You are on page 1of 14

PERSPECTIVE

Neurophysiological, Neuroimmunological, and


Neuroendocrine Basis of Pruritus
Martin Steinhoff1, John Bienenstock2, Martin Schmelz3, Marcus Maurer4, Ed Wei5 and Tamás Bı́ró6

Pruritus (itch) can be defined as an unpleasant cutaneous sensation associated with the immediate desire to
scratch. Recent findings have identified potential classes of endogenous ‘‘itch mediators’’ and establish a
modern concept for the pathophysiology of pruritus. First, there in no universal peripheral itch mediator, but
disease-specific sets of involved mediators. Second, numerous mediators of skin cells can activate and sensitize
pruritic nerve endings, and even modulate their growth. Our knowledge of itch processing in the spinal cord
and the involved centers in the central nervous system is rapidly growing. This review summarizes the current
information about the significance of neuron–skin interactions, ion channels, neuropeptides, proteases,
cannabinoids, opioids, kinins, cytokines, biogenic amines, neurotransmitters, and their receptors in the
pathobiology of pruritus. A deeper understanding of these circuits is required for the development of novel
antipruritic strategies.
Journal of Investigative Dermatology (2006) 126, 1705–1718. doi:10.1038/sj.jid.5700231

Introduction drug application, stress, and others. volved with a different impact among
Pruritus (itch) can be defined as an Current evidence clearly indicates the various pruritic diseases (e.g. atopic
unpleasant cutaneous sensation asso- existence of an interactive network dermatitis, urticaria, renal, and chole-
ciated with the immediate desire to between the skin and the peripheral static pruritus). However, the impor-
scratch. Teleologically, pruritus may as well as the central nervous system to tance of the central nervous ‘‘itch
be interpreted as part of the body’s regulate and respond to pruritic stimuli centers’’ under physiological and
defense mechanism by which we (Figure 1). It became evident that pathophysiological conditions is still at
dispose of potential dangerous organ- specified sensory nerves and their the beginning of being understood. This
isms or stimuli. Chronic or intense receptors are crucially involved in the review summarizes the current knowl-
scratching leads to the development pathophysiology of pruritus. Thus, prur- edge about the significance of various
of skin lesions and the release of itus is not merely a submodality of mediators and receptors in the patho-
inflammatory mediators that potentially pain, but an individual sensation of our physiology of pruritus. Owing to limited
induce or aggravate pruritus resulting sensory nerve system. space, several important papers could
in reinforcement of scratching. This Recent observations clearly expand not be cited. A prolonged reference
‘‘itch–scratch’’ cycle, unfortunately, is our knowledge of potential classes of version can be found in the supplement.
frequently resistant to topical and endogenous ‘‘itch mediators’’ (re-
systemic therapy (Figure 1, Table S1). viewed in Stander and Steinhoff, Neurophysiology of itch
Pruritus is one of the most common 2002; Yosipovitch et al., 2003) and Primary afferent pruriceptive neurons.
symptoms in dermatology and general established a modern concept of the According to the intensity hypothesis
medicine. It can be initiated during pathophysiology of pruritus (Table 1). of itch, low-level activation of un-
inflammation, cancer, metabolic dis- In other words, different peripheral itch specific nociceptors would induce
eases, infection, psychiatric diseases, mediators and receptors may be in- pruritus, whereas higher discharge fre-

1
Department of Dermatology, IZKF Münster, Ludwig Boltzmann-Institute for Immunobiology of the Skin, University Hospital Muenster, Muenster, Germany;
2
Department of Pathology and Molecular Medicine, The Brain-Body Institute, McMaster University, St Joseph’s Healthcare Hamilton, Ontario, Canada;
3
Department of Anesthesiology and Critical Care Medicine, Faculty of Clinical Medicine Mannheim, University of Heidelberg, Mannheim, Germany;
4
Department of Dermatology and Allergy, Charité University of Berlin, Berlin, Germany; 5School of Public Health, University of California, Berkeley, California,
USA and 6Department of Physiology, Medical and Health Science Center, Research Center for Molecular Medicine, University of Debrecen,
Debrecen, Hungary
Correspondence: Dr Martin Steinhoff, Department of Dermatology, IZKF Münster, Ludwig Boltzmann-Institute for Immunobiology of the Skin, University
Hospital Münster, Von-Esmarch-Straße 58, Münster D-48149, Germany. E-mail: msteinho@uni-muenster.de
Abbreviations: AD, Alzheimer disease; BDNF, brain-derived neurotrophic factor; CB, cannabinoid; CGRP, calcitonin gene-related peptide;
CRH, corticotropin-releasing hormone; ET-1, endothelin-1; MC, mast cell; NGF, nerve growth factor; PAR, proteinase-activated receptor; SP, substance P;
TNF-a, tumor necrosis factor alpha; TRP, transient receptor potential; VIP, vasoactive intestinal polypeptide
Received 19 July 2005; revised 30 December 2005; accepted 6 January 2006

& 2006 The Society for Investigative Dermatology www.jidonline.org 1705


M Steinhoff et al.
Basis of Pruritus

Langerhans cell histamine. They comprise about 20%


of the mechano-heat-insensitive class
of C-fibers.
Brain The pruritogenic potency of inflam-
Epidermis matory mediators is characterized by
Immune cells their ability to activate histamine-posi-
tive mechano-insensitive C-nocicep-
tors. However, concomitant activation
of mechano-sensitive and mechano-
insensitive histamine-negative nocicep-
tors will decrease the itch. Therefore,
Blood vessel
itch sensation is apparently based on
Protons, kinins,
proteases, amines, both, activity in the pruriceptors and
prostanoids, cytokines
Neuropetides absence of activity in the pain-mediat-
ing nociceptors.
DRG
Additional primary afferents involved
in producing itch. Histamine-sensitive
C-fibers have been found among the
mechano-insensitive afferent C-fibers.
Spinal cord
They are characterized by very high
Figure 1. Pathophysiology of pruritus. Exogenous as well as endogenous factors released by immune transcutaneous electrical thresholds and
cells, epithelial cells, or endothelial cells induce activation of signal cascades from the periphery via are involved in the generation of the
dorsal root ganglia (DRG) and spinal cord to the central nervous system (CNS). Activation of specific
axon reflex erythema (Schmelz et al.,
areas in the CNS results in perception of itch leading to a scratch response. By a direct axon reflex
mechanism, sensory nerve endings release neuropeptides (Table 1), which may aggravate the itch 2000a). Recently, focal electrical stimu-
response by stimulating release of pruritic mediators from MCs, endothelial cells, and epithelial cells. lation with low intensity but high fre-
Whether neuropeptides may also trigger the release of antipruritic agents from these cells is only poorly quency has been shown in humans to
understood. induce itch (Ikoma et al., 2005). Inter-
estingly, the electrically induced itch
was not accompanied by an axon reflex
quencies would provoke pain. How- by a particular low conduction velo- erythema, suggesting that the activated
ever, application of low concentrations city, large innervation territories, me- fibers do not belong to the histamine-
of algogens generally does not cause chanical unresponsiveness, and high sensitive pruriceptors described above.
itch, just less intense pain. Further, transcutaneous electrical thresholds
intraneural electrical microstimulation (Schmelz et al., 2003). In line with the Specific spinal pruriceptive neurons.
of human afferent nerves induces either large innervation territories of these Using the cat spinal cord, a specific
pain or, less commonly, pruritus. In- fibers, two-point discrimination for class of dorsal horn neurons projecting
creasing the stimulation frequency in- histamine-induced itch is poor (15 cm to the thalamus has been demonstrated,
creases the intensity of pain or itch, but in the upper arm). which respond strongly to histamine
no switch from pruritus to pain is The relative prevalence of the dif- administered to the skin by iontophor-
observed. Therefore, a dedicated neuro- ferent C-fiber types has been estimated esis (Andrew and Craig, 2001). The
nal system for encoding itch could be from recordings in the superficial per- time course of these responses was
predicted. C-fibers, responding to hista- oneal nerve (Schmidt et al., 1997). similar to that of itch in humans and
mine application in parallel to the itch About 80% are polymodal nociceptors, matched the responses of the periph-
ratings of subjects, have been discovered which respond to mechanical, heat, eral C-itch fibers. These units were also
among the group on mechano-insensi- and chemical stimuli. The remaining unresponsive to mechanical stimula-
tive C-afferents confirming that there is a 20% do not respond to mechanical tion and differed from the histamine-
specific pathway for itch (Figure 1, stimulation. These fibers are mainly insensitive nociceptive units in lamina I
Tables 1, 2, and S1). In contrast, the ‘‘mechano-insensitive nociceptors’’, of the spinal cord. In addition, their
most common type of C-fibers, ‘‘poly- which are activated by chemical sti- axons had a lower conduction velocity
modal’’ nociceptors are either insensi- muli, and can be sensitized to mechan- and anatomically distinct projections to
tive to histamine or only weakly ical stimulation in the presence of the thalamus. Thus, the combination of
activated by it (Schmelz et al., 2003). inflammation (Schmelz et al., 2000b). dedicated peripheral and central neu-
Thus, this fiber subtype cannot account This latter characteristic led to the rons with a unique response pattern to
for the prolonged itch induced by the name ‘‘sleeping nociceptor’’. Among pruritogenic mediators and anatomi-
intradermal application of histamine. the mechano-insensitive afferent C- cally distinct projections to the thala-
The histamine-sensitive or ‘‘itch’’ fibers, there is a subset of units, which mus provides the basis for a specific
fibers or pruriceptors are characterized have a strong and sustained response to neuronal pathway for itch.

1706 Journal of Investigative Dermatology (2006), Volume 126


M Steinhoff et al.
Basis of Pruritus

Table 1. Involvement of neuromediators in pruritus, pain, and burning (selected)


Neuromediator Receptor Source Target cells/function References

Acetylcholine Nicotinergic and muscarinergic Autonomic cholinergic Mediates itch in atopic Grando (1997); Schmelz et al.
Acetylcholine receptors nerves, keratinocytes, dermatitis patients; M3R (2000a, b)
lymphocytes, melanocytes probably involved in itch

Catecholamines, Adrenergic receptors Autonomic adrenergic Pain transmission Haüstein (1990); Hundley
noradrenaline nerves, keratinocytes, and Yosipovitch (2004)
melanocytes

Substance P Tachykinin (neurokinin) Sensory nerve fibers Low (physiologically relevant) Janiszewski et al. (1994);
receptor concentrations: priming of MCs; Okabe et al. (2000); Scholzen
release of TNF-a, histamine, and Luger (2004)
leukotriene B4, prostaglandin
D2 from MCs (agents involved
in pruritus and burning)

Neurokinin A Tachykinin (neurokinin) Sensory nerve fibers Up-regulation of keratinocyte Burbach et al. (2001)
receptor nerve growth factor expression

VIP Vasoactive intestinal peptide/ Sensory nerve fibers, Histamine release from MCs; Steinhoff et al. (2003b and
pituitary adenylate cyclase Merkel cells allodynia (no allodynia in AD!) references therein)
activating peptide (VPAC) intensifies acetylcholine
receptors induced itch in atopic dermatitis
patients (together with
acetylcholine)

Pituitary adenylate VPAC receptors Autonomic and sensory Involved in flush, vasodilatation, Delgado et al. (2001 and
cyclase-activating nerve fibers, lymphocytes, pain, neurodegeneration; references therein)
polypeptide dermal endothelial cells pruritus? Induces release of
histamine from MCs

CGRP CGRP receptors Sensory nerve fibers Pain transmission, prolongation Steinhoff et al. (2003b and
of itch latency following SP references therein); Brain and
injection (inhibitory effect on Grant (2004); Brain and
itching); sensitization of Williams (1988)
receptive endings; increase of
CGRP fibers in itchy skin
diseases

MSH Melanocortin receptors Melanocytes, keratinocytes, Releases histamine from MCs; Steinhoff et al. (2003b and
endothelial cells, pruritus references therein)
Langerhans cells, MCs,
fibroblasts, monocytes
CRH CRH-R1, -2 CRH-R1: keratinocytes, Release of histamine, cytokines, Skofitsch et al. (1995);
MCs TNF-a, vascular endothelial Slominski et al. (1995);
growth factor from MCs Theoharides et al. (1998);
CRH-R2: bone-marrow CRH-like-immunoreactivity on Slominski and Wortsman
MCs sensory nerves (rat) (2000); Venihaki et al. (2001);
Lytinas et al. (2003);
Kempuraj et al. (2004); Cao
et al. (2005)

Opioids m-, k-, d-opioid receptors (partly Nerves, keratinocytes Antipruritic effect of m-opioid Onigbogi et al. (2000);
receptor-independent cell antagonists (central effect) and Andrew and Craig (2001);
activation) k-opioid agonists (spinal cord Stander et al. (2003); Blunk
level); opioid agonists do not et al. (2004); Bigliardi-Qi
provoke itch upon injection or et al. (2005)
intradermal application;

Table 1 continued on following page

www.jidonline.org 1707
M Steinhoff et al.
Basis of Pruritus

Table 1. continued
Neuromediator Receptor Source Target cells/function References

m-opioid receptor upregulation


in atopic dermatitis
Endocannabinoids CB receptors Nerves, keratinocytes Antipruritic in the periphery Maccarrone et al. (2003);
Ibrahim et al. (2005); Stander
et al. (2005)

Endothelins Endothelin receptors A, B Endothelium, MCs Burning itch, degraded by Katugampola et al. (2000);
chymase via ETA receptor Maurer et al. (2004)
activation

Kinins Bradykinin receptors Endothelial cells, Bradykinin induces pain over Hayashi and Majima (1999)
immunocytes pruritus; bradykinin B2 receptor
antagonists reduce itch

Kallikreins, proteases Partly by proteinase-activated Keratinocytes, endothelial Massive itch behavior in mice Steinhoff et al. (2000);
receptors (PARs, tryptic cells, MCs, platelets overexpressing epidermal Steinhoff et al. (2003a, b);
enzymes) kallikrein 7; potential role of Lundequist et al. (2004); Ny
other kallikreins; chymase and Egelrud (2004); Steinhoff
degrades pruritic and antipruritic et al. (2005)
peptides; tryptase induces
inflammation and itch by a
neurogenic mechanism via PAR2

Neurotrophins (NGF, Specific receptors trk A: NGF; NGF: keratinocytes, MCs, NGF levels enhanced in atopic Kanda and Watanabe (2003);
NT-4, BDNF) trk B: NT-4, BDNF; trk C: NT-3 fibroblasts, eosinophils. dermatitis and epidermal barrier Kimata (2003); Groneberg
BDNF: eosinophils disruption; induces tryptase et al. (2005); Raap et al.
release from MCs; inducible by (2005)
histamine; trk A: enhanced in
keratinocytes during
inflammation; NT-4: enhanced
in AD; induces sprouting.
BDNF: increases eosinophil
chemotaxis levels in AD, and
inhibits apoptosis. Sensitization
of receptive nerve endings,
upregulation of neuronal
neuropeptides, and TPV1
1
VPAC, Vasoactive intestinal peptide/pituitary adenylate cyclase activating peptide.

Central itch processing. The itch-selec- anterior cingulate cortex, supplemen- pain have been described. In contrast
tive units in lamina I of the spinal cord tary motor area, and inferior parietal to pain, itch seems to be characterized
form a distinct pathway projecting to lobe predominantly in the left hemi- by a lack of secondary somatosensory
the posterior part of the ventromedial sphere (Mochizuki et al., 2003 and cortex activation on the parietal oper-
thalamic nucleus, which projects to the references therein). The significant co- culum and by left hemispheric dom-
dorsal insular cortex, a region that has activation of motor areas supports the inance (Drzezga et al., 2001). In
been shown to be involved in a variety familiar observation that itch is inher- addition, the periaqueductal gray mat-
of interoceptive modalities like thermo- ently linked to a desire to scratch. ter was activated only when painful
ception, visceral sensations, thirst, and The multiple activated sites in the brain and itching stimuli were simulta-
hunger. The supraspinal processing after itch induction argue against the neously applied. This activation was
of itch and its corresponding scratch existence of a single itch center and combined with reduced activity in the
response have recently been investi- reflect the multidimensionality of itch. anterior cingulate, dorsolateral prefron-
gated in man by functional positron Thus, a broad overlap of activated tal cortex, and parietal cortex, suggest-
emission tomography (Darsow et al., brain areas is evident for pain and itch ing that the periaqueductal gray matter
2000). Induction of itch by intradermal (Drzezga et al., 2001 and references might be involved in the central inhibi-
histamine injections and histamine therein). However, subtle differences in tion of itch by pain (Mochizuki et al.,
skin-prick-induced co-activation of the the activation pattern between itch and 2003).

1708 Journal of Investigative Dermatology (2006), Volume 126


M Steinhoff et al.
Basis of Pruritus

Table 2. TRP channels with potential role in the pathogenesis and therapy of itch and pain
Receptor Expression in the skin Activator/sensitizer Target cells/functions References

TRPV1 Sensory neurons Exogenous vanilloids (capsaicin, Responds to noxious heat, capsaicin, Biro et al. (1997); Caterina et al.
resiniferatoxin) endovanilloids, anandamide, (1997); Caterina and Julius
prostaglandins, desensitization (2001); Stander et al. (2003);
MCs T4431C Depletes neuropeptides from sensory Yosipovitch et al. (2003); Lazar
neurons et al. (2004); Biro et al. (2005);
Bodo et al. (2005); Xu et al.
Epidermal keratinocytes Acidosis Long-term antipruritic effect: suspends (2005)
interplay between sensory neurons and
MCs
Hair follicle keratinocytes Endovanilloids Modulates dendritic cell function
Langerhans cells Activated by histamine, Affects epidermal and hair follicle
bradykinin, eicosanoids, ATP, proliferation, differentiation, apoptosis
prostaglandins, neurotrophins, etc and cytokine release
Smooth muscle Camphor desensitizes TRPV1 via Increased expression in epidermal
protein kinase C keratinocytes of prurigo nodularis
patients
Sebocytes High Ca2+ permeability
Conductance B100 pS

TRPV2 Sensory neurons T4531C TRPV3 modulates TRPV1-mediated pain Stokes et al. (2004)
and itch
MCs

TRPV3 Sensory neurons T4311C TRPV3 knockouts deficit in response to Peier et al. (2002b); Moqrich
(interacting with TRPV1) innocuous (mostly TRPV3 range) and et al. (2005)
noxious heat (rather TRPV1 range)
Epidermal keratinocytes TRPV3 and TRPV4 may modulate
epidermal osmotic pressure

TRPV4 Sensory neurons T4251C TRPV4 is activated by eicosanoids Watanabe et al. (2003); Stokes
Epidermal keratinocytes Eicosanoids TRPV2 and TRPV4 stimulated by MCs in et al. (2004)
a protein kinase A-dependent manner
MCs

TRPM8 Sensory neurons T: 8–281C Antipruritic, anti-irritative (induce Wei and Seid (1983); Peier et al.
sensation of cooling) (2002a)
Menthol, icilin, and their analogs

TRPA1 Sensory neurons T: below 171C Noxious cold sensation Clapham (2003); Story et al.
Activated by mustard and Mechano-transduction of hair cells (2003), Bandell et al. (2004);
cinnamon oils, raw garlic, Tominaga and Caterina (2004);
sensitized by camphor, McKemy (2005); McPherson
cannabinoids, bradykinin et al. (2005); Reid (2005)
(coupled to bradykinin-2R)
Signaling via phospholipase C, —
protein kinase C

Skin–nerve interactions and itch neuronal network in the skin. It choline), certain neuropeptides (e.g.
The skin is highly innervated by pri- is further discussed that in the facial substance P (SP), calcitonin gene-
mary sensory nerves, postganglionic skin some parasympathetic nerves exist related peptide (CGRP), opioids,
cholinergic parasympathetic, and post- associated with flushing. These neurons cannabinoids (CB)), and certain neuro-
ganglionic sympathetic nerves, result- utilize, among others, classical neuro- trophins (nerve growth factor (NGF),
ing in a complex afferent/efferent transmitters (catecholamines, acetyl- neurotrophin-4).

www.jidonline.org 1709
M Steinhoff et al.
Basis of Pruritus

Certain subtypes of unmyelinated C- dermis influence skin-derived pruritus mediators such as endovanilloids,
fibers also project into the epidermis (Figure S1, Tables 1 and 2). endorphins, neuropeptides, proteases,
(Kennedy et al., 1994). Thus, neurome- and cytokines (Figure 2, Table 1). With
diators may directly communicate with Role of the epidermis in itch sensation. respect to the role of dry skin in
keratinocytes or Langerhans cells, and Itch sensations can be induced owing pruritus, Nojima et al. (2004) have
vice versa. For example, CBs stimulate to damage to the skin barrier function shown in a murine model that dry skin
the release of b-endorphin from kerati- in the dry skin (xerosis) and atopic induces enhanced c-fos expression,
nocytes, thereby activating sensory eczema as well as papulosquamous which reflects activation of spinal cord
neurons resulting in the modulation of diseases such as psoriasis and lichen neurons. SP as well as CRF decrease
pain (Ibrahim et al., 2005). From that it planus. In this context, itching most the electric potential in keratinocytes as
is evident that dysregulation of skin probably is not related to mast cells well as did skin barrier disruption.
function (pH changes, trauma, dis- (MCs). The epidermis itself is inner- Thus, ion channels including the
rupted barrier function, inflammation, vated by sensory nerve endings anato- voltage-gated, ATP-gated (Inoue et al.,
infection, UV light) can directly or mically associated with keratinocytes 2005), and transient receptor potential
indirectly stimulate sensory nerve and Langerhans cells. Recent studies (TRP)V-gated may be directly involved
endings, thereby inducing pruritus have demonstrated that – upon stimula- in the transmission of pruritus via
(Figure 2). Therefore, cell–nerve inter- tion – keratinocytes are capable of keratinocyte activation in the dry
actions in the epidermis as well as the releasing pruritic as well as antipruritic skin. Keratinocytes express a variety

Inflammation
 Vasodilatation
 Plasma extravasation
 Recruitment leukocytes

Itch CGRP R
(NKIR) etc.
4
Brain 3

SP
CGRP
2 Endogenous/
exogenous
proteinases

Dorsal root PAR2


ganglia

5
8 PAR2
9 Inflammatory
Central Peripheral cells
ending ending

Primary afferent
sensory neuron 1

Tryptase
Spinal cord − 6

CGRP
7
− +
Degradation Vasodilatation

Figure 2. PAR2 mediates protease-induced inflammation and pruritus by a neurogenic mechanism (modified from Steinhoff et al. (2000, 2003b)). (1) Tryptase
released from degranulated MCs activates PAR2 at the plasma membrane of sensory nerve endings. (2) Activation of PAR2 by tryptase, trypsins, various
kallikreins, probably exogenous proteases (bacteria, house-dust mite) stimulates the release of CGRP and the tachykinins SP and neurokinin A (NKA) from
sensory nerve endings. (3) CGRP interacts with the CGRP1 receptor to induce arteriolar dilation and hyperemia. (4) SP interacts with the neurokinin-1 receptor
on endothelial cells of post-capillary venules to cause gap formation and plasma extravasation. Hyperemia and plasma extravasation cause edema during
inflammation. (5) SP may stimulate degranulation of MCs, providing a positive feedback mechanism. (6) Tryptase degrades CGRP and terminates its effects. (7)
CGRP inhibits SP degradation by neutral endopeptidase and also enhances SP release, thereby amplifying the effects. (8) Mediators from MCs and other
inflammatory cells stimulate the release of vasoactive peptides from sensory nerves and also sensitize nerves. (9) At the spinal cord level, PAR2-induced
intracellular Ca mobilization leads to the release of CGRP (and SP) from central nerve endings, thereby activating neurokinin-1 receptor and CGRPRs to transit
itch responses to the central nervous system.

1710 Journal of Investigative Dermatology (2006), Volume 126


M Steinhoff et al.
Basis of Pruritus

of receptors (neuropeptide receptors, in host defense against bacterial infec- converting enzyme and by reducing
neurotrophin receptors (for NGF, neuro- tions (Maurer, 2002) and cardiovascu- the numbers of their binding sites.
trophin-4), CB receptors, proteinase- lar diseases, and is known to induce a Finally, several independent studies
activated receptor-2 (PAR2), and the burning and painful sensation as well using MC-deficient animals have
TRPV1 ion channel, all of which have as itch upon injection into human skin shown that the frequency and/or dura-
been demonstrated to be involved in (Katugampola et al., 2000). Interest- tion of scratching in response to itch-
transmitting itch sensations. Interest- ingly, degradation of ET-1 by MC inducing agents are not reduced in the
ingly, different trigger factors are chymase requires prior activation, absence of MCs (Hayashi et al., 2001).
capable of stimulating the release of which is, at least in part, provided for On the contrary, pruritus appears to be
pruritogenic or antipruritogenic factors by ET-1 itself by binding to endothelin- similar to or even stronger in geneti-
from keratinocytes. For example, pros- A receptors on MCs. In other words, cally MC-deficient KitW/KitW-v mice
taglandin E2 induces the release of when ET-1 induces damage (e.g. prur- than in normal mice. For example,
neurotrophin-4 from keratinocytes itus), it also starts an ‘‘automatic self- KitW/KitW-v mice exhibit a 20% in-
(Kanda et al., 2005). Both, the hista- destruction program’’, that is, endo- crease in scratching behavior induced
mine H1 and H2 receptors are ex- thelin-A-dependent release of ET-1- by histamine or SP as compared to
pressed by keratinocytes and may be degrading chymase from MCs. This normal Kit þ / þ mice (Hossen et al.,
involved in epidermal barrier dysfunc- novel MC function may be relevant 2003 and references therein), and both
tion (Ashida et al., 2001). In addition, for many skin conditions, in which the time and the incidence of scratch-
nociceptin may stimulate the release of neuropeptides are implicated such as ing responses induced by intracuta-
leukotriene B4 from keratinocytes via atopic dermatitis or psoriasis. neous injections of compound 48/80
opioid receptor-like 1 receptor (Andoh Why is the degradation of neuro- exceeded those of Kit þ / þ mice
et al., 2004). Moreover, Miyamoto peptides by MC proteases a likely (Inagaki et al., 2002).
et al. (2002) have shown that antago- mechanism of MC-mediated control Taken together, these findings sug-
nists of opioid receptors suppressed of pruritus? MCs undergo degranulation gest that MCs can be involved both
scratching behavior in a mouse model and protease release in response to in the activation as well as termination
of dry skin, most probably by a central several neuromediators including SP of pruritus. The use of novel mouse
effect. In summary, dry skin induces and CGRP (Bienenstock et al., 1987). models including techniques to surgi-
itch by stimulating the release of However, processing of peptide med- cally denervate defined skin areas
various pruritogenic mediators from iators by MC proteases does not require as well as mice deficient for neuro-
keratinocytes. Less, however, is known prior activation of MCs as some pro- peptides, neuropeptide-degrading pro-
about the potential release of endogen- teases are constitutively expressed on teases, and/or MCs will help to clarify
ous antipruritogenic factors from kera- cell surfaces, thus allowing for contin- these important questions.
tinocytes. uous regulation of neuropeptide levels
in the skin even in the absence of
MC-degranulating signals (Hagermark, NEUROPEPTIDES, NEUROTROPHINS
The MC–nerve connection: functions 1974). In addition, MCs may also AND RECEPTORS
beyond itch induction? The depicted contribute to the termination of neuro- Tachykinins: ‘‘the MC connection’’
MC–nerve interactions may not only genic inflammation and pruritus by One of the best – albeit probably not
promote but also terminate pruritus. regulating tissue expression of neutral most important – neuromediators stu-
This yet unproven hypothesis is sup- endopeptidases and angiotensin-con- died thus far is SP (Table 1). Upon
ported by several independent lines of verting enzyme, two zinc metallo- stimulation by exogenous or endogen-
evidence (Figure S1). First, MCs express proteinases that effectively control ous trigger factors, the above intro-
and release large amounts of proteases neuropeptide skin levels. Levels of duced slow-conducting histamine-
(tryptase, chymase, carboxypeptidase angiotensin-converting enzyme and sensitive C-fibers are not only capable
A, and cathepsins), which have been neutral endopeptidase in human skin of transporting the itch signal to the
shown to degrade/inactivate prurito- MCs were found to be increased in central nervous system (orthodromic)
genic peptides. For example, MC response to various proinflammatory but also release neuropeptides such as
enzymes have been implicated to mediators that are produced by MCs. SP and CGRP (antidromic). Because of
downregulate axon reflex-mediated Interestingly, those proinflammatory their anatomical association to cuta-
neurogenic inflammation in skin and mediators, particularly tumor necrosis neous nerves, MCs and their released
other tissues by cleaving SP, CGRP, factor alpha (TNF-a), are also known to products appear to play an important
and vasoactive intestinal polypeptide downregulate the expression of recep- role in the pathophysiology of itch
(VIP). Also, MC chymases efficiently tors for SP and corticotropin-releasing and inflammation (Steinhoff et al.,
degrade and terminate the activity of hormone (CRH). Thus, MC-derived 2003a, b). Independent of neurokinin-1
endothelin-1 (ET-1) (Maurer, 2002). mediators could also limit pruritus at receptor, intradermal application of SP
ET-1 (a neuropeptide that is also sites of inflammation by increasing the releases histamine from MCs, which
produced by, among others, endothe- clearance of neuropeptides via neutral acts as a pruritogen (Thomsen et al.,
lial cells and MCs) has been implicated endopeptidases and/or angiotensin- 2002). Moreover, SP enhances intra-

www.jidonline.org 1711
M Steinhoff et al.
Basis of Pruritus

dermal concentrations of nitric oxide, exert their effects by triggering opioid Central effects. It is common experi-
which may enhance SP-induced prur- receptors (m, d, k, and orphan recep- ence that itch sensation can be reduced
itus (Andoh and Kuraishi, 2003). Spe- tors) (Table 1). by painful sensations caused by
cific neurokinin-1 receptor effects of SP scratching or by various painful (ther-
on MCs include TNF upregulation, mal, mechanical, chemical) stimuli.
which in turn can sensitize pruriceptive Peripheral effects. With respect to itch, Cutaneous field stimulation has also
afferent endings. intradermally injected opioids activate been successfully used to inhibit hista-
Therefore, it is apparent that various MCs by a non-receptor-mediated mec- mine-induced itch for several hours in
neuropeptides may exert indirect ef- hanism. In contrast to morphine, the a relatively large (20 cm in diameter)
fects on sensory nerves by triggering highly potent m-opioid agonist, fentanyl area around a stimulated site suggest-
the release of pruritic agents from does not provoke any MC degranula- ing a central mode of action. Not only
various target cells such as MCs (hista- tion, even at concentrations having is itch inhibited by enhanced input of
mine, tryptase, chymase, TNF-a), en- m-agonistic effects exceeding those of pain stimuli, but vice versa inhibition of
dothelial cells (kinins, endothelin), morphine. Thus, one can conclude that pain processing may reduce its inhibi-
keratinocytes (prostanoids, NGF), and morphine-induced MC degranulation is tory effect, and thus enhance itch. This
immune cells (cytokines). This me- not mediated by m-opioid receptors. As phenomenon is particularly relevant to
chanism is probably the main cause high local concentrations of opioids are spinally administered m-opioid receptor
for itch in atopic dermatitis, psoriasis, required to degranulate MCs, itch agonists, which induce segmental an-
and prurigo nodularis (Ständer et al., induced by systemic administration in algesia often combined with segmental
2003). therapeutic opioids doses is probably pruritus (Onigbogi et al., 2000). This
In the past, it was believed that MC not owing to peripheral MC degranula- mechanism might well account for the
activation was all-or-nothing, with IgE tion, but to central mechanisms. There antipruritic effect of m-opioid antago-
cross-linking inducing the functional is also no evidence for direct neuronal nists (nalmefene, naloxone, and nal-
consequences of allergic reactions and excitation by peripherally applied trexone) observed in experimentally
anaphylaxis. However, the activity of opioids as potent opioid agonists, even evoked histamine-induced itch as
MCs in health and disease is clearly at high concentrations, do not provoke well as pruritus in different dermatoses
much more complex. Using the patch- itch (Blunk et al., 2004 and references (Metze et al., 1999; Heyer et al., 2002).
clamp technique, Janiszewski et al. therin). Thus, the impact of the ob- This phenomenon is particularly rele-
(1994) reported that MCs do not served increased expression of m-opioid vant to spinally administered m-opioid
respond electrophysiologically to very receptors in atopic dermatitis (Bigliardi- receptor agonists, which induce seg-
low (picomolar) concentrations of SP, Qi et al., 2005) is unclear. However, mental analgesia combined with a
but activation and delayed degranula- according to the inhibitory effects of naloxone-sensitive segmental pruritus
tion occurred after a second exposure. neuronal m-opioid receptors, it would in about half of the patients.
Therefore, MCs can be primed when be expected that their increased ex- Interestingly, while m-opioid recep-
exposed to physiologically relevant pression act antipruritic. Interestingly, tor antagonists significantly diminish
low concentrations of SP, and lower peripherally applied CBs suppress his- itch, in animal experiments, k-opioid
their thresholds to subsequent activa- tamine-induced pruritus, with inhibi- antagonists enhanced itch (Kamei
tion. Thus, if MCs are indeed primed tory CB receptors CB1 and CB2 being and Nagase, 2001). In line with these
by SP exposure, it would enable a found on skin nerves (Stander et al., results, the k-opioid agonist nalbuphine
subthreshold stimulus to initiate MC 2005). However, the inhibitory effects as well as the new developed k-opioid
activation (Figure S1). Furthermore, of peripheral CBs have been suggested receptor agonist, TRK-820, have been
secretion can occur without the to be – at least in part – mediated shown to reduce pruritus. As m- as well
evidence of degranulation, and even by a secondary release of endogenous as d-opioid receptors are expressed
molecules stored within the same opioids in the skin (Ibrahim et al., by cutaneous nerve fibers, a crucial
granules can be released and secreted 2005). Moreover, endogenous CBs, role of opioids in the modulation of
in a discriminatory pattern. Of course, such as anandamide, have also been itching can be expected.
this mechanism may also be true for shown to activate TRPV1 receptors (see
other mediators not studied thus far. below), which adds to the complex role
of CBs in the modulation of pruritus. Kinins, kallikreins
A very similar complex interaction has Pruritic activity of kinins has also been
Opioids been shown for nociceptin: it activates investigated decades ago (Table 1). It
Another group of neuropeptides, the the inhibitory opioid receptor-like 1 was shown that both the tryptic en-
opioids, have been used as analgetic receptor, whereas the direct neuronal zymes (such as kallikreins) and the
agents for more than 2000 years. So far, effect on nociception appears to be resulting peptide fragments (like the
more than 20 endogenous analogs inhibitory. Additionally, the secondary mainly pain-inducing bradykinin) can
have been described, which are sub- release of leukotriene B4 by nociceptin induce itch by activating histamine-
divided into three classes (endorphins, in the skin provokes itch behavior sensitive C-fibers (Schmelz et al.,
enkephalins, and dynorphins) and (Andoh et al., 2004). 2003). Furthermore, there is some

1712 Journal of Investigative Dermatology (2006), Volume 126


M Steinhoff et al.
Basis of Pruritus

evidence that bradykinin type-2 recep- mediator involved in stress response, references therein) (Figure 2, Table 1).
tor antagonists can reduce deoxycholic thereby modulating inflammation, Trypsin and MC chymase provoke
acid-induced itch behavior in rodents immunity, and pruritus. For example, itching and visible changes (edema,
(Hayashi and Majima, 1999). More- it was shown that CRH triggers flare) when injected intracutaneously,
over, in this model, inhibition of tissue release of several mediator involved at least in part via MC activation. In
kallikrein suppressed the itch indicat- in itch response (Lytinas et al., 2003), contrast, papain-induced pruritus
ing a major role for epidermal kallik- possibly via CRH-R1 (Cao et al., 2005) seems to be histamine-independent.
rein during itch responses. Recent (Table 1). A milestone in our understanding of
studies on itch mediators mainly focus Neurotrophins such as NGF and protease-mediated signaling was the
on the role of proteases in pruritus (see neurotrophin-4 have also been impli- cloning of the first proteinase-activated
also in the following chapters): in- cated in itch pathophysiology. NGF is receptor, PAR1. Although PAR1, PAR2,
creased kallikrein activity and reduced released by keratinocytes, MCs, and and PAR4 have been described in
kininogen levels were found in pruritic fibroblasts (Groneberg et al., 2005). neurons, a role of PARs in the patho-
popular eruptions. It is noteworthy that Activation of its high-affinity receptor physiology of itching has only been
intracutaneous injections of kallikrein (trk A) on sensory nerves leads to nerve shown for PAR2 (Vergnolle et al.,
did not provoke marked itch in rats. sprouting and sensitization. Serum 2003). Importantly, functional PAR2 is
However, massive itch behavior is levels of NGF are increased in atopic present on primary spinal afferents, and
observed in mice overexpressing epi- dermatitis patients, which induces releases neuropeptides upon stimula-
dermal kallikrein-7 (Ny and Egelrud, release of the pruritogenic mediator tion by tryptase (Steinhoff et al., 2000,
2004 and references therein). tryptase (Groneberg et al., 2005). Also, 2003a, b, 2005). From this observation,
MCs and keratinocytes of these patients one may speculate that proteinases
generate high levels of NGF, which can activate PAR2 on sensory neurons,
Other neuropeptides and neurotrophins be stimulated by histamine (Kanda and thereby triggering pruritus in skin dis-
Several mediators from the central Watanabe, 2003). Similarly, neuro- eases such as atopic dermatitis. Addi-
and peripheral nervous system, which trophin-4 levels were also found to tionally, keratinocyte-derived PAR2,
are involved in neuroimmune and be enhanced in atopic dermatitis which is upregulated in the epidermis
neuroendocrine interactions (Steinhoff (Grewe et al., 2000), and brain- of atopic dermatitis patients (Budden-
et al., 2003a, b and references therein). derived neurotrophic factor (BDNF) kotte et al., 2005) may mediate pruritus
Some of these mediators have been induces chemotaxis of eosinophils of induced by endogenous (trypsins, kal-
implicated in the pathophysiology of these patients (Raap et al., 2005). likreins) or exogenous proteases (bac-
pruritus (Table 1). For example, CGRP Together, these results are clearly in teria, house-dust mite). Indeed, in
modulates inflammation and pruritus favor of an important role of neuro- atopic dermatitis patients, the endogen-
(Brain and Grant, 2004), and prolongs trophins in the pathophysiology of ous PAR2 agonist tryptase was in-
itch latency following SP injection itching, although direct evidence is still creased up to four-fold and PAR2
suggesting an inhibitory effect of lacking. expression was markedly enhanced on
CGRP on SP-induced itching. How- primary afferent nerve fibers of lesional
ever, increased amounts of CGRP skin. In contrast, no significant differ-
were observed in nerve fibers of Proteases and their receptors ences in histamine concentrations were
pruritic diseases such as atopic derma- Another intriguing pruritogenic candi- observed between the diseased and
titis, nummular eczema, and prurigo dates are the proteases. These mole- healthy samples, suggesting that tryp-
nodularis. cules comprise about 5% of the human tase might be more important than
Intradermal application of vasoac- genome making them the largest pro- histamine for the transmission of itch
tive intestinal polypeptide, neurotensin, tein family within the human being. responses in atopic dermatitis. More-
and secretin also led to a histamine- Classically, proteases are still regarded over, intracutaneous injection of spe-
dependent itch response, associated as destructive enzymes, which break cific PAR2 agonists provoked sustained
with pruritus, whealing, and axon- down proteins or merely activate or and prolonged itch in such patients.
reflex erythema. In normal human inactivate peptides by processing mole- This observation may also explain why
skin, vasoactive intestinal polypeptide cules. However, little is known about non-sedative antihistamines are poorly
showed a comparable potency with the role of proteases as signaling effective in atopic dermatitis. Thus,
respect to pruritus as compared to SP. molecules during neuronal transmis- PAR2 activation on cutaneous sensory
Moreover, pituitary adenylate cyclase- sion. Already in the 1950s, proteases nerves and keratinocytes may be a
activating polypeptide, somatostatin, have been suggested to be ultimately novel pathway for the transmission of
and CRH were demonstrated to stimu- involved in itch responses, burning, itch responses during atopic dermatitis
late histamine release and MC degra- pain, and inflammation. Arthur and and probably other skin diseases.
nulation from human and rat skin Shelley, and later Rajka demonstrated Hence, PAR2 antagonists or protease
(reviewed in Steinhoff et al., 2003a, b that exogenous as well as endogenous inhibitors may be promising therapeu-
and references therein). CRH is well serine proteases are capable of indu- tic targets for the treatment of pruritus
known as an important neuroendocrine cing pruritus (Bodo et al., 2005 and (Figure 2).

www.jidonline.org 1713
M Steinhoff et al.
Basis of Pruritus

The TRP channel family in itch Taken together, these findings strongly suggests a major role of TRPV1 expres-
Recent findings suggest that itch med- implicate that TRPV1 is indeed a sion in non-neuronal cells in pruritus
iators exert their effects also by activat- central integrator molecule in the pain patients.
ing ion channels of the TRP channel and itch pathway.
family. TRP channels comprise six Prolonged or repeated vanilloid ap- TRPV2, TRPV3, and TRPV4: further
groups of molecules: the canonical plication results in a depletion of intriguing targets to be investigated.
(TRPC), the vanilloid (TRPV), the mela- neuropeptides such as SP in C-type Originally, these channels were also
statin (TRPM), the polycystin (TRPP), neurons, hence suspending the inter- described as cellular temperature sen-
and mucolipin (TRPML) subfamilies, play between skin sensory neurons and sor molecules as all are activated by
and the anhyrin (TRPA). In general, MCs. Indeed, topical capsaicin effec- increasing temperatures (4531C for
these molecules act as calcium-perme- tively prevented histamine-induced TRPV2; 4311C for TRPV3; and
able sensory transduction channels to itch under experimental conditions. In 4251C for TRPV4) (Peier et al.,
detect, for example, taste, warmth, addition, capsaicin is widely used in 2002b and references therein) (Table 2).
heat, cold, and osmotic/mechanical the therapy of pruritus in numerous Importantly, TRPV3 exerts a very simi-
stress both at cellular and multicellular skin diseases such as prurigo nodularis, lar neuronal expression pattern to that
(i.e., whole organism) levels (reviewed notalgia paresthetica, pruritus ani, he- of TRPV1. In addition, it was also
in Zhang et al., 2003 and references modialysis-related pruritus, uremic postulated that TRPV3 subunits might
therein). With respect to the develop- pruritus, etc. (Biro et al., 1997; re- form heteromultimeric structures by
ment and, most intriguingly, therapy of viewed in Yosipovitch et al., 2003). interacting with TRPV1 monomers
itch, recently certain temperature-sen- Furthermore, recent findings provide and therefore may act as signal co-
sitive members of the TRPV subfamily a new ‘‘hot’’ twist to the field further transducers and/or regulators of the
and the TRPM8 gained substantial highlighting the pathophysiological TRPV1-mediated pain and itch sensa-
interest (Figures 1 and 2; Tables 2 and S1). and therapeutic importance of TRPV1 tion. Indeed, mice lacking the TRPV3
signaling in itch. Namely, functional have strong deficits in response to both
TRPV1: a putative central role in the TRPV1 channels were described on innocuous (mostly TRPV3 range) and
pathogenesis and therapy of itch. numerous non-neuronal cells types noxious heat (rather TRPV1 range)
TRPV1 was originally described on including, of greatest importance, hu- (Moqrich et al., 2005).
C-type nociceptive sensory neurons man skin epidermal keratinocytes, der- Most intriguingly, functional TRPV3
(Caterina et al., 1997) as a molecular mal MCs, dendritic cells, and various and TRPV4 channels (similarly to
target for capsaicin, the pungent in- keratinocyte populations of the hair TRPV1) are also expressed at high
gredient of hot chili peppers. The follicle (Bodo et al., 2005 and refer- levels on epidermal keratinocytes (Pe-
activation of the receptor first excites ences therein). In addition, activation ier et al., 2002b and references therein;
these neurons by initiating ionic fluxes of TRPV1 – beside markedly affecting Moqrich et al., 2005). In addition,
and concomitant action potential firing proliferation, differentiation, and apop- TRPV4 was shown to be activated by
and neuropeptide release. At higher tosis – resulted in the upregulation of IL- such lipid peroxidation products as
doses and longer times, capsaicin 1b and tumor growth factor-b, whereas eicosanoids, which may also function
induces the desensitization the sensory IGF and HGF were downregulated in as TRPV1-activating pruritogenic sub-
afferents (Caterina and Julius, 2001 and the non-neuronal cells (Bodo et al., stances (Watanabe et al., 2003). Thus,
references therein). 2005 and references therein). sensitization and activation of TRP
In addition to capsaicin, TRPV1 can These novel results invite an attrac- channels could underly pruritic activity
also be activated/sensitized by numer- tive hypothesis with further potential of prostanoids and adding to their
ous endogenous substances collec- therapeutic implications (Figures 1 and complex role in pruritus induction.
tively referred to as ‘‘endovanilloids’’ 3; Tables 2 and S1). Namely, topically Finally, with respect to the central
(Table 2). The receptor was first shown applied capsaicin may not exclusively involvement of MCs in the initiation of
to be stimulated by low threshold target sensory neurons but also TRPV1- itch, it is worth noting that TRPV2 and
(4431C), heat and acidosis. Later, expressing MCs and keratinocytes, TRPV4 (along with TRPV1) are also
several other molecules were also thereby modulating the proposed neu- expressed by MCs (Stokes et al., 2004).
described to either directly and/or ronal–non-neuronal network. We do These authors also revealed that physi-
indirectly act on the TRPV1. These not know the complete pattern of cal and thermal activation of TRPV2 on
agents are, for example, eicosanoids, mediator changes following TRPV1 MCs resulted in a proinflammatory
bradykinin, prostaglandins, and various stimulation on non-neuronal cells. degranulation event, which depended
neurotrophins (such as NGF, neurotro- However, TRPV1 expression was dra- on the activity of the protein kinase A-
phin-3 and -4) (Lazar et al., 2004 and matically increased in epidermal kera- related signaling, one of the chief
references therein). It was also shown tinocytes of prurigo nodularis patients mechanisms in initiating sensitization
that histamine-induced excitation of and normalized after successfully treat- of TRPV1 (a key event in initiating itch
sensory neurons and PAR2 activation ing the characteristic nodular pruritic and pain, see above) as well.
(Amadesi et al., 2004) does involve lesions with topical capsaicin (Stander Although more studies are necessary
the activation/sensitization of TRPV1. et al., 2004). This example strongly to further define the itch-related ‘‘non-

1714 Journal of Investigative Dermatology (2006), Volume 126


M Steinhoff et al.
Basis of Pruritus

menthol, it did not smell or irritate the


eyes.
Comparisons continued after the
cloning of TRPM8 with calcium entry
Initiation of itch Terminiation of itch into cells as an index of activity. The
EC50 of icilin in TRPM8-transfected
cells was reported to be 200  to
800  less than menthol, but potencies
of the two cannot be directly matched
TRPV1 TRPV1
because the maximum efficacy of icilin
for calcium entry was greater than that
of menthol. Icilin also activates TRPA1
(ANKM1), another TRP channel, but at
a lower potency than TRPM8.
Specific Sensory nerve
In an animal model of scratching
receptors
Pruritogenic endings in hairless rats, provoked by a magne-
substances sium-deficient diet, a 2% icilin oint-
(histamine,
neuropeptides,
Capsaicin
ment reduced the degree of excoria-
kinins, etc.)
tions by 55–60% (E Wei and Meingass-
ner T, unpublished data). In this model,
Specific it was found that 2–3% icilin ointment,
receptors
TRPV1
elicited robust cooling sensations for
2–4 h without any irritancy. The onset
TRPV1 of maximal cooling occurred within
10–15 minutes after application.
The TRPM8 agonist menthol and
analogs were also examined (Behrendt
Non-neuronal
skin cells et al., 2004). The most active analogs
were comparable in activity to
()menthol and at least 16-fold less
Figure 3. The proposed dual action of capsaicin to terminate itch. Pruritogenic substances either active than icilin, with lower maximal
directly (via their specific receptors) or indirectly (via activation/sensitization of TRPV1) stimulate sensory
responses. Carboxamides exert activity
neurons and non-neuronal cell types (MCs, keratinocytes) of the skin to initiate itch (left part in blue
frame). Topically applied capsaicin may not exclusively target sensory neuron-specific TRPV1 and hence similar to icilin on TRPM8 in vitro.
deplete the neuropeptide content but also act on TRPV1-expressing MCs and keratinocytes, and, in turn, When applied to the skin, carboxa-
significantly alter the proposed neuronal–non-neuronal interaction network to terminate itch (right part in mides produce cold sensations lasting
red frame). from 30 minutes to 1 hour. Thus, car-
bocamides may serve as a model
for endogenous TRPM8 agonists. Thus,
with the identification of TRPM8 (and
thermosensor’’ role of these channels produce sensations of cold (Peier et al., TRPA1) (Table 2) and the novel chemi-
(e.g. in keratinocyte-specific cytokine 2002a and references therein). cal ligands for these ion channels,
and mediator synthesis and release), Icilin was first synthesized with the understanding the linkage of psychic
the close resemblance to TRPV1 with intent of finding a morphine-like an- and somatic (e.g. dry skin, temperature
respect to cell-specific expression, ac- algesic, but when tested in rodents it changes) adjuncts of skin discomfort to
tivation, sensitization, and to the in- produced unusual behavioral events. molecular, cellular, and sensory inputs
itiated cellular mechanisms highlights Icilin injected into the systemic circula- now seems more exact. Ultimately, the
their putative roles in itch pathophy- tion of fur-coated animals produces proposed mechanisms of chemical ac-
siology. rotational movements similar to those tion will have to be resolved by further
manifested by a dog when wet (‘‘wet tests in human subjects.
Icilin and the ‘‘cool’’ TRPM8 channel. dog shakes’’). Icilin initiates punctate
Another intriguing member of the TRP sensations of coolness, harmonizing Cytokines, IFNs
family is TRPM8, which is selectively with the idea of ‘‘cold spots’’, when Recent findings clearly indicate a
expressed in C-type sensory neurons the particles come into contact with direct role of cytokines and chemo-
(Figures 1 and 3; Tables 2 and S1). the various mucosal surfaces (Wei and kines on the regulation of primary
TRPM8 is thought to be a thermosensor Seid, 1983). Icilin was compared to afferent neurons via receptor activation
for coolness and cold (8–281C) and also menthol and, for in vivo effects such as (Steinhoff et al., 2003a, b and refer-
activated by chemicals – menthol, ‘‘wet shake behavior’’, it was 400  ences therein; Dillon et al., 2004)
menthol analogs, and icilin – which more active than menthol. Unlike (Figures 1 and 2 and S1).

www.jidonline.org 1715
M Steinhoff et al.
Basis of Pruritus

IL-2. Clinical observations suggest a these observations, it is intriguing to ity within the central nervous system
role of IL-2 as an inducer of pruritus. speculate that IL-31 is upregulated in when processing itch information.
High doses of recombinant IL-2, ap- pruritic but not in non-pruritic forms of With respect to novel therapies, the
plied to cancer patients, for example, chronic skin inflammation. Thus, IL-31 potential redundancy of many pruritic
are capable of provoking flush, vasodi- may be a new link between the signals complicates specific treatment
latation, and pruritus. Whether this immune and nerval system by regulat- (Figure 3, Table S1). Thus, it will be a
is a direct, receptor-mediated process ing inflammation as well as itch. This major task to identify convergent per-
or an indirect, for example, via MCs may also suggest that IL-31 and its ipheral and central targets in the itch
or endothelial cells, is still unknown. signaling pathway represent a novel pathway to increase the chances of
Accordingly, treatment of Alzheimer target for antipruritic therapy (Sonkoly clinical success.
disease (AD) patients with systemic or et al., 2006). Therefore, future research will have
topical immunosuppressants such as to clarify underlying mechanisms that
tacrolimus, pimecrolimus, or cyclos- induce, modulate, and transmit the itch
INF-g. In a double-blind study, a
porin A, for example, which inhibit the signal on a molecular level to finally
beneficial effect of IFN-g on itch
production of various cytokines includ- identify the essential molecules as
responses has been clearly demon-
ing IL-2, experience attenuation of targets for antipruritic therapy. More-
strated also in AD patients. Pruritus
pruritus. over, understanding perception and
was reduced by 50% even 1–2 years
Another mechanism of action with regulation of itch signals within the
after long-term treatment with recom-
respect to cytokine-induced itch may central nervous system will lead to
binant human IFN-g. However, the
be synergistic amplification or receptor novel strategies for the treatment of
mode of action and the receptor
transactivation. For example, bradyki- pruritus.
density of IFN receptors on sensory
nin appears to augment the effect of IL-
nerves in the skin is unknown. CONFLICT OF INTEREST
2-induced pruritus on sensory nerves.
The authors state no conflict of interest.
However, the latency preceding the
itch response after injection in AD Summary and perspectives
ACKNOWLEDGMENTS
patients suggests an indirect prurito- Itch research not only has proposed a This work was supported by grants from the IZKF
genic effect of IL-2 via other mediators. variety of potential itch mediators but Münster, SFB 492, SFB 293, DFG (STE 1014/2-1),
also identified non-neuronal cells con- Serono Germany, CE.R.I.E.S. Paris, and Rosacea
foundation (to M.S.). Owing to limited space for
tributing to the pathophysiology of references, an adequate version of important
IL-8. Recent observations suggested a
pruritus. Being confronted with a references can be looked up in the supplement.
role of IL-8 as a mediator of itch in AD
variety of potential pruritic mediators
patients. However, intracutaneous ap-
generated by close interaction of neu- SUPPLEMENTARY MATERIAL
plication of IL-8 was not sufficient to
ronal and non-neuronal cells three Supplementary References.
induce pruritus or erythema in human
major directions for future research Figure S1. Hypothetic mechanisms of mast cell
skin (Stander and Steinhoff, 2002 and
arise: functions in the control of pruritus.
references therein).
First – which is the crucial pruritic Table S1. Established therapies of itch and
mediator in a particular skin disease experimental strategies.
IL-31. IL-31 is a novel cytokine pre- and how can it be modulated/sup-
ferentially produced by T-helper type 2 pressed? Second – what is the basis for REFERENCES
cells, which induces both severe prur- an inflammatory process to release itch Amadesi S, Nie J, Vergnolle N, Cottrell GS, Grady
EF, Trevisani M et al. (2004) Protease-
ititis and dermatitis in mice. Dillon mediators and sensitize itch receptors activated receptor 2 sensitizes the capsaicin
et al. (2004) recently demonstrated that rather than to produce pain and sensi- receptor transient receptor potential vanil-
transgenic overexpression of the novel tization to pain? Third, which mechan- loid receptor 1 to induce hyperalgesia.
isms can be used to modulate neuronal J Neurosci 24:4300–12
cytokine IL-31 in T-lymphocytes in-
duces severe pruritus and dermatitis in itch processing in the periphery, the Andoh T, Kuraishi Y (2003) Nitric oxide enhances
substance P-induced itch-associated re-
mice. IL-31 signals via a heterodimeric spinal cord, and the central nervous sponses in mice. Br J Pharmacol 138:202–8
receptor composed of IL-31 receptor A system? The insight in anatomical and Andoh T, Yageta Y, Takeshima H, Kuraishi Y
and the oncostatin M receptor. physiological structures as provided by (2004) Intradermal nociceptin elicits itch-
Whether IL-31 exerts its effects via central imaging will help to identify the associated responses through leukotriene
involved central areas. Consequently, B(4) in mice. J Invest Dermatol 123:196–201
direct activation of the IL-31R on
sensory nerves or indirectly, for exam- both, animal studies and clinical trials Andrew D, Craig AD (2001) Spinothalamic
lamina I neurons selectively sensitive to
ple, via keratinocytes is unknown. The will further dissect the interaction and histamine: a central neural pathway for itch.
finding that keratinocytes express the communication between the central Nat Neurosci 4:72–7
IL-31 R suggests that IL-31 may induce and the peripheral nervous system Ashida Y, Denda M, Hirao T (2001) Histamine H1
pruritus through the induction of a yet under physiological and pathophysio- and H2 receptor antagonists accelerate
logical conditions. Here also, novel skin barrier repair and prevent epidermal
unknown keratinocyte-derived media-
hyperplasia induced by barrier disruption in
tor, which subsequently activates un- molecular imaging approaches may a dry environment. J Invest Dermatol 116:
myelinated C fibers in the skin. From help to better understand the complex- 261–5

1716 Journal of Investigative Dermatology (2006), Volume 126


M Steinhoff et al.
Basis of Pruritus

Behrendt HJ, Germann T, Gillen C, Hatt H, histamine-induced itch in the human cere- Inoue K, Denda M, Tozaki H, Fujishita K, Koizumi
Jostock R (2004) Characterization of the bral cortex: a correlation analysis with S, Inoue K (2005) Characterization of multi-
mouse cold-menthol receptor TRPM8 and dermal reactions. J Invest Dermatol 115: ple P2X receptors in cultured normal human
vanilloid receptor type-1 VR1 using a fluoro- 1029–33 epidermal keratinocytes. J Invest Dermatol
metric imaging plate reader (FLIPR) assay. Dillon SR, Sprecher C, Hammond A, Bilsborough 124:756–63
Br J Pharmacol 141:737–45 J, Rosenfeld-Franklin M, Presnell SR et al. Janiszewski J, Bienenstock J, Blennerhassett MG
Bienenstock J, Tomioka M, Matsuda H, Stead RH, (2004) Interleukin 31, a cytokine produced (1994) Picomolar doses of substance P
Quinonez G, Simon GT et al. (1987) The role by activated T cells, induces dermatitis in trigger electrical responses in mast cells
of mast cells in inflammatory processes: mice. Nat Immunol 5:752–60 without degranulation. Am J Physiol 267:
evidence for nerve/mast cell interactions. Drzezga A, Darsow U, Treede RD, Siebner H, C138–45
Int Arch Allergy Appl Immunol 82:238–43 Frisch M, Munz F et al. (2001) Central Kamei J, Nagase H (2001) Norbinaltorphimine, a
Bigliardi-Qi M, Lipp B, Sumanovski LT, Buechner activation by histamine-induced itch: analo- selective kappa-opioid receptor antagonist,
SA, Bigliardi PL (2005) Changes of epidermal gies to pain processing: a correlational induces an itch-associated response in mice.
mu-opiate receptor expression and nerve analysis of O-15 H2O positron emission Eur J Pharmacol 418:141–5
endings in chronic atopic dermatitis. Derma- tomography studies. Pain 92:295–305 Kanda N, Koike S, Watanabe S (2005) Prosta-
tology 210:91–9 Grewe M, Vogelsang K, Ruzicka T, Stege H, glandin E2 enhances neurotrophin-4 pro-
Biro T, Acs G, Acs P, Modarres S, Blumberg PM Krutmann J (2000) Neurotrophin-4 produc- duction via EP3 receptor in human
(1997) Recent advances in understanding tion by human epidermal keratinocytes: keratinocytes. J Pharmacol Exp Ther 315:
of vanilloid receptors: a therapeutic target increased expression in atopic dermatitis. 796–804
for treatment of pain and inflammation in J Invest Dermatol 114:1108–12 Kanda N, Watanabe S (2003) Histamine enhances
skin. J Investig Dermatol Symp Proc 2:56–60 Groneberg DA, Serowka F, Peckenschneider N, the production of nerve growth factor in
Biro T, Ko MC, Bromm B, Wei ET, Bigliardi P, Artuc M, Grutzkau A, Fischer A et al. (2005) human keratinocytes. J Invest Dermatol 121:
Siebenhaar F et al. (2005) How best to fight Gene expression and regulation of nerve 570–7
that nasty itch – from new insights into the growth factor in atopic dermatitis mast cells Katugampola R, Church MK, Clough GF (2000)
neuroimmunological, neuroendocrine, and and the human mast cell line-1. J Neuro- The neurogenic vasodilator response to
neurophysiological bases of pruritus to novel immunol 161:87–92 endothelin-1: a study in human skin in vivo.
therapeutic approaches. Exp Dermatol 14: Hagermark O (1974) Studies on experimental itch Exp Physiol 85:839–46
225–40 induced by kallikrein and bradykinin. Acta Kennedy WR, Wendelschafer-Crabb G, Brelje TC
Blunk JA, Schmelz M, Zeck S, Skov P, Likar R, Dermatol Venereol 54:397–400 (1994) Innervation and vasculature of human
Koppert W (2004) Opioid-induced mast Haüstein UF (1990) Adrenergic urticaria and sweat glands: an immunohistochemistry-la-
cell activation and vascular responses is not adrenergic pruritus. Acta Dermatol Venerol ser scanning confocal fluorescence micro-
mediated by mu-opioid receptors: an in vivo 70:82–4 scopy study. J Neurosci 14:6825–33
microdialysis study in human skin. Anesth
Analg 98:364–70 Hayashi I, Majima M (1999) Reduction of sodium Lazar J, Szabo T, Marincsak R, Kovacs L,
deoxycholic acid-induced scratching beha- Blumberg PM, Biro T (2004) Sensitization
Bodo E, Biro T, Telek A, Czifra G, Griger Z, Toth viour by bradykinin B2 receptor antagonists. of recombinant vanilloid receptor-1 by var-
BI et al. (2005) A hot new twist to hair Br J Pharmacol 126:197–204 ious neurotrophic factors. Life Sci 75:153–63
biology: involvement of vanilloid receptor-1
(VR1/TRPV1) signaling in human hair growth Hayashi K, Sato H, Kaise T, Ohmori K, Ishii A, Lytinas M, Kempuraj D, Huang M, Boucher W,
control. Am J Pathol 166:985–98 Sano J et al. (2001) Roles of mast cells and Esposito P, Theoharides TC (2003) Acute
sensory nerves in cutaneous vascular hyper- stress results in skin corticotropin-releasing
Brain SD, Grant AD (2004) Vascular actions of permeability and scratching behavior in- hormone secretion, mast cell activation and
calcitonin gene-related peptide and adreno- duced by poly-L-arginine in rats. Eur J vascular permeability, an effect mimicked by
medullin. Physiol Rev 84:903–34 Pharmacol 425:219–27 intradermal corticotropin-releasing hormone
Brain SD, Williams TJ (1988) Neuropharmacology Heyer G, Groene D, Martus P (2002) Efficacy of and inhibited by histamine-1 receptor an-
of peptides in skin. Semin Dermatol 7: naltrexone on acetylcholine-induced allo- tagonists. Int Arch Allergy Immunol 130:
278–83 kinesis in atopic eczema. Exp Dermatol 11: 224–31
Buddenkotte J, Stroh C, Engels IH, Moormann C, 448–55 Maurer BA (2002) Big thinking. Nature 415:
Shpacovitch VM, Seeliger S et al. (2005) Hossen MA, Sugimoto Y, Kayasuga R, Kamei C 489–91
Agonists of proteinase-activated receptor-2 (2003) Involvement of histamine H3 recep- Metze D, Reimann S, Beissert S, Luger T (1999)
stimulate upregulation of intercellular cell tors in scratching behaviour in mast cell- Efficacy and safety of naltrexone, an oral
adhesion molecule-1 in primary human deficient mice. Br J Dermatol 149:17–22 opiate receptor antagonist, in the treatment
keratinocytes via activation of NF-kappa B. of pruritus in internal and dermatological
J Invest Dermatol 124:38–45 Hundley SL, Yosipovitch G (2004) Mirtazapine for
reducing nocturnal itch in patients with diseases. J Am Acad Dermatol 41:533–9
Cao J, Papadopoulou N, Kempuraj D, Boucher chronic pruritus: a pilot study. J Am Acad Miyamoto T, Nojima H, Shinkado T, Nakahashi
WS, Sugimoto K, Cetrulo CL et al. (2005) Derm 50:889–91 T, Kuraishi Y (2002) Itch-associated response
Human mast cells express corticotropin- induced by experimental dry skin in mice.
releasing hormone (CRH) receptors and Ibrahim MM, Porreca F, Lai J, Albrecht PJ, Rice FL,
Khodorova A et al. (2005) CB2 cannabinoid Jpn J Pharmacol 88:285–92
CRH leads to selective secretion of vascular
endothelial growth factor. J Immunol 174: receptor activation produces antinociception Mochizuki H, Tashiro M, Kano M, Sakurada Y,
7665–75 by stimulating peripheral release of endo- Itoh M, Yanai K (2003) Imaging of central
genous opioids. Proc Natl Acad Sci USA itch modulation in the human brain using
Caterina MJ, Julius D (2001) The vanilloid 102:3093–8 positron emission tomography. Pain 105:
receptor: a molecular gateway to the pain 339–46
pathway. Annu Rev Neurosci 24:487–517 Ikoma A, Handwerker H, Miyachi Y, Schmelz M
(2005) Electrically evoked itch in humans. Moqrich A, Hwang SW, Earley TJ, Petrus MJ,
Caterina MJ, Schumacher MA, Tominaga M, Pain 113:148–54 Murray AN, Spencer KS et al. (2005)
Rosen TA, Levine JD, Julius D (1997) The Impaired thermosensation in mice lacking
capsaicin receptor: a heat-activated ion Inagaki N, Igeta K, Kim JF, Nagao M, Shiraishi N,
Nakamura N et al. (2002) Involvement of TRPV3, a heat and camphor sensor in the
channel in the pain pathway. Nature 389: skin. Science 307:1468–72
816–24 unique mechanisms in the induction of
scratching behavior in BALB/c mice by Nojima H, Cuellar JM, Simons CT, Carstens MI,
Darsow U, Drzezga A, Frisch M, Munz F, Weilke compound 48/80. Eur J Pharmacol 448: Carstens E (2004) Spinal c-fos expression
F, Bartenstein P et al. (2000) Processing of 175–83 associated with spontaneous biting in a

www.jidonline.org 1717
M Steinhoff et al.
Basis of Pruritus

mouse model of dry skin pruritus. Neurosci Schmidt R, Schmelz M, Ringkamp M, Handwer- Steinhoff M, Stander S, Seeliger S, Ansel JC,
Lett 361:79–82 ker HO, Torebjork HE (1997) Innervation Schmelz M, Luger T (2003b) Modern aspects
Ny A, Egelrud T (2004) Epidermal hyperproliferation territories of mechanically activated C noci- of cutaneous neurogenic inflammation. Arch
and decreased skin barrier function in mice ceptor units in human skin. J Neurophysiol Dermatol 139:1479–88
overexpressing stratum corneum chymotryptic 78:2641–8 Steinhoff M, Vergnolle N, Young SH, Tognetto M,
enzyme. Acta Dermatol Venereol 84:18–22 Sonkoly E, Muller A, Lauerma AI, Pivarcsi A, Soto Amadesi S, Ennes HS et al. (2000) Agonists
Onigbogi O, Ajayi AA, Ukponmwan OE (2000) H, Kemeny L et al. (2006) IL-31: A new link of proteinase-activated receptor 2 induce
Mechanisms of chloroquine-induced body- between T cells and pruritus in atopic skin inflammation by a neurogenic mechanism.
scratching behavior in rats: evidence of inflammation. J Allergy Clin Immunol 117: Nat Med 6:151–8
involvement of endogenous opioid peptides. 411–7 Stokes AJ, Shimoda LM, Koblan-Huberson M,
Pharmacol Biochem Behav 65:333–7 Ständer S, Moormann C, Schumacher M, Bud- Adra CN, Turner H (2004) A TRPV2-PKA
Peier AM, Moqrich A, Hergarden AC, Reeve AJ, denkotte J, Artuc M, Shpacovitch V et al. signaling module for transduction of physical
Andersson DA, Story GM et al. (2002a) A (2004) Expression of vanilloid receptor sub- stimuli in mast cells. J Exp Med 200:137–47
TRP channel that senses cold stimuli and type 1 in cutaneous sensory nerve fibers, Thomsen JS, Sonne M, Benfeldt E, Jensen SB,
menthol. Cell 108:705–15 mast cells, and epithelial cells of appendage Serup J, Menne T (2002) Experimental itch in
structures. Exp Dermatol 13:129–39 sodium lauryl sulphate-inflamed and normal
Peier AM, Reeve AJ, Andersson DA, Moqrich A,
Earley TJ, Hergarden AC et al. (2002b) A Ständer S, Schmelz M, Metze D, Luger T, skin in humans: a randomized, double-blind,
heat-sensitive TRP channel expressed in Rukwied R (2005) Distribution of cannabi- placebo-controlled study of histamine and
keratinocytes. Science 296:2046–9 noid receptor 1 (CB1) and 2 (CB2) on sensory other inducers of itch. Br J Dermatol 146:
nerve fibers and adnexal structures in human 792–800
Raap U, Goltz C, Deneka N, Bruder M, Renz H, skin. J Dermatol Sci 38:177–88
Kapp A et al. (2005) Brain-derived neuro- Vergnolle N, Ferazzini M, D’Andrea MR, Bud-
trophic factor is increased in atopic derma- Ständer S, Steinhoff M (2002) Pathophysiology of denkotte J, Steinhoff M (2003) Proteinase-
titis and modulates eosinophil functions pruritus in atopic dermatitis: an overview. activated receptors: novel signals for peri-
compared with that seen in nonatopic sub- Exp Dermatol 11:12–24 pheral nerves. Trends Neurosci 26:496–500
jects. J Allergy Clin Immunol 115:1268–75 Ständer S, Steinhoff M, Schmelz M, Weisshaar E, Watanabe H, Vriens J, Prenen J, Droogmans G,
Schmelz M, Michael K, Weidner C, Schmidt R, Metze D, Luger T (2003) Neurophysiology Voets T, Nilius B (2003) Anandamide and
Torebjork HE, Handwerker HO (2000a) of pruritus: cutaneous elicitation of itch. arachidonic acid use epoxyeicosatrienoic
Which nerve fibers mediate the axon reflex Arch Dermatol 139:1463–70 acids to activate TRPV4 channels. Nature
flare in human skin? Neuroreport 11:645–8 424:434–8
Steinhoff M, Buddenkotte J, Shpacovitch V,
Schmelz M, Schmid R, Handwerker HO, Tor- Rattenholl A, Moormann C, Vergnolle N Wei ET, Seid DA (1983) AG-3-5: a chemical
ebjork HE (2000b) Encoding of burning pain et al. (2005) Proteinase-activated receptors: producing sensations of cold. J Pharm
from capsaicin-treated human skin in two transducers of proteinase-mediated signaling Pharmacol 35:110–2
categories of unmyelinated nerve fibres. in inflammation and immune response. Yosipovitch G, Greaves MW, Schmelz M (2003)
Brain 123:560–71 Endocr Rev 26:1–43 Itch. Lancet 361:690–4
Schmelz M, Schmidt R, Weidner C, Hilliges M, Steinhoff M, Neisius U, Ikoma A, Fartasch M, Zhang Y, Hoon MA, Chandrashekar J, Mueller KL,
Torebjork HE, Handwerker HO (2003) Che- Heyer G, Skov PS et al. (2003a) Proteinase- Cook B, Wu D et al. (2003) Coding of sweet,
mical response pattern of different classes of activated receptor-2 mediates itch: a novel bitter, and umami tastes: different receptor
C-nociceptors to pruritogens and algogens. pathway for pruritus in human skin. cells sharing similar signaling pathways. Cell
J Neurophysiol 89:2441–8 J Neurosci 23:6176–80 112:293–301

1718 Journal of Investigative Dermatology (2006), Volume 126

You might also like