Biological and Pharmaceutical Bulletin
Online ISSN : 1347-5215
Print ISSN : 0918-6158
ISSN-L : 0918-6158
Current Topics
Potential New Therapeutic Targets for Pathological Pruritus
Yasushi Kuraishi
Author information
JOURNAL FREE ACCESS FULL-TEXT HTML

2013 Volume 36 Issue 8 Pages 1228-1234

Details
Abstract

Very few approved medications are indicated for the treatment of pruritus, and drug development for pruritic diseases is awaited. During the past two decades, progress has been made in understanding the molecular basis of the physiology and pathophysiology of pruritus. Newly identified potential targets for pathological pruritus include receptors (histamine H4 receptor, leukotriene B4 receptors, interleukin-31 receptor A, bombesin BB2 receptor, toll-like receptor 3, α-adrenoceptor, and opioid μ- and κ-receptors), channels (transient receptor potential (TRP) V3 and TRPA1 channels), and enzymes (histidine decarboxylase, sphingomyelin glucosylceramide deacylase, 5-lipoxygenase, leukotriene A4 hydrolase, and autotaxin). The development of specific, effective blockers and agonists/antagonists of these targets is awaited.

1. INTRODUCTION

Itching is a sensation that provokes a desire to scratch or rub. Itching arises from the superficial layers of the skin, the conjunctiva, and the mucous membranes adjoining the skin. It is a biowarning sensation, the biological purpose of which is to elicit scratching and to remove irritating objects (e.g., parasites, irritants, allergens, and small sharp objects) from these regions. Itching signals are transmitted from the periphery to the brain via the dorsal horn by primary sensory neurons and spinothalamic tract neurons. The suppression of both itching and scratching is important in the treatment of pruritic diseases, because unrelieved itching has a negative impact on quality of life and vigorous scratching worsens the conditions of itchy regions. Since scratching is induced by an itching sensation and by a spinal reflex (scratch reflex), the inhibition of the production of itch signals in the periphery is an ideal pharmacologic intervention in the treatment of pruriceptive itching. However, it may be difficult to ameliorate itching of all pruritic diseases through the peripheral action because of the presence of manifold itch mediators. Thus, the inhibition of the transmission of itch signals in the dorsal horn is also an important site of action of antipruritic drugs. Here I review the molecular targets that have been shown to be associated with pathological pruritus (Table 1).

Table 1. Potential New Therapeutic Targets for Pathological Pruritus
Mediator/stimulantEnzymeReceptor/channel
Periphery
Histamine (keratinocyte)Histidine decarboxylase (keratinocyte)H1 receptor (primary afferent)
Histamine (mast cell)Histidine decarboxylase (mast cell)H4 receptor (T lymphocyte, other cells)
Proteinase (mast cell, other cells)PAR2 receptor (primary afferent, keratinocyte)
Sphingosylphosphorylcholine (keratinocyte)Sphingomyelin glucosylceramide deacylase (keratinocyte)Not identified (primary afferent, keratinocyte)
LTB4 (keratinocyte)5-Lipoxygenase and LTA4 hydrolase (keratinocyte)BLT1 receptor (primary afferent)
LTB4 (mast cell)5-Lipoxygenase and LTA4 hydrolase (mast cell)BLT2 receptor (T lymphocyte)
Lysophosphatidic acid (blood)Autotaxin (liver?)Not determined (primary afferent?)
Interleukin-31 (T lymphocyte)Interleukin-31 receptor A and oncostatin M receptor (primary afferent, keratinocyte)
Warm temperature?TRPV3 (primary afferent, keratinocyte)
OxidantTRPA1 (primary afferent)
Dorsal horn
GRP (sensory neuron, dorsal horn neuron)BB2 receptor (dorsal horn neuron)
Toll-like receptor 3 (sensory neuron)
Noradrenaline (descending neuron)α1- and α2-receptor (sensory neurons, dorsal horn neuron)

LTA4, leukotriene A4; LTB4, leukotriene B4; GRP, gastrin-releasing peptide.

2. TARGETS IN THE PERIPHERY

2.1. Histamine and Histidine Decarboxylase

Histamine is a major itch mediator associated with mast cell-mediated acute responses in the skin and conjunctiva. Histamine H1 receptor antagonists and mast cell stabilizers are usually used to treat itching in immediate allergy.1) In animal experiments, H1 receptor antagonists exert a significant, although partial, inhibition of the itch-related response, hind-paw scratching, to passive cutaneous anaphylaxis and immediate pollen allergy in the conjunctiva.2,3)

Single topical application of sodium laurate, an alkaline surfactant, induces delayed scratching in mice, although topical application of a neutral surfactant or sodium hydroxide solution does not induce delayed scratching.4) Sodium laurate-induced delayed scratching is almost completely inhibited by a histamine H1 receptor antagonist, but it is not affected by deficiency in mast cells.4) In the normal skin, histamine is mainly present in mast cells in the dermis and a trace of histamine is present in the epidermis. Topical application of sodium laurate increases histamine production in the epidermal keratinocytes, which may be due to enhanced processing of 74-kDa to 53-kDa histidine decarboxylase, the latter of which is a key enzyme for histamine production, in the epidermal keratinocytes.4) Since there are no histamine-containing granules in the keratinocytes and histamine is spontaneously released without being stored, histidine decarboxylase is also a potential target for the prevention of alkaline surfactant-induced delayed itching. Consistent with this view, sodium laurate-induced delayed scratching is inhibited by the topical application of agents that suppress the increase in 53-kDa histidine decarboxylase in the keratinocytes.5)

Binding affinities of histamine H3 and H4 receptors for histamine are much higher than those of histamine H1 and H2 receptors.6) Intradermal injections of H3 receptor agonists increase scratching behaviors in mice,7) but it remains unknown whether the H3 receptor is involved in pathological itching. Systemic administration of a histamine H4 receptor antagonist reduces scratching induced by allergic dermatitis and passive cutaneous anaphylaxis in mice.810) Intradermal injection of an H4 receptor agonist elicits scratching, which is markedly inhibited by an H4 receptor antagonist and H4 receptor deficiency.9) Histamine-independent scratching is also inhibited by an H4 receptor antagonist.11) Thus, the histamine H4 receptor in the periphery and/or central nervous system is a potential target for pruritic diseases.

2.2. Proteinases and Proteinase-Activated Receptor 2 (PAR2)

Proteinases (endopeptidases), but not exopeptidases, have long been known to cause itching when injected intradermally in humans.12) Pruritogenic mechanisms of proteinases include the activation of the PAR2,13,14) the degranulation of mast cells,1517) and the production of pruritogenic peptides.18,19) The proteinase receptor PAR2 may be involved in itching and scratching in atopic dermatitis,20,21) urticaria,13,22) dermatophyte infection,12) and contact with cowhage seed pod.23) Some mast cells in the human skin express PAR2, the stimulation of which results in histamine release.24) However, histamine does not play an important role in proteinase receptor PAR2-mediated scratching in mice,16) and separate sensory neurons may play a role in histamine- and proteinase receptor-mediated itching.2528) The responses of dorsal horn neurons to intradermal injection of a PAR2 agonist, but not histamine, are enhanced under dry skin conditions.28) Tryptase, a PAR2 agonist proteinase, increases in mast cells in the lesional skin of chronic dermatitis.20,21) Thus, the proteinase receptor PAR2 is a potential therapeutic target for pruritic diseases, including H1 antagonist-resistant dermatitis. Proteinases are involved in itching in several pruritic diseases. However, it may be difficult for specific proteinase inhibitors to inhibit itching in a broad spectrum of pruritic diseases because there are many types of pruritogenic proteinases.

2.3. Lipid Mediators

Stratum corneum lipids, especially ceramides, play a pivotal role in maintaining the water barrier of the skin. Ceramides are produced from sphingomyelin and glucocerebroside by sphingomyelinase and glucorebrosidase, respectively, in the stratum corneum and the superficial layers of the epidermis. In patients with atopic dermatitis, the amount of ceramide in the stratum corneum decreases, and there is an inverse correlation between ceramide and sphingosylphosphorylcholine (SPC) levels in the lesional stratum corneum.29) In the epidermis of patients with atopic dermatitis, the activity of sphingomyelin glucosylceramide deacylase is increased, and sphingomyelin and glucocerebroside are converted to SPC and glucosylsphingosine, respectively, by this enzyme, which results in the decrease in ceramide, leading to skin dryness and the disruption of the skin barrier.30) SPC downregulates filaggrin gene transcription, which also leads to skin dryness and the disruption of the skin barrier.31) SPC is also increased in the lesional epidermis of mice with chronic allergic dermatitis.32) Intradermal injection of SPC, but not of sphingomyelin and sphingosine, elicits scratching in mice.3234) SPC may act on the epidermal keratinocytes, dendritic cells, primary sensory neurons, and mast cells in the skin.3436) Naturally occurring D-erythro SPC, but not L-threo SPC, induces scratching after intradermal injection, raising the possibility that SPC acts on a specific receptor to induce itching.33) However, SPC receptors remain unclear, although some candidates were reported. It is expected that the suppression of the increased activity of sphingomyelin glucosylceramide deacylase results in not only the decrease in SPC (relieving itch) but also the increase in ceramides in the stratum corneum and epidermis (improving skin dryness and barrier disruption) in patients with atopic dermatitis. Therefore, sphingomyelin glucosylceramide deacylase is a more attractive therapeutic target for SPC-mediated itching in atopic dermatitis.

SPC acts on keratinocytes to produce leukotriene (LT) B4, which is involved in SPC-induced scratching.34) LTB4 elicits hind-paw scratching at relatively low doses after administration to the skin and conjunctiva in mice.3,37) LTB4 was shown to be involved in scratching in mice with chronic allergic dermatitis and allergic conjunctivitis,3,32,38) but not in those with acute cutaneous allergy.39) There are two LTB4 receptor subtypes, BLT1 and BLT2, which have high and low binding affinities for LTB4, respectively.40) The BLT1 receptor is expressed mainly in leukocytes, and the BLT2 receptor is expressed more ubiquitously.40) Dorsal root ganglion neurons, mainly transient receptor potential (TRP) V1-positive small neurons, express the BLT1 receptor, and LTB4 administration increases intracellular Ca2+ ions in cultured dorsal ganglion neurons.41) When LTB4 production is localized in the epidermis (e.g., dermatophyte infection), BLT1 receptors on primary afferents may be involved in LTB4-mediated itching. LTB4 synthesis is catalyzed by 5-lipoxygenase and LTA4 hydrolase. Scratching induced by passive cutaneous anaphylaxis is suppressed by an LTA4 hydrolase inhibitor, suggesting that mast cell-derived LTB4 is involved in itching.42) When LTB4 is produced in the dermis (e.g., mast cells), it may act as a leukocyte chemoattractant via BLT2 receptor stimulation, and leukocyte-released superoxide may induce itching.43) Nonselective LTB4 antagonists are more desirable than the selective BLT1 or BLT2 antagonists because both BLT1 and BLT2 receptors can be involved in LTB4-mediated itching. A 5-lipoxygenase inhibitor relieves pruritus in Sjögren–Larsson syndrome44) and has a tendency to improve pruritus in atopic dermatitis.45) In animal experiments, a 5-lipoxygenase inhibitor inhibits scratching in mice with acute ocular or cutaneous allergy and chronic allergic dermatitis.3,38,39) Thus, 5-lipoxygenase and LTA4 hydrolase are also potential therapeutic targets for pruritus in allergic inflammation.

Intradermal injection of 12(S)-hydroperoxyeicosatetraenoic acid, a 12-lipoxygenase metabolite, induces scratching with a potency similar to that of LTB4.46) This action has been shown to be mediated by the BLT2 receptor.47)

Lysophosphatidic acid is a phospholipid mediator with many biological functions and disease implications. Lysophosphatidic acid is produced both in cells and biological fluids; it is predominantly produced by autotaxin (lysophospholipase D), a plasma enzyme in the blood.48) The serum levels of autotaxin activity and autotaxin protein are enhanced, and there is a significant correlation between the intensity of itch perception and serum autotaxin activity in patients with pruritic cholestasis.49) Biliary drainage treatment relieves cholestatic pruritus and serum autotaxin activity.49) The serum levels of histamine, tryptase, substance P, μ-opioid activity, and bile salt are not correlated with the intensity of itch perception.49) Intradermal injection of lysophosphatidic acid induces scratching in mice.49,50) Autotaxin activity in the serum is not increased in patients with other pruritic diseases, such as uremia, Hodgkin’s disease, or atopic dermatitis.51) These findings raise the possibility that lysophosphatidic acid and autotaxin are potential therapeutic targets for cholestatic pruritus.

2.4. Cytokines

Interleukin (IL)-31 is preferentially produced by activated CD4+ T cells. IL-31 transgenic mice show severe scratching and skin lesions.52) Subcutaneous infusion of IL-31 protein using a mini-osmotic pump also increases scratching.52) IL-31 mRNA increases in the skin of patients with atopic dermatitis53,54) and mice with chronic allergic dermatitis.55) Repeated intraperitoneal injections of anti-IL-31 monoclonal antibody decrease scratching behavior in mice with chronic allergic dermatitis.56) These findings taken together suggest that IL-31 is involved in promoting pruritus in chronic allergic dermatitis, especially atopic dermatitis. IL-31 signaling occurs through a functional complex of IL-31 receptor A with the oncostatin M receptor. Although oncostatin M receptor expression is ubiquitous, IL-31 receptor A has much higher expression in the dorsal root ganglia compared with other tissues.53) Oncostatin M receptor is expressed mainly in the nonpeptidergic unmyelinated sensory neurons,57) and IL-31 receptor A is expressed in the small-sized neurons expressing oncostatin M receptor.58) Therefore, it is possible that IL-31 acts on pruriceptive primary afferents as an itch mediator. However, since the stimulation of toll-like receptor 2 upregulates IL-31 receptor A with oncostatin M receptor in cultured human keratinocytes,59) epidermal keratinocytes may also play a role in IL-31-mediated itching in atopic dermatitis. Thus, IL-31 and IL-31 receptor A may be potential therapeutic targets for pruritus in atopic dermatitis.

2.5. Channels

Primary sensory neurons expressing TRPV1 channels play an important role in itch signaling.60) TRPV1 channels are indispensable to current generation following the stimulation of itch-related G-protein-coupled receptors. Histamine stimulation of H1 receptor produces TRPV1 currents through the phospholipase A2–12-lipoxygenase and phospholipase Cβ signaling systems.60,61) Similarly, proteinase stimulation of the PAR2 receptor produces TRPV1 currents through a phospholipase C–protein kinase C pathway.62) Therefore, TRPV1 channel blockers may relieve itching in a broad spectrum of pruritic diseases. However, the TRPV1 channel may not be an adequate therapeutic target for pruritic diseases because it plays a key role in sensing heat pain, and systemic administration of TRPV1 channel blockers may suppress the withdrawal reflex induced by noxious heat stimulation.

The TRPV3 channel is activated at innocuous warm temperature and is expressed in primary sensory neurons and keratinocytes.63,64) A gain-of-function mutation in the TRPV3 channel causes spontaneous scratching and dermatitis in mice.65) In contrast, a deficiency of the TRPV3 channel suppresses spontaneous scratching in mice with skin dryness, without affecting cutaneous barrier disruption and a decrease in stratum corneum hydration.66) Thus, the TRPV3 channel may be a potential therapeutic target for pruritus of xeroderma.

TRPA1 channels are expressed in a subset of TRPV1-positive sensory neurons and are involved in cold sensing.67,68) The TRPA1 channel has been shown to be involved in itch signaling. For example, chloroquine (an antimalaria drug) and bovine adrenal medulla peptide 8–22 (a pruritogenic opioid peptide) act on Mas-related G protein-coupled receptor (Mrgpr) A3 and MrgprC11, respectively, on primary sensory neurons to induce scratching in mice, and TRPA1 is required for the Mrgpr-mediated signaling.69) TRPA1 currents are sensitized by PAR2 receptor stimulation.70) TRPA1 has been shown to be involved in oxidant-induced scratching.71) Although the TRPA1 channel seems to be an interesting target for pruritus, it remains unknown whether it is involved in pathological pruritus.

3. TARGETS IN THE CENTRAL NERVOUS SYSTEM

3.1. Itch Transmitters

Neuromedin B and gastrin-releasing peptide (GRP), mammalian homologues of amphibian bombesin, have high binding affinities for bombesin BB1 and BB2 receptors, respectively. GRP is expressed in a small subset of peptidergic primary afferent neurons and a subset of neurons in the superficial dorsal horn.72,73) Intrathecal administration of a bombesin-saporin conjugate produces the selective ablation of BB2 receptor-expressing spinal neurons, without effect on BB1 receptor-expressing spinal neurons.74) This treatment markedly reduces scratching responses to intradermal injections of many pruritogenic substances such as histamine, compound 48/80, serotonin, endothelin-1, PAR2 agonist, and chloroquine.75) BB2 receptor mutation and intrathecal injection of a BB2 receptor antagonist reduce scratching responses of mice to intradermal injections of compound 48/80, PAR2 agonist, and chloroquine, whereas these manipulations do not affect pain responses.73) Serum levels of GRP correlate with the itching score in patients with atopic dermatitis.76) GRP-positive nerve fibers are increased in the skin of patients with atopic dermatitis and mice with chronic allergic dermatitis,76,77) and GRP released from primary afferents may degranulate mast cells.78) GRP and BB2 receptor seem to be interesting targets for pruritus, although their roles in pathological pruritus remain unclear. In addition, glutamate rather than GRP has been shown to be the principal excitatory transmitter between C-fiber primary afferents and BB2 receptor-expressing dorsal horn neurons.79) Neuromedin B is mainly expressed in small-sized sensory neurons.72) Intracerebroventricular injections of neuromedin B (and of GRP) elicits scratching in rats,80) but intrathecal injection of neuromedin B elicits pain-like behaviors.81)

Toll-like receptors mediate innate immune responses and recognize pathogen-associated molecule patterns. Although toll-like receptors are mainly expressed by immune cells, some members are expressed in primary sensory neurons and have been shown to be implicated in itching. Toll-like receptor 3 is expressed in a subset of small-sized dorsal root ganglion neurons that are positive for GRP and the TRPV1 channel.82) A deficiency of toll-like receptor 3 reduces scratching responses to compound 48/80 and chloroquine and spontaneous scratching in mice with dry skin.82) Toll-like receptor 3 deficiency inhibits long-term potentiation in the dorsal horn neurons, suggesting involvement in the regulation of central sensitization.82) Toll-like receptor 7 has been implicated in itching induced by imiquimod, an immune response modifier.83)

3.2. Itch-Inhibitory System

Itch perception is suppressed by various stimuli from the external environment; for example, itching is inhibited by counterstimuli including noxious stimuli, innocuous vibration, and warming stimulation applied to the itching area and vibration of the opposite side of the body.84,85) Distraction with noncontact stimulation also inhibits itching in humans86) and scratching behaviors in mice.87,88) These findings suggest the presence of itch-inhibitory systems in the central nervous system.

Subpopulations of wide dynamic range neurons and nociceptive-specific neurons in the superficial dorsal horn mainly receive itch signals from the periphery.8991) Histamine-evoked firing of primate spinothalamic tract neurons and murine superficial dorsal horn neurons is inhibited by scratching of the skin.92,93) The spontaneous activity of superficial dorsal horn neurons increases in mice with chronic dry skin, and the increased activity is also suppressed by scratching.94) This suppression is blocked by local administration of antagonists of glycine or γ-aminobutyric acid,94) and a subset of inhibitory interneurons in the dorsal horn is involved in the inhibitory regulation of itching.95) The scratching-induced suppression is inhibited by cold block and spinal transection at the upper spinal cord level, suggesting that this suppression is regulated by the descending inhibitory system.94)

Depletion of noradrenaline in the spinal cord enhances scratching behaviors induced by acute allergy and pruritogen injection in mice.96,97) Intrathecal injection of a nonselective α-adrenoceptor antagonist, but not of selective α1- or α2-adrenoceptor antagonists, also enhances the scratching behaviors.96,98) These findings suggest that the descending noradrenergic system exerts tonic inhibition on itch signaling in the spinal cord. Both α1- and α2-adrenoceptor antagonists act on the spinal cord to inhibit pruritogen-induced scratching.97,98) Thus, the descending noradrenergic system and α1/2-adrenoceptors in the spinal cord are potential therapeutic targets for pruritus treatment.

3.3. Opioids

An adverse effect of opioid analgesics (μ-opioid receptor agonists) includes pruritus, which is relieved by opioid μ-receptor antagonists.99) Opioid μ-receptor antagonists have been used in the treatment of pruritus in dermatologic and systemic diseases.100) Nalfurafine, a selective opioid κ-receptor agonist, has recently been approved for the treatment of chronic uremic pruritus in Japan.101) Thus, opioid μ- and κ-receptors are potential therapeutic targets for pruritus. In the spinal cord, the opioid μ-receptor isoform MOR1D heterodimerizes with the bombesin BB2 receptor, and the stimulation of MOR1D by morphine produces an effect similar to the stimulation of the BB2 receptor.102) Analgesia induced by intracisternal injection of morphine is antagonized by naloxonazine, an opioid μ1-receptor antagonist, but scratching induced by intracisternal morphine is not inhibited by naloxonazine.103) These opioid μ-receptor subtypes are potential therapeutic targets for pruritus.

4. CONCLUSION

Very few approved medications are indicated for the treatment of pruritus. There are no uniformly effective interventions to treat itching in all pruritic diseases, because no single mediator or mechanism is responsible for all pruritus. Therefore, precise information is needed on the details of the mechanisms of itching in pruritic diseases. During the past two decades, progress has been made in understanding the molecular basis of the physiology and pathophysiology of pruritus, although further research is needed. Specific, effective blockers and agonist/antagonists of newly identified targets are indispensable to corroborate their significance in the treatment of pathological pruritus, and their development is awaited.

REFERENCES
  • 1)  Abelson MB, McLaughlin JT, Gomes PJ. Antihistamines in ocular allergy: are they all created equal? Curr. Allergy Asthma Rep., 11, 205–211 (2011).
  • 2)  Inagaki N, Nakamura N, Nagao M, Musoh K, Kawasaki H, Nagai H. Participation of histamine H1 and H2 receptors in passive cutaneous anaphylaxis-induced scratching behavior in ICR mice. Eur. J. Pharmacol., 367, 361–371 (1999).
  • 3)  Andoh T, Sakai K, Urashima M, Kitazawa K, Honma A, Kuraishi Y. Involvement of leukotriene B4 in itching in a mouse model of ocular allergy. Exp. Eye Res., 98, 97–103 (2012).
  • 4)  Inami Y, Andoh T, Sasaki A, Kuraishi Y. Topical surfactant-induced pruritus: involvement of histamine released from epidermal keratinocytes. J. Pharmacol. Exp. Ther., 344, 459–466 (2013).
  • 5)  Inami Y, Andoh T, Kuraishi Y. Prevention of topical surfactant-induced itch-related responses by chlorogenic acid through the inhibition of increased histamine production in the epidermis. J. Pharmacol. Sci., 121, 242–245 (2013).
  • 6)  Thurmond RL, Gelfand EW, Dunford PJ. The role of histamine H1 and H4 receptors in allergic inflammation: the search for new antihistamines. Nat. Rev. Drug Discov., 7, 41–53 (2008).
  • 7)  Sugimoto Y, Iba Y, Nakamura Y, Kayasuga R, Kamei C. Pruritus-associated response mediated by cutaneous histamine H3 receptors. Clin. Exp. Allergy, 34, 456–459 (2004).
  • 8)  Rossbach K, Wendorff S, Sander K, Stark H, Gutzmer R, Werfel T, Kietzmann M, Bäumer W. Histamine H4 receptor antagonism reduces hapten-induced scratching behaviour but not inflammation. Exp. Dermatol., 18, 57–63 (2009).
  • 9)  Dunford PJ, Williams KN, Desai PJ, Karlsson L, McQueen D, Thurmond RL. Histamine H4 receptor antagonists are superior to traditional antihistamines in the attenuation of experimental pruritus. J. Allergy Clin. Immunol., 119, 176–183 (2007).
  • 10)  Ohsawa Y, Hirasawa N. The antagonism of histamine H1 and H4 receptors ameliorates chronic allergic dermatitis via anti-pruritic and anti-inflammatory effects in NC/Nga mice. Allergy, 67, 1014–1022 (2012).
  • 11)  Yamaura K, Oda M, Suwa E, Suzuki M, Sato H, Ueno K. Expression of histamine H4 receptor in human epidermal tissues and attenuation of experimental pruritus using H4 receptor antagonist. J. Toxicol. Sci., 34, 427–431 (2009).
  • 12)  Arthur RP, Shelley WB. The role of proteolytic enzymes in the production of pruritus in man. J. Invest. Dermatol., 25, 341–346 (1955).
  • 13)  Ui H, Andoh T, Lee JB, Nojima H, Kuraishi Y. Potent pruritogenic action of tryptase mediated by PAR-2 receptor and its involvement in anti-pruritic effect of nafamostat mesilate in mice. Eur. J. Pharmacol., 530, 172–178 (2006).
  • 14)  Andoh T, Takayama Y, Yamakoshi T, Lee JB, Sano A, Shimizu T, Kuraishi Y. Involvement of serine protease and proteinase-activated receptor 2 in dermatophyte-associated itch in mice. J. Pharmacol. Exp. Ther., 343, 91–96 (2012).
  • 15)  Hägermark Ö, Rajka G, Bergqvist U. Experimental itch in human skin elicited by rat mast cell chymase. Acta Derm. Venereol., 52, 125–128 (1972).
  • 16)  Tsujii K, Andoh T, Lee JB, Kuraishi Y. Activation of proteinase-activated receptors induces itch-associated response through histamine-dependent and -independent pathways in mice. J. Pharmacol. Sci., 108, 385–388 (2008).
  • 17)  Takahagi S, Shindo H, Watanabe M, Kameyoshi Y, Hide M. Refractory chronic urticaria treated effectively with the protease inhibitors, nafamostat mesilate and camostat mesilate. Acta Derm. Venereol., 90, 425–426 (2010).
  • 18)  Andoh T, Yoshida T, Lee JB, Kuraishi Y. Cathepsin E induces itch-related response through the production of endothelin-1 in mice. Eur. J. Pharmacol., 686, 16–21 (2012c).
  • 19)  Costa R, Manjavachi MN, Motta EM, Marotta DM, Juliano L, Torres HA, Pesquero JB, Calixto JB. The role of kinin B1 and B2 receptors in the scratching behaviour induced by proteinase-activated receptor-2 agonists in mice. Br. J. Pharmacol., 159, 888–897 (2010).
  • 20)  Steinhoff M, Neisius U, Ikoma A, Fartasch M, Heyer G, Skov PS, Luger TA, Schmelz M. Proteinase-activated receptor-2 mediates itch: a novel pathway for pruritus in human skin. J. Neurosci., 23, 6176–6180 (2003).
  • 21)  Tsujii K, Andoh T, Ui H, Lee JB, Kuraishi Y. Involvement of tryptase and proteinase-activated receptor-2 in spontaneous itch-associated response in mice with atopy-like dermatitis. J. Pharmacol. Sci., 109, 388–395 (2009).
  • 22)  Zhu Y, Peng C, Xu JG, Liu YX, Zhu QG, Liu JY, Li FQ, Wu JH, Hu JH. Participation of proteinase-activated receptor-2 in passive cutaneous anaphylaxis-induced scratching behavior and the inhibitory effect of tacrolimus. Biol. Pharm. Bull., 32, 1173–1176 (2009).
  • 23)  Reddy VB, Iuga AO, Shimada SG, LaMotte RH, Lerner EA. Cowhage-evoked itch is mediated by a novel cysteine protease: a ligand of protease-activated receptors. J. Neurosci., 28, 4331–4335 (2008).
  • 24)  Moormann C, Artuc M, Pohl E, Varga G, Buddenkotte J, Vergnolle N, Brehler R, Henz BM, Schneider SW, Luger TA, Steinhoff M. Functional characterization and expression analysis of the proteinase-activated receptor-2 in human cutaneous mast cells. J. Invest. Dermatol., 126, 746–755 (2006).
  • 25)  Davidson S, Zhang X, Yoon CH, Khasabov SG, Simone DA, Giesler GJ Jr. The itch-producing agents histamine and cowhage activate separate populations of primate spinothalamic tract neurons. J. Neurosci., 27, 10007–10014 (2007).
  • 26)  Nakano T, Andoh T, Lee JB, Kuraishi Y. Different dorsal horn neurons responding to histamine and allergic itch stimuli. Neuroreport, 19, 723–726 (2008).
  • 27)  Namer B, Carr R, Johanek LM, Schmelz M, Handwerker HO, Ringkamp M. Separate peripheral pathways for pruritus in man. J. Neurophysiol., 100, 2062–2069 (2008).
  • 28)  Akiyama T, Carstens MI, Carstens E. Enhanced responses of lumbar superficial dorsal horn neurons to intradermal PAR-2 agonist but not histamine in a mouse hindpaw dry skin itch model. J. Neurophysiol., 105, 2811–2817 (2011).
  • 29)  Okamoto R, Arikawa J, Ishibashi M, Kawashima M, Takagi Y, Imokawa G. Sphingosylphosphorylcholine is upregulated in the stratum corneum of patients with atopic dermatitis. J. Lipid Res., 44, 93–102 (2003).
  • 30)  Imokawa G. A possible mechanism underlying the ceramide deficiency in atopic dermatitis: expression of a deacylase enzyme that cleaves the N-acyl linkage of sphingomyelin and glucosylceramide. J. Dermatol. Sci., 55, 1–9 (2009).
  • 31)  Choi H, Kim S, Kim HJ, Kim KM, Lee CH, Shin JH, Noh M. Sphingosylphosphorylcholine down-regulates filaggrin gene transcription through NOX5-based NADPH oxidase and cyclooxygenase-2 in human keratinocytes. Biochem. Pharmacol., 80, 95–103 (2010).
  • 32)  Andoh T, Haza S, Saito A, Kuraishi Y. Involvement of leukotriene B4 in spontaneous itch-related behaviour in NC mice with atopic dermatitis-like skin lesions. Exp. Dermatol., 20, 894–898 (2011).
  • 33)  Kim HJ, Kim H, Han ES, Park SM, Koh JY, Kim KM, Noh MS, Kim JJ, Lee CH. Characterizations of sphingosylphosphorylcholine-induced scratching responses in ICR mice using naltrexon, capsaicin, ketotifen and Y-27632. Eur. J. Pharmacol., 583, 92–96 (2008).
  • 34)  Andoh T, Saito A, Kuraishi Y. Leukotriene B4 mediates sphingosylphosphorylcholine-induced itch-associated responses in mouse skin. J. Invest. Dermatol., 129, 2854–2860 (2009).
  • 35)  Ceballos A, Sabatté J, Nahmod K, Martínez D, Salamone G, Vermeulen M, Maggini J, Salomón H, Geffner J. Sphingosylphosphorylcholine activates dendritic cells, stimulating the production of interleukin-12. Immunology, 121, 328–336 (2007).
  • 36)  Kim HJ, Kim KM, Koh JY, Noh MS, Park MK, Lee HJ, Kim SY, Lee CH. Sphingosylphosphorylcholine induces degranulation of mast cells in the skin and plasma exudation in the ears of mice. J. Dermatol. Sci., 57, 57–59 (2010).
  • 37)  Andoh T, Kuraishi Y. Intradermal leukotriene B4, but not prostaglandin E2, induces itch-associated responses in mice. Eur. J. Pharmacol., 353, 93–96 (1998).
  • 38)  Tsukumo Y, Harada D, Manabe H. Pharmacological characterization of itch-associated response induced by repeated application of oxazolone in mice. J. Pharmacol. Sci., 113, 255–262 (2010).
  • 39)  Kuraishi Y, Ohtsuka E, Nakano T, Kawai S, Andoh T, Nojima H, Kamimura K. Possible involvement of 5-lipoxygenase metabolite in itch-associated response of mosquito allergy in mice. J. Pharmacol. Sci., 105, 41–47 (2007).
  • 40)  Yokomizo T, Kato K, Terawaki K, Izumi T, Shimizu T. A second leukotriene B4 receptor, BLT2. A new therapeutic target in inflammation and immunological disorders. J. Exp. Med., 192, 421–432 (2000).
  • 41)  Andoh T, Kuraishi Y. Expression of BLT1 leukotriene B4 receptor on the dorsal root ganglion neurons in mice. Brain Res. Mol. Brain Res., 137, 263–266 (2005).
  • 42)  Tsuji F, Aono H, Tsuboi T, Murakami T, Enomoto H, Mizutani K, Inagaki N. Role of leukotriene B4 in 5-lipoxygenase metabolite- and allergy-induced itch-associated responses in mice. Biol. Pharm. Bull., 33, 1050–1053 (2010).
  • 43)  Fernandes ES, Vong CT, Quek S, Cheong J, Awal S, Gentry C, Aubdool AA, Liang L, Bodkin JV, Bevan S, Heads R, Brain SD. Superoxide generation and leukocyte accumulation: key elements in the mediation of leukotriene B4-induced itch by transient receptor potential ankyrin 1 and transient receptor potential vanilloid 1. FASEB J., 27, 1664–1673 (2013).
  • 44)  Willemsen MA, Lutt MA, Steijlen PM, Cruysberg JR, van der Graaf M, Nijhuis-van der Sanden MW, Pasman JW, Mayatepek E, Rotteveel JJ. Clinical and biochemical effects of zileuton in patients with the Sjögren–Larsson syndrome. Eur. J. Pediatr., 160, 711–717 (2001).
  • 45)  Woodmansee DP, Simon RA. A pilot study examining the role of zileuton in atopic dermatitis. Ann. Allergy Asthma Immunol., 83, 548–552 (1999).
  • 46)  Kim DK, Kim HJ, Sung KS, Kim H, Cho SA, Kim KM, Lee CH, Kim JJ. 12(S)-HPETE induces itch-associated scratchings in mice. Eur. J. Pharmacol., 554, 30–33 (2007).
  • 47)  Kim HJ, Kim DK, Kim H, Koh JY, Kim KM, Noh MS, Lee S, Kim S, Park SH, Kim JJ, Kim SY, Lee CH. Involvement of the BLT2 receptor in the itch-associated scratching induced by 12(S)-lipoxygenase products in ICR mice. Br. J. Pharmacol., 154, 1073–1078 (2008).
  • 48)  Aoki J, Inoue A, Okudaira S. Two pathways for lysophosphatidic acid production. Biochim. Biophys. Acta, 1781, 513–518 (2008).
  • 49)  Kremer AE, Martens JJ, Kulik W, Ruëff F, Kuiper EM, van Buuren HR, van Erpecum KJ, Kondrackiene J, Prieto J, Rust C, Geenes VL, Williamson C, Moolenaar WH, Beuers U, Oude Elferink RP. Lysophosphatidic acid is a potential mediator of cholestatic pruritus. Gastroenterology, 139, 1008–1018, e1 (2010).
  • 50)  Hashimoto T, Ohata H, Momose K. Itch-scratch responses induced by lysophosphatidic acid in mice. Pharmacology, 72, 51–56 (2004).
  • 51)  Kremer AE, van Dijk R, Leckie P, Schaap FG, Kuiper EM, Mettang T, Reiners KS, Raap U, van Buuren HR, van Erpecum KJ, Davies NA, Rust C, Engert A, Jalan R, Oude Elferink RP, Beuers U. Serum autotaxin is increased in pruritus of cholestasis, but not of other origin, and responds to therapeutic interventions. Hepatology, 56, 1391–1400 (2012).
  • 52)  Dillon SR, Sprecher C, Hammond A, Bilsborough J, Rosenfeld-Franklin M, Presnell SR, Haugen HS, Maurer M, Harder B, Johnston J, Bort S, Mudri S, Kuijper JL, Bukowski T, Shea P, Dong DL, Dasovich M, Grant FJ, Lockwood L, Levin SD, LeCiel C, Waggie K, Day H, Topouzis S, Kramer J, Kuestner R, Chen Z, Foster D, Parrish-Novak J, Gross JA. Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice. Nat. Immunol., 5, 752–760 (2004).
  • 53)  Sonkoly E, Muller A, Lauerma AI, Pivarcsi A, Soto H, Kemeny L, Alenius H, Dieu-Nosjean MC, Meller S, Rieker J, Steinhoff M, Hoffmann TK, Ruzicka T, Zlotnik A, Homey B. IL-31: a new link between T cells and pruritus in atopic skin inflammation. J. Allergy Clin. Immunol., 117, 411–417 (2006).
  • 54)  Nobbe S, Dziunycz P, Mühleisen B, Bilsborough J, Dillon SR, French LE, Hofbauer GF. IL-31 expression by inflammatory cells is preferentially elevated in atopic dermatitis. Acta Derm. Venereol., 92, 24–28 (2012).
  • 55)  Takaoka A, Arai I, Sugimoto M, Honma Y, Futaki N, Nakamura A, Nakaike S. Involvement of IL-31 on scratching behavior in NC/Nga mice with atopic-like dermatitis. Exp. Dermatol., 15, 161–167 (2006).
  • 56)  Grimstad O, Sawanobori Y, Vestergaard C, Bilsborough J, Olsen UB, Grønhøj-Larsen C, Matsushima K. Anti-interleukin-31-antibodies ameliorate scratching behaviour in NC/Nga mice: a model of atopic dermatitis. Exp. Dermatol., 18, 35–43 (2009).
  • 57)  Tamura S, Morikawa Y, Miyajima A, Senba E. Expression of oncostatin M receptor β in a specific subset of nociceptive sensory neurons. Eur. J. Neurosci., 17, 2287–2298 (2003).
  • 58)  Bando T, Morikawa Y, Komori T, Senba E. Complete overlap of interleukin-31 receptor A and oncostatin M receptor beta in the adult dorsal root ganglia with distinct developmental expression patterns. Neuroscience, 142, 1263–1271 (2006).
  • 59)  Kasraie S, Niebuhr M, Baumert K, Werfel T. Functional effects of interleukin 31 in human primary keratinocytes. Allergy, 66, 845–852 (2011).
  • 60)  Imamachi N, Park GH, Lee H, Anderson DJ, Simon MI, Basbaum AI, Han SK. TRPV1-expressing primary afferents generate behavioral responses to pruritogens via multiple mechanisms. Proc. Natl. Acad. Sci. U.S.A., 106, 11330–11335 (2009).
  • 61)  Shim WS, Tak MH, Lee MH, Kim M, Kim M, Koo JY, Lee CH, Kim M, Oh U. TRPV1 mediates histamine-induced itching via the activation of phospholipase A2 and 12-lipoxygenase. J. Neurosci., 27, 2331–2337 (2007).
  • 62)  Dai Y, Moriyama T, Higashi T, Togashi K, Kobayashi K, Yamanaka H, Tominaga M, Noguchi K. Proteinase-activated receptor 2-mediated potentiation of transient receptor potential vanilloid subfamily 1 activity reveals a mechanism for proteinase-induced inflammatory pain. J. Neurosci., 24, 4293–4299 (2004).
  • 63)  Peier AM, Reeve AJ, Andersson DA, Moqrich A, Earley TJ, Hergarden AC, Story GM, Colley S, Hogenesch JB, McIntyre P, Bevan S, Patapoutian A. A heat-sensitive TRP channel expressed in keratinocytes. Science, 296, 2046–2049 (2002).
  • 64)  Xu H, Ramsey IS, Kotecha SA, Moran MM, Chong JA, Lawson D, Ge P, Lilly J, Silos-Santiago I, Xie Y, DiStefano PS, Curtis R, Clapham DE. TRPV3 is a calcium-permeable temperature-sensitive cation channel. Nature, 418, 181–186 (2002).
  • 65)  Yoshioka T, Imura K, Asakawa M, Suzuki M, Oshima I, Hirasawa T, Sakata T, Horikawa T, Arimura A. Impact of the Gly573Ser substitution in TRPV3 on the development of allergic and pruritic dermatitis in mice. J. Invest. Dermatol., 129, 714–722 (2009).
  • 66)  Yamamoto-Kasai E, Imura K, Yasui K, Shichijou M, Oshima I, Hirasawa T, Sakata T, Yoshioka T. TRPV3 as a therapeutic target for itch. J. Invest. Dermatol., 132, 2109–2112 (2012).
  • 67)  Kobayashi K, Fukuoka T, Obata K, Yamanaka H, Dai Y, Tokunaga A, Noguchi K. Distinct expression of TRPM8, TRPA1, and TRPV1 mRNAs in rat primary afferent neurons with adelta/c-fibers and colocalization with trk receptors. J. Comp. Neurol., 493, 596–606 (2005).
  • 68)  Wang H, Woolf CJ. Pain TRPs. Neuron, 46, 9–12 (2005).
  • 69)  Wilson SR, Gerhold KA, Bifolck-Fisher A, Liu Q, Patel KN, Dong X, Bautista DM. TRPA1 is required for histamine-independent, Mas-related G protein-coupled receptor-mediated itch. Nat. Neurosci., 14, 595–602 (2011).
  • 70)  Dai Y, Wang S, Tominaga M, Yamamoto S, Fukuoka T, Higashi T, Kobayashi K, Obata K, Yamanaka H, Noguchi K. Sensitization of TRPA1 by PAR2 contributes to the sensation of inflammatory pain. J. Clin. Invest., 117, 1979–1987 (2007).
  • 71)  Liu T, Ji RR. Oxidative stress induces itch via activation of transient receptor potential subtype ankyrin 1 in mice. Neurosci. Bull., 28, 145–154 (2012).
  • 72)  Fleming MS, Ramos D, Han SB, Zhao J, Son YJ, Luo W. The majority of dorsal spinal cord gastrin releasing peptide is synthesized locally whereas neuromedin B is highly expressed in pain- and itch-sensing somatosensory neurons. Mol. Pain, 8, 52 (2012).
  • 73)  Sun YG, Chen ZF. A gastrin-releasing peptide receptor mediates the itch sensation in the spinal cord. Nature, 448, 700–703 (2007).
  • 74)  Mishra SK, Holzman S, Hoon MA. A nociceptive signaling role for neuromedin B. J. Neurosci., 32, 8686–8695 (2012).
  • 75)  Sun YG, Zhao ZQ, Meng XL, Yin J, Liu XY, Chen ZF. Cellular basis of itch sensation. Science, 325, 1531–1534 (2009).
  • 76)  Kagami S, Sugaya M, Suga H, Morimura S, Kai H, Ohmatsu H, Fujita H, Tsunemi Y, Sato S. Serum gastrin-releasing peptide levels correlate with pruritus in patients with atopic dermatitis. J. Invest. Dermatol., 133, 1673–1675 (2013).
  • 77)  Tominaga M, Ogawa H, Takamori K. Histological characterization of cutaneous nerve fibers containing gastrin-releasing peptide in NC/Nga mice: an atopic dermatitis model. J. Invest. Dermatol., 129, 2901–2905 (2009).
  • 78)  Andoh T, Kuwazono T, Lee JB, Kuraishi Y. Gastrin-releasing peptide induces itch-related responses through mast cell degranulation in mice. Peptides, 32, 2098–2103 (2011).
  • 79)  Koga K, Chen T, Li XY, Descalzi G, Ling J, Gu J, Zhuo M. Glutamate acts as a neurotransmitter for gastrin releasing peptide-sensitive and insensitive itch-related synaptic transmission in mammalian spinal cord. Mol. Pain, 7, 47 (2011).
  • 80)  Su PY, Ko MC. The role of central gastrin-releasing peptide and neuromedin B receptors in the modulation of scratching behavior in rats. J. Pharmacol. Exp. Ther., 337, 822–829 (2011).
  • 81)  Cridland RA, Henry JL. Bombesin, neuromedin C and neuromedin B given intrathecally facilitate the tail flick reflex in the rat. Brain Res., 584, 163–168 (1992).
  • 82)  Liu T, Berta T, Xu ZZ, Park CK, Zhang L, Lü N, Liu Q, Liu Y, Gao YJ, Liu YC, Ma Q, Dong X, Ji RR. TLR3 deficiency impairs spinal cord synaptic transmission, central sensitization, and pruritus in mice. J. Clin. Invest., 122, 2195–2207 (2012).
  • 83)  Liu T, Xu ZZ, Park CK, Berta T, Ji RR. Toll-like receptor 7 mediates pruritus. Nat. Neurosci., 13, 1460–1462 (2010).
  • 84)  Melzack R, Schecter B. Itch and vibration. Science, 147, 1047–1048 (1965).
  • 85)  Ward L, Wright E, McMahon SB. A comparison of the effects of noxious and innocuous counterstimuli on experimentally induced itch and pain. Pain, 64, 129–138 (1996).
  • 86)  Leibovici V, Magora F, Cohen S, Ingber A. Effects of virtual reality immersion and audiovisual distraction techniques for patients with pruritus. Pain Res. Manag., 14, 283–286 (2009).
  • 87)  Tohda C, Yamaguchi T, Kuraishi Y. Intracisternal injection of opioids induces itch-associated response through μ-opioid receptors in mice. Jpn. J. Pharmacol., 74, 77–82 (1997).
  • 88)  Yamaguchi T, Maekawa T, Nishikawa Y, Nojima H, Kaneko M, Kawakita T, Miyamoto T, Kuraishi Y. Characterization of itch-associated responses of NC mice with mite-induced chronic dermatitis. J. Dermatol. Sci., 25, 20–28 (2001).
  • 89)  Andrew D, Craig AD. Spinothalamic lamina I neurons selectively sensitive to histamine: a central neural pathway for itch. Nat. Neurosci., 4, 72–77 (2001).
  • 90)  Jinks SL, Carstens E. Responses of superficial dorsal horn neurons to intradermal serotonin and other irritants: comparison with scratching behavior. J. Neurophysiol., 87, 1280–1289 (2002).
  • 91)  Omori Y, Andoh T, Shirakawa H, Ishida H, Hachiga T, Kuraishi Y. Itch-related responses of dorsal horn neurons to cutaneous allergic stimulation in mice. Neuroreport, 20, 478–481 (2009).
  • 92)  Davidson S, Zhang X, Khasabov SG, Simone DA, Giesler GJ Jr. Relief of itch by scratching: state-dependent inhibition of primate spinothalamic tract neurons. Nat. Neurosci., 12, 544–546 (2009).
  • 93)  Akiyama T, Tominaga M, Carstens MI, Carstens EE. Site-dependent and state-dependent inhibition of pruritogen-responsive spinal neurons by scratching. Eur. J. Neurosci., 36, 2311–2316 (2012).
  • 94)  Akiyama T, Iodi Carstens M, Carstens E. Transmitters and pathways mediating inhibition of spinal itch-signaling neurons by scratching and other counterstimuli. PLoS ONE, 6, e22665 (2011).
  • 95)  Ross SE, Mardinly AR, McCord AE, Zurawski J, Cohen S, Jung C, Hu L, Mok SI, Shah A, Savner EM, Tolias C, Corfas R, Chen S, Inquimbert P, Xu Y, McInnes RR, Rice FL, Corfas G, Ma Q, Woolf CJ, Greenberg ME. Loss of inhibitory interneurons in the dorsal spinal cord and elevated itch in Bhlhb5 mutant mice. Neuron, 65, 886–898 (2010).
  • 96)  Gotoh Y, Omori Y, Andoh T, Kuraishi Y. Tonic inhibition of allergic itch signaling by the descending noradrenergic system in mice. J. Pharmacol. Sci., 115, 417–420 (2011).
  • 97)  Gotoh Y, Andoh T, Kuraishi Y. Noradrenergic regulation of itch transmission in the spinal cord mediated by α-adrenoceptors. Neuropharmacology, 61, 825–831 (2011).
  • 98)  Gotoh Y, Andoh T, Kuraishi Y. Clonidine inhibits itch-related response through stimulation of α2-adrenoceptors in the spinal cord in mice. Eur. J. Pharmacol., 650, 215–219 (2011).
  • 99)  Reich A, Szepietowski JC. Opioid-induced pruritus: an update. Clin. Exp. Dermatol., 35, 2–6 (2010).
  • 100)  Phan NQ, Bernhard JD, Luger TA, Ständer S. Antipruritic treatment with systemic μ-opioid receptor antagonists: a review. J. Am. Acad. Dermatol., 63, 680–688 (2010).
  • 101)  Phan NQ, Lotts T, Antal A, Bernhard JD, Ständer S. Systemic kappa opioid receptor agonists in the treatment of chronic pruritus: a literature review. Acta Derm. Venereol., 92, 555–560 (2012).
  • 102)  Liu XY, Liu ZC, Sun YG, Ross M, Kim S, Tsai FF, Li QF, Jeffry J, Kim JY, Loh HH, Chen ZF. Unidirectional cross-activation of GRPR by MOR1D uncouples itch and analgesia induced by opioids. Cell, 147, 447–458 (2011).
  • 103)  Andoh T, Yageta Y, Konno M, Yamaguchi-Miyamoto T, Takahata H, Nojima H, Nemoto H, Kuraishi Y. Evidence for separate involvement of different μ-opioid receptor subtypes in itch and analgesia induced by supraspinal action of opioids. J. Pharmacol. Sci., 106, 667–670 (2008).
 
© 2013 The Pharmaceutical Society of Japan
feedback
Top