The Itch-Scratch Cycle: A Review of the Mechanisms

The Itch-Scratch Cycle: A Review of the Mechanisms

Authors

  • Giulia Rinaldi St. George’s University & Hospital of London, UK

Keywords:

itch, prurigo, scratch, pruritus

Abstract

Background: Despite being one of the most common presenting dermatological symptoms, itching continues to perplex health care professionals because it is notoriously difficult to control.

Objective: This review gathers evidence to answer the 2-part question, “Why do we itch and scratch?” by exploring the history of itchy disease, the neurobiology of itch, and the 4 different clinical origins of itch: pruritogenic, neurological, neuropathic, and psychological.

Results: The automated scratching reflex and its biological and psychological reasons for existence are complicated and poorly understood. Currently, there are a myriad of treatments available for individuals suffering from this condition; however, many remain symptomatic.

Conclusions: The itch-scratch cycle is a complex pain-like sensation with a reflex-like response. In the future, continued exploration into the mechanisms behind itch and scratch may open the doors for new therapeutic interventions.

References

Ikoma A, Steinhoff M, Ständer S, Yosipovitch G, Schmelz M. The neurobiology of itch. Nat Rev Neurosci. 2006;7(7):535-547.

von Hospenthal T. Lessons for the NHS: Commissioning a Dermatology Service. Based on Case Studies from England. British Association of Dermatologists. 2013. Available from: https://www.bad.org.uk/shared/get-file.ashx?itemtype=document&id=1009. Accessed January 4, 2018.

Garibyan L, Rheingold CG, Lerner EA. Understanding the pathophysiology of itch. Dermatol Ther. 2013;26(2):84–91.

Nelson KE, Masters Williams CF. Infectious Disease Epidemiology: Theory and Practice. Boston: Jones and Bartlett Publishers; 2001:1-29.

Suetonius. The Twelve Caesars. Graves R, ed. Penguin Classics; 2007, 95 pp.

Lyell A. Daniel Turner (1667–1740) LRCP London (1711) M.D. honorary, Yale (1723) surgeon, physician and pioneer dermatologist. Int J Dermatol. 1982;21(3):162-170.

Hazen HH. Diseases of the Skin. St Louis: CV Mosby; 1915. Available from: https://archive.org/details/diseasesskin00hazegoog. Accessed January 4, 2018.

Kini SP, DeLong LK, Veledar E, McKenzie-Brown AM, Schaufele M, Chen SC. The impact of pruritus on quality of life: the skin equivalent of pain. Arch Dermatol. 2011;147(10):1153–1156.

Zachariae R, Lei U, Haedersdal M, Zachariae C. Itch severity and quality of life in patients with pruritus: preliminary validity of a Danish adaptation of the itch severity scale. Acta Derm Venereol. 2012;92(5):508–514.

Erturk IE, Arican O, Omurlu IK, Sut N. Effect of the pruritus on the quality of life: a preliminary study. Ann Dermatol. 2012;24(4):406–412.

Hong J, Buddenkotte J, Berger TG, Steinhoff M. Management of itch in atopic dermatitis. Semin Cutan Med Surg. 2011;30(2):71–86.

Yosipovitch G, Greaves MW, Schmelz M. Itch. Lancet. 2003;361(9358):690–694.

Bernhard JD. Itch and pruritus: what are they, and how should itches be classified? Dermatol Ther. 2005;18(4):288–291.

Broadbent JL. Observations on histamine-induced pruritus and pain. Br J Pharmacol Chemother. 1955;10(2):183–185.

Shim WS, Oh U. Histamine-induced itch and its relationship with pain. Mol Pain. 2008;4:29.

Quilliam JP. Substances producing pain and Itch. Proc R Soc Med. 1965;58(3):215–216.

Bulca S, Bayramgurler D, Odyakmaz Demirsoy E, et al. Comparison of effects of 5 and 10 mg oral desloratadine and levocetirizine on histamine-induced wheal and flare response in healthy volunteers. J Dermatolog Treat. 2013;24(6):473–476.

Klein PA, Clark RA. An evidence-based review of the efficacy of antihistamines in relieving pruritus in atopic dermatitis. Arch Dermatol. 1999;135(12):1522–1525.

Han L, Dong X. Itch mechanisms and circuits. Annu Rev Biophys. 2014;43:331–355.

Murata Y, Song M, Kikuchi H, et al. Phase 2a, randomized, double-blind, placebo-controlled, multicenter, parallel-group study of a H4 R-antagonist (JNJ-39758979) in Japanese adults with moderate atopic dermatitis. J Dermatol. 2015;42(2):129–139.

Grundmann S, Ständer S. Chronic pruritus: clinics and treatment. Ann Dermatol. 2011;23(1):1–11.

Imaizumi A, Kawakami T, Murakami F, Soma Y, Mizoguchi M. Effective treatment of pruritus in atopic dermatitis using H1 antihistamines (second-generation antihistamines): changes in blood histamine and tryptase levels. J Dermatol Sci. 2003;33(1):23–29.

Steinhoff M, Neisius U, Ikoma A, et al. Proteinase-activated receptor-2 mediates itch: a novel pathway for pruritus in human skin. J Neurosci. 2003;23(15):6176–6180.

Sikand P, Shimada SG, Green BG, LaMotte RH. Similar itch and nociceptive sensations evoked by punctate cutaneous application of capsaicin, histamine and cowhage. Pain. 2009;144(1–2):66–75.

Andoh T, Kuraishi Y. Antipruritic mechanisms of topical E6005, a phosphodiesterase 4 inhibitor: inhibition of responses to proteinase-activated receptor 2 stimulation mediated by increase in intracellular cyclic AMP. J Dermatol Sci. 2014;76(3):206–213.

Mishra SK, Hoon MA. The cells and circuitry for itch responses in mice. Science. 2013;340(6135):968–971.

Ellis CN, Berberian B, Sulica VI, et al. A double-blind evaluation of topical capsaicin in pruritic psoriasis. J Am Acad Dermatol. 1993;29(3):438–442.

Simonsen E, Komenda P, Lerner B, et al. Treatment of uremic pruritus: a systematic review. Am J Kidney Dis. 2017;70(5):638–655.

Breneman DL, Cardone JS, Blumsack RF, Lather RM, Searle EA, Pollack VE. Topical capsaicin for treatment of hemodialysis-related pruritus. J Am Acad Dermatol. 1992;26(1):91–94.

Pereira MP, Luling H, Dieckhofer A, Steinke S, Zeidler C, Ständer S. Brachioradial pruritus and notalgia paraesthetica: a comparative observational study of clinical presentation and morphological pathologies. Acta Derm Venereol. 2018;98(1):82–88.

Kido-Nakahara M, Buddenkotte J, Kempkes C, et al. Neural peptidase endothelin-converting enzyme 1 regulates endothelin 1-induced pruritus. J Clin Invest. 2014;124(6):2683–2695.

Paus R, Schmelz M, Bíró T, Steinhoff M. Frontiers in pruritus research: scratching the brain for more effective itch therapy. J Clin Invest. 2006;116(5):1174–1186.

Arai I, Tsuji M, Takeda H, Akiyama N, Saito S. A single dose of interleukin-31 (IL-31) causes continuous itch-associated scratching behaviour in mice. Exp Dermatol. 2013;22(10):669–671.

Furue M, Yamamura K, Kido-Nakahara M, Nakahara T, Fukui Y. Emerging role of interleukin-31 and interleukin-31 receptor in pruritus in atopic dermatitis. Allergy. 2018;73(1):29–36.

Jang H, Matsuda A, Jung K, et al. Skin pH is the master switch of kallikrein 5-mediated skin barrier destruction in a murine atopic dermatitis model. J Invest Dermatol. 2016;136(1):127-135.

Furio L, de Veer S, Jaillet M, et al. Transgenic kallikrein 5 mice reproduce major cutaneous and systemic hallmarks of Netherton syndrome. J Exp Med. 2014;211(3):499–513.

Carstens EE, Carstens MI, Simons CT, Jinks SL. Dorsal horn neurons expressing NK-1 receptors mediate scratching in rats. Neuroreport. 2010;21(4):303–308.

Toyoda M, Nakamura M, Makino T, Hino T, Kagoura M, Morohashi M. Nerve growth factor and substance P are useful plasma markers of disease activity in atopic dermatitis. Br J Dermatol. 2002;147(1):71–9.

Zhang Z, Zheng W, Xie H, et al. Up-regulated expression of substance P in CD8+ T cells and NK1R on monocytes of atopic dermatitis. J Transl Med. 2017;15(1):93.

Lee MR, Shumack S. Prurigo nodularis: a review. Australas J Dermatol. 2005;46(4):211–220.

Liu XY, Liu ZC, Sun YG, et al. Unidirectional cross-activation of GRPR by MOR1D uncouples itch and analgesia induced by opioids. Cell. 2011;147(2):447–458.

Tefferi A, Fonseca R. Selective serotonin reuptake inhibitors are effective in the treatment of polycythemia vera-associated pruritus. Blood. 2002;99(1):2627.

Jinks SL, Carstens E. Responses of superficial dorsal horn neurons to intradermal serotonin and other irritants: comparison with scratching behavior. J Neurophysiol. 2002;87(3):1280–1289.

Rausl A, Nordlind K, Wahlgren CF. Pruritic and vascular responses induced by serotonin in patients with atopic dermatitis and in healthy controls. Acta Derm Venereol. 2013;93(3):277–280.

Stander S, Bockenholt B, Schurmeyer-Horst F, et al. Treatment of chronic pruritus with the selective serotonin re-uptake inhibitors paroxetine and fluvoxamine: results of an open-labelled, two-arm proof-of-concept study. Acta Derm Venereol. 2009;89(1):45–51.

Schworer H, Hartmann H, Ramadori G. Relief of cholestatic pruritus by a novel class of drugs: 5-hydroxytryptamine type 3 (5-HT3) receptor antagonists: effectiveness of ondansetron. Pain. 1995;61(1):33–37.

Kremer AE, Feramisco J, Reeh PW, Beuers U, Oude Elferink RPJ. Receptors, cells and circuits involved in pruritus of systemic disorders. Biochim Biophys Acta. 2014;1842(7):869–892.

Gupta MA, Guptat AK. The use of antidepressant drugs in dermatology. J Eur Acad Dermatol Venereol. 2001;15(6):512–518.

Kremer AE, Martens JJWW, Kulik W, et al. Lysophosphatidic acid is a potential mediator of cholestatic pruritus. Gastroenterology. 2010;139(3):1008–1018, 1018.e1.

Kremer AE, Gonzales E, Schaap FG, Oude Elferink RPJ, Jacquemin E, Beuers U. Serum autotaxin activity correlates with pruritus in pediatric cholestatic disorders. J Pediatr Gastroenterol Nutr. 2016;62(4):530–535.

Kremer AE, van Dijk R, Leckie P, et al. Serum autotaxin is increased in pruritus of cholestasis, but not of other origin, and responds to therapeutic interventions. Hepatology. 2012;56(4):1391–1400.

Thornton JR, Losowsky MS. Opioid peptides and primary biliary cirrhosis. BMJ. 1988;297(6662):1501–1504.

Bergasa NV, Talbot TL, Alling DW, et al. A controlled trial of naloxone infusions for the pruritus of chronic cholestasis. Gastroenterology. 1992;102(2):544–549.

Joshi GG, Thakur BS, Sircar S, Namdeo A, Jain AK. Role of intravenous naloxone in severe pruritus of acute cholestasis. Indian J Gastroenterol. 2009;28(5):180–182.

Mansour-Ghanaei F, Taheri A, Froutan H, et al. Effect of oral naltrexone on pruritus in cholestatic patients. World J Gastroenterol. 2006;12(7):1125–1128.

Combs SA, Teixeira JP, Germain MJ. Pruritus in kidney disease. Semin Nephrol. 2015;35(4):383–391.

Kumagai H, Ebata T, Takamori K, et al. Efficacy and safety of a novel κ-agonist for managing intractable pruritus in dialysis patients. Am J Nephrol. 2012;36(2):175–183.

Solak Y, Biyik Z, Atalay H, et al. Pregabalin versus gabapentin in the treatment of neuropathic pruritus in maintenance haemodialysis patients: a prospective, crossover study. Nephrology (Carlton). 2012;17(8):710–717.

Lau T, Leung S, Lau W. Gabapentin for uremic pruritus in hemodialysis patients: a qualitative systematic review. Can J Kidney Health Dis. 2016;3:14.

Stumpf A, Ständer S. Neuropathic itch: diagnosis and management. Dermatol Ther. 2013;26(2):104–109.

Maciel AAW, Cunha PR, Laraia IO, Trevisan F. Efficacy of gabapentin in the improvement of pruritus and quality of life of patients with notalgia paresthetica. An Bras Dermatol. 2014;89(4):570–575.

PD Y, NE W. Efficacy of gabapentin in the management of pruritus of unknown origin. Arch Dermatol. 2005;141(12):1507–1509.

Oaklander AL. Neuropathic itch. Semin Cutan Med Surg. 2011;30(2):87–92.

Suárez AL, Feramisco JD, Koo J, Steinhoff M. Psychoneuroimmunology of psychological stress and atopic dermatitis: pathophysiologic and therapeutic updates. Acta Derm Venereol. 2012;92(1):7–15.

Schut C, Bosbach S, Gieler U, Kupfer J. Personality traits, depression and itch in patients with atopic dermatitis in an experimental setting: a regression analysis. Acta Derm Venereol. 2014;94(1):20–25.

Hsu DT, Sanford BJ, Meyers KK, et al. Response of the μ-opioid system to social rejection and acceptance. Mol Psychiatry. 2013;18(11):1211–1217.

Lutz P-E, Kieffer BL. Opioid receptors: distinct roles in mood disorders. Trends Neurosci. 2013;36(3):195–206.

Ehrich E, Turncliff R, Du Y, et al. Evaluation of opioid modulation in major depressive disorder. Neuropsychopharmacology. 2015;40(6):1448–1455.

Geracioti TDJ, Carpenter LL, Owens MJ, et al. Elevated cerebrospinal fluid substance p concentrations in posttraumatic stress disorder and major depression. Am J Psychiatry. 2006;163(4):637–643.

Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. J Clin Invest. 2010;120(11):3760–3772.

Lawson SN. Phenotype and function of somatic primary afferent nociceptive neurones with C-, Aδ- or Aα/β-fibres. Exp Physiol. 2002;87(2):239–244.

Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellular and molecular mechanisms of pain. Cell. 2009;139(2):267–284.

Ward L, Wright E, McMahon SB. A comparison of the effects of noxious and innocuous counterstimuli on experimentally induced itch and pain. Pain. 1996;64(1):129–138.

Tuckett RP. Itch evoked by electrical stimulation of the skin. J Invest Dermatol. 1982;79(6):368–373.

Handwerker HO, Forster C, Kirchhoff C. Discharge patterns of human C-fibers induced by itching and burning stimuli. J Neurophysiol. 1991;66(1):307–315.

Ochoa J, Torebjork E. Sensations evoked by intraneural microstimulation of C nociceptor fibres in human skin nerves. J Physiol. 1989;415:583–599.

Miyamoto T, Nojima H, Shinkado T, Nakahashi T, Kuraishi Y. Itch-associated response induced by experimental dry skin in mice. Jpn J Pharmacol. 2002;88(3):285–292.

Yu YQ, Barry DM, Hao Y, Liu XT, Chen ZF. Molecular and neural basis of contagious itch behavior in mice. Science. 2017;355(6329):1072–1076.

Wilson SR, Nelson AM, Batia L, et al. The ion channel TRPA1 is required for chronic itch. J Neurosci. 2013;33(22):9283–9294.

Spradley JM, Davoodi A, Carstens MI, Carstens E. Opioid modulation of facial itch- and pain-related responses and grooming behavior in rats. Acta Derm Venereol. 2012;92(5):515–520.

Matesic LE, Copeland NG, Jenkins NA. Itchy mice: the identification of a new pathway for the development of autoimmunity. Curr Top Microbiol Immunol. 2008;321:185–200.

Shimada SG, LaMotte RH. Behavioral differentiation between itch and pain in mouse. Pain. 2008;139(3):681–387.

Kuraishi Y, Nagasawa T, Hayashi K, Satoh M. Scratching behavior induced by pruritogenic but not algesiogenic agents in mice. Eur J Pharmacol. 1995;275(3):229–233.

Andrew D, Craig AD. Spinothalamic lamina I neurons selectively sensitive to histamine: a central neural pathway for itch. Nat Neurosci. 2001;4(1):72–77.

Sun Y-G, Chen Z-F. A gastrin-releasing peptide receptor mediates the itch sensation in the spinal cord. Nature. 2007;448(7154):700–703.

Han L, Ma C, Liu Q, et al. A subpopulation of nociceptors specifically linked to itch. Nat Neurosci. 2013;16(2):174–182.

Sun YG, Zhao ZQ, Meng XL, Yin J, Liu XY, Chen ZF. Cellular basis of itch sensation. Science. 2009;325(5947):1531–1534.

Schmelz M, Schmidt R, Bickel A, Handwerker HO, Torebjork HE. Specific C-receptors for itch in human skin. J Neurosci. 1997;17(20):8003–8008.

Namer B, Carr R, Johanek LM, Schmelz M, Handwerker HO, Ringkamp M. Separate peripheral pathways for pruritus in man. J Neurophysiol. 2008;100(4):2062–2069.

Johanek LM, Meyer RA, Hartke T, et al. Psychophysical and physiological evidence for parallel afferent pathways mediating the sensation of itch. J Neurosci. 2007;27(28):7490–7497.

Davidson S, Zhang X, Khasabov SG, et al. Pruriceptive spinothalamic tract neurons: physiological properties and projection targets in the primate. J Neurophysiol. 2012;108(6):1711–1723.

Davidson S, Zhang X, Yoon CH, Khasabov SG, Simone DA, Giesler GJJ. The itch-producing agents histamine and cowhage activate separate populations of primate spinothalamic tract neurons. J Neurosci. 2007;27(37):10007–10014.

Mochizuki H, Sadato N, Saito DN, et al. Neural correlates of perceptual difference between itching and pain: a human fMRI study. Neuroimage. 2007;36(3):706–717.

Papoiu ADP, Coghill RC, Kraft RA, Wang H, Yosipovitch G. A tale of two itches: common features and notable differences in brain activation evoked by cowhage and histamine induced itch. Neuroimage. 2012;59(4):3611–3623.

LaMotte RH, Dong X, Ringkamp M. Sensory neurons and circuits mediating itch. Nat Rev Neurosci. 2014;15(1):19–31.

Ji RR. Neuroimmune interactions in itch: do chronic itch, chronic pain, and chronic cough share similar mechanisms? Pulm Pharmacol Ther. 2015;35(Supplement C):81–86.

Mei J. The mechanisms and perception of itch. Yale Scientific. 2011. Available from: http://www.yalescientific.org/2011/05/the-mechanisms-and-perception-of-itch/. Accessed January 4, 2018.

Alexander JO. The physiology of itch. Parasitol Today. 1986;2(12):345–351.

Zhao ZQ, Liu XY, Jeffry J, et al. Descending control of itch transmission by the serotonergic system via 5-HT1A-facilitated GRP-GRPR signaling. Neuron. 2014;84(4):821–834.

Murota H, Katayama I. Exacerbating factors of itch in atopic dermatitis. Allergol Int. 2017;66(1):8–13.

Leknes SG, Bantick S, Willis CM, Wilkinson JD, Wise RG, Tracey I. Itch and motivation to scratch: an investigation of the central and peripheral correlates of allergen- and histamine-induced itch in humans. J Neurophysiol. 2007;97(1):415–422.

Patel T, Ishiuji Y, Yosipovitch G. Nocturnal itch: why do we itch at night? Acta Derm Venereol. 2007;87(4):295–298.

Lavery MJ, Stull C, Kinney MO, Yosipovitch G. Nocturnal pruritus: the battle for a peaceful night's sleep. Int J Mol Sci. 2016;17(3):425.

Strine TW, Chapman DP. Associations of frequent sleep insufficiency with health-related quality of life and health behaviors. Sleep Med. 2005;6(1):23–27.

Nocturnal pruritus: Prevalence, characteristics, and impact on ItchyQoL in a chronic itch population. J Am Acad Dermatol. 2017;76(6):AB179.

Warlich B, Fritz F, Osada N, et al. Health-related quality of life in chronic pruritus: an analysis related to disease etiology, clinical skin conditions and itch intensity. Dermatology. 2015;231(3):253–259.

Phan NQ, Blome C, Fritz F, et al. Assessment of pruritus intensity: prospective study on validity and reliability of the visual analogue scale, numerical rating scale and verbal rating scale in 471 patients with chronic pruritus. Acta Derm Venereol. 2012;92(5):502–507.

Millington GWM, Collins A, Lovell CR, et al. British Association of Dermatologists' guidelines for the investigation and management of generalized pruritus in adults without an underlying dermatosis, 2018. Br J Dermatol. 2018;178(1):34–60.

JE N. Hemochromatosis and pruritus. Ann Intern Med. 1983;98(6):1026.

Valsecchi R, Cainelli T. Generalized pruritus: a manifestation of iron deficiency. Arch Dermatol. 1983;119(8):630.

Le Gall-Ianotto C, Misery L. Pruritus in hematological diseases (including aquagenic pruritus). In: Misery L, Ständer S, eds. Pruritus. Cham, Switzerland: Springer; 2016:271–281.

Levy C. Management of pruritus in patients with cholestatic liver disease. Gastroenterol Hepatol (NY). 2011;7(9):615–617.

Greenberger PA. Chronic urticaria: new management options. World Allergy Organ J. 2014;7(1):31.

Zhang P, Wu MX. A clinical review of phototherapy for psoriasis. Lasers Med Sci. 2017/10/24. 2018;33(1):173–180.

Song J, Xian D, Yang L, Xiong X, Lai R, Zhong J. Pruritus: progress toward pathogenesis and treatment. Biomed Res Int. 2018;2018:9625936.

Gangemi S, Quartuccio S, Casciaro M, Trapani G, Minciullo PL, Imbalzano E. Interleukin 31 and skin diseases: a systematic review. Allergy Asthma Proc. 2017;38(6):401–408.

Ruzicka T, Hanifin JM, Furue M, et al. Anti–interleukin-31 receptor A antibody for atopic dermatitis. N Engl J Med. 2017;376(9):826–835.

Nemoto O, Furue M, Nakagawa H, Shiramoto M, Hanada R, Matsuki S, et al. The first trial of CIM331, a humanized antihuman interleukin-31 receptor A antibody, in healthy volunteers and patients with atopic dermatitis to evaluate safety, tolerability and pharmacokinetics of a single dose in a randomized, double-blind, placebo-controlled study. Br J Dermatol. 2016;174(2):296–304.

Yosipovitch G, Ständer S, Kerby MB, et al. Serlopitant for the treatment of chronic pruritus: results of a randomized, multicenter, placebo-controlled phase 2 clinical trial. J Am Acad Dermatol. 2018;78(5):882–891.e10.

Heitman A, Xiao C, Cho Y, Polymeropoulos C, Birznieks G, Polymeropoulos M. Tradipitant improves worst itch and disease severity in patients with chronic pruritus related to atopic dermatitis. J Am Acad Dermatol. 2018;79(3):AB300.

Beck LA, Thaci D, Hamilton JD, et al. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. N Engl J Med. 2014;371(2):130–139.

Odhiambo JA, Williams HC, Clayton TO, Robertson CF, Asher MI; ISAAC Phase III Study Group. Global variations in prevalence of eczema symptoms in children from ISAAC Phase Three. J Allergy Clin Immunol. 2009;124(6):1251–8.e23.

Dalgard FJ, Gieler U, Tomas-Aragones L, et al. The psychological burden of skin diseases: a cross-sectional multicenter study among dermatological out-patients in 13 European countries. J Invest Dermatol. 2015;135(4):984–991.

Downloads

Published

2019-04-30

Issue

Section

Review

How to Cite

1.
The Itch-Scratch Cycle: A Review of the Mechanisms. Dermatol Pract Concept [Internet]. 2019 Apr. 30 [cited 2024 Apr. 19];9(2):90-7. Available from: https://dpcj.org/index.php/dpc/article/view/dermatol-pract-concept-articleid-dp0902a03

Share