Intranasal CRMP2-Ubc9 Inhibitor Regulates NaV1.7 to Alleviate Trigeminal Neuropathic Pain

Dysregulation of voltage-gated sodium NaV1.7 channels in sensory neurons contributes to chronic pain conditions, including trigeminal neuropathic pain. We previously reported that chronic pain results in part from increased SUMOylation of collapsin response mediator protein 2 (CRMP2), leading to an increased CRMP2/NaV1.7 interaction and increased functional activity of NaV1.7. Targeting this feed-forward regulation, we developed compound 194, which inhibits CRMP2 SUMOylation mediated by the SUMO-conjugating enzyme Ubc9. We further demonstrated that 194 effectively reduces the functional activity of NaV1.7 channels in dorsal root ganglia neurons and alleviated inflammatory and neuropathic pain. Here, we employed a comprehensive array of investigative approaches, encompassing biochemical, pharmacological, genetic, electrophysiological, and behavioral analyses, to assess the functional implications of NaV1.7 regulation by CRMP2 in trigeminal ganglia (TG) neurons. We confirmed the expression of Scn9a, Dpysl2, and UBE2I within TG neurons. Furthermore, we found an interaction between CRMP2 and NaV1.7, with CRMP2 being SUMOylated in these sensory ganglia. Disrupting CRMP2 SUMOylation with compound 194 uncoupled the CRMP2/NaV1.7 interaction, impeded NaV1.7 diffusion on the plasma membrane, and subsequently diminished NaV1.7 activity. Compound 194 also led to a reduction in TG neuron excitability. Finally, when intranasally administered to rats with chronic constriction injury of the infraorbital nerve (CCI-ION), 194 significantly decreased nociceptive behaviors. Collectively, our findings underscore the critical role of CRMP2 in regulating NaV1.7 within TG neurons, emphasizing the importance of this indirect modulation in trigeminal neuropathic pain.

[1]  Heather N. Allen,et al.  Neuropilin-1 is essential for vascular endothelial growth factor A-mediated increase of sensory neuron activity and development of pain-like behaviors. , 2023, Pain.

[2]  G. Zamponi,et al.  Electrophysiological and computational analysis of Cav3.2 channel variants associated with familial trigeminal neuralgia , 2022, Molecular Brain.

[3]  Aida Calderón-Rivera,et al.  Cell specific regulation of NaV1.7 activity and trafficking in rat nodose ganglia neurons , 2022, Neurobiology of pain.

[4]  Romain F. Laine,et al.  TrackMate 7: integrating state-of-the-art segmentation algorithms into tracking pipelines , 2022, Nature Methods.

[5]  Peng Zhang,et al.  Prolonged PGE2 treatment increased TTX-sensitive but not TTX-resistant sodium current in trigeminal ganglionic neurons , 2022, Neuropharmacology.

[6]  G. Zamponi,et al.  CaV3.2 calcium channels contribute to trigeminal neuralgia. , 2022, Pain.

[7]  R. Khanna,et al.  Small molecule targeting NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces pain in chronic constriction injury (CCI) rats , 2022, Channels.

[8]  R. Khanna,et al.  Small molecule targeting NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces and prevents pain chronification in a mouse model of oxaliplatin-induced neuropathic pain , 2021, Neurobiology of Pain.

[9]  A. Hohmann,et al.  Selective targeting of NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces pain in rodents , 2021, Science Translational Medicine.

[10]  R. Khanna,et al.  CRMP2 Is Involved in Regulation of Mitochondrial Morphology and Motility in Neurons , 2021, Cells.

[11]  Vimala N. Bharadwaj,et al.  Intranasal Administration for Pain: Oxytocin and Other Polypeptides , 2021, Pharmaceutics.

[12]  R. Khanna,et al.  Non-SUMOylated CRMP2 decreases NaV1.7 currents via the endocytic proteins Numb, Nedd4-2 and Eps15 , 2021, Molecular brain.

[13]  J. Gerrard,et al.  Exome Sequencing Implicates Impaired GABA Signaling and Neuronal Ion Transport in Trigeminal Neuralgia , 2020, iScience.

[14]  R. Khanna,et al.  Studies on CRMP2 SUMOylation-deficient transgenic mice identify sex-specific NaV1.7 regulation in the pathogenesis of chronic neuropathic pain , 2020, bioRxiv.

[15]  R. Benoliel,et al.  Differential gene expression changes in the dorsal root versus trigeminal ganglia following peripheral nerve injury in rats , 2020, European journal of pain.

[16]  Donal N. Gorman,et al.  Lack of Detection of the Analgesic Properties of PF‐05089771, a Selective Nav1.7 Inhibitor, Using a Battery of Pain Models in Healthy Subjects , 2019, Clinical and translational science.

[17]  S. Dib-Hajj,et al.  Building sensory axons: Delivery and distribution of NaV1.7 channels and effects of inflammatory mediators , 2019, Science Advances.

[18]  R. Khanna,et al.  Mining the Nav1.7 interactome: Opportunities for chronic pain therapeutics. , 2019, Biochemical pharmacology.

[19]  A. M. Rush,et al.  The role of Nav1.7 in human nociceptors: insights from human induced pluripotent stem cell–derived sensory neurons of erythromelalgia patients , 2019, Pain.

[20]  M. Khanna,et al.  Blocking CRMP2 SUMOylation reverses neuropathic pain , 2018, Molecular Psychiatry.

[21]  Yue Wang,et al.  Inhibition of the Ubc9 E2 SUMO-conjugating enzyme–CRMP2 interaction decreases NaV1.7 currents and reverses experimental neuropathic pain , 2018, Pain.

[22]  M. Khanna,et al.  Betulinic acid, derived from the desert lavender Hyptis emoryi, attenuates paclitaxel-, HIV-, and nerve injury–associated peripheral sensory neuropathy via block of N- and T-type calcium channels , 2018, Pain.

[23]  A. McDonnell,et al.  Efficacy of the Nav1.7 blocker PF-05089771 in a randomised, placebo-controlled, double-blind clinical study in subjects with painful diabetic peripheral neuropathy , 2018, Pain.

[24]  R. Khanna,et al.  Cdk5-mediated CRMP2 phosphorylation is necessary and sufficient for peripheral neuropathic pain , 2018, Neurobiology of pain.

[25]  K. Messlinger,et al.  Current understanding of trigeminal ganglion structure and function in headache , 2018, Cephalalgia : an international journal of headache.

[26]  Yue Wang,et al.  Homology‐guided mutational analysis reveals the functional requirements for antinociceptive specificity of collapsin response mediator protein 2‐derived peptides , 2018, British journal of pharmacology.

[27]  R. Khanna,et al.  CRMP2 and voltage-gated ion channels: potential roles in neuropathic pain , 2018, Neuronal signaling.

[28]  M. Khanna,et al.  Chemical shift perturbation mapping of the Ubc9-CRMP2 interface identifies a pocket in CRMP2 amenable for allosteric modulation of Nav1.7 channels , 2018, Channels.

[29]  Ki Duk Park,et al.  CRISPR/Cas9 editing of Nf1 gene identifies CRMP2 as a therapeutic target in neurofibromatosis type 1-related pain that is reversed by (S)-Lacosamide , 2017, Pain.

[30]  Yue Wang,et al.  Dissecting the role of the CRMP2–neurofibromin complex on pain behaviors , 2017, Pain.

[31]  J. Olesen,et al.  Whole transcriptome expression of trigeminal ganglia compared to dorsal root ganglia in Rattus Norvegicus , 2017, Neuroscience.

[32]  M. Khanna,et al.  A single structurally conserved SUMOylation site in CRMP2 controls NaV1.7 function , 2017, Channels.

[33]  M. Khanna,et al.  Hierarchical CRMP2 posttranslational modifications control NaV1.7 function , 2016, Proceedings of the National Academy of Sciences.

[34]  Ki Duk Park,et al.  Efficacy of (S)-lacosamide in preclinical models of cephalic pain , 2016, Pain reports.

[35]  J. Wood,et al.  Nav1.7 and other voltage-gated sodium channels as drug targets for pain relief , 2016, Expert opinion on therapeutic targets.

[36]  M. Bruchez,et al.  Kinetically Tunable Photostability of Fluorogen-Activating Peptide-Fluorogen Complexes. , 2015, Chemphyschem : a European journal of chemical physics and physical chemistry.

[37]  M. Khanna,et al.  A membrane-delimited N-myristoylated CRMP2 peptide aptamer inhibits CaV2.2 trafficking and reverses inflammatory and postoperative pain behaviors , 2015, Pain.

[38]  Cai-yue Liu,et al.  The role of large-conductance, calcium-activated potassium channels in a rat model of trigeminal neuropathic pain , 2015, Cephalalgia : an international journal of headache.

[39]  Guy M. Hagen,et al.  ThunderSTORM: a comprehensive ImageJ plug-in for PALM and STORM data analysis and super-resolution imaging , 2014, Bioinform..

[40]  Stephen G Waxman,et al.  Regulating excitability of peripheral afferents: emerging ion channel targets , 2014, Nature Neuroscience.

[41]  Jared G. Smith,et al.  The psychosocial and affective burden of posttraumatic neuropathy following injuries to the trigeminal nerve. , 2013, Journal of orofacial pain.

[42]  T. Kanazawa,et al.  Suppression of ATP-induced excitability in rat small-diameter trigeminal ganglion neurons by activation of GABAB receptor , 2013, Brain Research Bulletin.

[43]  Sarah M. Wilson,et al.  CRMP2 Protein SUMOylation Modulates NaV1.7 Channel Trafficking* , 2013, The Journal of Biological Chemistry.

[44]  T. Kanazawa,et al.  Brain-derived neurotrophic factor enhances the excitability of small-diameter trigeminal ganglion neurons projecting to the trigeminal nucleus interpolaris/caudalis transition zone following masseter muscle inflammation , 2013, Molecular pain.

[45]  S. Dib-Hajj,et al.  The NaV1.7 sodium channel: from molecule to man , 2012, Nature Reviews Neuroscience.

[46]  Euiseong Kim,et al.  Hypoesthesia after IAN block anesthesia with lidocaine: management of mild to moderate nerve injury , 2012, Restorative dentistry & endodontics.

[47]  Y. Wang,et al.  Prevention of posttraumatic axon sprouting by blocking collapsin response mediator protein 2-mediated neurite outgrowth and tubulin polymerization , 2012, Neuroscience.

[48]  S. Meroueh,et al.  CRMP-2 peptide mediated decrease of high and low voltage-activated calcium channels, attenuation of nociceptor excitability, and anti-nociception in a model of AIDS therapy-induced painful peripheral neuropathy , 2012, Molecular pain.

[49]  S. Meroueh,et al.  Suppression of inflammatory and neuropathic pain by uncoupling CRMP-2 from the presynaptic Ca2+ channel complex , 2011, Nature Medicine.

[50]  G. Klasser,et al.  The prevalence of persistent pain after nonsurgical root canal treatment. , 2011, Quintessence international.

[51]  T. Renton,et al.  Trigeminal nerve injuries in relation to the local anaesthesia in mandibular injections , 2010, BDJ.

[52]  X. Michalet Mean square displacement analysis of single-particle trajectories with localization error: Brownian motion in an isotropic medium. , 2010, Physical review. E, Statistical, nonlinear, and soft matter physics.

[53]  W. Frey,et al.  Trigeminal pathways deliver a low molecular weight drug from the nose to the brain and orofacial structures. , 2010, Molecular pharmaceutics.

[54]  W. Frey,et al.  Intranasal delivery to the central nervous system: mechanisms and experimental considerations. , 2010, Journal of pharmaceutical sciences.

[55]  J. Hodges,et al.  Frequency of persistent tooth pain after root canal therapy: a systematic review and meta-analysis , 2010, BDJ.

[56]  R. Khanna,et al.  Regulation of N-type voltage-gated calcium channels (Cav2.2) and transmitter release by collapsin response mediator protein-2 (CRMP-2) in sensory neurons , 2009, Journal of Cell Science.

[57]  R. Khanna,et al.  An Atypical Role for Collapsin Response Mediator Protein 2 (CRMP-2) in Neurotransmitter Release via Interaction with Presynaptic Voltage-gated Calcium Channels* , 2009, The Journal of Biological Chemistry.

[58]  J. Henley,et al.  Protein SUMOylation in neuropathological conditions. , 2009, Drug news & perspectives.

[59]  Martin Koltzenburg,et al.  ProTx-II, a Selective Inhibitor of NaV1.7 Sodium Channels, Blocks Action Potential Propagation in Nociceptors , 2008, Molecular Pharmacology.

[60]  James A J Fitzpatrick,et al.  Fluorogen-activating single-chain antibodies for imaging cell surface proteins , 2008, Nature Biotechnology.

[61]  S. Hoka,et al.  Intranasal lidocaine 8% spray for second-division trigeminal neuralgia. , 2006, British journal of anaesthesia.

[62]  Miller H. Smith,et al.  Nerve injuries after dental injection: a review of the literature. , 2006, Journal.

[63]  E. Valmaseda-Castellón,et al.  Frequency and evolution of lingual nerve lesions following lower third molar extraction. , 2006, Journal of oral and maxillofacial surgery : official journal of the American Association of Oral and Maxillofacial Surgeons.

[64]  P. Goadsby,et al.  Calcium channels modulate nociceptive transmission in the trigeminal nucleus of the cat , 2005, Neuroscience.

[65]  B. Vasconcelos,et al.  Lingual nerve damage after mandibular third molar surgery: a randomized clinical trial. , 2005, Journal of oral and maxillofacial surgery : official journal of the American Association of Oral and Maxillofacial Surgeons.

[66]  A. Plückthun,et al.  A mutation designed to alter crystal packing permits structural analysis of a tight‐binding fluorescein–scFv complex , 2005, Protein science : a publication of the Protein Society.

[67]  H. Schaible,et al.  Effects of N-, P/Q- and L-type Calcium Channel Blockers on Nociceptive Neurones of the Trigeminal Nucleus with Input from the Dura , 2004, Cephalalgia : an international journal of headache.

[68]  R. Benoliel,et al.  Application of a pro-inflammatory agent to the orbital portion of the rat infraorbital nerve induces changes indicative of ongoing trigeminal pain , 2002, Pain.

[69]  E. Valmaseda-Castellón,et al.  Inferior alveolar nerve damage after lower third molar surgical extraction: a prospective study of 1117 surgical extractions. , 2001, Oral surgery, oral medicine, oral pathology, oral radiology, and endodontics.

[70]  J. Gregg Neuropathic complications of mandibular implant surgery: review and case presentations. , 2000, Annals of the Royal Australasian College of Dental Surgeons.

[71]  T. Nurmikko,et al.  Lamotrigine (Lamictal) in refractory trigeminal neuralgia: results from a double-blind placebo controlled crossover trial , 1997, Pain.

[72]  H. Kawamoto,et al.  Characterization of heat-hyperalgesia in an experimental trigeminal neuropathy in rats , 1997, Experimental Brain Research.

[73]  B. Vos,et al.  Behavioral evidence of trigeminal neuropathic pain following chronic constriction injury to the rat's infraorbital nerve , 1994, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[74]  B. van der Dijs,et al.  Pimozide therapy for trigeminal neuralgia. , 1989, Archives of neurology.

[75]  G. Fromm,et al.  Baclofen in the treatment of trigeminal neuralgia: Double‐blind study and long‐term follow‐up , 1984, Annals of neurology.

[76]  J. Ballantyne,et al.  Painful Traumatic Trigeminal Neuropathy , 2014 .

[77]  P. Svensson,et al.  Seven-year follow-up of patients diagnosed with atypical odontalgia: a prospective study. , 2013, Journal of orofacial pain.

[78]  Y. Sharav,et al.  Peripheral painful traumatic trigeminal neuropathy: clinical features in 91 cases and proposal of novel diagnostic criteria. , 2012, Journal of orofacial pain.

[79]  M. Takeda,et al.  Prostaglandin E2 potentiates the excitability of small diameter trigeminal root ganglion neurons projecting onto the superficial layer of the cervical dorsal horn in rats , 2006, Experimental Brain Research.

[80]  R. Gracely,et al.  Effect of dexamethasone and dipyrone on lingual and inferior alveolar nerve hypersensitivity following third molar extractions: preliminary report. , 2004, Journal of orofacial pain.

[81]  R. Young,et al.  Unmyelinated axons in the trigeminal motor root of human and cat , 1979, The Journal of comparative neurology.