The evidence for pharmacological treatment of neuropathic pain. Pain 150:573–581.
    1. Gupta-Rossi N, Ortica S, Meas-Yedid V, Heuss S, Moretti J, Olivo-Marin JC, Israël A. (2011) The adaptor-associated kinase 1, AAK1, is a positive regulator of the Notch pathway. J Biol Chem 286:18720–18730.
    1. Hargreaves K, Dubner R, Brown F, Flores C, Joris J. (1988) A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 32:77–88.
    1. Hayashida K, Obata H, Nakajima K, Eisenach JC. (2008) Gabapentin acts within the locus coeruleus to alleviate neuropathic pain. Anesthesiology 109:1077–1084.
    1. Hayashida K, Parker R, Eisenach JC. (2007) Oral gabapentin activates spinal cholinergic circuits to reduce hypersensitivity after peripheral nerve injury and interacts synergistically with oral donepezil. Anesthesiology 106:1213–1219.
    1. Hylden JL, Wilcox GL. (1980) Intrathecal morphine in mice: a new technique. Eur J Pharmacol 67:313–316.
    1. Jackson AP, Flett A, Smythe C, Hufton L, Wettey FR, Smythe E. (2003) Clathrin promotes incorporation of cargo into coated pits by activation of the AP2 adaptor micro2 kinase. J Cell Biol 163:231–236.
    1. Kearns AE, Donohue MM, Sanyal B, Demay MB. (2001) Cloning and characterization of a novel protein kinase that impairs osteoblast differentiation in vitro. J Biol Chem 276:42213–42218.
    1. Kim SH, Chung JM. (1992) An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 50:355–363.
    1. Kittler JT, Chen G, Kukhtina V, Vahedi-Faridi A, Gu Z, Tretter V, Smith KR, McAinsh K, Arancibia-Carcamo IL, Saenger W, et al. (2008) Regulation of synaptic inhibition by phospho-dependent binding of the AP2 complex to a YECL motif in the GABAA receptor gamma2 subunit. Proc Natl Acad Sci USA 105:3616–3621.
    1. Kuai L, Ong SE, Madison JM, Wang X, Duvall JR, Lewis TA, Luce CJ, Conner SD, Pearlman DA, Wood JL, et al. (2011) AAK1 identified as an inhibitor of neuregulin-1/ErbB4-dependent neurotrophic factor signaling using integrative chemical genomics and proteomics. Chem Biol 18:891–906.
    1. Laird JM, Bennett GJ. (1993) An electrophysiological study of dorsal horn neurons in the spinal cord of rats with an experimental peripheral neuropathy. J Neurophysiol 69:2072–2085.
    1. Liu C, Walker JM. (2006) Effects of a cannabinoid agonist on spinal nociceptive neurons in a rodent model of neuropathic pain. J Neurophysiol 96:2984–2994.
    1. McCarson KE, Enna SJ. (2014) GABA pharmacology: the search for analgesics. Neurochem Res 39:1948–1963.
    1. Mestre C, Pélissier T, Fialip J, Wilcox G, Eschalier A. (1994) A method to perform direct transcutaneous intrathecal injection in rats. J Pharmacol Toxicol Methods 32:197–200.
    1. Mogil JS. (2009) Animal models of pain: progress and challenges. Nat Rev Neurosci 10:283–294.
    1. Montana MC, Conrardy BA, Cavallone LF, Kolber BJ, Rao LK, Greco SC, Gereau RW., 4th (2011) Metabotropic glutamate receptor 5 antagonism with fenobam: examination of analgesic tolerance and side effect profile in mice. Anesthesiology 115:1239–1250.
    1. Morrow TJ (2004) Animal models of painful diabetic neuropathy: the STZ rat model. Curr Protoc Neurosci Chapter 9:Unit 9.18.
    1. Obata H, Saito S, Koizuka S, Nishikawa K, Goto F. (2005) The monoamine-mediated antiallodynic effects of intrathecally administered milnacipran, a serotonin noradrenaline reuptake inhibitor, in a rat model of neuropathic pain. Anesth Analg 100:1406–1410, table of contents.
    1. Powell DR, DaCosta CM, Gay J, Ding ZM, Smith M, Greer J, Doree D, Jeter-Jones S, Mseeh F, Rodriguez LA, et al. (2013) Improved glycemic control in mice lacking Sglt1 and Sglt2. Am J Physiol Endocrinol Metab 304:E117–E130.
    1. Santos-Nogueira E, Redondo Castro E, Mancuso R, Navarro X. (2012) Randall-Selitto test: a new approach for the detection of neuropathic pain after spinal cord injury. J Neurotrauma 29:898–904.
    1. Shi B, Conner SD, Liu J. (2014) Dysfunction of endocytic kinase AAK1 in ALS. Int J Mol Sci 15:22918–22932.
    1. Snedecor SJ, Sudharshan L, Cappelleri JC, Sadosky A, Desai P, Jalundhwala Y, Botteman M. (2014) Systematic review and meta-analysis of pharmacological therapies for pain associated with postherpetic neuralgia and less common neuropathic conditions. Int J Clin Pract 68:900–918.
    1. Stone LS, German JP, Kitto KF, Fairbanks CA, Wilcox GL. (2014) Morphine and clonidine combination therapy improves therapeutic window in mice: synergy in antinociceptive but not in sedative or cardiovascular effects. PLoS One 9:e109903.
    1. Tang T, Li L, Tang J, Li Y, Lin WY, Martin F, Grant D, Solloway M, Parker L, Ye W, et al. (2010) A mouse knockout library for secreted and transmembrane proteins. Nat Biotechnol 28:749–755.
    1. Tanimoto-Mori S, Nakazato-Imasato E, Toide K, Kita Y. (2008) Pharmacologic investigation of the mechanism underlying cold allodynia using a new cold plate procedure in rats with chronic constriction injuries. Behav Pharmacol 19:85–90.
    1. Teasell RW, Mehta S, Aubut JA, Foulon B, Wolfe DL, Hsieh JT, Townson AF, Short C, Spinal Cord Injury Rehabilitation Evidence Research Team (2010) A systematic review of pharmacologic treatments of pain after spinal cord injury. Arch Phys Med Rehabil 91:816–831.
    1. Traub LM. (2009) Tickets to ride: selecting cargo for clathrin-regulated internalization. Nat Rev Mol Cell Biol 10:583–596.
    1. Ultanir SK, Hertz NT, Li G, Ge WP, Burlingame AL, Pleasure SJ, Shokat KM, Jan LY, Jan YN. (2012) Chemical genetic identification of NDR1/2 kinase substrates AAK1 and Rabin8 uncovers their roles in dendrite arborization and spine development. Neuron 73:1127–1142.
    1. Wang G, Dai D, Chen X, Yuan L, Zhang A, Lu Y, Zhang P. (2014) Upregulation of neuregulin-1 reverses signs of neuropathic pain in rats. Int J Clin Exp Pathol 7:5916–5921.
    1. Watzman N, Barry H, 3rd, Buckley JP, Kinnard WJ., Jr (1964) Semiautomatic System for Timing Rotarod Performance. J Pharm Sci 53:1429–1430.
    1. Woolfe G, MacDonald AD. (1944) The evaluation of the analgesic action of pethidine hydrochloride (Demerol). J Pharmacol Exp Ther 80:300–307.
    1. Xie K, Qiao F, Sun Y, Wang G, Hou L. (2015) Notch signaling activation is critical to the development of neuropathic pain. BMC Anesthesiol 15:41.
    1. Yaksh TL, Ozaki G, McCumber D, Rathbun M, Svensson C, Malkmus S, and Yaksh MC (2001) An automated flinch detecting system for use in the formalin nociceptive bioassay. J Appl Physiol (1985) 90:2386-2402.
    1. Ye GL, Savelieva KV, Vogel P, Baker KB, Mason S, Lanthorn TH, Rajan I. (2015) Ligation of mouse L4 and L5 spinal nerves produces robust allodynia without major motor function deficit. Behav Brain Res 276:99–110.
    1. Zambrowicz BP, Sands AT. (2003) Knockouts model the 100 best-selling drugs: will they model the next 100? Nat Rev Drug Discov 2:38–51.
    1. Zeilhofer HU, Ralvenius WT, Acuña MA. (2015) Restoring the spinal pain gate: GABA(A) receptors as targets for novel analgesics. Adv Pharmacol 73:71–96.

Source: PubMed

3
Abonnieren