Screening of candidate analgesics using a patient-derived human iPSC model of nociception identifies putative compounds for therapeutic treatment

Jack R. Thornton , Alberto Capurro , Sally Harwood , Thomas C Henderson , Adrienne Unsworth , Franziska Görtler , Sushma Nagaraja-Grellscheid , Vsevolod Telezhkin , Majlinda Lako , Evelyne Sernagor , Lyle Armstrong

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (5) : e70339

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (5) : e70339 DOI: 10.1002/ctm2.70339
RESEARCH ARTICLE

Screening of candidate analgesics using a patient-derived human iPSC model of nociception identifies putative compounds for therapeutic treatment

Author information +
History +
PDF

Abstract

Background and purpose: In this study, we applied an induced pluripotent stem cell (iPSC)-based model of inherited erythromelalgia (IEM) to screen a library of 281 small molecules, aiming to identify candidate pain-modulating compounds.

Experimental approach: Human iPSC-derived sensory neuron-like cells, which exhibit action potentials in response to noxious stimulation, were evaluated using whole-cell patch-clamp and microelectrode array (MEA) techniques.

Key results: Sensory neuron-like cells derived from individuals with IEM showed spontaneous electrical activity characteristic of genetic pain disorders. The drug screen identified four compounds (AZ106, AZ129, AZ037 and AZ237) that significantly decreased spontaneous firing with minimal toxicity. The calculated IC50 values indicate the potential efficacy of these compounds. Electrophysiological analysis confirmed the compounds’ ability to reduce action potential generation in IEM patient-specific iPSC-derived sensory neuron-like cells.

Conclusions and implications: Our screening approach demonstrates the reproducibility and effectiveness of human neuronal disease modelling offering a promising avenue for discovering new analgesics. These findings address a critical gap in current therapeutic strategies for both general and neuropathic pain, warranting further investigation. This study highlights the innovative use of patient-derived iPSC sensory neuronal models in pain research and emphasises the potential for personalised medicine in developing targeted analgesics.

Keywords

analgesic candidates / drug screening / electrophysiology / induced pluripotent stem cells (iPSCs) / inherited erythromelalgia (IEM) / sensory neuron

Cite this article

Download citation ▾
Jack R. Thornton, Alberto Capurro, Sally Harwood, Thomas C Henderson, Adrienne Unsworth, Franziska Görtler, Sushma Nagaraja-Grellscheid, Vsevolod Telezhkin, Majlinda Lako, Evelyne Sernagor, Lyle Armstrong. Screening of candidate analgesics using a patient-derived human iPSC model of nociception identifies putative compounds for therapeutic treatment. Clinical and Translational Medicine, 2025, 15(5): e70339 DOI:10.1002/ctm2.70339

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Van Hecke O, Torrance N, Smith BH. Chronic pain epidemiology and its clinical relevance. Br J Anaesth. 2013; 111(1): 13-18.

[2]

Zidarov D, Visca R, Gogovor A, Ahmed S. Performance and quality indicators for the management of non-cancer chronic pain: a scoping review protocol. BMJ Open. 2016; 6(2): e01048.

[3]

Lynch ME. The need for a Canadian pain strategy. Pain Res Manag. 2011; 16(2): 77-80.

[4]

Khouzam RH. Chronic pain and its management in primary care. South Med J. 2000; 93(10): 946-952.

[5]

Borsook D, Youssef AM, Simons L, Elman I, Eccleston C. When pain gets stuck: the evolution of pain chronification and treatment resistance. Pain. 2018; 159(12): 2421-2436.

[6]

Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D. Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. European Journal of Pain. 2006; 10(4): 287.

[7]

Percie du Sert N, Rice AS. Improving the translation of analgesic drugs to the clinic: animal models of neuropathic pain. Br J Pharmacol. 2014; 171(12): 2951-2963.

[8]

Eijkelkamp N, Linley JE, Baker MD, et al. Neurological perspectives on voltage-gated sodium channels. Brain. 2012; 135(9): 2585-2612.

[9]

Theile JW, Cummins TR. Recent developments regarding voltage-gated sodium channel blockers for the treatment of inherited and acquired neuropathic pain syndromes. Front Pharmacol. 2011; 2: 54.

[10]

Cao L, McDonnell A, Nitzsche A, et al. Pharmacological reversal of a pain phenotype in iPSC-derived sensory neurons and patients with inherited erythromelalgia. Sci Transl Med. 2016; 8(335): 335ra56.

[11]

Jarvis MF, Honore P, Shieh CC, et al. A-803467, a potent and selective Nav1.8 sodium channel blocker, attenuates neuropathic and inflammatory pain in the rat. Proc Natl Acad Sci U S A. 2007; 104(20): 8520-8525.

[12]

McDonnell A, Collins S, Ali Z, 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. Pain. 2018; 159(8): 1465-1476.

[13]

Patel R, Dickenson AH. Mechanisms of the gabapentinoids and α 2 δ-1 calcium channel subunit in neuropathic pain. Pharmacol Res Perspect. 2016; 4(2): e00205.

[14]

Meents JE, Bressan E, Sontag S, et al. The role of Nav1.7 in human nociceptors: insights from human induced pluripotent stem cell-derived sensory neurons of erythromelalgia patients. Pain. 2019; 160(6): 1327-1341.

[15]

Kuete CF, Granja-Vazquez R, Truong V, et al. Profiling human iPSC-derived sensory neurons for analgesic drug screening using a multi-electrode array. bioRxiv. 2024. Preprint]. 2024.11.18.623405.

[16]

Hiranuma M, Okuda Y, Fujii Y, Richard JP, Watanabe T. Characterization of human iPSC-derived sensory neurons and their functional assessment using multi electrode array. Sci Rep. 2024; 14(1): 6011.

[17]

Tay SH, Pang JKS, Ng W, et al. iPSC-derived human sensory neurons reveal a subset of TRPV1 antagonists as anti-pruritic compounds. Sci Rep. 2024; 14(1): 31182.

[18]

Black BJ, Ghazal RE, Lojek N, Williams V, Rajput JS, Lawson JM. Phenotypic screening of prospective analgesics among FDA-approved compounds using an iPSC-based model of acute and chronic inflammatory nociception. Adv Sci (Weinh). 2024; 11(11): e2303724.

[19]

Atmaramani R, Pancrazio JJ, Black BJ. Adaptation of robust Z' factor for assay quality assessment in microelectrode array-based screening using adult dorsal root ganglion neurons. J Neurosci Methods. 2020; 339: 108699.

[20]

Odawara A, Shibata M, Ishibashi Y, Nagafuku N, Matsuda N, In SuzukiI. Vitro pain assay using human iPSC-derived sensory neurons and microelectrode array. Toxicol Sci. 2022; 188(1): 131-141.

[21]

Nimbalkar S, Guo X, Colón A, et al. Development of a functional human induced pluripotent stem cell-derived nociceptor MEA system as a pain model for analgesic drug testing. Front Cell Dev Biol. 2023; 11: 1011145.

[22]

Buskin A, Zhu L, Chichagova V, et al. Disrupted alternative splicing for genes implicated in splicing and ciliogenesis causes PRPF31 retinitis pigmentosa. Nat Commun. 2018; 9(1): 4234.

[23]

Melguizo-Sanchis D, Xu Y, Taheem D, et al. Modelling of severe aplastic anaemia reveals impaired differentiation and telomere shortening in blood progenitors. Cell Death Dis. 2018; 9(2): 128.

[24]

Wang M, Wang J, Tsui AYP, et al. Mechanisms of peripheral neurotoxicity associated with four chemotherapy drugs using human induced pluripotent stem cell-derived peripheral neurons. Toxicol In Vitro. 2021; 77: 105233.

[25]

Telezhkin V, Schnell C, Yarova P, et al. Forced cell cycle exit and modulation of GABAA, CREB, and GSKβ signalling promote functional maturation of induced pluripotent stem cell-derived neurons. Am J Physiol Cell Physiol. 2016; 310(7): C520-C541.

[26]

Telezhkin V, Straccia M, Yarova P, et al. Kv7 channels are upregulated during striatal neuron development and promote maturation of human iPSC-derived neurons. Pflugers Arch. 2018; 470(9): 1359-1376.

[27]

Anders S, Huber W. Differential expression analysis for sequence count data. Genome Biol. 2010; 11(10): R106.

[28]

Jung M, Dourado M, Maksymetz J, et al. Cross-species transcriptomic atlas of dorsal root ganglia reveals species-specific programs for sensory function. Nat Commun. 2023; 14(1): 366.

[29]

Hao Y, Stuart T, Kowalski MH, et al. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat Biotechnol. 2024; 42: 293-304.

[30]

Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods. 2019; 16: 1289-1296.

[31]

Wickham H, Averick M, Bryan J, et al. Welcome to the Tidyverse. Journal of Open Source Software. 2019; 4(43): 1686.

[32]

Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15: 550.

[33]

Ritchie ME, Phipson B, Wu D, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research. 2015; 43(7).

[34]

Chambers SM, Qi Y, Mica Y, et al. Combined small-molecule inhibition accelerates developmental timing and converts human pluripotent stem cells into nociceptors. Nat Biotechnol. 2012 Jul 1; 30(7): 715-720.

[35]

Young GT, Gutteridge A, Fox HDE, et al. Characterizing human stem cell derived sensory neurons at the single-cell level reveals their ion channel expression and utility in pain research. Molecular Therapy. 2014; 22(8): 1530-1543.

[36]

Weir GA, Middleton SJ, Clark AJ, et al. Using an engineered glutamate-gated chloride channel to silence sensory neurons and treat neuropathic pain at the source. Brain. 2017; 140(10): 2570-2585.

[37]

Schwartzentruber J, Foskolou S, Kilpinen H, et al. Molecular and functional variation in iPSC-derived sensory neurons. Nature Genetics. 2018; 50(1): 54-61.

[38]

Ito T, Ito T, Sakai A, Maruyama M, et al. Dorsal root ganglia homeobox downregulation in primary sensory neurons contributes to neuropathic pain in rats. Mol Pain. 2020: 16: 1744806920904462.

[39]

Jo S, Bean BP. Lidocaine binding enhances inhibition of Nav1.7 channels by the sulfonamide PF-05089771. Mol Pharmacol. 2020; 97(6): 377-383.

[40]

Zou M, Li S, Klein WH, Xiang M. Brn3a/Pou4f1 regulates dorsal root ganglion sensory neuron specification and axonal projection into the spinal cord. Dev Biol. 2012; 364(2): 114-127.

[41]

Megat S, Ray PR, Tavares-Ferreira D, et al. Differences between dorsal root and trigeminal ganglion nociceptors in mice revealed by translational profiling. J Neurosci. 2019; 39(35): 6829-6847.

[42]

Burnstock G. P2X receptors in sensory neurones. Br J Anaesth. 2000; 84(4): 476-488.

[43]

Sheets PL, Jackson JO,, Waxman SG, Dib-Hajj SD, Cummins TR. A Nav1.7 channel mutation associated with hereditary erythromelalgia contributes to neuronal hyperexcitability and displays reduced lidocaine sensitivity. J Physiol. 2007; 581(3): 1019-1031. Pt.

[44]

Cojen H, O'Leary ME. Single-cell analysis of sodium channel expression in dorsal root ganglion neurons. Molecular and Cellular Neuroscience. 2011; 46(1): 159-166.

[45]

Chang W, Berta T, Kim YH, Lee S, Lee SY, Ji RR. Expression and role of voltage-gated sodium channels in human dorsal root ganglion neurons with special focus on Nav1.7, species differences, and regulation by paclitaxel. Neurosci Bull. 2018; 34(1): 4-12.

[46]

Gorham L, Just S, Doods H. Somatostatin 4 receptor activation modulates TRPV1 [correction of TRPV1] currents in dorsal root ganglion neurons. Neurosci Lett. 2014; 573: 25-29.

[47]

Fiorino DF, Garcia-Gruzman M. Muscarinic pain pharmacology: realising the promise of novel analgesics by overcoming old challenges. Handb Exp Pharmacol. 2012; 208: 191-221.

[48]

Kecskés A, Pohóczky K, Kecskés M, et al. Characterization of neurons expressing the novel analgesic drug target somatostatin receptor 4 in mouse and human brains. Int j Mol Sci. 2020; 21(20): 7788.

[49]

Kecskés A, Pohóczky K, Kecskés M, et al. Characterization of neurons expressing the novel analgesic drug target somatostatin receptor 4 agonists are potential analgesics for neuropathic pain. Int J Mol Sci. 2019; 20(24): 6245.

[50]

Abdel-Magid AF. Treating pain with somatostatin receptor subtype 4 agonists. ACS Med Chem Lett. 2015; 6(2): 110-111.

[51]

Liu QQ, Yao XX, Gao SH, et al. Role of 5-HT receptors in neuropathic pain: potential therapeutic implications. Pharmacol Res. 2020; 159: 104949.

[52]

Liu C, Liu S, Lu X, Tao F. Role of descending dopaminergic pathways in pain modulation. Curr Neuropharmacol. 2019; 17(12): 1176-1182.

[53]

Ruiz-Cantero MC, González-Cano R, Tejada , et al. Sigma-1 receptor: a drug target for the modulation of neuroimmune and neuroglial interactions during chronic pain. Pharmacol Res. 2021; 163: 105339.

[54]

Hough LB, Rice FL. H3 receptors and pain modulation: peripheral, spinal, and brain interactions. J Pharmacol Exp Ther. 2011; 336(1): 30-37.

[55]

Ingólfsson HI, Thakur P, Herold KF, et al. Phytochemicals perturb membranes and promiscuously alter protein function. ACS Chem Biol. 2014; 9(8): 1788-1798.

[56]

Ford ZK, Reker AN, Chen S, Kadakia F, Bunk A, Davidson S. Cannabinoid receptor 1 expression in human dorsal root ganglia and CB13-induced bidirectional modulation. Front Pain Res (Lausanne). 2021; 2: 721332.

[57]

Stein C. Opioid Receptors on Peripheral Sensory Neurons. Madame Curie Bioscience Database. Landes Bioscience; 2000-2013.

[58]

Siebenga P, van Amerongen G, Hay JL, et al. Lack of detection of the analgesic properties of PF-05089771, a selective Nav 1.7 inhibitor, using a battery of pain models in healthy subjects. Clin Transl Sci. 2020; 13(2): 318-324.

[59]

Blair NT, Bean BP. Roles of tetrodotoxin (TTX)-sensitive Na+ current, TTX-resistant Na+ current, and Ca2+ current in the action potentials of nociceptive sensory neurons. The Journal of Neuroscience. 2002; 22(23): 10277-10290.

[60]

Renganathan M, Cummins TR, Waxman SG. Contribution of Na(v)1.8 sodium channels to action potential electrogenesis in DRG neurons. J Neurophysiol. 2001; 86(2): 629-640.

[61]

Cummins TR, Howe JR, Waxman SG. Slow closed-state inactivation: a novel mechanism underlying ramp currents in cells expressing the hNE/PN1 sodium channel. J Neurosci. 1998 Dec 1; 18(23): 9607-9619.

[62]

Pramod AB, Foster J, Carvelli L, Henry LK. SLC6 transporters: structure, function, regulation, disease association and therapeutics. Mol Aspects Med. 2013; 34(2-3): 197-219.

[63]

Du X, Hao H, Yang Y, et al. Local GABAergic signalling within signaling within sensory ganglia controls peripheral nociceptive transmission. J Clin Invest. 2017; 127(5): 1741-1756.

[64]

Sałat K, Podkowa A, Malikowska N, et al. Novel, highly potent and in vivo active inhibitor of GABA transporter subtype 1 with anticonvulsant, anxiolytic, antidepressant and antinociceptive properties. Neuropharmacology. 2017; 113(A): 331-342.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

8

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/