Content » Vol 97, Issue 8

Investigative Report

Involvement of Leukotriene B4 Released from Keratinocytes in Itch-associated Response to Intradermal Interleukin-31 in Mice

Tsugunobu Andoh, Ayako Harada and Yasushi Kuraishi

Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan

ABSTRACT

A recent study suggests that interleukin-31 (IL-31) exerts its effect via indirect mechanisms rather than through direct stimulation of cutaneous nerves. How-ever, the underlying peripheral mechanisms of IL-31-induced itch in the skin remain unclear. Therefore, the present study investigated the peripheral mechanisms underlying IL-31-induced itch in mice. IL-31-induced itch-related response was inhibited by anti-allergic drugs (tranilast and azelastine), but not by an H1 histamine receptor antagonist (terfenadine). Furthermore, a 5-lipoxygenase inhibitor (zileuton), but not a cyclooxygenase inhibitor (indomethacin), and a leuko-triene B4 (LTB4) receptor antagonist (CMHVA) attenuated the action of IL-31. IL-31 receptor-immunoreactivity was observed in the epidermis and primary sensory neurones. IL-31 receptor mRNA was expressed in mouse keratinocytes and dorsal root ganglia neurones. IL-31 increased the production of LTB4 in mouse keratinocytes. These results suggest that IL-31 elicits itch not only through direct action on primary sensory neurones, but also by inducing LTB4 production in keratinocytes.

Key words: itch; leukotriene B4; interleukin-31; keratinocytes; scratching; dorsal root ganglia.

Accepted May 16, 2017; Epub ahead of print May 17, 2017

Acta Derm Venereol 2017; 97: xx–xx.

Corr: Tsugunobu Andoh, Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-1094, Japan. E-mail: andoht@pha.u-toyama.ac.jp

INTRODUCTION

Itch (or pruritus), an unpleasant skin sensation that evokes the desire to scratch, is the most common symptom of dermatitis (e.g. atopic dermatitis) and certain systemic disorders (e.g. cholestasis). Histamine from the mast cells has been thought to play an essential role in itch. H1 histamine receptor antagonists are the drugs of first choice for the treatment of itch. However, many severe pruritic diseases, except acute urticaria, respond poorly to H1 histamine receptor antagonists (1–3). Therefore, the underlying mechanisms and mediators of itch in most pruritic diseases are unclear.

Interleukin-31 (IL-31), a cytokine released from T cells (especially, CD4+-Th2 cells), signals through a receptor complex comprising IL-31RA and oncostatin M receptor β (OSMRβ) (4, 5). IL-31 plays a major role in the induction of chronic inflammation in diseases such as dermatitis, allergic rhinitis and asthma (6). In particular, the expression of IL-31 mRNA is elevated in the skin of patients with atopic dermatitis (7, 8) as well as in animal models of atopic dermatitis (9) and contact dermatitis (10). IL-31 induces skin inflammation (5) and scratching, an itch-associated response (11). However, in animal models of atopic dermatitis, the administration of anti-IL-31 antibody inhibited spontaneous scratching, but not dermatitis (12). These findings suggest that IL-31 is a mediator of itch.

The peripheral mechanisms of IL-31-induced pruritus are not completely clear. A recent study showed that IL-31 increases the intracellular Ca2+ ion concentration in the primary cultures of mouse dorsal root ganglia (DRG) neurones, suggesting that IL-31 directly activates primary afferents and induces itch sensation (13). However, IL-31 receptor complex is expressed not only in DRG neurones, but also in several immune cells (e.g. activated monocytes, macrophages, eosinophils, and basophils) and keratinocytes (4). A recent study showed that IL-31 exerts its pruritic effect indirectly via keratinocytes and secondary mediators rather than through the stimulation of its receptors on cutaneous nerves (14). In addition, our previous studies on the peripheral mechanisms of itch have shown the important role of keratinocytes and their itch mediators, including leukotriene B4 (LTB4) (15–18). Therefore, this study investigated the involvement of LTB4 in peripheral mechanism of IL-31-induced itch.

MATERIALS AND METHODS
Animals

Male ICR mice aged 4–11 weeks were used in this study. The mice were purchased from Japan SLC (Shizuoka, Japan). For one part of the experiment, neonatal mice were used for the isolation of cutaneous keratinocytes. Neonatal mice were obtained from late pregnant mice purchased from Japan SLC (Shizuoka). They were housed in a room with controlled temperature (21–23°C), humidity (45–65%), and light (on from 07.00h to 19.00h). Food and water were freely available. All experimental procedures involving animals were approved by the Committee for Animal Experiments at the University of Toyama and conducted in accordance with the guidelines of the Japanese Pharmacological Society.

Drugs

Recombinant mouse IL-31 was prepared in Chugai Pharmaceutical Co., Ltd (Shizuoka, Japan). Briefly, IL-31 was purified from the supernatant of mouse IL-31-transformed Chinese hamster ovary cells using hydroxyapatite column (Bio-Rad Laboratories, Inc., Hercules, CA, USA), anion-exchange column (GE healthcare, Chicago, IL, USA), and gel filtration column (GE healthcare). IL-31 was dissolved in phosphate buffered-saline (PBS) and administered intradermally. Naltrexone hydrochloride (Sigma-Aldrich, St Louis, MO, USA) (18–20) was dissolved in physiological saline and administered subcutaneously 15 min before IL-31 injection. Terfenadine (16, 18, 20) and tranilast (Sigma-Aldrich) (21) were dissolved in 0.5% sodium carboxymethyl cellulose and administered orally 30 and 60 min before IL-31 injection, respectively. Azelastine hydrochloride (Sigma-Aldrich) (16, 18, 22) was dissolved in tap water and administered orally 30 min before IL-31 injection. Indomethacin (Sigma-Aldrich) (15, 18), zileuton (Ono Pharmaceutical Co. Ltd, Osaka, Japan) (15, 18, 23), and CMHVA (5-[2-(2-carboxyethyl)-3-[6-(4-methoxyphenyl)-5E-hexenyl]oxyphenoxy] valeric acid) (Ono Pharmaceutical Co. Ltd) (15, 16, 18, 24, 25)) were dissolved in 0.5% sodium carboxymethyl cellulose and administered orally 30, 60, and 60 min before IL-31 injection, respectively.

Behavioural tests

The mouse hair was clipped over the rostral part of the back the day before the experiment. For acclimation, the mice were housed individually in the observation cage (an acrylic cage composing 4 cells (13 × 9 × 35 cm)) for at least 1 h. Immediately after the intradermal injection, the mice were put back into the same cells, and their behaviour was videotaped for 1 h by personnel kept out of the observation room. Scratching by the hind paws at the injection site was observed during video playback (26). The mice stretched either the left or the right hind paw, leaned their head towards the paws, rapidly scratched themselves, and brought their toenails to their mouth. A series of these movements was counted as one bout of scratching (15).

Immunohistochemistry

Under anaesthesia with pentobarbital (80 mg/kg, intraperitoneal), the mice were transcardially perfused with PBS and then 4% paraformaldehyde (PFA). The rostral back skin was isolated and post-fixed with 4% PFA at 4°C overnight. The skin was immersed in 30% sucrose solution at 4°C for at least 2 days. The tissue was embedded in Tissue-Tek® optimum cutting temperature (OCT) compound (Sakura Fineteck Co., Ltd, Tokyo) and stored at −80°C until use. The frozen samples were sectioned at 20 μm with a cryostat (Leica, Wetzlar, Germany). After being washed with PBS 3 times, the sections were treated with 0.3% Triton X-100 in PBS and then with 0.25% foetal bovine serum to block non-specific immunoglobulin binding. The sections were incubated with goat anti-IL-31 receptor A (IL-31RA) antibody (AF2107; 1:500, R&D Systems, Inc., Minneapolis, MN, USA) and rabbit anti-protein gene product 9.5 antibody (RA95101; 1:2000, Ultraclone Ltd, Isle of Wight, UK) at 4°C overnight. After washing, the preparations were incubated with Alexa Fluor 594-conjugated anti-rabbit IgG and Alexa Fluor 488-conjugated anti-goat IgG antibodies (Life Technologies, Carlsbad, CA, USA) for 1 h at room temperature. The fluorescence signals were observed using a confocal laser-scanning microscope (Bio-Rad Laboratories Inc., Hercules, USA).

Primary cultures of murine keratinocytes and dorsal ganglion neurones

The skin from neonatal mice was removed and treated with 0.05% collagenase A (Roche Diagnostics GmbH, Mannheim, Germany) dissolved in serum-free MCDB 153 medium (Sigma-Aldrich) containing 0.67% 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (Dojindo, Kumamoto, Japan), 0.12% sodium bicarbonate (Wako Pure Chemical Industries Ltd, Osaka, Japan), 0.01% penicillin G (Meiji Seika Pharma Co., Ltd, Tokyo, Japan), and 0.006% kanamycin (Wako Pure Chemical Industries Ltd) at 4°C overnight. The epidermal sheets were peeled gently from the underlying dermis. The keratinocytes were dissociated by gently shaking the serum-free MCDB 153 medium containing the epidermal sheet. After washing with keratinocyte growth medium (CELLnTEC Advanced Cell Systems AG, Bern, Switzerland), the keratinocytes were cultured in collagen-coated 24-well or 6-cm-diameter plates.

The bilateral DRGs at C1–T13 levels were removed from the 6-week-old mice. DRG was treated with 0.25% collagenase (Wako Pure Chemical Industries Ltd) in serum-free Dulbecco’s Modified Eagle’s medium (DMEM) containing antibiotics (100 units/ml penicillin and 100 μg/ml streptomycin) at 37°C for 1 h and was shaken gently for dissociation. The dissociated cells were treated with 0.25% trypsin-EDTA at 37°C for 15 min and then the cells were washed with DMEM containing antibiotics. The cells suspended in DMEM containing antibiotics, 10% foetal bovine serum, and 50 μM cytosine β-D-arabinofuranoside (Sigma-Aldrich), were cultured in poly-D-lysine-coated 6-cm-diameter plates.

Reverse transcription and PCR

The total RNA from the primary cultures of murine keratinocytes and DRG neurones was extracted by using GeneElute Mammalian Total RNA miniprep kit (Sigma-Aldrich). The total RNA (1 μg) was reverse transcribed into the cDNA using oligo (dT)16 primer and Reverscript III® (Wako Pure Chemical Industries Ltd). PCR was performed as described previously (16). The primer sequences used were as follows: IL-31RA (sense) 5’-tgctgctatggcatgaagtc-3’; IL-31RA (antisense) 5’-tcaagtgccaacaagacagc-3’, OSMRβ (sense) 5’-taaaccaggcgtccgttac-3’; OSMRβ (antisense) 5’-aaggttttccacggtgagtg-3’, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (sense) 5’-ccaaggtcatccatgacaac-3’; GAPDH (antisense) 5’-ttactccttggaggccatgt-3’. The reaction product was separated on 2% agarose gel and stained with ethidium bromide.

Measurement of LTB4

To measure LTB4 production in keratinocytes, the culture medium (200 μl) was collected (removed) from the primary cultures of murine keratinocytes 10 min after IL-31 administration and assayed for LTB4 with an enzyme immunoassay (EIA) kit (Cayman Chemical, Ann Arbor, MI, USA). Zileuton and the vehicle (dime-thyl sulphoxide, final concentration 0.001%) were treated 60 min before IL-31 application. Anti-IL-31RA antibody (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) and non-specific IgG (Sigma-Aldrich) were treated 30 min before IL-31 application. The remaining keratinocytes were treated with 1% Triton X-100 and used for protein determination by using a protein assay kit (Bio-Rad Laboratories, Inc.). The amount of LTB4 was normalized to the amount of protein.

Data processing

Data are presented as means ± standard error of the mean (SEM). The statistical significance was analysed using the Student’s t-test (2 groups), one-way analysis of variance (ANOVA), or 2-way repeated measures-ANOVA followed by a post hoc Holm–Šidák test (3 or more groups). A p-value < 0.05 was considered statistically significant.

RESULTS
Scratching behaviour induced by IL-31

An intradermal injection of IL-31 (100 ng/site), but not vehicle, into the rostral back elicited hind-paw scratching directed towards the injection site (Fig. 1A). This effect peaked during the first 10 min. Compared with the treatment of vehicle (PBS) control, IL-31 (100 ng/site)-induced scratching increased significantly from 10 min after the injection, and the significant effects continued for at least 50 min (Fig. 1A). The dose-response curve for IL-31 was bell-shaped (Fig. 1B).


Fig. 1. Scratching induced after the intradermal injection of interleukin (IL)-31 in mice. (A) The time course of scratching after vehicle (upper panel) and IL-31 (lower panel, 100 ng/site) injection. (B) The dose-response curve for the scratch-inducing effects of IL-31. The mice were given an intradermal injection of IL-31 or vehicle (PBS). Data are presented as mean and standard error of the mean (SEM) (n = 8–16). *p < 0.05 compared with that of PBS (Holm–Šidák test). PBS: phosphate buffered-saline.

Effects of various drugs on IL-31-induced scratching

The effects of systemic pretreatment on the scratching induced by an intradermal injection of IL-31 (100 ng/site) were examined by using several agents. The μ-opioid receptor antagonist naltrexone hydrochloride (1 and 10 mg/kg) significantly inhibited IL-31-induced scratching (Fig. 2). The H1 histamine receptor antagonist, terfenadine (30 mg/kg), did not exhibit any effect on scratching (Fig. 2). The anti-allergic agents tranilast (100 and 300 mg/kg; Fig. 2) and azelastine hydrochloride (30 mg/kg; Fig. 2) inhibit IL-31-induced scratching. The 5-lipoxygenase inhibitor, zileuton (30 and 100 mg/kg), inhibited IL-31-induced scratching, whereas the cyclooxygenase inhibitor indomethacin (10 mg/kg) did not (Fig. 3). The LTB4 receptor antagonist CMHVA (10, 30 and 100 mg/kg) also attenuated IL-31-induced scratching (Fig. 3). Naltrexone hydrochloride (10 mg/kg), tranilast (30 mg/kg), azelastine hydrochloride (30 mg/kg), zileuton (100 mg/kg, p.o.), and CMHVA (100 mg/kg) did not affect the spontaneous locomotor activity, suggesting that these drugs at the dosage used do not have sedative effects (Fig. S1).


Fig. 2. Effects of various drugs on interleukin (IL)-31-induced scratching in mice. IL-31 (100 ng/site) was injected intradermally. Naltrexone hydrochloride (naltrexone) was injected subcutaneously 15 min before IL-31 injection. Terfenadine, tranilast, or azelastine hydrochloride (azelastine) was administered orally 30, 60 or 30 min, respectively before IL-31 injection. Vehicle (VH)1 (saline), VH2 (0.5% sodium carboxymethyl cellulose) and VH3 (tap water) denote the vehicle administered. Data are presented as mean and standard error of the mean (SEM) (n=5–8). *p<0.05 when compared with that of VH (Student’s t-test or Holm–Šidák test).


Fig. 3. Effects of arachidonic acid metabolism-related drugs on interleukin (IL)-31-induced scratching in mice. IL-31 (100 ng/site) was injected intradermally. Indomethacin, zileuton, or CMHVA was administered orally 30, 60, or 60 min, respectively before IL-31 injection. Vehicle (VH) (0.5% sodium carboxymethyl cellulose) denotes the vehicle administered. Data are presented as mean and SEM (n = 5–8). *p < 0.05 compared with that of VH (Holm–Šidák test).

Distribution of IL-31 receptor A in the mouse skin, and the expression of IL-31 receptor mRNA in the mouse skin, dorsal root gangleon (DRG), and primary cultures of DRG neurones and keratinocytes

IL-31RA-immunoreactivity was mainly localized in the epidermis and PGP-9.5-immunoreactive primary afferents (Fig. 4A).


Fig. 4. Distribution of interleukin-31 receptor A (IL-31RA) in the mouse skin and the expression of IL-31 receptors mRNA. (A) The typical examples of the distribution of IL-31 receptor A (IL-31RA) and primary afferent neurones in the mouse skin. IL-31RA (green) and PGP-9.5 (red, a marker of neurone) were immunostained in the rostral back skin. Arrowheads indicate primary sensory neurones, and the dotted lines indicates the boundary between the dermis and epidermis. Scale bar = 100 μm. (B) The expression of IL-31 receptor (IL-31RA and OSMRβ) mRNA in the mouse skin, dorsal root ganglion (DRG), and primary cultures of DRG neurones and keratinocytes. IL-31RA, OSMRβ, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) mRNAs were determined using RT-PCR.

The RT-PCR revealed the substantial expression of IL-31RA and OSMRβ mRNA in the DRG and skin (Fig. 4B). In addition, the primary cultures of DRG neurones and keratinocytes also expressed both IL-31RA and OSMRβ mRNA (Fig. 4B).

IL-31-induced production of LTB4 in primary cultures of mouse keratinocytes

A bath-application of IL-31 (0.2 and 2 μg/ml) to the primary cultures of mouse keratinocytes significantly increased the production of LTB4 (Fig. 5A). The effect of IL-31 (2 μg/ml) was almost abolished by the 5-lipoxygenase inhibitor zileuton (10 μM; Fig. 5A). In addition, anti-IL-31RA antibody (10 μg/ml) significantly inhibited IL-31-induced LTB4 production (Fig. 5B).


Fig. 5. Interleukin (IL)-31-induced leukotriene B4 (LTB4) production in the primary cultures of keratinocytes. (A) IL-31-induced production of LTB4 and effect of 5-lipoxygenase inhibitor zileuton on LTB4 production. Zileuton (10 μM) or the vehicle (VH1) was treated 60 min before IL-31 or the vehicle (VH2) application. The concentration of LTB4 in the tested medium was determined 10 min after IL-31 application. Data are presented as mean and standard error of the mean (SEM) (n = 18). *p <  0.05 compared with that of VH1+VH2 (Holm–Šidák test). (B) Effect of anti-IL-31RA on IL-31-induced LTB4 production. Anti-31RA antibody (anti-31RA, 10 μg/ml) or non-specific immunoglobulin G (IgG) (nIgG, 10 μg/ml) were treated 30 min before IL-31 (2 μg/ml) or VH2. The concentration of LTB4 in the tested medium was determined 10 min after IL-31 application. Data are presented as mean and SEM (n = 6). *p < 0.05 compared with that of nIgG+VH2 (Holm–Šidák test).

DISCUSSION

This study aimed to demonstrate peripheral mechanisms of IL-31-induced itch and found that IL-31-induced itch through the production of LTB4 in epidermal keratinocytes.

An intradermal injection of IL-31 elicited scratching at the injection site. Next, it was examined whether scratching in mice was an itch-related behaviour. The scratching was inhibited by the μ-opioid receptor antagonist naltrexone. μ-Opioid receptor antagonists inhibit the scratching induced by several pruritogens (e.g. substance P, sphingosylphosphorylcholine, serotonin, gastrin-releasing peptide, and α-melanocyte-stimulating hormone) (18–20, 27, 28), dermatoses (e.g. skin allergy, atopic dermatitis and xerosis) in rodents (29–31), and itching/scratching in humans with pruritic diseases (e.g. cholestasis, urticaria and atopic dermatitis) (32, 33). μ-Opioid receptor antagonists suppress itch-associated behaviour without inhibiting the pain-related behaviour (34, 35). The opioid antagonists may inhibit itching/scratching by acting on the μ-opioid receptors in the central nervous system, especially in the lower brainstem (36–39). Thus, it can be suggested that IL-31-induced scratching is an itch-associated response in mice.

It has been reported that IL-31 acts directly on primary afferent neurones (13). In this study, although an intradermal administration of IL-31 elicited scratching with the effect peaking during the first 10-min period, the significant effect of IL-31 continued for another 50 min after the attainment of peak effect, suggesting that IL-31 affects not only directly, but also indirectly, primary afferent neurones.

The H1 histamine receptor antagonist terfenadine (30 mg/kg) inhibited histamine-induced scratching in mice (30). However, the same dosage of terfenadine did not affect IL-31-induced scratching, suggesting that histamine may not play an important role in scratching. However, anti-allergic drugs (tranilast and azelastine) attenuated IL-31-induced scratching. Tranilast has an inhibitory action on the production of LTB4 (40). In addition to H1 histamine receptor antagonistic activity, azelastine has an inhibitory effect on LTB4 production and LTB4 receptor antagonistic activity (22). The IL-31-induced scratching was inhibited by 5-lipoxygenase inhibitor zileuton and LTB4 receptor antagonist CMHVA. However, the cyclooxygenase inhibitor indomethacin did not show any effect on scratching. An intradermal injection of LTB4, but not prostaglandin E2, elicits scratching in mice (41). Taken together, these findings suggest that LTB4 is involved in IL-31-induced scratching.

5-Lipoxygenase is a key enzyme involved in the production of LTB4 (42). In the skin, 5-lipoxygenase is expressed mainly in the keratinocytes (43) and mast cells (44). IL-31 receptors are expressed in the keratinocytes, but not in the mast cells (4, 6, 45; Fig. 4 and Fig. S2A). In addition, an intradermal injection of IL-31 elicited scratching in mast cell-deficient mice and the normal littermates (Fig. S2B). Therefore, it is suggested that the mast cells are not involved in IL-31-induced LTB4 production. Our previous report showed that an intradermal injection of proteinase-activated receptor-2 (PAR2) agonist SLIGRL-NH2 (peptidergic agonist) induced LTB4 production (46). PAR2 is mainly expressed in the keratinocytes in the skin (25). These findings suggest that intradermal pruritogens including IL-31 act on the epidermal keratinocytes. In this study, IL-31 induced LTB4 production in the primary cultures of mouse keratinocytes via IL-31 receptor. Thus, it can be suggested that cutaneous keratinocytes are the main LTB4 producing cells. The mechanisms underlying IL-31-induced LTB4 production in keratinocytes remain unclear. IL-31 activates 3 signalling pathways: Jak/STAT pathway, PI3K/AKT pathway, and MAPK pathway (47). However, whether these pathways are involved in the production of LTB4 remain unknown. IL-31 also increases the intracellular Ca2+ concentration in keratinocytes (48). The increase in intracellular free Ca2+ ions activates phospholipase A2, a key enzyme for the production of arachidonic acid (49). Arachidonic acid, a substrate of 5-lipoxygenase, is involved in the production of LTB4. Thus, as a possible mechanism, IL-31-induced Ca2+ influx is involved in the production of LTB4.

LTB4 directly activates DRG neurones and induces scratching (41, 50). The LTB4 receptors, BLT1 and BLT2, have high and low binding affinities for LTB4, respectively (51). The LTB4 receptor antagonist CMHVA has a similar affinity for both receptors (51). The DRG neurones and skin in the normal mice expressed BLT1 receptor mRNA, but not BLT2 receptors (50). Most (81%) of the BLT1-immunoreactive DRG neurones are transient receptor potential vanilloid 1 (TRPV1, a marker of unmyelinated C-fibre neurones) (50). Therefore, it is suggested that BLT1 receptor is involved in LTB4-mediated IL31-induced scratching. A recent study showed that BLT2 antagonist, but not BLT1 receptor antagonist, inhibits LTB4-induced scratching (52). However, our previous study has been shown that an intradermal injection of BLT2 receptor agonist (12(S)-hydroxy-(5Z, 8E, 10E)-heptadecatrienoic acid and CAY10538) did not induce scratching in mice (data not shown). Therefore, further study on the role of BLT2 in scratching will be necessary.

In this study, IL-31 receptors were expressed in the DRG neurones/primary afferents, suggesting that IL-31 directly activates the primary afferents. Cevikbas et al. showed that IL-31 acts on IL-31RA(+)/TRPV1(+) neurones (13). In addition, IL-31-induced scratching is attenuated in TRPV1-deficient mice (13). Our previous reports have shown that TRPV1-deficiency or -desensitization in primary sensory neurones abolish the scratching induced by pruritogens (e.g. substance P and serotonin) (27, 28) and allergy (e.g. mosquito allergy) (53). Taken together, TRPV1-positive sensory neurones play an important role in the itch signalling induced by IL-31 and LTB4. IL-31 acts not only on IL-31RA(+)/TRPV1(+) neurones, but also on IL-31RA(+)/transient receptor channel potential cation channel ankyrin subtype 1 (TRPA1)(+) neurones (13). Fernandes et al. (52) showed that LTB4-induced scratching is attenuated in both TRPV1- and TRPA1-deficient mice. Therefore, it is suggested that TRPA1-positive sensory neurones also play an important role in IL-31- and LTB4-induced scratching.

In conclusion, IL-31 elicits itch not only through direct action on primary sensory neurones, but also by inducing LTB4 production in keratinocytes.

ACKNOWLEDGEMENTS

The authors thank Chugai Pharmaceutical Co., Ltd for providing the mouse IL-31. This work was supported by a research grant from Chugai Pharmaceutical Co. Ltd., and partially supported by the Platform Project for Supporting in Drug Discovery and Life Science Research Platform for Drug Discovery, Informatics, and Structural Life Science from the Japan Agency for Medical Research and Development (AMED).

REFERENCES
  1. Krause L, Shuster S. Mechanism of action of antipruritic drugs. Br Med J 1983; 28: 1199–1200.
    View article    Google Scholar
  2. Savin JA, Dow R, Harlow BJ, Massey H, Yee HF. The effect of a new non-sedative H1-receotor antagonist (LN2964) on the itching and scratching of patients with atopic eczema. Clin Exp Dermatol 1986; 11: 600–602.
    View article    Google Scholar
  3. Wahlgren CF, Hägermark Ö, Bergstorm R. The antipruritic effect of sedative and non-sedative antihistamine in atopic dermatitis. Br J Dermatol 1990; 22: 545–551.
    View article    Google Scholar
  4. Cornelissen C, Lüscher-Firzlaff J, Baron JM, Lüscher B. Signaling by IL-31 and functional consequences. Eur J Cell Biol 2012; 91: 552–566.
    View article    Google Scholar
  5. Dillon SR, Sprecher C, Hammond A, Bilsborough J, Rosenfeld-Franklin M, Presnell SR, et al. Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice. Nat Immunol 2004; 5: 752–760.
    View article    Google Scholar
  6. Rabenhorst A, Hartmann K. Interleukin-31: a novel diagnostic marker of allergic diseases. Curr Allergy Asthma Rep 2014; 14: 423.
    View article    Google Scholar
  7. Bilsborough J, Leung DY, Maurer M, Howell M, Boguniewicz M, Yao L, et al. IL-31 is associated with cutaneous lymphocyte antigen-positive skin homing T cells in patients with atopic dermatitis. J Allergy Clin Immunol 2006; 117: 418–425.
    View article    Google Scholar
  8. Sonkoly E, Muller A, Lauerma AI, Pivarcsi A, Soto H, Kemeny L, et al. IL-31: a new link between T cells and pruritus in atopic skin inflammation. J Allergy Clin Immunol 2006, 117: 411–417.
    View article    Google Scholar
  9. Takaoka A, Arai I, Sugimoto M, Yamaguchi A, Tanaka M, Nakaike S. Expression of IL-31 gene transcripts in NC/Nga mice with atopic dermatitis. Eur J Pharmacol 2005; 516: 180–181.
    View article    Google Scholar
  10. 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 2006; 15: 161–167.
    View article    Google Scholar
  11. 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: 669–671.
    View article    Google Scholar
  12. 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 2009; 18: 35–43.
    View article    Google Scholar
  13. Cevikbas F, Wang X, Akiyama T, Kempkes C, Savinko T, Antal A, et al. A sensory neuron-expressed IL-31 receptor mediates T helper cell-dependent itch: Involvement of TRPV1 and TRPA1. J Allergy Clin Immunol 2014; 133: 448–460.
    View article    Google Scholar
  14. Hawro T, Saluja R, Weller K, Altrichter S, Metz M, Maurer M. Interleukin-31 does not induce immediate itch in atopic dermatitis patients and healthy controls after skin challenge. Allergy 2014; 69: 113–117.
    View article    Google Scholar
  15. Andoh T, Katsube N, Maruyama M, Kuraishi Y. Involvement of leukotriene B4 in substance P-induced itch-associated response in mice. J Invest Dermatol 2001; 117: 1621–1626.
    View article    Google Scholar
  16. Andoh T, Yageta Y, Takeshima H, Kuraishi Y. Intradermal nociceptin elicits itch-associated responses through leukotriene B4 in mice. J Invest Dermatol 2004; 123: 196–201.
    View article    Google Scholar
  17. Andoh T, Nishikawa Y, Yamaguchi-Miyamoto T, Nojima H, Narumiya S, Kuraishi Y. Thromboxane A2 induces itch-associated responses through TP receptors in the skin in mice. J Invest Dermatol 2007; 127: 2042–2047.
    View article    Google Scholar
  18. Andoh T, Saito A, Kuraishi Y. Leukotriene B4 mediates sphingosylphosphorylcholine-induced itch-associated responses in mouse skin. J Invest Dermatol 2009; 129: 2854–2860.
    View article    Google Scholar
  19. Andoh T, Kuwazono T, Lee JB, Kuraishi Y. Gastrin-releasing peptide induces itch-related responses through mast cell degranulation in mice. Peptides 2011; 32: 2098–2103.
    View article    Google Scholar
  20. Shimizu K, Andoh T, Yoshihisa Y, Shimizu T. Histamine released from epidermal keratinocytes plays a role in ?-melanocyte-stimulating hormone-induced itching in mice. Am J Pathol 2015; 185: 3003–3010.
    View article    Google Scholar
  21. Sugimoto Y, Umakoshi K, Nojiri N, Kamei C. Effects of histamine H1 receptor antagonists on compound 48/80-induced scratching behavior in mice. Eur J Pharmacol 1998; 351: 1–5.
    View article    Google Scholar
  22. Andoh A, Kuraishi Y. Inhibitory effects of azelastine on substance P-induced itch-associated response in mice. Eur J Pharmacol 2002; 436: 235–239.
    View article    Google Scholar
  23. Carter GW, Young PR, Albert DH, Bouska J, Dyer R, Bell RL, et al. 5-lipoxygenase inhibitory activity of zileuton. J Pharmacol Exp Ther 1991; 256: 929–937.
    View article    Google Scholar
  24. Kishikawa K, Matsunaga N, Maruyama T, Seo R, Toda M, Miyamoto T, Kawasaki A. ONO-LB-457: a novel and orally active leukotriene B4 receptor antagonist. Adv Prostaglandin Thromboxane Leukot Res 1991; 21A: 407–410.
    View article    Google Scholar
  25. 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 2009; 109: 388–395.
    View article    Google Scholar
  26. Kuraishi Y, Nagasawa T, Hayashi K, Satoh M. Scratching behavior induced by pruritogenic but not algesiogenic agents in mice. Eur J Pharmacol 1995; 275: 229–233.
    View article    Google Scholar
  27. Andoh T, Nagasawa T, Satoh M, Kuraishi Y. Substance P induction of itch-associated response mediated by cutaneous NK1 tachykinin receptors in mice. J Pharmacol Exp Ther 1998; 286: 1140–1145.
    View article    Google Scholar
  28. Yamaguchi T, Nagasawa T, Satoh M, Kuraishi Y. Itch-associated response induced by intradermal serotonin through 5-HT2 receptors in mice. Neurosci Res 1999; 35: 77–83.
    View article    Google Scholar
  29. 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: 285–292.
    View article    Google Scholar
  30. Ohtsuka E, Kawai S, Ichikawa T, Nojima H, Kitagawa K, Shirai Y, et al. Roles of mast cells and histamine in mosquito bite-induced allergic itch-associated responses in mice. Jpn J Pharmacol 2001; 86: 97–105.
    View article    Google Scholar
  31. Yamaguchi T, Maekawa T, Nishikawa Y, Nojima H, Kaneko M, Kawakita T, et al. Characterization of itch-associated responses of NC mice with mite-induced chronic dermatitis. J Dermatol Sci 2001; 25: 20–28.
    View article    Google Scholar
  32. Bergasa NV, Alling DW, Talbot TL, Swain MG, Yurdaydin C, Turner ML, et al. Effects of naloxone infusions in patients with the pruritus of cholestasis. A double-blind, randomized, controlled trial. Ann Intern Med 1995; 123: 161–167.
    View article    Google Scholar
  33. Monroe EW. Efficacy and safety of nalmefene in patients with severe pruritus caused by chronic urticaria and atopic dermatitis. J Am Acad Dermatol 1989; 21: 135–136.
    View article    Google Scholar
  34. Akiyama T, Carstens MI, Carstens E. Differential itch- and pain-related behavioral responses and µ-opioid modulation in mice. Acta Derm Venereol 2010; 90: 575–581.
    View article    Google Scholar
  35. Gotoh Y, Andoh T, Kuraishi Y. Noradrenergic regulation of itch transmission in the spinal cord mediated by ?-adrenoceptors. Neuropharmacology 2011; 61: 825–831.
    View article    Google Scholar
  36. Thomas DA, Williams GM, Iwata K, Kenshalo DR Jr, Dubner R. Dubner Multiple effects of morphine on facial scratching in monkeys. Anesth Analg 1993; 77: 933–935.
    View article    Google Scholar
  37. Tohda C, Yamaguchi T, Kuraishi Y. Intracisternal injection of opioids induces itch-associated response through ?-opioid receptors in mice. Jpn J Pharmacol 1997; 74: 77–82.
    View article    Google Scholar
  38. Andoh T, Yageta Y, Konno M, Yamaguchi-Miyamoto T, Takahata H, Nojima H, et al. Evidence for separate involvement of different mu-opioid receptor subtypes in itch and analgesia induced by supraspinal action of opioids. J Pharmacol Sci 2008; 106: 667–670.
    View article    Google Scholar
  39. Kuraishi Y, Yageta Y, Konno M, Andoh T, Yamaguchi-Miyamoto T, Nojima H. Intracisternal, but not intrathecal, injection of naloxone inhibits cutaneous itch-related response in mice. Biol Pharm Bull 2008; 31: 2143–2145.
    View article    Google Scholar
  40. Yamamura H, Kohno SW, Ohata K, Koda A, Kawai M, Horiba M. Inhibition of anaphylactic chemical mediator release by tranilast. Arerugi 1987; 36: 937–942.
    View article    Google Scholar
  41. Andoh A, Kuraishi Y. Intradermal leukotriene B4, but not prostaglandin E2, induces itch-associated responses in mice. Eur J Pharmacol 1998; 353: 93–96.
    View article    Google Scholar
  42. Murphy RC, Gijón MA. Biosynthesis and metabolism of leukotrienes. Biochem J 2007; 405: 379–395.
    View article    Google Scholar
  43. 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 2001; 20: 894–898.
    View article    Google Scholar
  44. Lin TY, London CA. Characterization and modulation of canine mast cell derived eicosanoids. Vet Immunol Immunopathol 2010; 135: 118–127.
    View article    Google Scholar
  45. Kasraie S, Niebuhr M, Baumert K, Werfel T. Functional effects of interleukin 31 in human primary keratinocytes. Allergy 2011; 66: 845–852.
    View article    Google Scholar
  46. 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: 206–213.
    View article    Google Scholar
  47. Zhang Q, Putheti P, Zhou Q, Liu Q, Gao W. Structures and biological functions of IL-31 and IL-31 receptors. Cytokine Growth Factor Rev 2008; 19: 347–356.
    View article    Google Scholar
  48. Lee CH, Hong CH, Yu WT, Chuang HY, Huang SK, Chen GS, et al. Mechanistic correlations between two itch biomarkers, cytokine interleukin-31 and neuropeptide ?-endorphin, via STAT3/calcium axis in atopic dermatitis. Br J Dermatol 2012; 167: 794–803.
    View article    Google Scholar
  49. Clark JD, Milona N, Knopf JL. Purification of a 110-kilodalton cytosolic phospholipase A2 from the human monocytic cell line U937. Proc Natl Acad Sci USA 1990; 87: 7708–7712.
    View article    Google Scholar
  50. Andoh T, Kuraishi Y. Expression of BLT1 leukotriene B4 receptor on the dorsal root ganglion neurons in mice. Brain Res Mol Brain Res 2005; 137: 263–266.
    View article    Google Scholar
  51. Yokomizo T, Izumi T, Shimizu T. Leukotriene B4: metabolism and signal transduction. Arch Biochem Biophys 2001; 385: 231–241.
    View article    Google Scholar
  52. Fernandes ES, Vong CT, Quek S, Cheong J, Awal S, Gentry C, et al. 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 2013; 27: 1664–1673.
    View article    Google Scholar
  53. Nakano T, Andoh T, Sasaki A, Nojima H, Kuraishi Y. Different roles of capsaicin-sensitive and H1 histamine receptor-expressing sensory neurons in itch of mosquito allergy in mice. Acta Derm Venereol 2008; 88: 449–454.
    View article    Google Scholar
Supplementary content
Figure S1
Figure S2