Challenges of neuropathic pain: focus on diabetic neuropathy

Daniela C Rosenberger, Vivian Blechschmidt, Hans Timmerman, André Wolff, Rolf-Detlef Treede, Daniela C Rosenberger, Vivian Blechschmidt, Hans Timmerman, André Wolff, Rolf-Detlef Treede

Abstract

Neuropathic pain is a frequent condition caused by a lesion or disease of the central or peripheral somatosensory nervous system. A frequent cause of peripheral neuropathic pain is diabetic neuropathy. Its complex pathophysiology is not yet fully elucidated, which contributes to underassessment and undertreatment. A mechanism-based treatment of painful diabetic neuropathy is challenging but phenotype-based stratification might be a way to develop individualized therapeutic concepts. Our goal is to review current knowledge of the pathophysiology of peripheral neuropathic pain, particularly painful diabetic neuropathy. We discuss state-of-the-art clinical assessment, validity of diagnostic and screening tools, and recommendations for the management of diabetic neuropathic pain including approaches towards personalized pain management. We also propose a research agenda for translational research including patient stratification for clinical trials and improved preclinical models in relation to current knowledge of underlying mechanisms.

Keywords: Neuroinflammation; Painful diabetic neuropathy; Personalized pain management; Quantitative sensory testing; Spinal sensitization; Stratification in clinical trials.

Conflict of interest statement

R.D.T. reports grants from DFG, non-financial support from Astellas Pharma GmbH (Munich, Germany), during the conduct of the study; grants from Bayer, DFG, EU, personal fees from Astellas, Bayer, Decision Resources, Grünenthal, GSK, Hydra, and Pfizer, all outside the submitted work. The other authors report no conflicts of interest in this work.

Figures

Fig. 1
Fig. 1
Selection of peripheral and central mechanisms contributing to neuropathic pain. AMPA-R/NMDA-R ionotropic glutamate receptors, AP action potential, ATP adenosine triphosphate, BDNF brain-derived neurotrophic factor, CCL2/FKN chemokines, CCR2/CX3CR1 chemokine receptors, CGRP calcitonin gene-related peptide, GABA gamma-aminobutyric acid, Gly Glycin, FKN fractalkine (CX3CL1), IL-1β interleukin 1β, IL-6 interleukin 6, KCC2 chloride potassium symporter, MMP matrix metalloproteinase, NK1-R neurokinin 1 receptor, NO nitric oxide, p-p38 MAPK phosphorylated p38 mitogen-activated protein kinase, PG prostaglandins, SP substance P, TNFα tumor necrosis factor-alpha, TNF-R tumor necrosis factor receptor, trkB tyrosine kinase B, TRPV1 transient receptor potential vanilloid 1, VGSC voltage-gated sodium channel
Fig. 2
Fig. 2
Selection of structural and functional alterations in diabetic neuropathy. AGE advanced glycation end products, ROS reactive oxygen species
Fig. 3
Fig. 3
Pathophysiology of painless and painful diabetic neuropathy: diabetes mellitus leads to several pathological changes in neuronal, immune and vascular cells that can lead to structural and functional alterations of the nervous system that can result in diabetic neuropathy (see Fig. 2). Several factors contribute to the development of neuropathic pain in diabetic neuropathy. AGE advanced glycation end products, HIF-1α hypoxia-induced factor 1α, PKC protein kinase C, TRPA1 transient receptor potential ankyrin 1, VGSC voltage-gated sodium channel, vWF von Willebrand factor

References

    1. Abbott CA, Garrow AP, Carrington AL, Morris J, Van Ross ER, Boulton AJ, North-West diabetes foot care s Foot ulcer risk is lower in South-Asian and African-Caribbean compared with European diabetic patients in the U.K.: the North–West diabetes foot care study. Diabetes Care. 2005;28:1869–1875. doi: 10.2337/diacare.28.8.1869.
    1. Abbott CA, Malik RA, van Ross ER, Kulkarni J, Boulton AJ. Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the U.K. Diabetes Care. 2011;34:2220–2224. doi: 10.2337/dc11-1108.
    1. Ahmad J. The diabetic foot. Diabetes Metab Syndr. 2016;10:48–60. doi: 10.1016/j.dsx.2015.04.002.
    1. Aiyer R, Barkin RL, Bhatia A. Treatment of neuropathic pain with venlafaxine: a systematic review. Pain Med. 2017;18:1999–2012. doi: 10.1093/pm/pnw261.
    1. Alam U, Jeziorska M, Petropoulos IN, Asghar O, Fadavi H, Ponirakis G, Marshall A, Tavakoli M, Boulton AJM, Efron N, Malik RA. Diagnostic utility of corneal confocal microscopy and intra-epidermal nerve fibre density in diabetic neuropathy. PLoS One. 2017;12(7):e0180175. doi: 10.1371/journal.pone.0180175.
    1. Albers JW, et al. Effect of prior intensive insulin treatment during the Diabetes Control and Complications Trial (DCCT) on peripheral neuropathy in type 1 diabetes during the Epidemiology of Diabetes Interventions and Complications (EDIC) Study. Diabetes Care. 2010;33:1090–1096. doi: 10.2337/dc09-1941.
    1. Albrecht DS, et al. Neuroinflammation of the spinal cord and nerve roots in chronic radicular pain patients. Pain. 2018;159:968–977. doi: 10.1097/j.pain.0000000000001171.
    1. Allegri M, Baron R, Hans G, Correa-Illanes G, Mayoral Rojals V, Mick G, Serpell M. A pharmacological treatment algorithm for localized neuropathic pain. Curr Med Res Opin. 2016;32(2):377–384. doi: 10.1185/03007995.2015.1129321.
    1. Alleman CJ, Westerhout KY, Hensen M, Chambers C, Stoker M, Long S, van Nooten FE. Humanistic and economic burden of painful diabetic peripheral neuropathy in Europe: a review of the literature. Diabetes Res Clin Pract. 2015;109:215–225. doi: 10.1016/j.diabres.2015.04.031.
    1. Ametov AS, et al. The sensory symptoms of diabetic polyneuropathy are improved with alpha-lipoic acid: the SYDNEY trial. Diabetes Care. 2003;26:770–776. doi: 10.2337/diacare.26.3.770.
    1. Amir R, et al. The role of sodium channels in chronic inflammatory and neuropathic pain. J Pain. 2006;7:S1–29. doi: 10.1016/j.jpain.2006.01.444.
    1. Andersson C, Guttorp P, Sarkka A. Discovering early diabetic neuropathy from epidermal nerve fiber patterns. Stat Med. 2016;35:4427–4442. doi: 10.1002/sim.7009.
    1. Andersen ST, et al. Risk factors for incident diabetic polyneuropathy in a cohort with screen-detected Type 2 diabetes followed for 13 years: ADDITION-Denmark. Diabetes Care. 2018;41:1068–1075. doi: 10.2337/dc17-2062.
    1. Apfel SC, et al. Positive neuropathic sensory symptoms as endpoints in diabetic neuropathy trials. J Neurol Sci. 2001;189:3–5. doi: 10.1016/s0022-510x(01)00584-6.
    1. Archer AG, Roberts VC, Watkins PJ. Blood flow patterns in painful diabetic neuropathy. Diabetologia. 1984;27:563–567. doi: 10.1007/BF00276968.
    1. Attal N, Fermanian C, Fermanian J, Lanteri-Minet M, Alchaar H, Bouhassira D. Neuropathic pain: are there distinct subtypes depending on the aetiology or anatomical lesion? Pain. 2008;138:343–353. doi: 10.1016/j.pain.2008.01.006.
    1. Attal N, et al. EFNS guidelines on the pharmacological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010;17:1113–e1188. doi: 10.1111/j.1468-1331.2010.02999.x.
    1. Attal N, et al. Assessing symptom profiles in neuropathic pain clinical trials: can it improve outcome? Eur J Pain. 2011;15:441–443. doi: 10.1016/j.ejpain.2011.03.005.
    1. Attal N, Bouhassira D. Pharmacotherapy of neuropathic pain: which drugs, which treatment algorithms? Pain. 2015;156(Suppl 1):S104–14. doi: 10.1097/01.j.pain.0000460358.01998.15.
    1. Attal N, Bouhassira D, Baron R. Diagnosis and assessment of neuropathic pain through questionnaires. Lancet Neurol. 2018;17:456–466. doi: 10.1016/S1474-4422(18)30071-1.
    1. Backonja MM, Coe CL, Muller DA, Schell K. Altered cytokine levels in the blood and cerebrospinal fluid of chronic pain patients. J Neuroimmunol. 2008;195:157–163. doi: 10.1016/j.jneuroim.2008.01.005.
    1. Backonja MM, et al. Value of quantitative sensory testing in neurological and pain disorders: NeuPSIG consensus. Pain. 2013;154:1807–1819. doi: 10.1016/j.pain.2013.05.047.
    1. Backryd E, Lind AL, Thulin M, Larsson A, Gerdle B, Gordh T. High levels of cerebrospinal fluid chemokines point to the presence of neuroinflammation in peripheral neuropathic pain: a cross-sectional study of 2 cohorts of patients compared with healthy controls. Pain. 2017;158:2487–2495. doi: 10.1097/j.pain.0000000000001061.
    1. Balasubramanyan S, Stemkowski PL, Stebbing MJ, Smith PA. Sciatic chronic constriction injury produces cell-type-specific changes in the electrophysiological properties of rat substantia gelatinosa neurons. J Neurophysiol. 2006;96:579–590. doi: 10.1152/jn.00087.2006.
    1. Barbosa M, Saavedra A, Severo M, Maier C, Carvalho D. Validation and reliability of the portuguese version of the Michigan neuropathy screening instrument. Pain Pract. 2017;17:514–521. doi: 10.1111/papr.12479.
    1. Baron R. Mechanisms of disease: neuropathic pain—a clinical perspective. Nat Clin Pract Neurol. 2006;2:95–106. doi: 10.1038/ncpneuro0113.
    1. Baron R, Binder A, Wasner G. Neuropathic pain: diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol. 2010;9:807–819. doi: 10.1016/S1474-4422(10)70143-5.
    1. Baron R, Forster M, Binder A. Subgrouping of patients with neuropathic pain according to pain-related sensory abnormalities: a first step to a stratified treatment approach. Lancet Neurol. 2012;11:999–1005. doi: 10.1016/S1474-4422(12)70189-8.
    1. Baron R, et al. Peripheral neuropathic pain: a mechanism-related organizing principle based on sensory profiles. Pain. 2017;158:261–272. doi: 10.1097/j.pain.0000000000000753.
    1. Bastyr EJ, 3rd, Price KL, Bril V, Group MS. Development and validity testing of the neuropathy total symptom score-6: questionnaire for the study of sensory symptoms of diabetic peripheral neuropathy. Clin Ther. 2005;27:1278–1294. doi: 10.1016/j.clinthera.2005.08.002.
    1. Bee LA, Dickenson AH. Descending facilitation from the brainstem determines behavioural and neuronal hypersensitivity following nerve injury and efficacy of pregabalin. Pain. 2008;140:209–223. doi: 10.1016/j.pain.2008.08.008.
    1. Beggs S, Salter MW. Stereological and somatotopic analysis of the spinal microglial response to peripheral nerve injury. Brain Behav Immun. 2007;21:624–633. doi: 10.1016/j.bbi.2006.10.017.
    1. Belfer I, Dai F. Phenotyping and genotyping neuropathic pain. Curr Pain Headache Rep. 2010;14:203–212. doi: 10.1007/s11916-010-0110-1.
    1. Bennett DL, Woods CG. Painful and painless channelopathies. Lancet Neurol. 2014;13:587–599. doi: 10.1016/s1474-4422(14)70024-9.
    1. Bernier LP, Ase AR, Seguela P. P2X receptor channels in chronic pain pathways. Br J Pharmacol. 2018;175:2219–2230. doi: 10.1111/bph.13957.
    1. Bierhaus A, et al. Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in diabetic neuropathy. Nat Med. 2012;18:926–933. doi: 10.1038/nm.2750.
    1. Binder A. Human surrogate models of neuropathic pain: validity and limitations. Pain. 2016;157(Suppl 1):S48–52. doi: 10.1097/j.pain.0000000000000460.
    1. Blesneac I, et al. Rare NaV1.7 variants associated with painful diabetic peripheral neuropathy. Pain. 2018;159:469–480. doi: 10.1097/j.pain.0000000000001116.
    1. Borsook D, Kussman BD, George E, Becerra LR, Burke DW. Surgically induced neuropathic pain: understanding the perioperative process. Ann Surg. 2013;257:403–412. doi: 10.1097/SLA.0b013e3182701a7b.
    1. Bosma RL, et al. Brain dynamics and temporal summation of pain predicts neuropathic pain relief from ketamine infusion. Anesthesiology. 2018;129:1015–1024. doi: 10.1097/aln.0000000000002417.
    1. Bouhassira D, Attal N. Diagnosis and assessment of neuropathic pain: the saga of clinical tools. Pain. 2011;152:S74–83. doi: 10.1016/j.pain.2010.11.027.
    1. Bouhassira D, Attal N, Fermanian J, Alchaar H, Gautron M, Masquelier E, Rostaing S, Lanteri-Minet M, Collin E, Grisart J, Boureau F. Development and validation of the neuropathic pain symptom. Inventory Pain. 2004;108(3):248–257. doi: 10.1016/j.pain.2003.12.024.
    1. Bouhassira D, et al. Comparison of pain syndromes associated with nervous or somatic lesions and development of a new neuropathic pain diagnostic questionnaire (DN4) Pain. 2005;114:29–36. doi: 10.1016/j.pain.2004.12.010.
    1. Bouhassira D, Lantéri-Minet M, Attal N, Laurent B, Touboul C. Prevalence of chronic pain wih neuropathic characteristics in the general population. Pain. 2008;136:S380–387. doi: 10.1016/j.pain.2007.08.013.
    1. Bouhassira D, Letanoux M, Hartemann A. Chronic pain with neuropathic characteristics in diabetic patients: a French cross-sectional study. PLoS ONE. 2013;8(9):e74195. doi: 10.1371/journal.pone.0074195.
    1. Bouhassira D, et al. Neuropathic pain phenotyping as a predictor of treatment response in painful diabetic neuropathy: data from the randomized, double-blind, COMBO-DN study. Pain. 2014;155:2171–2179. doi: 10.1016/j.pain.2014.08.020.
    1. Boulton AJ. The diabetic foot—an update. Foot Ankle Surg. 2008;14:120–124. doi: 10.1016/j.fas.2008.05.004.
    1. Bril V, Perkins BA. Validation of the Toronto Clinical Scoring system for diabetic polyneuropathy. Diabetes Care. 2002;25:2048–2052. doi: 10.2337/diacare.25.11.2048.
    1. Bril V, Tomioka S, Buchanan RA, Perkins BA, mTCNS Study Group Reliability and validity of the modified Toronto Clinical Neuropathy Score in diabetic sensorimotor polyneuropathy. Diabet Med. 2009;26:240–246. doi: 10.1111/j.1464-5491.2009.02667.x.
    1. Bril V, et al. Evidence-based guideline: treatment of painful diabetic neuropathy: report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Neurology. 2011;76:1758–1765. doi: 10.1212/WNL.0b013e3182166ebe.
    1. Brown JJ, Pribesh SL, Baskette KG, Vinik AI, Colberg SR. A comparison of screening tools for the early detection of peripheral neuropathy in adults with and without type 2 diabetes. J Diabetes Res. 2017;2017:1467213. doi: 10.1155/2017/1467213.
    1. Burke D, Fullen BM, Stokes D, Lennon O. Neuropathic pain prevalence following spinal cord injury: a systematic review and meta-analysis. Eur J Pain. 2017;21:29–44. doi: 10.1002/ejp.905.
    1. Busserolles J, Tsantoulas C, Eschalier A, Lopez Garcia JA. Potassium channels in neuropathic pain: advances, challenges, and emerging ideas. Pain. 2016;157(Suppl 1):S7–14. doi: 10.1097/j.pain.0000000000000368.
    1. Callaghan BC, Cheng HLT, Stables CL, Smith AL, Feldman EL. Diabetic neuropathy: clinical manifestations and current treatments. Lancet Neurol. 2012;11:521–534. doi: 10.1016/S1474-4422(12)70065-0.
    1. Callaghan BC, Little AA, Feldman EL, Hughes RAC. Enhanced glucose control for preventing and treating diabetic neuropathy. Cochrane Database Syst Rev. 2012 doi: 10.1002/14651858.CD007543.pub2.
    1. Calles-Escandon J, et al. Effect of intensive compared with standard glycemia treatment strategies on mortality by baseline subgroup characteristics: the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Diabetes Care. 2010;33:721–727. doi: 10.2337/dc09-1471.
    1. Cameron NE, Eaton SE, Cotter MA, Tesfaye S. Vascular factors and metabolic interactions in the pathogenesis of diabetic neuropathy. Diabetologia. 2001;44:1973–1988. doi: 10.1007/s001250100001.
    1. Campbell JN, Meyer RA. Mechanisms of neuropathic pain. Neuron. 2006;52:77–92. doi: 10.1016/j.neuron.2006.09.021.
    1. Campbell CM, et al. Randomized control trial of topical clonidine for treatment of painful diabetic neuropathy. Pain. 2012;153:1815–1823. doi: 10.1016/j.pain.2012.04.014.
    1. Caselli A, Rich J, Hanane T, Uccioli L, Veves A. Role of C-nociceptive fibers in the nerve axon reflex-related vasodilation in diabetes. Neurology. 2003;60:297–300. doi: 10.1212/01.wnl.0000040250.31755.f9.
    1. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816–824. doi: 10.1038/39807.
    1. Challa SR. Surgical animal models of neuropathic pain: Pros and Cons. Int J Neurosci. 2015;125:170–174. doi: 10.3109/00207454.2014.922559.
    1. Chalk C, Benstead TJ, Moore F. Aldose reductase inhibitors for the treatment of diabetic polyneuropathy. Cochrane Database Syst Rev. 2007 doi: 10.1002/14651858.CD004572.pub2.
    1. Chaparro LE, Wiffen PJ, Moore RA, Gilron I. Combination pharmacotherapy for the treatment of neuropathic pain in adults. Cochrane Database Syst Rev. 2012 doi: 10.1002/14651858.CD008943.pub2.
    1. Chaplan SR, et al. Neuronal hyperpolarization-activated pacemaker channels drive neuropathic pain. J Neurosci. 2003;23:1169–1178. doi: 10.1523/JNEUROSCI.23-04-01169.2003.
    1. Cheliout-Heraut F, et al. Exploration of small fibers for testing diabetic neuropathies. Jt Bone Spine. 2005;72:412–415. doi: 10.1016/j.jbspin.2004.10.003.
    1. Chen L, Huang LY. Protein kinase C reduces Mg2+ block of NMDA-receptor channels as a mechanism of modulation. Nature. 1992;356:521–523. doi: 10.1038/356521a0.
    1. Chen J, et al. The alpha2delta-1-NMDA receptor complex is critically involved in neuropathic pain development and gabapentin therapeutic actions. Cell Rep. 2018;22:2307–2321. doi: 10.1016/j.celrep.2018.02.021.
    1. Cheng HT, et al. Increased axonal regeneration and swellings in intraepidermal nerve fibers characterize painful phenotypes of diabetic neuropathy. J Pain. 2013;14:941–947. doi: 10.1016/j.jpain.2013.03.005.
    1. Choi SR, et al. Spinal D-serine increases PKC-dependent GluN1 phosphorylation contributing to the sigma-1 receptor-induced development of mechanical allodynia in a mouse model of neuropathic pain. J Pain. 2017;18:415–427. doi: 10.1016/j.jpain.2016.12.002.
    1. Clark AK, Malcangio M. Microglial signalling mechanisms: cathepsin S and fractalkine. Exp Neurol. 2012;234:283–292. doi: 10.1016/j.expneurol.2011.09.012.
    1. Clark AK, Malcangio M. Fractalkine/CX3CR1 signaling during neuropathic pain. Front Cell Neurosci. 2014;8:121. doi: 10.3389/fncel.2014.00121.
    1. Colleoni M, Sacerdote P. Murine models of human neuropathic pain. Biochem Biophys Acta. 2010;1802:924–933. doi: 10.1016/j.bbadis.2009.10.012.
    1. Colloca L, et al. Neuropathic pain. Nat Rev Dis Primers. 2017;3:17002. doi: 10.1038/nrdp.2017.2.
    1. Collins SL, Moore RA, McQuayHj WP. Antidepressants and anticonvulsants for diabetic neuropathy and postherpetic neuralgia: a quantitative systematic review. J Pain Symptom Manag. 2000;20:449–458. doi: 10.1016/s0885-3924(00)00218-9.
    1. Cooper TE, et al. Morphine for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017;5:CD011669. doi: 10.1002/14651858.CD011669.pub2.
    1. Cornblath DR, Chaudhry V, Carter K, Lee D, Seysedadr M, Miernicki M, Joh T. Total neuropathy score: validation and reliability study. Neurology. 1999;53:1660–1664. doi: 10.1212/wnl.53.8.1660.
    1. Costigan M, Scholz J, Woolf CJ. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci. 2009;32:1–32. doi: 10.1146/annurev.neuro.051508.135531.
    1. Coull JA, et al. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature. 2005;438:1017–1021. doi: 10.1038/nature04223.
    1. Coward K, et al. Immunolocalization of SNS/PN3 and NaN/SNS2 sodium channels in human pain states. Pain. 2000;85:41–50. doi: 10.1016/S0304-3959(99)00251-1.
    1. Cox JJ, et al. An SCN9A channelopathy causes congenital inability to experience pain. Nature. 2006;444:894–898. doi: 10.1038/nature05413.
    1. Crawford B, Bouhassira D, Wong A, Dukes E. Conceptual adequacy of the neuropathic pain symptom inventory in six countries. Health Qual Life Outcomes. 2008;18(6):62. doi: 10.1186/1477-7525-6-62.
    1. Cruccu G, Truini A. Sensory profiles: a new strategy for selecting patients in treatment trials for neuropathic pain. Pain. 2009;146:5–6. doi: 10.1016/j.pain.2009.07.004.
    1. Cruccu G, Truini A. A review of neuropathic pain: from guidelines to clinical practice. Pain Ther. 2017;6:35–42. doi: 10.1007/s40122-017-0087-0.
    1. Cruccu G, et al. Recommendations for the clinical use of somatosensory-evoked potentials. Clin Neurophysiol. 2008;119:1705–1719. doi: 10.1016/j.clinph.2008.03.016.
    1. Cruccu G, et al. EFNS guidelines on neuropathic pain assessment: revised 2009. Eur J Neurol. 2010;17:1010–1018. doi: 10.1111/j.1468-1331.2010.02969.x.
    1. Cruccu G, et al. Trigeminal neuralgia: new classification and diagnostic grading for practice and research. Neurology. 2016;87:220–228. doi: 10.1212/WNL.0000000000002840.
    1. Cummins TR, Sheets PL, Waxman SG. The roles of sodium channels in nociception: implications for mechanisms of pain. Pain. 2007;131:243–257. doi: 10.1016/j.pain.2007.07.026.
    1. Curtin CM, Kenney D, Suarez P, Hentz VR, Hernandez-Boussard T, Mackey S, Carroll IR. A double-blind placebo randomized controlled trial of minocycline to reduce pain after carpal tunnel and trigger finger release. J Hand Surg Am. 2017;42:166–174. doi: 10.1016/j.jhsa.2016.12.011.
    1. Davis KD, et al. Brain imaging tests for chronic pain: medical, legal and ethical issues and recommendations. Nat Rev Neurol. 2017;13:624–638. doi: 10.1038/nrneurol.2017.122.
    1. De Keyser R, van den Broeke EN, Courtin A, Dufour A, Mouraux A. Event-related brain potentials elicited by high-speed cooling of the skin: a robust and non-painful method to assess the spinothalamic system in humans. Clin Neurophysiol. 2018;129:1011–1019. doi: 10.1016/j.clinph.2018.02.123.
    1. Delpont B, Blanc C, Osseby GV, Hervieu-Begue M, Giroud M, Bejot Y. Pain after stroke: a review. Rev Neurol (Paris) 2018;174:671–674. doi: 10.1016/j.neurol.2017.11.011.
    1. Demant DT, et al. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain. 2014;155:2263–2273. doi: 10.1016/j.pain.2014.08.014.
    1. Deng Y, Luo L, Hu Y, Fang K, Liu J. Clinical practice guidelines for the management of neuropathic pain: a systematic review. BMC Anesthesiol. 2016;16:12. doi: 10.1186/s12871-015-0150-5.
    1. Derry S, Rice AS, Cole P, Tan T, Moore RA. Topical capsaicin (high concentration) for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017;1:CD011669. doi: 10.1002/14651858.CD007393.pub4.
    1. Derry S, Bell RF, Straube S, Wiffen PJ, Aldington D, Moore RA. Pregabalin for neuropathic pain in adults. Cochrane Database Syst Rev. 2019;1:CD007076. doi: 10.1002/14651858.CD007076.pub3.
    1. Devigili G, et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain. 2008;131:1912–1925. doi: 10.1093/brain/awn093.
    1. Devor M. Ectopic discharge in Abeta afferents as a source of neuropathic pain. Exp Brain Res. 2009;196:115–128. doi: 10.1007/s00221-009-1724-6.
    1. Dewanjee S, et al. Molecular mechanism of diabetic neuropathy and its pharmacotherapeutic targets. Eur J Pharmacol. 2018;833:472–523. doi: 10.1016/j.ejphar.2018.06.034.
    1. Di Stefano G, et al. Diagnostic accuracy of laser-evoked potentials in diabetic neuropathy. Pain. 2017;158:1100–1107. doi: 10.1097/j.pain.0000000000000889.
    1. Dib-Hajj SD, Cummins TR, Black JA, Waxman SG. Sodium channels in normal and pathological pain. Annu Rev Neurosci. 2010;33:325–347. doi: 10.1146/annurev-neuro-060909-153234.
    1. Doth AH, Hansson PT, Jensen MP, Taylor RS. The burden of neuropathic pain: a systematic review and meta-analysis of health utilities. Pain. 2010;149:338–344. doi: 10.1016/j.pain.2010.02.034.
    1. Drdla-Schutting R, Benrath J, Wunderbaldinger G, Sandkuhler J. Erasure of a spinal memory trace of pain by a brief, high-dose opioid administration. Science (New York, NY) 2012;335:235–238. doi: 10.1126/science.1211726.
    1. Dull MM, et al. Methylglyoxal causes pain and hyperalgesia in human through C-fiber activation. Pain. 2019 doi: 10.1097/j.pain.0000000000001644.
    1. Duehmke RM, Derry S, Wiffen PJ, Bell RF, Aldington D, Moore RA. Tramadol for neuropathic pain in adults. Cochrane Database Syst Rev. 2017;6:CD011669. doi: 10.1002/14651858.CD003726.pub4.
    1. Dworkin RH, et al. Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain. 2007;132:237–251. doi: 10.1016/j.pain.2007.08.033.
    1. Dworkin RH, et al. Recommendations for the pharmacological management of neuropathic pain: an overview and literature update. Mayo Clin Proc. 2010;85:S3–14. doi: 10.4065/mcp.2009.0649.
    1. Dworkin RH, et al. Interventional management of neuropathic pain: NeuPSIG recommendations. Pain. 2013;154:2249–2261. doi: 10.1016/j.pain.2013.06.004.
    1. Dyck PJ. Detection, characterization, and staging of polyneuropathy: assessed in diabetics. Muscle Nerve. 1988;11:21–32. doi: 10.1002/mus.880110106.
    1. Dyck PJ, et al. Human diabetic endoneurial sorbitol, fructose, and myo-inositol related to sural nerve morphometry. Ann Neurol. 1980;8:590–596. doi: 10.1002/ana.410080608.
    1. Dyck PJ, Karnes JL, O'Brien PC, Litchy WJ, Low PA, Melton LJ., 3rd The Rochester Diabetic Neuropathy Study: reassessment of tests and criteria for diagnosis and staged severity. Neurology. 1992;42:1164–1170. doi: 10.1212/wnl.42.6.1164.
    1. Dyck PJ, et al. The prevalence by staged severity of various types of diabetic neuropathy, retinopathy, and nephropathy in a population-based cohort: the Rochester Diabetic Neuropathy Study. Neurology. 1993;43:817–824. doi: 10.1212/wnl.43.4.817.
    1. Dyck PJ, et al. Signs and symptoms versus nerve conduction studies to diagnose diabetic sensorimotor polyneuropathy: Cl vs NPhys trial. Muscle Nerve. 2010;42:157–164. doi: 10.1002/mus.21661.
    1. Dyck PJ, Carter RE, Litchy WJ. Modeling nerve conduction criteria for diagnosis of diabetic polyneuropathy. Muscle Nerve. 2011;44:340–345. doi: 10.1002/mus.22074.
    1. Edwards JL, Vincent AM, Cheng HT, Feldman EL. Diabetic neuropathy: mechanisms to management. Pharmacol Ther. 2008;120:1–34. doi: 10.1016/j.pharmthera.2008.05.005.
    1. Edwards RR, et al. Patient phenotyping in clinical trials of chronic pain treatments: IMMPACT recommendations. Pain. 2016;157:1851–1871. doi: 10.1097/j.pain.0000000000000602.
    1. Ellis RJ, et al. Continued high prevalence and adverse clinical impact of human immunodeficiency virus-associated sensory neuropathy in the era of combination antiretroviral therapy: the CHARTER Study. Arch Neurol. 2010;67:552–558. doi: 10.1001/archneurol.2010.76.
    1. England JD, et al. Distal symmetrical polyneuropathy: a definition for clinical research. A report of the American Academy of Neurology, the American Association of Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Arch Phys Med Rehabil. 2005;86:167–174. doi: 10.1016/j.apmr.2004.09.011.
    1. Epping R, Verhagen AP, Hoebink EA, Rooker S, Scholten-Peeters GGM. The diagnostic accuracy and test–retest reliability of the Dutch PainDETECT and the DN4 screening tools for neuropathic pain in patients with suspected cervical or lumbar radiculopathy. Musculoskelet Sci Pract. 2017;30:72–79. doi: 10.1016/j.msksp.2017.05.010.
    1. Estacion M, et al. NaV1.7 gain-of-function mutations as a continuum: A1632E displays physiological changes associated with erythromelalgia and paroxysmal extreme pain disorder mutations and produces symptoms of both disorders. J Neurosci. 2008;28:11079–11088. doi: 10.1523/jneurosci.3443-08.2008.
    1. European Medicines Agency. EMA/CHMP/970057/2011: guideline on the clinical development of medicinal products intended for the treatment of pain. . Accessed 15 Sep 2019
    1. Ezenwa M, Yao Y, Suarez M, Zhao Z, Carrasco J, Angulo V, Shuey D, Roach K, Wang Z, Molokie R, Wilkie D. (187) Normative values for quantitative sensory testing in African Americans. J Pain. 2016;17:4. doi: 10.1016/j.jpain.2016.01.090.
    1. Farooqi MA, et al. Validation of cooling detection threshold as a marker of sensorimotor polyneuropathy in type 2 diabetes. J Diabetes Complications. 2016;30:716–722. doi: 10.1016/j.jdiacomp.2015.12.023.
    1. Feldman EL, Stevens MJ, Thomas PK, Brown MB, Canal N, Greene DA. A practical two-step quantitative clinical and electrophysiological assessment for the diagnosis and staging of diabetic neuropathy. Diabetes Care. 1994;17:1281–1289. doi: 10.2337/diacare.17.11.1281.
    1. Feldman EL, Nave KA, Jensen TS, Bennett DLH. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron. 2017;93:1296–1313. doi: 10.1016/j.neuron.2017.02.005.
    1. Feldman EL, et al. Diabetic neuropathy. Nat Rev Dis Primers. 2019;5:41. doi: 10.1038/s41572-019-0092-1.
    1. Fernyhough P. Mitochondrial dysfunction in diabetic neuropathy: a series of unfortunate metabolic events. Curr DiabRep. 2015;15:89. doi: 10.1007/s11892-015-0671-9.
    1. Fernyhough P, McGavock J. Mechanisms of disease: mitochondrial dysfunction in sensory neuropathy and other complications in diabetes. Handb Clin Neurol. 2014;126:353–377. doi: 10.1016/b978-0-444-53480-4.00027-8.
    1. Fields HL, Rowbotham M, Baron R. Postherpetic neuralgia: irritable nociceptors and deafferentation. Neurobiol Dis. 1998;5:209–227. doi: 10.1006/nbdi.1998.0204.
    1. Finnerup NB, Sindrup SH, Jensen TS. The evidence for pharmacological treatment of neuropathic pain. Pain. 2010;150:573–581. doi: 10.1016/j.pain.2010.06.019.
    1. Finnerup NB, et al. Neuropathic pain needs systematic classification. Eur J Pain. 2013;17:953–956. doi: 10.1002/j.1532-2149.2012.00282.x.
    1. Finnerup NB, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14:162–173. doi: 10.1016/S1474-4422(14)70251-0.
    1. Finnerup NB, et al. Neuropathic pain: an updated grading system for research and clinical practice. Pain. 2016;157:1599–1606. doi: 10.1097/j.pain.0000000000000492.
    1. Foley PL, et al. Prevalence and natural history of pain in adults with multiple sclerosis: systematic review and meta-analysis. Pain. 2013;154:632–642. doi: 10.1016/j.pain.2012.12.002.
    1. Forbes HJ, Thomas SL, Smeeth L, Clayton T, Farmer R, Bhaskaran K, Langan SM. A systematic review and meta-analysis of risk factors for postherpetic neuralgia. Pain. 2016;157:30–54. doi: 10.1097/j.pain.0000000000000307.
    1. Forstenpointner J, Otto J, Baron R. Individualized neuropathic pain therapy based on phenotyping: are we there yet? Pain. 2018;159:569–575. doi: 10.1097/j.pain.0000000000001088.
    1. Frazier WA, Angeletti RH, Bradshaw RA. Nerve growth factor and insulin. Science (New York, NY) 1972;176:482–488. doi: 10.1126/science.176.4034.482.
    1. Freo U, Romualdi P, Kress HG. Tapentadol for neuropathic pain: a review of clinical studies. J Pain Res. 2019;12:1537–1551. doi: 10.2147/JPR.S190162.
    1. Freynhagen R, Strojek K, Griesing T, Whalen E, Balkenohl M. Efficacy of pregabalin in neuropathic pain evaluated in a 12-week, randomised, double-blind, multicentre, placebo-controlled trial of flexible- and fixed-dose regimens. Pain. 2005;115:254–263. doi: 10.1016/j.pain.2005.02.032.
    1. Freynhagen R, Baron R, Gockel U, Tolle TR. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin. 2006;22:1911–1920. doi: 10.1185/030079906X132488.
    1. Freynhagen R, et al. Current understanding of the mixed pain concept: a brief narrative review. Curr Med Res Opin. 2019;35:1011–1018. doi: 10.1080/03007995.2018.1552042.
    1. Fukuoka M, Sakurai K, Ohta T, Kiyoki M, Katayama I. Tacalcitol, an active vitamin D3, induces nerve growth factor production in human epidermal keratinocytes. Skin Pharmacol Appl Skin Physiol. 2001;14:226–233. doi: 10.1159/000056351.
    1. Fullerton B, Jeitler K, Seitz M, Horvath K, Berghold A, Siebenhofer A. Intensive glucose control versus conventional glucose control for type 1 diabetes mellitus. Cochrane Database Syst Rev. 2014 doi: 10.1002/14651858.CD009122.pub2.
    1. Gallagher HC, Gallagher RM, Butler M, Buggy DJ, Henman MC. Venlafaxine for neuropathic pain in adults. Cochrane Database Syst Rev. 2015 doi: 10.1002/14651858.CD011091.pub2.
    1. Gao F, Zheng ZM. Animal models of diabetic neuropathic pain. Exp Clin Endocrinol Diabetes. 2014;122:100–106. doi: 10.1055/s-0033-1363234.
    1. Gasparotti R, Padua L, Briani C, Lauria G. New technologies for the assessment of neuropathies. Nat Rev Neurol. 2017;13:203–216. doi: 10.1038/nrneurol.2017.31.
    1. German National Disease Management Guideline for Diabetic Neuropathy: Bundesärztekammer (BÄK), Kassenärztliche Bundesvereinigung (KBV), Arbeitsgemeinschaft der Wissenschaftli-chen Medizinischen Fachgesellschaften (AWMF). Nationale VersorgungsLeitlinie Neuropathie bei Diabetes im Erwachsenenalter—Langfassung, 1. Auflage. Version 5. 2011. . Accessed 17 Sep 2019. 10.6101/AZQ/000302
    1. Gibbons CH. Treatment-induced neuropathy of diabetes. Curr Diab Rep. 2017;17:127. doi: 10.1007/s11892-017-0960-6.
    1. Gibbons CH. Treatment induced neuropathy of diabetes—long term implications in type 1 diabetes. J Diabetes Complications. 2017;31:715–720. doi: 10.1016/j.jdiacomp.2017.01.010.
    1. Gibbons CH, Freeman R. Treatment-induced neuropathy of diabetes: an acute, iatrogenic complication of diabetes. Brain. 2015;138:43–52. doi: 10.1093/brain/awu307.
    1. Gierthmuhlen J, Baron R. Neuropathic pain. Semin Neurol. 2016;36:462–468. doi: 10.1055/s-0036-1584950.
    1. Gierthmühlen J, Maier C, Baron R, Tölle T, Treede RD, Birbaumer N, Huge V, Koroschetz J, Krumova EK, Lauchart M, Maihöfner C, Richter H, Westermann A, DFNS Study Group Sensory signs in complex regional pain syndrome and peripheral nerve injury. Pain. 2012;153:765–774. doi: 10.1016/j.pain.2011.11.009.
    1. Gilron I, Jensen TS, Dickenson AH. Combination pharmacotherapy for management of chronic pain: from bench to bedside. Lancet Neurol. 2013;12(11):1084–1095. doi: 10.1016/S1474-4422(13)70193-5.
    1. Gimbel JS, Richards P, Portenoy RK. Controlled-release oxycodone for pain in diabetic neuropathy: a randomized controlled trial. Neurology. 2003;60:927–934. doi: 10.1212/01.wnl.0000057720.36503.2c.
    1. Gold MS, Gebhart GF. Nociceptor sensitization in pain pathogenesis. Nat Med. 2010;16:1248–1257. doi: 10.1038/nm.2235.
    1. Gold MS, Reichling DB, Shuster MJ, Levine JD. Hyperalgesic agents increase a tetrodotoxin-resistant Na+ current in nociceptors. Proc Natl Acad Sci USA. 1996;93(3):1108–1112. doi: 10.1073/pnas.93.3.1108.
    1. Gonzalez-Duarte A, Lem-Carrillo M, Guerrero-Torres L. Normative values of quantitative sensory testing in Hispanic Latino population. Brain Behav. 2016;6:e00466. doi: 10.1002/brb3.466.
    1. Goodman CW, Brett AS. Gabapentin and Pregabalin for pain—is increased prescribing a cause for concern? N Engl J Med. 2017;377:411–414. doi: 10.1056/NEJMp1704633.
    1. Gore M, Brandenburg NA, Dukes E, Hoffman DL, Tai KS, Stacey B. Pain severity in diabetic peripheral neuropathy is associated with patient functioning, symptom levels of anxiety and depression, and sleep. J Pain Symptom Manag. 2005;30:374–385. doi: 10.1016/j.jpainsymman.2005.04.009.
    1. Gracely RH, Lynch SA, Bennett GJ. Painful neuropathy: altered central processing maintained dynamically by peripheral input. Pain. 1992;51:175–194. doi: 10.1016/0304-3959(92)90259-E.
    1. Granovsky Y. Conditioned pain modulation: a predictor for development and treatment of neuropathic pain. Curr Pain Headache Rep. 2013;17:361. doi: 10.1007/s11916-013-0361-8.
    1. Granovsky Y, Nahman-Averbuch H, Khamaisi M, Granot M. Efficient conditioned pain modulation despite pain persistence in painful diabetic neuropathy. Pain Rep. 2017;2:e592. doi: 10.1097/pr9.0000000000000592.
    1. Gregory NS, Harris AL, Robinson CR, Dougherty PM, Fuchs PN, Sluka KA. An overview of animal models of pain: disease models and outcome measures. J Pain. 2013;14:1255–1269. doi: 10.1016/j.jpain.2013.06.008.
    1. Grewal AS, Bhardwaj S, Pandita D, Lather V, Sekhon BS. Updates on aldose reductase inhibitors for management of diabetic complications and non-diabetic diseases. Mini Rev Med Chem. 2016;16:120–162. doi: 10.2174/1389557515666150909143737.
    1. Grote CW, Wright DE. A role for insulin in diabetic neuropathy. Front Neurosci. 2016;10:581. doi: 10.3389/fnins.2016.00581.
    1. Gruber-Schoffnegger D, Drdla-Schutting R, Honigsperger C, Wunderbaldinger G, Gassner M, Sandkuhler J. Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-alpha and IL-1beta is mediated by glial cells. J Neurosci. 2013;33:6540–6551. doi: 10.1523/jneurosci.5087-12.2013.
    1. Haanpaa ML, Backonja MM, Bennett MI, Bouhassira D, Cruccu G, Hansson PT, Jensen TS, Kauppila T, Rice ASC, Smith BH, Treede RD, Baron R. Assessment of neuropathic pain in primary care. Am J Med. 2009;122:S13–S21. doi: 10.1016/j.amjmed.2009.04.006.
    1. Haanpaa M, et al. NeuPSIG guidelines on neuropathic pain assessment. Pain. 2011;152:14–27. doi: 10.1016/j.pain.2010.07.031.
    1. Hains BC, Waxman SG. Sodium channel expression and the molecular pathophysiology of pain after SCI. Prog Brain Res. 2007;161:195–203. doi: 10.1016/S0079-6123(06)61013-3.
    1. Hameed S. Nav1.7 and Nav1.8: role in the pathophysiology of pain. Mol Pain. 2019;15:1744806919858801. doi: 10.1177/1744806919858801.
    1. Hansson P. Neuropathic pain: clinical characteristics and diagnostic workup. Eur J Pain. 2002;6(Suppl A):47–50. doi: 10.1053/eujp.2001.0322.
    1. Haroutounian S, Nikolajsen L, Bendtsen TF, Finnerup NB, Kristensen AD, Hasselstrom JB, Jensen TS. Primary afferent input critical for maintaining spontaneous pain in peripheral neuropathy. Pain. 2014;155:1272–1279. doi: 10.1016/j.pain.2014.03.022.
    1. Hatch MN, Cushing TR, Carlson GD, Chang EY. Neuropathic pain and SCI: identification and treatment strategies in the 21st century. J Neurol Sci. 2018;384:75–83. doi: 10.1016/j.jns.2017.11.018.
    1. Hearn L, Derry S, Phillips T, Moore RA, Wiffen PJ. Imipramine for neuropathic pain in adults. Cochrane Database Syst Rev. 2014 doi: 10.1002/14651858.CD010769.pub2.
    1. Hearn L, Moore RA, Derry S, Wiffen PJ, Phillips T. Desipramine for neuropathic pain in adults. Cochrane Database Syst Rev. 2014 doi: 10.1002/14651858.CD011003.pub2.
    1. Hebert HL, Veluchamy A, Torrance N, Smith BH. Risk factors for neuropathic pain in diabetes mellitus. Pain. 2017;158:560–568. doi: 10.1097/j.pain.0000000000000785.
    1. Herder C, et al. Proinflammatory cytokines predict the incidence and progression of distal sensorimotor polyneuropathy: KORA F4/FF4 study. Diabetes Care. 2017;40:569–576. doi: 10.2337/dc16-2259.
    1. Hildebrand ME, et al. Potentiation of synaptic GluN2B NMDAR currents by Fyn kinase is gated through BDNF-mediated disinhibition in spinal pain processing. Cell Rep. 2016;17:2753–2765. doi: 10.1016/j.celrep.2016.11.024.
    1. Hirayasu K, et al. Difference in normal limit values of nerve conduction parameters between Westerners and Japanese people might need to be considered when diagnosing diabetic polyneuropathy using a Point-of-Care Sural Nerve Conduction Device (NC-stat(R)/DPNCheck) J Diabetes Investig. 2018;9:1173–1181. doi: 10.1111/jdi.12818.
    1. Hoeijmakers JG, Faber CG, Merkies IS, Waxman SG. Painful peripheral neuropathy and sodium channel mutations. Neurosci Lett. 2015;596:51–59. doi: 10.1016/j.neulet.2014.12.056.
    1. Hoffmann T, Kistner K, Carr RW, Nassar MA, Reeh PW, Weidner C. Reduced excitability and impaired nociception in peripheral unmyelinated fibers from Nav1.9-null mice. Pain. 2017;158:58–67. doi: 10.1097/j.pain.0000000000000723.
    1. Holiner I, et al. Validity of the neurological examination in diagnosing diabetic peripheral neuropathy. Pediatr Neurol. 2013;49:171–177. doi: 10.1016/j.pediatrneurol.2013.03.014.
    1. Hsieh ST. Pathology and functional diagnosis of small-fiber painful neuropathy. Acta Neurol Taiwan. 2010;19:82–89.
    1. Huang Q, Chen Y, Gong N, Wang YX. Methylglyoxal mediates streptozotocin-induced diabetic neuropathic pain via activation of the peripheral TRPA1 and Nav1.8 channels. Metab Clin Exp. 2016;65:463–474. doi: 10.1016/j.metabol.2015.12.002.
    1. Hwang YT, Davies G. 'Insulin neuritis' to 'treatment-induced neuropathy of diabetes': new name, same mystery. Pract Neurol. 2016;16:53–55. doi: 10.1136/practneurol-2015-001215.
    1. IASP (2011) Part III: Pain terms. A current list with definitions and notes on usage. International Association for the Study of Pain (IASP). Pain terms. . Accessed 15 Mar 2019
    1. IDF Diabetes Atlas, 8th ed. . Accessed 30 Oct 2019
    1. Inoue K. Purinergic signaling in microglia in the pathogenesis of neuropathic pain. Proc Jpn Acad Ser B Phys Biol Sci. 2017;93:174–182. doi: 10.2183/pjab.93.011.
    1. Inoue K, Tsuda M. Microglia in neuropathic pain: cellular and molecular mechanisms and therapeutic potential. Nat Rev Neurosci. 2018;19:138–152. doi: 10.1038/nrn.2018.2.
    1. Inyang KE, Szabo-Pardi T, Wentworth E, McDougal TA, Dussor G, Burton MD, Price TJ. The antidiabetic drug metformin prevents and reverses neuropathic pain and spinal cord microglial activation in male but not female mice. Pharmacol Res. 2019;139:1–16. doi: 10.1016/j.phrs.2018.10.027.
    1. Ismail-Beigi F, et al. Effect of intensive treatment of hyperglycaemia on microvascular outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. Lancet. 2010;376:419–430. doi: 10.1016/S0140-6736(10)60576-4.
    1. Jaggi AS, Jain V, Singh N. Animal models of neuropathic pain. Fundam Clin Pharmacol. 2011;25:1–28. doi: 10.1111/j.1472-8206.2009.00801.x.
    1. Jain R, Jain S, Raison CL, Maletic V. Painful diabetic neuropathy is more than pain alone: examining the role of anxiety and depression as mediators and complicators. Curr Diab Rep. 2011;11:275–284. doi: 10.1007/s11892-011-0202-2.
    1. Jay GW, Barkin RL. Neuropathic pain: etiology, pathophysiology, mechanisms, and evaluations. Dis Mon. 2014;60:6–47. doi: 10.1016/j.disamonth.2013.12.001.
    1. Jensen TS, Finnerup NB. Allodynia and hyperalgesia in neuropathic pain: clinical manifestations and mechanisms. Lancet Neurol. 2014;13:924–935. doi: 10.1016/s1474-4422(14)70102-4.
    1. Jensen TS, Bach FW, Kastrup J, Dejgaard A, Brennum J. Vibratory and thermal thresholds in diabetics with and without clinical neuropathy. Acta Neurol Scand. 1991;84:326–333. doi: 10.1111/j.1600-0404.1991.tb04963.x.
    1. Jensen TS, Madsen CS, Finnerup NB. Pharmacology and treatment of neuropathic pains. Curr Opin Neurol. 2009;22:467–474. doi: 10.1097/WCO.0b013e3283311e13.
    1. Jensen TS, Baron R, Haanpaa M, Kalso E, Loeser JD, Rice AS, Treede RD. A new definition of neuropathic pain. Pain. 2011;152:2204–2205. doi: 10.1016/j.pain.2011.06.017.
    1. Ji RR, Kohno T, Moore KA, Woolf CJ. Central sensitization and LTP: do pain and memory share similar mechanisms? Trends Neurosci. 2003;26:696–705. doi: 10.1016/j.tins.2003.09.017.
    1. Ji RR, Berta T, Nedergaard M. Glia and pain: is chronic pain a gliopathy? Pain. 2013;154(Suppl 1):S10–28. doi: 10.1016/j.pain.2013.06.022.
    1. Ji RR, Nackley A, Huh Y, Terrando N, Maixner W. Neuroinflammation and central sensitization in chronic and widespread pain. Anesthesiology. 2018;129:343–366. doi: 10.1097/aln.0000000000002130.
    1. Jiang B, Zhang Y, Zhao J, She C, Zhou X, Dong Q, Wang P. Effects of localized X-ray irradiation on peripheral nerve regeneration in transected sciatic nerve in rats. Radiat Res. 2017;188:455–462. doi: 10.1667/RR14799.1.
    1. Jin SX, Zhuang ZY, Woolf CJ, Ji RR. p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci. 2003;23:4017–4022. doi: 10.1523/JNEUROSCI.23-10-04017.2003.
    1. Karst M, Salim K, Burstein S, Conrad I, Hoy L, Schneider U. Analgesic effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial. JAMA. 2003;290:1757–1762. doi: 10.1001/jama.290.13.1757.
    1. Kavitha KV, Tiwari S, Purandare VB, Khedkar S, Bhosale SS, Unnikrishnan AG. Choice of wound care in diabetic foot ulcer: a practical approach. World J Diabetes. 2014;5:546–556. doi: 10.4239/wjd.v5.i4.546.
    1. Keller AF, Beggs S, Salter MW, De Koninck Y. Transformation of the output of spinal lamina I neurons after nerve injury and microglia stimulation underlying neuropathic pain. Mol Pain. 2007;3:27. doi: 10.1186/1744-8069-3-27.
    1. Kennedy DL, Kemp HI, Ridout D, Yarnitsky D, Rice AS. Reliability of conditioned pain modulation: a systematic review. Pain. 2016;157:2410–2419. doi: 10.1097/j.pain.0000000000000689.
    1. Khangura RK, Sharma J, Bali A, Singh N, Jaggi AS. An integrated review on new targets in the treatment of neuropathic pain. Korean J Physiol Pharmacol. 2019;23:1–20. doi: 10.4196/kjpp.2019.23.1.1.
    1. Kim SK, et al. Cortical astrocytes rewire somatosensory cortical circuits for peripheral neuropathic pain. J Clin Investig. 2016;126:1983–1997. doi: 10.1172/jci82859.
    1. Klein T, Magerl W, Hopf HC, Sandkuhler J, Treede RD. Perceptual correlates of nociceptive long-term potentiation and long-term depression in humans. J Neurosci. 2004;24:964–971. doi: 10.1523/jneurosci.1222-03.2004.
    1. Klein T, Magerl W, Rolke R, Treede RD. Human surrogate models of neuropathic pain. Pain. 2005;115:227–233. doi: 10.1016/j.pain.2005.03.021.
    1. Kobayashi M, Zochodne DW. Diabetic neuropathy and the sensory neuron: new aspects of pathogenesis and their treatment implications. J Diabetes Investig. 2018;9:1239–1254. doi: 10.1111/jdi.12833.
    1. Kopf S, et al. Deep phenotyping neuropathy: an underestimated complication in patients with pre-diabetes and type 2 diabetes associated with albuminuria. Diabetes Res Clin Pract. 2018;146:191–201. doi: 10.1016/j.diabres.2018.10.020.
    1. Kosek E, et al. Do we need a third mechanistic descriptor for chronic pain states? Pain. 2016;157:1382–1386. doi: 10.1097/j.pain.0000000000000507.
    1. Kotani N, et al. Cerebrospinal fluid interleukin 8 concentrations and the subsequent development of postherpetic neuralgia. Am J Med. 2004;116:318–324. doi: 10.1016/j.amjmed.2003.10.027.
    1. Krause SJ, Backonja MM. Development of a neuropathic pain questionnaire. Clin J Pain. 2003;19:306–314. doi: 10.1097/00002508-200309000-00004.
    1. Krishnan ST, Rayman G. The LDIflare: a novel test of C-fiber function demonstrates early neuropathy in type 2 diabetes. Diabetes Care. 2004;27:2930–2935. doi: 10.2337/diacare.27.12.2930.
    1. Lacagnina MJ, Watkins LR, Grace PM. Toll-like receptors and their role in persistent pain. Pharmacol Ther. 2018;184:145–158. doi: 10.1016/j.pharmthera.2017.10.006.
    1. La Cesa S, et al. How to diagnose neuropathic pain? The contribution from clinical examination, pain questionnaires and diagnostic tests. Neurol Sci. 2015;36:2169–2175. doi: 10.1007/s10072-015-2382-z.
    1. Lai J, Hunter JC, Porreca F. The role of voltage-gated sodium channels in neuropathic pain. Curr Opin Neurobiol. 2003;13:291–297. doi: 10.1016/S0959-4388(03)00074-6.
    1. Lai J, Porreca F, Hunter JC, Gold MS. Voltage-gated sodium channels and hyperalgesia. Annu Rev Pharmacol Toxicol. 2004;44:371–397. doi: 10.1146/annurev.pharmtox.44.101802.121627.
    1. Laiteerapong N, Ham SA, Gao Y, Moffet HH, Liu JY, Huang ES, Karter AJ. The legacy effect in Type 2 diabetes: impact of early glycemic control on future complications (the diabetes and aging study) Diabetes Care. 2019;42:416–426. doi: 10.2337/dc17-1144.
    1. Latremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain. 2009;10:895–926. doi: 10.1016/j.jpain.2009.06.012.
    1. Lauria G, et al. European Federation of Neurological Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis of small fiber neuropathy. Report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society. Eur J Neurol. 2010;17:903–912, e944–909. doi: 10.1111/j.1468-1331.2010.03023.x.
    1. Leone C, et al. Cooling the skin for assessing small-fibre function. Pain. 2019;160:1967–1975. doi: 10.1097/j.pain.0000000000001584.
    1. Lenz FA, Kwan HC, Martin R, Tasker R, Richardson RT, Dostrovsky JO. Characteristics of somatotopic organization and spontaneous neuronal activity in the region of the thalamic principal sensory nucleus in patients with spinal cord transection. J Neurophysiol. 1994;72:1570–1587. doi: 10.1152/jn.1994.72.4.1570.
    1. Liyanage PL, Lekamwasam S, Weerarathna TP. Validity of the Diabetic Neuropathy Score and Diabetic Neuropathy Examination score as screening tools for the detection of distal symmetrical diabetic neuropathy. J Diabetes. 2012;4:264–265. doi: 10.1111/j.1753-0407.2012.00201.x.
    1. Loeser JD, Treede RD. The Kyoto protocol of IASP basic pain terminology. Pain. 2008;137:473–477. doi: 10.1016/j.pain.2008.04.025.
    1. Love S. An experimental study of peripheral nerve regeneration after x-irradiation. Brain. 1983;106(Pt 1):39–54. doi: 10.1093/brain/106.1.39.
    1. Low PA, Singer W. Treatment-induced neuropathy of diabetes: an energy crisis? Brain. 2015;138:2–3. doi: 10.1093/brain/awu327.
    1. Lu WY, Xiong ZG, Lei S, Orser BA, Dudek E, Browning MD, MacDonald JF. G-protein-coupled receptors act via protein kinase C and Src to regulate NMDA receptors. Nat Neurosci. 1999;2:331–338. doi: 10.1038/7243.
    1. Lunn MP, Hughes RA, Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database Syst Rev. 2014 doi: 10.1002/14651858.CD007115.pub3.
    1. Luo ZD, Chaplan SR, Higuera ES, Sorkin LS, Stauderman KA, Williams ME, Yaksh TL. Upregulation of dorsal root ganglion (alpha)2(delta) calcium channel subunit and its correlation with allodynia in spinal nerve-injured rats. J Neurosci. 2001;21:1868–1875. doi: 10.1523/JNEUROSCI.21-06-01868.2001.
    1. Lucchetta M, Pazzaglia C, Padua L, Briani C. Exploring neuropathic symptoms in a large cohort of Italian patients with different peripheral nervous system diseases. Neurol Sci. 2011;32(3):423–426. doi: 10.1007/s10072-011-0498-3.
    1. Magerl W, Krumova EK, Baron R, Tolle T, Treede RD, Maier C. Reference data for quantitative sensory testing (QST): refined stratification for age and a novel method for statistical comparison of group data. Pain. 2010;151:598–605. doi: 10.1016/j.pain.2010.07.026.
    1. Magrinelli F, et al. The association between serum cytokines and damage to large and small nerve fibers in diabetic peripheral neuropathy. J Diabetes Res. 2015;2015:547834. doi: 10.1155/2015/547834.
    1. Malik RA, et al. Sural nerve pathology in diabetic patients with minimal but progressive neuropathy. Diabetologia. 2005;48:578–585. doi: 10.1007/s00125-004-1663-5.
    1. Mallick-Searle T, Snodgrass B, Brant JM. Postherpetic neuralgia: epidemiology, pathophysiology, and pain management pharmacology. J Multidiscip Healthc. 2016;9:447–454. doi: 10.2147/JMDH.S106340.
    1. Manchikanti L, Singh V. Managing phantom pain. Pain Phys. 2004;7:365–375.
    1. Marchand F, Perretti M, McMahon SB. Role of the immune system in chronic pain. Nat Rev Neurosci. 2005;6:521–532. doi: 10.1038/nrn1700.
    1. Margolis RB, Tait RC, Krause SJ. A rating system for use with patient pain drawings. Pain. 1986;24:57–65. doi: 10.1016/0304-3959(86)90026-6.
    1. Martin MM. Diabetic neuropathy; a clinical study of 150 cases. Brain. 1953;76:594–624. doi: 10.1093/brain/76.4.594.
    1. Martinez V, et al. The efficacy of a glial inhibitor, minocycline, for preventing persistent pain after lumbar discectomy: a randomized, double-blind, controlled study. Pain. 2013;154:1197–1203. doi: 10.1016/j.pain.2013.03.028.
    1. Mathieson S, Maher CG, Terwee CB, Folly de Campos T, Lin CW. Neuropathic pain screening questionnaires have limited measurement properties. A systematic review. J Clin Epidemiol. 2015;68:957–966. doi: 10.1016/j.jclinepi.2015.03.010.
    1. Max MB. Towards physiologically based treatment of patients with neuropathic pain. Pain. 1990;42:131–137. doi: 10.1016/0304-3959(90)91156-d.
    1. May A. Chronic pain may change the structure of the brain. Pain. 2008;137:7–15. doi: 10.1016/j.pain.2008.02.034.
    1. McMahon SB, Malcangio M. Current challenges in glia-pain biology. Neuron. 2009;64:46–54. doi: 10.1016/j.neuron.2009.09.033.
    1. Meacham K, Shepherd A, Mohapatra DP, Haroutounian S. Neuropathic pain: central vs. peripheral mechanisms. Curr Pain Headache Rep. 2017;21:28. doi: 10.1007/s11916-017-0629-5.
    1. Mehra S, Tavakoli M, Kallinikos PA, Efron N, Boulton AJ, Augustine T, Malik RA. Corneal confocal microscopy detects early nerve regeneration after pancreas transplantation in patients with type 1 diabetes. Diabetes Care. 2007;30:2608–2612. doi: 10.2337/dc07-0870.
    1. Meijer JW, van Sonderen E, Blaauwwiekel EE, Smit AJ, Groothoff JW, Eisma WH, Links TP. Diabetic neuropathy examination: a hierarchical scoring system to diagnose distal polyneuropathy in diabetes. Diabetes Care. 2000;23:750–753. doi: 10.2337/diacare.23.6.750.
    1. Meijer JW, Smit AJ, Sonderen EV, Groothoff JW, Eisma WH, Links TP. Symptom scoring systems to diagnose distal polyneuropathy in diabetes: the Diabetic Neuropathy Symptom score. Diabet Med. 2002;19:962–965. doi: 10.1046/j.1464-5491.2002.00819.x.
    1. Meijer JW, et al. Clinical diagnosis of diabetic polyneuropathy with the diabetic neuropathy symptom and diabetic neuropathy examination scores. Diabetes Care. 2003;26:697–701. doi: 10.2337/diacare.26.3.697.
    1. Meijer JW, Smit AJ, Lefrandt JD, van der Hoeven JH, Hoogenberg K, Links TP. Back to basics in diagnosing diabetic polyneuropathy with the tuning fork! Diabetes Care. 2005;28:2201–2205. doi: 10.2337/diacare.28.9.2201.
    1. Meng H, Johnston B, Englesakis M, Moulin DE, Bhatia A. Selective cannabinoids for chronic neuropathic pain: a systematic review and meta-analysis. Anesth Analg. 2017;125:1638–1652. doi: 10.1213/ANE.0000000000002110.
    1. Mick G, Baron R, Finnerup NB, Hans G, Kern KU, Brett B, Dworkin RH. What is localized neuropathic pain? A first proposal to characterize and define a widely used term. Pain Manag. 2011 doi: 10.2217/pmt.11.77.
    1. Mickle AD, Shepherd AJ, Mohapatra DP. Sensory TRP channels: the key transducers of nociception and pain. Progress Mol Biol Transl Sci. 2015;131:73–118. doi: 10.1016/bs.pmbts.2015.01.002.
    1. Mickle AD, Shepherd AJ, Mohapatra DP. Nociceptive TRP channels: sensory detectors and transducers in multiple pain pathologies. Pharmaceuticals (Basel, Switzerland) 2016 doi: 10.3390/ph9040072.
    1. Mika J. Modulation of microglia can attenuate neuropathic pain symptoms and enhance morphine effectiveness. Pharmacol Rep. 2008;60:297–307.
    1. Milligan ED, Sloane EM, Watkins LR. Glia in pathological pain: a role for fractalkine. J Neuroimmunol. 2008;198:113–120. doi: 10.1016/j.jneuroim.2008.04.011.
    1. Minett MS, Falk S, Santana-Varela S, Bogdanov YD, Nassar MA, Heegaard AM, Wood JN. Pain without nociceptors? Nav1.7-independent pain mechanisms. Cell Rep. 2014;6:301–312. doi: 10.1016/j.celrep.2013.12.033.
    1. Moghtaderi A, Bakhshipour A, Rashidi H. Validation of Michigan neuropathy screening instrument for diabetic peripheral neuropathy. Clin Neurol Neurosurg. 2006;108:477–481. doi: 10.1016/j.clineuro.2005.08.003.
    1. Moisset X, Bouhassira D. Brain imaging of neuropathic pain. NeuroImage. 2007;37(Suppl 1):S80–88. doi: 10.1016/j.neuroimage.2007.03.054.
    1. Moore KA, Kohno T, Karchewski LA, Scholz J, Baba H, Woolf CJ. Partial peripheral nerve injury promotes a selective loss of GABAergic inhibition in the superficial dorsal horn of the spinal cord. J Neurosci. 2002;22:6724–6731. doi: 10.1523/JNEUROSCI.22-15-06724.2002.
    1. Moore RA, Wiffen PJ, Derry S, Toelle T, Rice AS. Gabapentin for chronic neuropathic pain and fibromyalgia in adults. Cochrane Database Syst Rev. 2014 doi: 10.1002/14651858.CD007938.pub3.
    1. Moore RA, Derry S, Aldington D, Cole P, Wiffen PJ. Amitriptyline for neuropathic pain in adults. Cochrane Database Syst Rev. 2015 doi: 10.1002/14651858.CD008242.pub3.
    1. Moulin D, et al. Pharmacological management of chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Pain Res Manag. 2014;19:328–335. doi: 10.1155/2014/754693.
    1. Nardelli P, Khan J, Powers R, Cope TC, Rich MM. Reduced motoneuron excitability in a rat model of sepsis. J Neurophysiol. 2013;109:1775–1781. doi: 10.1152/jn.00936.2012.
    1. Nawroth PP, et al. The quest for more research on painful diabetic neuropathy. Neuroscience. 2018;387:28–37. doi: 10.1016/j.neuroscience.2017.09.023.
    1. Nebuchennykh M, Loseth S, Lindal S, Mellgren SI. The value of skin biopsy with recording of intraepidermal nerve fiber density and quantitative sensory testing in the assessment of small fiber involvement in patients with different causes of polyneuropathy. J Neurol. 2009;256:1067–1075. doi: 10.1007/s00415-009-5065-y.
    1. Ni HD, et al. Glial activation in the periaqueductal gray promotes descending facilitation of neuropathic pain through the p38 MAPK signaling pathway. J Neurosci Res. 2016;94:50–61. doi: 10.1002/jnr.23672.
    1. NICE (2013) Clinical guideline: neuropathic pain-pharmacological management. . Accessed July 15 2019
    1. North RY, et al. Electrophysiological and transcriptomic correlates of neuropathic pain in human dorsal root ganglion neurons. Brain. 2019;142:1215–1226. doi: 10.1093/brain/awz063.
    1. Nugraha B, et al. The IASP classification of chronic pain for ICD-11: functioning properties of chronic pain. Pain. 2019;160:88–94. doi: 10.1097/j.pain.0000000000001433.
    1. O'Brien PD, Sakowski SA, Feldman EL. Mouse models of diabetic neuropath. ILAR J. 2014;54:259–272. doi: 10.1093/ilar/ilt052.
    1. Olaleye D, Perkins BA, Bril V. Evaluation of three screening tests and a risk assessment model for diagnosing peripheral neuropathy in the diabetes clinic. Diabetes Res Clin Pract. 2001;54:115–128. doi: 10.1016/s0168-8227(01)00278-9.
    1. Pantalone KM, et al. Effect of glycemic control on the Diabetes Complications Severity Index score and development of complications in people with newly diagnosed type 2 diabetes. J Diabetes. 2018;10:192–199. doi: 10.1111/1753-0407.12613.
    1. Papanas N, Ziegler D. Efficacy of alpha-lipoic acid in diabetic neuropathy. Expert Opin Pharmacother. 2014;15:2721–2731. doi: 10.1517/14656566.2014.972935.
    1. Park J, Park HJ. Botulinum toxin for the treatment of neuropathic pain. Toxins. 2017 doi: 10.3390/toxins9090260.
    1. Pascal MMV, et al. DOLORisk: study protocol for a multi-centre observational study to understand the risk factors and determinants of neuropathic pain. Wellcome Open Res. 2018;3:63. doi: 10.12688/wellcomeopenres.14576.2.
    1. Patel R, Dickenson AH. Neuronal hyperexcitability in the ventral posterior thalamus of neuropathic rats: modality selective effects of pregabalin. J Neurophysiol. 2016;116:159–170. doi: 10.1152/jn.00237.2016.
    1. Peltier A, Goutman SA, Callaghan BC. Painful diabetic neuropathy. BMJ. 2014;348:g1799. doi: 10.1136/bmj.g1799.
    1. Perkins BA, Olaleye D, Zinman B, Bril V. Simple screening tests for peripheral neuropathy in the diabetes clinic. Diabetes Care. 2001;24:250–256. doi: 10.2337/diacare.24.2.250.
    1. Pfau DB, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): reference data for the trunk and application in patients with chronic postherpetic neuralgia. Pain. 2014;155:1002–1015. doi: 10.1016/j.pain.2014.02.004.
    1. Pinzur MS, Slovenkai MP, Trepman E, Shields NN, Diabetes Committee of American Orthopaedic F. Ankle S. Guidelines for diabetic foot care: recommendations endorsed by the Diabetes Committee of the American Orthopaedic Foot and Ankle Society. Foot Ankle Int. 2005;26:113–119. doi: 10.1177/107110070502600112.
    1. Ponirakis G, et al. The diagnostic accuracy of Neuropad for assessing large and small fibre diabetic neuropathy. Diabet Med. 2014;31:1673–1680. doi: 10.1111/dme.12536.
    1. Ponirakis G, et al. NerveCheck: an inexpensive quantitative sensory testing device for patients with diabetic neuropathy. Diabetes Res Clin Pract. 2016;113:101–107. doi: 10.1016/j.diabres.2015.12.023.
    1. Pop-Busui R, et al. Impact of glycemic control strategies on the progression of diabetic peripheral neuropathy in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) Cohort Diabetes care. 2013;36:3208–3215. doi: 10.2337/dc13-0012.
    1. Pop-Busui R, et al. Diabetic neuropathy: a position statement by the American Diabetes Association. Diabetes Care. 2017;40:136–154. doi: 10.2337/dc16-2042.
    1. Prabodha LBL, Sirisena ND, Dissanayake VHW. Susceptible and prognostic genetic factors associated with diabetic peripheral neuropathy: a comprehensive literature review. Int J Endocrinol. 2018;2018:8641942. doi: 10.1155/2018/8641942.
    1. Qaseem A, Wilt TJ, Kansagara D, Horwitch C, Barry MJ, Forciea MA, Clinical Guidelines Committee of the American College of P Hemoglobin A1c targets for glycemic control with pharmacologic therapy for nonpregnant adults with type 2 diabetes mellitus: a guidance statement update from the American College of Physicians. Ann Intern Med. 2018;168:569–576. doi: 10.7326/M17-0939.
    1. Quattrini C, et al. Surrogate markers of small fiber damage in human diabetic neuropathy. Diabetes. 2007;56:2148–2154. doi: 10.2337/db07-0285.
    1. Quattrini C, Jeziorska M, Boulton AJ, Malik RA. Reduced vascular endothelial growth factor expression and intra-epidermal nerve fiber loss in human diabetic neuropathy. Diabetes Care. 2008;31:140–145. doi: 10.2337/dc07-1556.
    1. Raghavendra V, Tanga F, DeLeo JA. Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J Pharmacol Exp Ther. 2003;306:624–630. doi: 10.1124/jpet.103.052407.
    1. Rahman M, Griffin SJ, Rathmann W, Wareham NJ. How should peripheral neuropathy be assessed in people with diabetes in primary care? A population-based comparison of four measures. Diabet Med. 2003;20:368–374. doi: 10.1046/j.1464-5491.2003.00931.x.
    1. Rahman W, D'Mello R, Dickenson AH. Peripheral nerve injury-induced changes in spinal alpha(2)-adrenoceptor-mediated modulation of mechanically evoked dorsal horn neuronal responses. J Pain. 2008;9:350–359. doi: 10.1016/j.jpain.2007.11.010.
    1. Raputova J, et al. Sensory phenotype and risk factors for painful diabetic neuropathy: a cross-sectional observational study. Pain. 2017;158:2340–2353. doi: 10.1097/j.pain.0000000000001034.
    1. Rauck RL, Shaibani A, Biton V, Simpson J, Koch B. Lacosamide in painful diabetic peripheral neuropathy: a phase 2 double-blind placebo-controlled study. Clin J Pain. 2007;23:150–158. doi: 10.1097/01.ajp.0000210957.39621.b2.
    1. Ren K, Dubner R. Interactions between the immune and nervous systems in pain. Nat Med. 2010;16:1267–1276. doi: 10.1038/nm.2234.
    1. Rice AS, Smith BH, Blyth FM. Pain and the global burden of disease. Pain. 2016;157:791–796. doi: 10.1097/j.pain.0000000000000454.
    1. Rodriguez-Gutierrez R, Lipska KJ, McCoy RG. Intensive glycemic control in type 2 diabetes mellitus—a balancing act of latent benefit and avoidable harm: a teachable moment. JAMA Intern Med. 2016;176:300–301. doi: 10.1001/jamainternmed.2015.8320.
    1. Rolke R, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): standardized protocol and reference values. Pain. 2006;123:231–243. doi: 10.1016/j.pain.2006.01.041.
    1. Roth T, van Seventer R, Murphy TK. The effect of pregabalin on pain-related sleep interference in diabetic peripheral neuropathy or postherpetic neuralgia: a review of nine clinical trials. Curr Med Res Opin. 2010;26:2411–2419. doi: 10.1185/03007995.2010.516142.
    1. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007 doi: 10.1002/14651858.CD005454.pub2.
    1. Sadosky A, Mardekian J, Parsons B, Hopps M, Bienen EJ, Markman J. Healthcare utilization and costs in diabetes relative to the clinical spectrum of painful diabetic peripheral neuropathy. J Diabetes Complicat. 2015;29:212–217. doi: 10.1016/j.jdiacomp.2014.10.013.
    1. Safarpour Y, Jabbari B. Botulinum toxin treatment of pain syndromes—an evidence based review. Toxicon. 2018;147:120–128. doi: 10.1016/j.toxicon.2018.01.017.
    1. Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017;23:1018–1027. doi: 10.1038/nm.4397.
    1. Sanchez-Ramirez GM, Caram-Salas NL, Rocha-Gonzalez HI, Vidal-Cantu GC, Medina-Santillan R, Reyes-Garcia G, Granados-Soto V. Benfotiamine relieves inflammatory and neuropathic pain in rats. Eur J Pharmacol. 2006;530:48–53. doi: 10.1016/j.ejphar.2005.11.016.
    1. Sandkuhler J. Understanding LTP in pain pathways. Mol Pain. 2007;3:9. doi: 10.1186/1744-8069-3-9.
    1. Sartor CD, Oliveira MD, Campos V, Ferreira J, Sacco ICN. Cross-cultural adaptation and measurement properties of the Brazilian Version of the Michigan Neuropathy Screening Instrument. Braz J Phys Ther. 2018;22:222–230. doi: 10.1016/j.bjpt.2017.10.004.
    1. Schafers M, Svensson CI, Sommer C, Sorkin LS. Tumor necrosis factor-alpha induces mechanical allodynia after spinal nerve ligation by activation of p38 MAPK in primary sensory neurons. J Neurosci. 2003;23:2517–2521. doi: 10.1523/JNEUROSCI.23-07-02517.2003.
    1. Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci. 2007;10:1361–1368. doi: 10.1038/nn1992.
    1. Scholz J, et al. Blocking caspase activity prevents transsynaptic neuronal apoptosis and the loss of inhibition in lamina II of the dorsal horn after peripheral nerve injury. J Neurosci. 2005;25:7317–7323. doi: 10.1523/jneurosci.1526-05.2005.
    1. Scholz J, et al. The IASP classification of chronic pain for ICD-11: chronic neuropathic pain. Pain. 2019;160:53–59. doi: 10.1097/j.pain.0000000000001365.
    1. Schratzberger P, et al. Reversal of experimental diabetic neuropathy by VEGF gene transfer. J Clin Investig. 2001;107:1083–1092. doi: 10.1172/jci12188.
    1. Schreiber AK, Nones CF, Reis RC, Chichorro JG, Cunha JM. Diabetic neuropathic pain: physiopathology and treatment. World J Diabetes. 2015;6:432–444. doi: 10.4239/wjd.v6.i3.432.
    1. Schuh-Hofer S, Fischer J, Unterberg A, Treede RD, Ahmadi R. Spinal cord stimulation modulates descending pain inhibition and temporal summation of pricking pain in patients with neuropathic pain. Acta Neurochir. 2018;160:2509–2519. doi: 10.1007/s00701-018-3669-7.
    1. Schwartz S, et al. A pooled analysis evaluating the efficacy and tolerability of tapentadol extended release for chronic, painful diabetic peripheral neuropathy. Clin Drug Investig. 2015;35:95–108. doi: 10.1007/s40261-014-0249-3.
    1. Selvarajah D, Awadh M, Gandhi R, Wilkinson ID, Tesfaye S. Alterations in somatomotor network functional connectivity in painful diabetic neuropathy—a resting state functional magnetic resonance imaging study. Diabetes. 2018 doi: 10.2337/db18-61-OR.
    1. Selvarajah D, Heiberg-Gibbons F, Wilkinson ID, Gandhi R, Tesfaye S. A magnetic resonance imaging volumetry study of regional brain atrophy in diabetic peripheral neuropathy. Diabetes. 2018 doi: 10.2337/db18-550-P.
    1. Serra J, et al. Microneurographic identification of spontaneous activity in C-nociceptors in neuropathic pain states in humans and rats. Pain. 2012;153:42–55. doi: 10.1016/j.pain.2011.08.015.
    1. Shen FY, et al. Alleviation of neuropathic pain by regulating T-type calcium channels in rat anterior cingulate cortex. Mol Pain. 2015;11:7. doi: 10.1186/s12990-015-0008-3.
    1. Shillo P, et al. Nerve and vascular biomarkers in skin biopsies differentiate painful from painless advanced diabetic peripheral neuropathy. Diabetologia. 2017;60:S71–S72.
    1. Shillo P, et al. Reduced vitamin D levels in painful diabetic peripheral neuropathy. Diabetic Med J Br Diabetic Assoc. 2019;36:44–51. doi: 10.1111/dme.13798.
    1. Shillo PR, Selvarajah D, Greig M, Rao GD, Edden RAE, Wilkinson ID, Tesfaye S. Painless diabetic peripheral neuropathy is characterised by reduced thalamic gamma-aminobutyric acid (GABA) Diabetic Med. 2016;33:15–16. doi: 10.1111/dme.5_13047.
    1. Shun CT, et al. Skin denervation in type 2 diabetes: correlations with diabetic duration and functional impairments. Brain. 2004;127:1593–1605. doi: 10.1093/brain/awh180.
    1. Singh VP, Bali A, Singh N, Jaggi AS. Advanced glycation end products and diabetic complications. Korean J Physiol Pharmacol. 2014;18:1–14. doi: 10.4196/kjpp.2014.18.1.1.
    1. Sloan G, et al. A new look at painful diabetic neuropathy. Diabetes Res Clin Pract. 2018;144:177–191. doi: 10.1016/j.diabres.2018.08.020.
    1. Smith BH, Torrance N. Epidemiology of neuropathic pain and its impact on quality of life. Curr Pain Headache Rep. 2012;16:191–198. doi: 10.1007/s11916-012-0256-0.
    1. Snyder MJ, Gibbs LM, Lindsay TJ. Treating painful diabetic peripheral neuropathy: an update. Am Fam Physician. 2016;94:227–234.
    1. Sorensen L, Molyneaux L, Yue DK. The relationship among pain, sensory loss, and small nerve fibers in diabetes. Diabetes Care. 2006;29:883–887. doi: 10.2337/diacare.29.04.06.dc05-2180.
    1. Sorge RE, et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci. 2015;18:1081–1083. doi: 10.1038/nn.4053.
    1. Spallone V. Might genetics play a role in understanding and treating diabetic polyneuropathy? Diabetes Metab Res Rev. 2017 doi: 10.1002/dmrr.2882.
    1. Spallone V, Morganti R, D'Amato C, Greco C, Cacciotti L, Marfia GA. Validation of DN4 as a screening tool for neuropathic pain in painful diabetic polyneuropathy. Diabet Med. 2012;29:578–585. doi: 10.1111/j.1464-5491.2011.03500.x.
    1. St John Smith E. Advances in understanding nociception and neuropathic pain. J Neurol. 2018;265:231–238. doi: 10.1007/s00415-017-8641-6.
    1. Stolar M. Glycemic control and complications in type 2 diabetes mellitus. Am J Med. 2010;123:S3–11. doi: 10.1016/j.amjmed.2009.12.004.
    1. Stracke H, Gaus W, Achenbach U, Federlin K, Bretzel RG. Benfotiamine in diabetic polyneuropathy (BENDIP): results of a randomised, double blind, placebo-controlled clinical study. Exp Clin Endocrinol Diabetes. 2008;116:600–605. doi: 10.1055/s-2008-1065351.
    1. Sumitani M, Ueda H, Hozumi J, Inoue R, Kogure T, Yamada Y, Kogure T. Minocycline does not decrease intensity of neuropathic pain intensity, but does improve its affective dimension. J Pain Palliate Care Pharmacother. 2016;30:31–35. doi: 10.3109/15360288.2014.1003674.
    1. Sullivan KA, Lentz SI, Roberts JL, Jr, Feldman EL. Criteria for creating and assessing mouse models of diabetic neuropathy. Curr Drug Targets. 2008;9:3–13. doi: 10.2174/138945008783431763.
    1. Sun C, et al. IL-17 contributed to the neuropathic pain following peripheral nerve injury by promoting astrocyte proliferation and secretion of proinflammatory cytokines. Mol Med Rep. 2017;15:89–96. doi: 10.3892/mmr.2016.6018.
    1. Tampin B, Briffa NK, Goucke R, Slater H. Identification of neuropathic pain in patients with neck/upper limb pain: application of a grading system and screening tools. Pain. 2013;154(12):2813–2822. doi: 10.1016/j.pain.2013.08.018.
    1. Tan T, Barry P, Reken S, Baker M, Guideline Development G. Pharmacological management of neuropathic pain in non-specialist settings: summary of NICE guidance. BMJ. 2010;340:c1079. doi: 10.1136/bmj.c1079.
    1. Tandrup T, Woolf CJ, Coggeshall RE. Delayed loss of small dorsal root ganglion cells after transection of the rat sciatic nerve. J Comp Neurol. 2000;422:172–180. doi: 10.1002/(SICI)1096-9861(20000626)422:2<172::AID-CNE2>;2-H.
    1. Taylor AM, Mehrabani S, Liu S, Taylor AJ, Cahill CM. Topography of microglial activation in sensory- and affect-related brain regions in chronic pain. J Neurosci Res. 2017;95:1330–1335. doi: 10.1002/jnr.23883.
    1. Tappe-Theodor A, Kuner R. Studying ongoing and spontaneous pain in rodents—challenges and opportunities. Eur J Neurosci. 2014;39:1881–1890. doi: 10.1111/ejn.12643.
    1. Tesfaye S, Harris ND, Wilson RM, Ward JD. Exercise-induced conduction velocity increment: a marker of impaired peripheral nerve blood flow in diabetic neuropathy. Diabetologia. 1992;35:155–159. doi: 10.1007/BF00402548.
    1. Tesfaye S, et al. Vascular risk factors and diabetic neuropathy. N Engl J Med. 2005;352:341–350. doi: 10.1056/NEJMoa032782.
    1. Tesfaye S, et al. Diabetic neuropathies: update on definitions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care. 2010;33:2285–2293. doi: 10.2337/dc10-1303.
    1. Tesfaye S, Selvarajah D, Gandhi R, Greig M, Shillo P, Fang F, Wilkinson ID. Diabetic peripheral neuropathy may not be as its name suggests: evidence from magnetic resonance imaging. Pain. 2016;157(Suppl 1):S72–80. doi: 10.1097/j.pain.0000000000000465.
    1. Thacker MA, et al. CCL2 is a key mediator of microglia activation in neuropathic pain states. Eur J Pain. 2009;13:263–272. doi: 10.1016/j.ejpain.2008.04.017.
    1. Themistocleous AC, et al. The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy. Pain. 2016;157:1132–1145. doi: 10.1097/j.pain.0000000000000491.
    1. Thrainsdottir S, et al. Endoneurial capillary abnormalities presage deterioration of glucose tolerance and accompany peripheral neuropathy in man. Diabetes. 2003;52:2615–2622. doi: 10.2337/diabetes.52.10.2615.
    1. Timmerman H, et al. Investigating the validity of the DN4 in a consecutive population of patients with chronic pain. PLoS ONE. 2017;12:e0187961. doi: 10.1371/journal.pone.0187961.
    1. Timmerman H, et al. Avoiding Catch-22: validating the PainDETECT in a population of patients with chronic pain. BMC Neurol. 2018;18:91. doi: 10.1186/s12883-018-1094-4.
    1. Timmerman H, Wilder-Smith OH, Steegers MA, Vissers KC, Wolff AP. The added value of bedside examination and screening QST to improve neuropathic pain identification in patients with chronic pain. J Pain Res. 2018;11:1307–1318. doi: 10.2147/JPR.S154698.
    1. Torrance N, Elliott AM, Lee AJ, Smith BH. Severe chronic pain is associated with increased 10 year mortality. A cohort record linkage study. Eur J Pain. 2010;14:380–386. doi: 10.1016/j.ejpain.2009.07.006.
    1. Torrance N, Ferguson JA, Afolabi E, Bennett MI, Serpell MG, Dunn KM, Smith BH. Neuropathic pain in the community: more under-treated than refractory? Pain. 2013;154:690–699. doi: 10.1016/j.pain.2012.12.022.
    1. Tracy JA, Dyck PJ. The spectrum of diabetic neuropathies. Phys Med Rehabil Clin N Am. 2008;19(1–26):v. doi: 10.1016/j.pmr.2007.10.010.
    1. Treede RD. The role of quantitative sensory testing in the prediction of chronic pain. Pain. 2019;160:S66–S69. doi: 10.1097/j.pain.0000000000001544.
    1. Treede RD, et al. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology. 2008;70:1630–1635. doi: 10.1212/01.wnl.0000282763.29778.59.
    1. Treede RD, Rief W, Barke A, Aziz Q, Bennett MI, Benoliel R, Cohen M, Evers S, Finnerup NB, First MB, Giamberardino MA, Kaasa S, Korwisi B, Kosek E, Lavandʼhomme P, Nicholas M, Perrot S, Scholz J, Schug S, Smith BH, Svensson P, Vlaeyen JWS, Wang SJ. Chronic pain as a symptom or a disease: the IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11) Pain. 2019;160:19–27. doi: 10.1097/j.pain.0000000000001384.
    1. Trouvin AP, Perrot S, Lloret-Linares C. Efficacy of venlafaxine in neuropathic pain: a narrative review of optimized treatment. Clin Ther. 2017;39:1104–1122. doi: 10.1016/j.clinthera.2017.05.347.
    1. Truini A, et al. Does the epidermal nerve fibre density measured by skin biopsy in patients with peripheral neuropathies correlate with neuropathic pain? Pain. 2014;155:828–832. doi: 10.1016/j.pain.2014.01.022.
    1. Truini A, et al. A cross-sectional study investigating frequency and features of definitely diagnosed diabetic painful polyneuropathy. Pain. 2018;159:2658–2666. doi: 10.1097/j.pain.0000000000001378.
    1. Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, Inoue K. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature. 2003;424:778–783. doi: 10.1038/nature01786.
    1. Tsuda M, Inoue K, Salter MW. Neuropathic pain and spinal microglia: a big problem from molecules in "small" glia. Trends Neurosci. 2005;28:101–107. doi: 10.1016/j.tins.2004.12.002.
    1. Tuveson B, Leffler AS, Hansson P. Heterotopic noxious conditioning stimulation (HNCS) reduced the intensity of spontaneous pain, but not of allodynia in painful peripheral neuropathy. Eur J Pain. 2007;11:452–462. doi: 10.1016/j.ejpain.2006.06.007.
    1. UKPSD Study Group U Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group Lancet. 1998;352:837–853. doi: 10.1016/S0140-6736(98)07019-6.
    1. United Nations (2019) Revision of world population prospects. . Accessed 12 07 2019
    1. Van Acker K, et al. Prevalence and impact on quality of life of peripheral neuropathy with or without neuropathic pain in type 1 and type 2 diabetic patients attending hospital outpatients clinics. Diabetes Metab. 2009;35:206–213. doi: 10.1016/j.diabet.2008.11.004.
    1. van den Born JC, Hammes HP, Greffrath W, van Goor H, Hillebrands JL. Gasotransmitters in vascular complications of diabetes. Diabetes. 2016;65:331–345. doi: 10.2337/db15-1003.
    1. van Hecke O, Austin SK, Khan RA, Smith BH, Torrance N. Neuropathic pain in the general population: a systematic review of epidemiological studies. Pain. 2014;155:654–662. doi: 10.1016/j.pain.2013.11.013.
    1. Vanelderen P, et al. Effect of minocycline on lumbar radicular neuropathic pain: a randomized, placebo-controlled, double-blind clinical trial with amitriptyline as a comparator. Anesthesiology. 2015;122:399–406. doi: 10.1097/ALN.0000000000000508.
    1. Vinik EJ, Hayes RP, Oglesby A, Bastyr E, Barlow P, Ford-Molvik SL, Vinik AI. The development and validation of the Norfolk QOL-DN, a new measure of patients' perception of the effects of diabetes and diabetic neuropathy. Diabetes Technol Ther. 2005;7:497–508. doi: 10.1089/dia.2005.7.497.
    1. Vinik A, Rosenstock J, Sharma U, Feins K, Hsu C, Merante D, Investigators D-AUUPIS. Efficacy and safety of mirogabalin (DS-5565) for the treatment of diabetic peripheral neuropathic pain: a randomized, double-blind, placebo- and active comparator-controlled, adaptive proof-of-concept phase 2 study. Diabetes Care. 2014;37:3253–3261. doi: 10.2337/dc14-1044.
    1. Vinik AI, et al. Capsaicin 8% patch repeat treatment plus standard of care (SOC) versus SOC alone in painful diabetic peripheral neuropathy: a randomised, 52-week, open-label, safety study. BMC Neurol. 2016;16:251. doi: 10.1186/s12883-016-0752-7.
    1. Vlckova-Moravcova E, Bednarik J, Dusek L, Toyka KV, Sommer C. Diagnostic validity of epidermal nerve fiber densities in painful sensory neuropathies. Muscle Nerve. 2008;37:50–60. doi: 10.1002/mus.20889.
    1. Vo T, Rice AS, Dworkin RH. Non-steroidal anti-inflammatory drugs for neuropathic pain: how do we explain continued widespread use? Pain. 2009;143:169–171. doi: 10.1016/j.pain.2009.03.013.
    1. Vollert J, et al. Quantitative sensory testing using DFNS protocol in Europe: an evaluation of heterogeneity across multiple centers in patients with peripheral neuropathic pain and healthy subjects. Pain. 2016;157:750–758. doi: 10.1097/j.pain.0000000000000433.
    1. Vollert J, et al. Pathophysiological mechanisms of neuropathic pain: comparison of sensory phenotypes in patients and human surrogate pain models. Pain. 2018;159:1090–1102. doi: 10.1097/j.pain.0000000000001190.
    1. von Hehn CA, Baron R, Woolf CJ. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron. 2012;73:638–652. doi: 10.1016/j.neuron.2012.02.008.
    1. Wall PD, Gutnick M. Ongoing activity in peripheral nerves: the physiology and pharmacology of impulses originating from a neuroma. Exp Neurol. 1974;43:580–593. doi: 10.1016/0014-4886(74)90197-6.
    1. Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful diabetic neuropathy. J Pain. 2015;16:616–627. doi: 10.1016/j.jpain.2015.03.008.
    1. Wang H, et al. Fulranumab for treatment of diabetic peripheral neuropathic pain: a randomized controlled trial. Neurology. 2014;83:628–637. doi: 10.1212/WNL.0000000000000686.
    1. Wang ZT, Yu G, Wang HS, Yi SP, Su RB, Gong ZH. Changes in VGLUT2 expression and function in pain-related supraspinal regions correlate with the pathogenesis of neuropathic pain in a mouse spared nerve injury model. Brain Res. 2015;1624:515–524. doi: 10.1016/j.brainres.2015.08.010.
    1. Watanabe K, et al. Altered cerebral blood flow in the anterior cingulate cortex is associated with neuropathic pain. J Neurol Neurosurg Psychiatry. 2018;89:1082–1087. doi: 10.1136/jnnp-2017-316601.
    1. Watson JC, Sandroni P. Central neuropathic pain syndromes. Mayo Clin Proc. 2016;91:372–385. doi: 10.1016/j.mayocp.2016.01.017.
    1. Watson CP, Moulin D, Watt-Watson J, Gordon A, Eisenhoffer J. Controlled-release oxycodone relieves neuropathic pain: a randomized controlled trial in painful diabetic neuropathy. Pain. 2003;105:71–78. doi: 10.1016/s0304-3959(03)00160-x.
    1. Weintrob N, Amitay I, Lilos P, Shalitin S, Lazar L, Josefsberg Z. Bedside neuropathy disability score compared to quantitative sensory testing for measurement of diabetic neuropathy in children, adolescents, and young adults with type 1 diabetes. J Diabetes Complicat. 2007;21:13–19. doi: 10.1016/j.jdiacomp.2005.11.002.
    1. Wiffen PJ, Derry S, Bell RF, Rice AS, Tolle TR, Phillips T, Moore RA. Gabapentin for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017;6:CD007938. doi: 10.1002/14651858.CD007938.pub4.
    1. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004;27:1047–1053. doi: 10.2337/diacare.27.5.1047.
    1. Wolff RF, Bala MM, Westwood M, Kessels AG, Kleijnen J. 5% lidocaine medicated plaster in painful diabetic peripheral neuropathy (DPN): a systematic review. Swiss Med Wkly. 2010;140:297–306.
    1. Wood JN, Boorman JP, Okuse K, Baker MD. Voltage-gated sodium channels and pain pathways. J Neurobiol. 2004;61:55–71. doi: 10.1002/neu.20094.
    1. Woolf CJ, Salter MW. Neuronal plasticity: increasing the gain in pain. Science (New York, NY) 2000;288:1765–1769. doi: 10.1126/science.288.5472.1765.
    1. Woolf CJ, Shortland P, Coggeshall RE. Peripheral nerve injury triggers central sprouting of myelinated afferents. Nature. 1992;355:75–78. doi: 10.1038/355075a0.
    1. World Health Organization (2001) International classification of functioning, disability and health: ICF. World Health Organization, Geneva. ICD 11. . Accessed 15 Aug 2019
    1. Wymer JP, Simpson J, Sen D, Bongardt S, Lacosamide SPSG. Efficacy and safety of lacosamide in diabetic neuropathic pain: an 18-week double-blind placebo-controlled trial of fixed-dose regimens. Clin J Pain. 2009;25:376–385. doi: 10.1097/AJP.0b013e318196d2b6.
    1. Xanthos DN, Sandkuhler J. Neurogenic neuroinflammation: inflammatory CNS reactions in response to neuronal activity. Nat Rev Neurosci. 2014;15:43–53. doi: 10.1038/nrn3617.
    1. Xiong Q, Lu B, Ye H, Wu X, Zhang T, Li Y. The diagnostic value of neuropathy symptom and change score, neuropathy impairment score and michigan neuropathy screening instrument for diabetic peripheral neuropathy. Eur Neurol. 2015;74:323–327. doi: 10.1159/000441449.
    1. Yagihashi S, Mizukami H, Sugimoto K. Mechanism of diabetic neuropathy: where are we now and where to go? J Diabetes Investig. 2011;2:18–32. doi: 10.1111/j.2040-1124.2010.00070.x.
    1. Yang XD, Fang PF, Xiang DX, Yang YY. Topical treatments for diabetic neuropathic pain. Exp Ther Med. 2019;17:1963–1976. doi: 10.3892/etm.2019.7173.
    1. Yarnitsky D, Granot M, Nahman-Averbuch H, Khamaisi M, Granovsky Y. Conditioned pain modulation predicts duloxetine efficacy in painful diabetic neuropathy. Pain. 2012;153:1193–1198. doi: 10.1016/j.pain.2012.02.021.
    1. Yuan RY, Sheu JJ, Yu JM, Chen WT, Tseng IJ, Chang HH, Hu CJ. Botulinum toxin for diabetic neuropathic pain: a randomized double-blind crossover trial. Neurology. 2009;72:1473–1478. doi: 10.1212/.
    1. Zamponi GW, Lewis RJ, Todorovic SM, Arneric SP, Snutch TP. Role of voltage-gated calcium channels in ascending pain pathways. Brain Res Rev. 2009;60:84–89. doi: 10.1016/j.brainresrev.2008.12.021.
    1. Zeilhofer HU, Wildner H, Yevenes GE. Fast synaptic inhibition in spinal sensory processing and pain control. Physiol Rev. 2012;92:193–235. doi: 10.1152/physrev.00043.2010.
    1. Zelman DC, Gore M, Dukes E, Tai KS, Brandenburg N. Validation of a modified version of the Brief Pain Inventory for painful diabetic peripheral neuropathy. J Vasc Nurs. 2005;23:97–104. doi: 10.1016/j.jvn.2005.06.004.
    1. Zhang J, Mense S, Treede RD, Hoheisel U. Prevention and reversal of latent sensitization of dorsal horn neurons by glial blockers in a model of low back pain in male rats. J Neurophysiol. 2017;118:2059–2069. doi: 10.1152/jn.00680.2016.
    1. Zhou M, Chen N, He L, Yang M, Zhu C, Wu F. Oxcarbazepine for neuropathic pain. Cochrane Database Syst Rev. 2017;12:CD007963. doi: 10.1002/14651858.CD007963.pub3.
    1. Zhuang ZY, Kawasaki Y, Tan PH, Wen YR, Huang J, Ji RR. Role of the CX3CR1/p38 MAPK pathway in spinal microglia for the development of neuropathic pain following nerve injury-induced cleavage of fractalkine. Brain Behav Immun. 2007;21:642–651. doi: 10.1016/j.bbi.2006.11.003.
    1. Ziegler D, et al. Oral treatment with alpha-lipoic acid improves symptomatic diabetic polyneuropathy: the SYDNEY 2 trial. Diabetes Care. 2006;29:2365–2370. doi: 10.2337/dc06-1216.
    1. Ziegler D, Movsesyan L, Mankovsky B, Gurieva I, Abylaiuly Z, Strokov I. Treatment of symptomatic polyneuropathy with actovegin in type 2 diabetic patients. Diabetes Care. 2009;32:1479–1484. doi: 10.2337/dc09-0545.
    1. Ziegler D, Keller J, Maier C, Pannek J, German Diabetes A. Diabetic neuropathy. Exp Clin Endocrinol Diabetes. 2014;122:406–415. doi: 10.1055/s-0034-1366435.
    1. Ziegler D, Landgraf R, Lobmann R, Reiners K, Rett K, Schnell O, Strom A. Painful and painless neuropathies are distinct and largely undiagnosed entities in subjects participating in an educational initiative (PROTECT study) Diabetes Res Clin Pract. 2018;139:147–154. doi: 10.1016/j.diabres.2018.02.043.
    1. Zin CS, Nissen LM, Smith MT, O'Callaghan JP, Moore BJ. An update on the pharmacological management of post-herpetic neuralgia and painful diabetic neuropathy. CNS Drugs. 2008;22:417–442. doi: 10.2165/00023210-200822050-00005.
    1. Zorina-Lichtenwalter K, Parisien M, Diatchenko L. Genetic studies of human neuropathic pain conditions: a review. Pain. 2018;159:583–594. doi: 10.1097/j.pain.0000000000001099.
    1. Zur E. Topical treatment of neuropathic pain using compounded medications. Clin J Pain. 2014;30:73–91. doi: 10.1097/AJP.0b013e318285d1ba.

Source: PubMed

3
Suscribir