Você está na página 1de 11

PRURITUS SECTION 2

Cutaneous Neurophysiology 5 CHAPTER

Cutaneous Neurophysiology
Gil Yosipovitch and Alexandru DP Papoiu

to the skin, tracheal mucous membrane and several mucocutaneous


Chapter Contents junctions (e.g. conjunctivae). Interestingly, the nerves in the deeper
Pruritus pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99 layer of the reticular dermis and subcutaneous fat do not transmit itch.
Mediators of pruritus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
Treatment of pruritus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105 PRURITUS PATHWAYS
Significant advances in our understanding of itch neurophysiology
have been achieved in the past decade. Examples include identifica-
tion of specific nerve fibers that transmit itch peripherally and
Key features centrally, as well as the discovery of new itch mediators4. While
the concept of pruritus as a sensory modality entirely separate from
Two subsets of pruritoceptive C neurons (responding to histamine
pain was not generally appreciated until the mid-twentieth century,
and cowhage, respectively) have been identified, and these
studies have clearly identified individual histamine-sensitive and non-
parallel pathways activate distinct spinothalamic tract neurons
histaminergic C nerve fibers that transmit itch5,6. C nerve fibers that
Itch mediators act both peripherally (e.g. histamine, proteases, transmit itch have exceptionally slow conduction velocities (28cm/
interleukin-31) and centrally (e.g. opioids, gastrin-releasing peptide second), innervate unusually wide territories, and represent 5% of
receptors) via several mechanisms the total C fibers.
There is an overlap between chronic itch and chronic pain, Histamine-sensitive C fibers are sensitive to thermal as well as pru-
including activation of multiple brain areas and associated ritogenic stimuli but not to mechanical stimuli; in contrast, the vast
neuromediators and receptors (e.g. nerve growth factor, majority of C fibers are sensitive to mechanical and heat stimuli but
neurotrophin 4, transient receptor potential vanilloid [TRPV] are entirely insensitive to histamine7. The co-responsiveness of itch-
receptor) transmitting C neurons to temperature changes is clinically relevant,
Significant cross-talk between cutaneous nerve fibers and the as raising the temperature of the skin lowers the threshold of these
stratum corneum is a possible mechanism for pruritus associated receptors to pruritogenic stimuli8; this provides an explanation for
with impaired barrier function (e.g. xerosis, atopic dermatitis) aggravation of pruritus in a warm environment. However, the fact that
oral antihistamines are ineffective in the treatment of most types of
Combination therapies with typical and systemic drugs that
pruritus suggests that other fibers have important roles in the sensation
reduce itch sensitization by counteracting the responsible
of itch9.
mediators represent promising treatment strategies
Indeed, a distinct parallel pathway of non-histaminergic C nerve
fibers that transmit itch was recently discovered in the peripheral
nervous system of humans and the spinothalamic tract of primates10,11.
INTRODUCTION These fibers are activated by spicules of the ubiquitous tropical legume
cowhage (Mucuna pruriens), which is known to induce an intense
The skin is a sensory organ with a dense network of highly specialized sensation of itch when rubbed, inserted or injected into the skin,
afferent sensory nerves that convey sensations such as pain, itch, without producing a histaminergic axon reflex12. The mechanism by
touch, temperature, vibration and pressure (Table 5.1); efferent auto- which cowhage induces itch involves the release of a protease (mucu-
nomic nerve branches are also present. Neuropeptides such as nerve nain; contained within the spicules) that activates proteinase-activated
growth factor (NGF) and substance P are secreted from nerve fibers, receptor (PAR)-2 and PAR-413. Cowhage-sensitive fibers are not itch-
with multiple effects that include immune modulation. Itch (pruritus) specific, transmitting a burning sensation together with itch and also
is the dominant symptom of many cutaneous diseases. Almost all sensitive to mechanical and other stimuli. PAR-2 receptors have a
inflammatory skin disorders can result in pruritus, which is often per- major role in mediating itch in patients with atopic dermatitis14 (see
ceived by the patient as the most unendurable symptom of his or her Ch. 12), explaining the hypersensitivity to mechanical stimuli (e.g.
condition. Pruritus can also occur in association with systemic disease contact with wool) in these individuals. The non-histaminergic, poly-
(e.g. renal failure or cholestasis; see Ch. 6), psychiatric conditions (see modal C nerve fibers stimulated by mucunain may also have clinical
Ch. 7) and damage to nerve fibers1,2. Itch is a multidimensional phe- relevance in chronic itch.
nomenon with sensory discriminative, cognitive, evaluative and moti- Gastrin-releasing peptide receptor (GRPR)-positive neurons that
vational components. In most instances, itch results from interactions transmit itch but not pain are found in the spinal cord of mice; their
that involve the brainskin axis. role in itch pathophysiology in humans remains to be determined15,16.
Pruritus has many similarities to pain. Both are unpleasant sensory Toll-like receptor 7 (TLR-7) was recently found to be expressed in C
experiences that can impair quality of life in affected individuals. fibers and to have a role in itch (especially that elicited by non-
However, the behavioral reaction patterns differ pain elicits a reflex histaminergic pruritogens) but not pain sensations in mice17.
withdrawal, whereas itch leads to a scratching response. Despite being Another important observation is that the perceived sensation of
an extremely common complaint3 and a sensation so rudimentary that pruritus can vary greatly in quality1822. Patients may describe burning,
almost every two- or four-footed creature experiences it, medical science pricking, insects crawling on the skin, or even a tickle, but the neuro
is still struggling to understand the mechanisms of itch and how it can physiologic and psychologic correlates of these qualitative differences
be inhibited. have not yet been elucidated. Recent information obtained from itch
The connection between itch and scratching is so close that in some questionnaires has enabled a better understanding of the different char- 99
languages the same word refers to both itch and scratch. Itch is restricted acteristics of itch and awaits translation into biologic explanations.
PRIMARY AFFERENT NEURONS THAT INNERVATE THE SKIN NEUROANATOMY OF ITCH

Fiber Diameter Myelination Conduction Responds to


SECTION velocity
Cerebral cortex:
2 A-beta (A) Large + >30m/s Light touch
Moving stimuli Anterior cingulate
+
Pruritus

A-delta (A) Small 230m/s Pain (nociceptors) Somatosensory I


Thermal
Mechanical Somatosensory II
Chemical, including
pruritogens
Thalamus:
C Small <2m/s Pain (nociceptors)
Itch*, histamine-sensitive Medial dorsal
Itch*, cowhage-sensitive
Ventral posterior
Thermal
Mechanical
*~5% of total C fibers transmit itch.

Separate C fibers carry both pruritogenic and thermal stimuli, but not mechanical stimuli.

Transmit pruritus accompanied by a burning sensation; also sensitive to mechanical
stimuli.

Table 5.1 Primary afferent neurons that innervate the skin.


Cerebral
aqueduct
Midbrain:
Cowhage-
sensitive Periaqueductal
Central Pathways to Higher Nervous System C fiber gray matter

Centers transmitting
itch
In the spinal cord, nociceptor C neurons synapse with secondary trans- Ascending lamina I
Histamine- spinothalamic fibers
mission neurons in the gray matter of the dorsal horn (Fig. 5.1). These sensitive
neurons then cross over and ascend in the lateral spinothalamic tract C fiber Descending
to the thalamus. Studies using microneurography identified a subclass transmitting inhibitory fibers
of lamina I spinothalamic tract neurons that are excited by iontopho- itch Dorsal root
retically administered histamine23. Later studies have found that these ganglion
nerve fibers are also able to transmit pain upon stimulation by capsaicin Spinal cord:
and bradykinin7. Neurophysiology experiments in primates have dis-
Dorsal horn
covered that cowhage-induced itch stimulates a distinct population of
spinothalamic tract neurons that are not involved in the transmission C fiber Lateral
transmitting
of histamine-mediated itch11. spinothalamic
pain tract
Much of our current understanding of supraspinal processing of itch
and the corresponding scratch response is derived from studies in
human subjects utilizing positron emission tomography (PET) and
Fig. 5.1 Neuroanatomy of itch. Itch and pain transmission occur via
functional MRI during stimulation with histamine and (more recently) unmyelinated C nerve fibers that excite lamina I neurons in the dorsal horn of
cowhage2428. In healthy individuals, induction of itch in this manner the spinal cord. Two subsets of pruritoceptive C-fiber neurons (which respond
can elicit co-activation of the anterior and posterior cingulate cortex, to histamine and cowhage, respectively) are conducted through distinct lateral
precuneus, somatosensory areas I and II, supramarginal gyrus, inferior spinothalamic pathways, with projections to the thalamus. Processing of itch
parietal lobe and insula-claustrum complex28. In one study, atopic der- through either pathway activates several regions of the brain, which are similar
matitis patients exhibited significantly more activation of the posterior to those involved in pain (see Table 5.4). Concomitant painful stimuli can
cingulate cortex and precuneus (a cortical area involved in integrative reduce the sensation of itch, possibly by a descending inhibitory mechanism
tasks such as visuospatial imagery, episodic memory retrieval and self- resulting from activation of the periaqueductal gray matter. In mice, gastrin-
awareness) than did healthy controls29. This highlights the emotional releasing peptide receptor-positive neurons transmit itch (but not pain) within
the spinal cord; the role of such neurons in humans remains to be determined.
and affective processing of itch experience in atopic individuals, in Adapted from Yosipovitch G. Pruritus: an update. Curr Probl Dermatol. 2003;15:13764.
whom the degree of brain activation correlated with itch intensity and
the severity of the atopic disease29. In healthy subjects, scratching has
been found to inhibit the activation of the cingulate cortex and to
activate the prefrontal cortex and cerebellum. The cerebellum may have
a role in coordination of the itchscratch cycle30,31. Scratching inhibits
histamine-evoked activity of spinothalamic neurons in primates, but Pruritus Receptor Units
not spontaneous activity or that stimulated by pain32. Removal of the epidermis abolishes the perception of itch, suggesting
Activation of multiple brain areas suggests a lack of a single itch that pruritus receptor units (which have yet to be identified) are located
center, emphasizing the multidimensionality of the itch sensation. predominantly within this layer. Light microscopic and ultrastructural
Pain demonstrates a similar pattern of brain activation involving many studies of human skin have shown the existence of intraepidermal
of the same cortical regions33. nerve fibers with free non-specialized nerve endings extending to the
Seeing other people scratching can induce sensations of itch and an stratum granulosum35. Recent studies have identified a subclass of C
urge to scratch34. However, the central nervous mechanisms of this nerve fibers in the epidermis that express MAS-related G protein-
transmission, which appear to be beyond conscious control, are poorly coupled receptors (MRGPRs) and are involved in chloroquine-induced
understood. Psychophysical studies have shown that the perception of pruritus36. The cell bodies of these neurons (located in the dorsal root
itch intensifies in atopic dermatitis patients when they are exposed to ganglia) also express gastrin-related peptide (GRP), which binds the
visual cues of itch, which also trigger a scratching response34. More GRPRs that are known to relay itch-specific information in the spinal
studies are needed to verify the working hypothesis that itch conta- cords of mice15 (see above).
100 gion involves processing in associative cortical networks such as Keratinocytes express a variety of neural mediators and receptors that
mirror neurons of the prefrontal cortex. appear to be involved in the sensation of itch (see Table 5.2). These
Histamines pruritic action can be potentiated by prostaglandin E1
MAJOR MEDIATORS OF PRURITUS: RELATIVE POTENCIES WITH REGARD and E2. Evidence for histamine as the main mediator of pruritus is
TO PRURITUS AND PAIN limited to a few skin diseases, including acute and chronic urticaria
and mastocytosis (e.g. urticaria pigmentosa). H1 antihistamines are
Mediator Pruritus Pain CHAPTER
usually effective in these disorders.

Histamine
PRIMARY MEDIATORS
+++* +*
Recognition of the histamine H4 receptor has expanded our under-
standing of the physiologic actions of histamine. The H4 receptor is
5
expressed by neurons and bone marrow-derived cells (e.g. eosinophils,

Cutaneous Neurophysiology
Tryptase +++ +
mast cells, dendritic cells, monocytes, CD8+ T cells). It mediates
Cathepsin S +++ ? chemotaxis in the latter group and is thought to have a role in the
Interleukin-31 +++ ? inflammation and pruritus of atopic dermatitis. H4 antagonists are
under development and have been shown to alleviate experimental
SECONDARY MEDIATORS
pruritus. In animals models of itch, the effects of H4 antagonists are
Prostaglandin E1,2 + + synergistic with those of centrally acting H1 antihistamines (e.g.
Substance P
+ + diphenhydramine)39,40.
-Opioid receptor agonists ++
Gastrin-Releasing Peptide
Nerve growth factor + +
As noted above, itch-specific GRPR-positive neurons in the dorsal horn
Interleukin-2 +++ +/ of the spinal cord have been identified in mice15. Although named for
*Superficial (intraepidermal) injection of histamine causes pruritus; deep dermal injection its role in regulating gastrointestinal functions, the GRP ligand of
causes pain. GRPRs is widely expressed in the CNS and peripheral sensory neurons,

Lowers threshold to pruritus induced by other mediators.
including MRGPR-expressing C-fiber neurons that terminate in the
Actions partly due to histamine release from mast cells.
epidermis.
Table 5.2 Major mediators of pruritus: relative potencies with regard to
pruritus and pain. +/, little or no activity; +, weak activity; ++, moderately Proteinases
active; +++, highly active; , no activity. Human dermal mast cells produce two proteases: tryptase and chymase.
The proximity of dermal mast cells to afferent C-neuron terminals
within the skin suggests a functional relationship between these two
cell types. Mast cell-derived tryptase contributes to the neurogenic
inflammatory response14. The activated mast cell releases tryptase
include opioids, proteases, substance P, NGF and neurotrophin 4 as
(along with other mediators, including histamine), which in turn acti-
well as their respective receptors, including - and -opioid receptors,
vates PAR-2, a G protein-coupled receptor present on C- fiber terminals
PAR-2, neurotrophic tyrosine kinase receptor type 1 (NTRK1 [TRKA]),
(see Fig. 5.2); specifically, self-activation of PAR-2 occurs after cleav-
and transient receptor potential vanilloid ion channels (particularly
age exposes a tethered ligand domain. Kallikrein and cathepsins in the
TRPV1 and TRPV3). Keratinocytes also have ATP voltage-gated ion
skin have also been implicated in itch induction via activation of PAR-2.
channels and adenosine receptor ligands similar to those observed in
The activated C fibers transmit this information to the CNS, where it
C nerve fibers involved in pain transmission. These structural similari-
induces itch. In addition, activation leads to local release of neuropep-
ties to nerve fibers suggest that keratinocytes may be involved in the
tides, including substance P and calcitonin gene-related peptide, which
transduction and generation of itch. PAR-2 receptors are thought to be
induce neurogenic inflammation41.
involved in the itch of atopic dermatitis and mediate cowhage-induced
Studies using dermal microdialysis have shown that tryptase levels
itch (see above). Cathepsin S, an endogenous protease, also induces itch
are elevated fourfold in non-lesional forearm skin of atopic dermatitis
via PAR-2 and PAR-4 receptors37.
patients14. Expression of PAR-2, the receptor for tryptase, is significantly
elevated in the epidermis and cutaneous nerve fibers of eczematous
MEDIATORS OF PRURITUS lesions, while non-lesional skin demonstrates a less pronounced
increase in PAR-2. It is noteworthy that proteinase (e.g. cathepsin B)
Various mediators that act centrally and/or peripherally are involved in activity can also be found in common allergens (e.g. grass pollen, house
pruritus, including histamine, proteases, substance P, opiates, NGF and dust mites)42, and Staphylococcus aureus can induce secretion of pro-
prostaglandins. In inflammatory skin diseases such as urticaria, pro teases; both of these exogenous factors are known to aggravate atopic
inflammatory mediators produce pruritus and also cause other signs of dermatitis and itch.
inflammation (e.g. erythema due to vasodilation, increased vascular The role of epidermal serine proteases in eliciting itch is supported
permeability). The relative potencies of the major mediators with regard by a mouse model where overexpression of a serine protease led to
to these responses (including pruritus) are listed in Table 5.2. Some of severe itch and scratching (Table 5.3) as well as the serine protease
these mediators cause pruritus indirectly by evoking release of hista- inhibitor deficiency (leading to excess epidermal protease activity) that
mine and other mediators (e.g. substance P and several opioid peptides) underlies Netherton syndrome, an autosomal recessive ichthyosiform
from mast cells or by potentiating the actions of other mediators (e.g. disorder with prominent pruritus and atopic manifestations (see Ch.
prostaglandins E1 and E2). 57). These observations suggest that interactions between proteases
and receptors on C fibers play important roles in itch and cutaneous
Histamine inflammation.
Histamine is the archetypal mediator of signs and symptoms of inflam-
mation, including pruritus. In the skin, histamine is contained prima- Opioid Peptides
rily within the granules of dermal mast cells. Histamine can be released The concept of central pruritus (and the possible involvement of
from mast cells upon activation of a range of receptors, including the pruritic mediators located within the CNS) is becoming increasingly
high-affinity IgE receptor (FcRI), the KIT receptor for stem cell factor, recognized in both cutaneous and systemic diseases, with implications
receptors for neuropeptides (e.g. substance P, NGF) and complement for treatment. The perception of pruritus is modified by endogenous
C5a. In IgE-mediated acute urticaria, histamine is released when a opiates via central and peripheral opioid receptors. It has been suggested
specific antigen cross-links adjacent FcRIs via their -chains. In that generalized pruritus is induced by an imbalance between the
autoimmune chronic urticaria, similar cross-linking occurs via func- - and -opioid systems. Activation of -opioid receptors stimulates
tional circulating IgG or (less commonly) IgE autoantibodies that react itch perception, whereas -opioid receptor stimulation inhibits
with epitopes expressed on the -chain of FcRI38 (see Fig. 18.3). His- -receptor effects, both centrally and peripherally43,44.
tologically, dermal mast cells and unmyelinated neurons are closely Morphine and other endogenous and exogenous -opioid receptor
juxtaposed (Fig. 5.2), raising the possibility of a close (synapsis-like) agonists are known to cause generalized pruritus1,2,4446. Morphine also 101
functional relationship between the immune and nervous systems. causes pruritus and erythema when injected intradermally. This
TYPICAL CUTANEOUS NERVE AND ACTIVATION OF C NERVE FIBERS IN ITCH TRANSMISSION

SECTION Typical cutaneous nerve Spinal cord

2 Dorsal root
ganglion
Pruritus

Peripheral
nerve Myelin

A
Myelinated
A
Cell body

Sympathetic
C C Mediator release by C fiber and mast cell
postganglionic

A Primary activation B Secondary activation


Substance P

H
Lower pH NK1 Itch
K+ Substance P receptor
H1
Prostaglandins receptor
Stimulus Mast cell
Impulses generated in stimulated
terminal propagate to other terminal Tryptase
branches as well as the spinal cord TNF-
Epidermis

D Augmentation by TNF- and tryptase E Augmentation by NGF F Induction and attenuation by the TRPV1 ion channel

Mast cell Chemotaxis and Mast cell Capsaicin, noxious heat,


Mast cell survival of mast cells lower pH, eicosanoids
TNF- (e.g. prostaglandins),
NGF
H neurotrophins (e.g. NGF)
Tryptase
Late: Late: substance P depletion
More substance P suspended
released More tryptase
interplay
released Early: substance P release
between
More substance P C fibers,
Cleaved, self-
released mast cells &
activated PAR2
TNF- keratinocytes
receptor
PAR2 receptor Early:
Sensitized Heterologous
C fibers release of sensitization Tryptase
Itch Keratinocyte
puritogenic
TRKA mediators
H
TRPV1 Early itch

Epidermal Sensitized Late: desensitization


hyperplasia C fibers Sprouting Keratinocyte & termination of itch

G Pathway of cowhage-induced itch H Transduction of itch by MRGPRs

PAR2 Keratinocyte Keratinocyte


Cathepsin S
BAM8-22
TRPV1
Cowhage Chloroquine MRGPR
(mucunain)

TRPA1
TRPA1

Cowhage-sensitive, nonhistaminergic,
Itch polymodal C fibers Itch C fibers

Fig. 5.2 See following page for legend.

102
Fig. 5.2 Typical cutaneous nerve and activation of C nerve fibers in itch response is only partially inhibited by the -opioid receptor antagonist
transmission. There are two categories of axons in a typical cutaneous nerve: naloxone but is substantially inhibited by topical pretreatment of the
primary afferent A, A and C fibers (cell bodies in dorsal root ganglion) and injected skin with the potent H1 antihistamine doxepin47. In contrast
sympathetic postganglionic fibers (cell bodies in sympathetic ganglion). to morphine, the highly potent -opioid agonist fentanyl does not
Separate C fibers (~5% of total) are known to carry pruritogenic stimuli via two CHAPTER
induce mast cell degranulation, even when applied in high doses.
pathways: (1) histamine-induced itch that is transmitted by mechanically
insensitive, capsaicin-sensitive (via transient receptor potential vanilloid
receptor 1 [TRPV1]) fibers; and (2) cowhage (mucunain)-induced itch
Therefore, opioids appear to induce itch via two possible mechanisms:
(1) degranulation of cutaneous mast cells48; and (2) activation of
5
-opioid receptors with a direct central and peripheral pruritogenic

Cutaneous Neurophysiology
accompanied by a burning sensation that is transmitted by polymodal fibers
(sensitive to capsaicin, mechanical and other stimuli). A In primary (direct) effect4851. Morphine-induced itch was recently shown to result from
activation of C fibers, the stimulus (e.g. impaired barrier function, activation of a heterodimeric receptor composed of the -opioid recep-
environmental changes or inflammation) induces lower pH, potassium release, tor and the gastrin-releasing peptide receptor; inhibition of the latter
or synthesis of prostaglandins (the latter increasing sensitivity of C-fiber blocked opioid-related itch but not analgesia, an observation that may
terminals to a number of substances). Environmental factors that increase have therapeutic relevance51a.
(more likely to result in itch) or decrease (more likely to result in pain) stratum Nociceptin, the endogenous peptide ligand for the opioid receptor-like
corneum pH can lead to C fiber activation. B In secondary activation, impulses
generated in the stimulated terminal propagate not only to the spinal cord but
1 (ORL1) receptor, has also been implicated in cutaneous inflamma-
also to other terminal branches (antidromic reflex arc), where they induce tion, pain and pruritus52. Investigations using a mouse model suggested
release of substance P. C Substance P primes mast cells via neurokinin-1 (NK1) that nociceptin interacts with ORL1 receptors expressed on keratino
receptors. Activated mast cells release histamine (H), which causes transmission cytes, leading to the production of leukotriene B4 that induces scratch-
of itch via H1 receptors on histamine-sensitive C fibers. Mast cells also secrete ing53. Nociceptin-induced scratching was significantly inhibited by
tumor necrosis factor (TNF)- and tryptase upon stimulation by substance P treatment with systemic naloxone.
and nerve growth factor (NGF), respectively. D TNF- sensitizes C fibers.
Tryptase cleaves the extracellular portion of proteinase-activated receptor-2
(PAR2) on C-fiber terminals, exposing a tethered ligand domain that binds and
Substance P
self-activates PAR2. This results in transmission of itch by the C fibers as well Substance P, a neuropeptide with a widespread distribution in peri
as release of additional substance P. E NGF released by mast cells and pheral nerves and the CNS, intensifies itch perception. Levels of sub-
keratinocytes (with increased production stimulated by histamine) activates stance P in the serum of patients with atopic dermatitis are elevated
tropomyosin-related kinase receptor A (TRKA) on C fibers, mast cells and and correlate with disease severity54. Intradermal injection of substance
keratinocytes. This induces C-fiber sprouting, sensitization and increased P provokes itch as well as elements of neurogenic inflammation such
substance P release; mast cell chemotaxis, survival and increased tryptase as erythema and the wheal-and-flare reaction. Substance P is synthe-
release; and epidermal hyperplasia. Of note, TRKA is also referred to as
sized in the cell bodies of C neurons, transported towards the peripheral
neurotrophic tyrosine kinase receptor type 1 (NTRK1). F The TRPV1 ion channel
on C fibers, keratinocytes and mast cells is activated by capsaicin, heat, low pH, nerve terminals, and released by antidromic depolarization to cause
eicosanoids and neurotrophins; although this initially stimulates transmission vasodilation and increased vascular permeability (see Fig. 5.2).
of itch and release of pruritogenic mediators, it may eventually lead to Although endogenously released substance P does not degranulate
desensitization, neuropeptide depletion and attenuation of itch. The mast cells in healthy human skin55 or cause any sensation at physio-
transmission of histamine-induced itch requires the presence of TRPV1. logic concentrations56, direct communication between nerve fibers and
G Cowhage-induced itch occurs through the release of mucunain, a protease mast cells via substance P has been verified57. High concentrations of
that activates PAR2 and PAR4 receptors (the latter not yet identified in the substance P can cause immediate mast cell degranulation, whereas low
skin); these receptors can also be activated by endogenous proteases such as concentrations specifically activate neurokinin-1 (NK-1) receptors on
cathepsin S. Activation of PAR2/4 sensitizes TRPV1 and TRP ankyrin 1 (TRPA1) mast cells, leading to sensitization of these cells and increased produc-
channels, resulting in cross-talk and itch transmission. H C nerve fibers
tion of tumor necrosis factor (TNF)-58. In turn, TNF- sensitizes
(including those responsive to histamine) in the epidermis also express
MAS-related G protein-coupled receptors (MRGPRs), which are stimulated by nociceptive nerve endings, producing a self-amplifying loop between
chloroquine or bovine adrenal medulla 8-22 peptide (BAM8-22; a neurons and mast cells.
proteolytically cleaved product of proenkephalin A). TRPA1 is required for Substance P co-localizes with other neurotransmitters such as sero-
transduction of itch by MRGPRs. tonin, dopamine and calcitonin gene-related peptide, and it can act as

MURINE MODELS OF ITCH

Model Clinical and laboratory features Pathogenesis/comments


Transgenic mice overexpressing PAR2 Epidermal hyperplasia and scaling as well as pruritus
and scratching
Transgenic mice overexpressing a serine protease Pruritus and scratching
TRPV1-deficient mice and Pirt-deficient mice Lack of response to pruritogens Pirt normally plays an important role in sensing itch
(histaminergic and non-histaminergic; also pain) via
TRPV1 channels (and also TRPV1-independent itch)
MRGPR-deficient mice and TRPA1-deficient mice Lack of scratching response to chloroquine and MGGPRs are normally activated by chloroquine and
BAM8-22, but preserved response to histamine BAM8-22, so the lack of response in TRPA1-deficient
mice implicates this receptor in downstream
signaling
GRPR-deficient mice Lacking of scratching response to pruritogens, but GRPR is expressed in the dorsal spinal cord
preserved response to painful stimuli
Bhlhb5-deficient mice Enhanced scratching response to pruritogens Selective loss of inhibitory interneurons in the
dorsal horn that regulate pruritus
-Opioid receptor-deficient mice Thinner epidermis, higher density of epidermal nerve
endings, and less scratching
Transgenic mice overexpressing interleukin-31 Pruritus, scratching, excoriations and alopecia
Table 5.3 Murine models of itch. Bhlhb5, basic helix-loop-helix family member B5; GRPR, gastrin-releasing peptide receptor; MRGPR, Mas-related G protein-
coupled receptors; PAR2, protease-activated receptor 2; Pirt, phosphoinositide-interacting regulator of transient receptor potential channels; TRPV1, transient 103
receptor potential vanilloid 1.
a neuromodulator. The relationship between substance P and keratino- mutation in TRPV3 results in chronic itch, scratching and an atopic-
cytes expressing vanilloid receptors remains to be elucidated. like dermatitis76. In contrast, TRP melastatin 8 (TRPM8) on C nerve
fibers functions as a thermosensor for cool (<28C, 82F) and is acti-
SECTION
Neurotrophins vated by menthol and icilin, which provide a cooling sensation that
has been used to relieve itch.
2 Neurotrophins are factors that regulate the growth and function of
nerve cells. The prototypic neurotrophic factor is nerve growth factor
TRP ankyrin 1 (TRPA1), a polymodal nociceptor, was recently found
to be a downstream mediator of histamine-independent itch stimulated
(NGF). Other members of this family include neurotrophins 3, 4 and
by MRGPRs (present in a subset of epidermal C fibers; see above) in
Pruritus

5 and brain-derived neurotrophic factor (BDNF)59. Increased levels of


mice77. Like TRPM8, TRPA1 can be activated by menthol.
epidermal NGF correlate with proliferation of terminal cutaneous
nerves and upregulation of neuropeptides (e.g. substance P)60. NGF is
known to induce sprouting of nerve fibers, sensitization of nerve Other Peripheral Mediators of Itch
endings, axonal transport in dorsal root ganglia cells, and increased Neurotransmitters
expression of neuropeptides (e.g. substance P)61. Neurotransmitters that play a role in itch include acetylcholine, an
Keratinocytes express high levels of NGF, and NGF not only is important neurotransmitter in the autonomic nervous system. Intra-
required for survival and regeneration of sensory neurons but also dermal injection of acetylcholine typically induces pain; however, in
controls the responsiveness of such neurons to external stimuli62,63. patients with chronic itch, it induces itch. In murine models of itch,
Patients with atopic dermatitis have increased expression of NGF activation of the muscarinic receptor 3 triggers pruritus78. Currently
in cutaneous mast cells, keratinocytes and fibroblasts64, and their there are no data regarding the role of norepinephrine, epinephrine or
plasma levels of NGF correlate with disease activity. There is growing dopamine in itch transmission.
evidence that NGF acts as a signaling molecule between mast cells and
keratinocytes in allergic skin diseases. Mast cell-derived histamine Peptidases
induces keratinocytes to increase NGF production, and the latter may
promote infiltration of mast cells into inflamed skin65,66. Upregulation Mast cells that express neuropeptides also generate neuropeptide-
of other neurotrophins, such as neurotrophin 4, has also been observed degrading peptidases such as angiotensin-converting enzyme (ACE) and
in keratinocytes from atopic dermatitis patients67. neural endopeptidases. The role they play in itch is unclear; however,
medications such as ACE inhibitors, which can induce pruritus without
a rash, may induce itch by inhibiting the activity of these degrading
Prostanoids enzymes.
In the skin, prostaglandins enhance histamine-induced itch68,69. Pros-
taglandins are the products of the transformation of the essential fatty Other mediators with potential roles in itch
acid arachidonic acid by cyclooxygenase-1 (COX-1) or cyclooxygenase-2 Serotonin has been implicated as an inducer of itch in murine models;
(COX-2). They are not by themselves pruritogenic when injected into however, in humans it is a very mild pruritogen. Nitric oxide is another
skin. However, intradermal injection of prostaglandin E1 (PGE1) factor that may induce itch via neurogenic inflammation79. The bovine
enhances pruritus due to histamine subsequently injected into the adrenal medulla 8-22 (BAM8-22) peptide, a proteolytically cleaved
same site70. It appears that only itch-mediating neurons that display product of proenkephalin A, is a potent activator of MRGPRs and can
lasting activation following exposure to histamine are excited by PGE2, stimulate itch, usually accompanied by a stinging or burning sensa-
and mechanosensitive fibers are unresponsive to both histamine and tion80. Chloroquine can also induce itch by activating MRGPRs, which
PGE2. are expressed by subset of C nerve fibers in the epidermis (see above).
PGE2 has also been shown to have a direct, low-level pruritogenic
effect both in healthy individuals and atopic dermatitis patients (without
inducing protein extravasation). This suggests that prostanoids periph- Immune Cells as Itch Mediators and Modulators
eral action is not solely via histamine and that prostanoids may potenti- Interactions between the nervous and immune systems in the skin
ate pruritus via other effects on nerve fibers. have important roles in itch induction81. Neuropeptides released by
Oral administration of aspirin, a cyclooxygenase inhibitor, does not cutaneous sensory nerves (e.g. substance P, calcitonin gene-related
ameliorate pruritus except in polycythemia vera71; however, topical peptide, vasointestinal peptide) can activate transcription factors and
application of aspirin may reduce pruritus in patients with chronic regulate the expression of adhesion molecules and proinflammatory
localized itch72. The role of other eicosanoids, including leukotrienes cytokines, thereby modulating immune and inflammatory responses41.
and 12-hydroxyeicosatetraenoic acid (12-HETE), in the pathogenesis of These neuropeptides also influence cellular proliferation and differen-
pruritus is unclear. In a mouse model, leukotriene B4 provoked tiation, tissue repair, and antigen presentation involving a number of
scratching53. cell types, including keratinocytes, mast cells, dermal microvascular
endothelial cells, and Langerhans cells. This interaction is bidirec-
Mediators that Activate Transient Receptor tional, as cytokines and chemokines are also able to regulate primary
nerve afferents via receptor activation.
Potential Receptors IL-2 is produced by activated T lymphocytes and causes pruritus
Neuromediators that activate ion channels belonging to the transient when injected intradermally82. High-dose IL-2 administered intrave-
receptor potential (TRP) family are also involved in the sensation of nously to patients with cancer (including stage IV melanoma) causes
itch. TRP vanilloid 1 (TRPV1) is a vanilloid receptor located on C nerve intense generalized pruritus. Moreover, treatment with topical cal-
fibers, dermal mast cells, dendritic cells and keratinocytes (see Fig. cineurin inhibitors, which block the production of IL-2, can result in
5.2)73. This receptor is activated by capsaicin, endogenous substances decreased itch. IL-2-induced pruritus is not reduced by antihistamines
such as cannabinoids (e.g. anandamide), prostaglandins and various nor nonsteroidal anti-inflammatory drugs, and whether it represents a
neurotrophins, as well as by acidosis and temperatures above 43C direct receptor-mediated effect or an indirect effect (e.g. via mast cells
(109F; mediates heat pain). Mechano-insensitive pruritoceptive or endothelial cells) remains to be determined.
C-nerve pathways can be activated by capsaicin, indicating that TRPV1 IL-31 is produced by T helper 2 (Th2) cells and induces pruritus by
is expressed, and the experimental induction of histamine- and modulating the function of sensory neurons. IL-31 may exert its pru-
serotonin-mediated itch requires cooperation of TRPV1 receptors74. In ritogenic effect via activation of the IL-31 receptor (IL-31R, a het-
addition, stimulation of TRPV1-positive nerve fibers releases multiple erodimer composed of the oncostatin M receptor [OSMR] protein and
pruritoceptive mediators (e.g. interleukins [ILs], neuropeptides). Of the IL-31 receptor A) on keratinocytes, which could subsequently stim-
note, cannabinoid receptors CB1 and CB2 are also found in the epider- ulate C fibers in the skin; the IL-31R is also found on cutaneous C
mis, and a cannabinoid agonist has been shown to inhibit histamine- fibers and in dorsal root ganglia. Signaling via the IL-31R and OSMR
induced itch75. (a heterodimer composed of the OSMR protein and a gp130 subunit)
TRPV3 is a thermosensor sensitive to temperatures >33C (91F; can result in keratinocyte proliferation, differentiation and apoptosis,
104 detects warmth) that is expressed in keratinocytes and dorsal root as well as cutaneous inflammation83. In addition, mutations in the gene
ganglion neurons in humans. In mice, a gain-of-function missense encoding the OSMR protein underlie familial primary localized
cutaneous amyloidosis, an autosomal dominant disorder characterized area of inflammation (including that induced by histamine iontophore-
by chronic localized itching and scratching that results in deposition of sis). It is similar to the phenomenon in chronic pain termed punctate
keratin-derived amyloid in the dermis84. Higher levels of IL-31 expres- hyperalgesia.
sion are found in the skin of patients with atopic dermatitis and prurigo In patients with chronic itch, painful stimuli (e.g. electrical stimuli,
CHAPTER
nodularis85,86, suggesting an important role for this cytokine in the heat pain) may be perceived as itch9395. An analogous phenomenon can
intense itching that characterizes these conditions.
TNF- is known to sensitize nociceptive nerve endings via its effect
occur in patients with chronic pain, in whom histamine iontophoresis
is perceived as painful96. These findings indicate that pain-induced
5
on TNF receptors (see Fig. 5.2); however, its role in itch is unclear. inhibition of pruritus may be compromised in patients with chronic

Cutaneous Neurophysiology
Although targeted TNF- inhibitors (see Ch. 128) do not directly itch. This may also explain why scratching aggravates itch in patients
decrease pruritus, thalidomide (which has anti-TNF- effects) can be with chronic itch and thereby induces a vicious itchscratch cycle.
effective in treating the itch associated with prurigo nodularis87.
IL-6 is expressed by immune, nerve and Schwann cells88, and levels Itch Related to Impaired Skin Barrier Function
of this cytokine are elevated in prurigo nodularis lesions89.
Itch is a common symptom of xerotic skin and is aggravated during
the winter in cold climates when the relative humidity falls indoors.
Chronic Itch Damage to the stratum corneum and the impaired barrier function that
Chronic itch may occur in the setting of pruritoceptive itch (originating results can induce itch even without inflammation. Environmental
from skin disease) or neuropathic itch (due to pathology in the nervous changes (e.g. in pH, temperature and humidity) may serve as triggers
system) and can also have systemic or psychiatric causes90. It often has that activate C nerve fibers to transmit the sensation of itch. Cross-talk
a significant effect on patients quality of life. Chronic itch and chronic between the stratum corneum and nerve fibers may explain the pruritus
pain share a number of features, with both potentially involving periph- associated with impaired barrier function. Studies have demonstrated
eral and central sensitization91 (Table 5.4). that keratinocytes release neuromediators upon damage to the stratum
corneum barrier, and nerve fibers sprout in the epidermis in response
Peripheral sensitization in chronic itch to this damage. In a murine model of dry skin, enhanced c-fos expres-
An increased density of cutaneous nerve endings is observed in some sion in the CNS was observed, reflecting an activation of axons97,98.
forms of chronic itch91. Patients with chronic itch in the setting of Additional evidence comes from a murine model of atopic dermatitis
atopic dermatitis have increased neurotrophin levels in involved skin, in which barrier damage was associated with significantly increased
including NGF and neurotrophin 467. Chronic localized pain is associ- scratching, and scratching activity diminished when the barrier was
ated with elevated levels of the same neurotrophins, which are known repaired99. Serine proteases that activate PAR2 (thereby stimulating
to sensitize nociceptive neurons59. itch) are secreted in response to an increasing (alkaline) stratum
corneum pH, which is commonly noted during barrier damage. This
Central sensitization in chronic itch suggests that environmental factors that increase stratum corneum pH
Chronic itch leads to sensitization of second-order neurons within the may increase itch perception (see Fig. 5.2). In contrast, a low-pH extra-
dorsal horn (see Fig. 5.1), thereby leading to increased sensitivity to cellular environment causes C nociceptors to induce pain.
itch. There are two forms of increased sensitivity to itch: alloknesis
and punctate hyperknesis. In alloknesis, stimuli that normally do not Senescent Skin and Itch
induce itch (e.g. touch, gentle warming) do so in the skin that sur- Itch is a particularly frequent dermatologic symptom in individuals
rounds a pruritic area92. This phenomenon is analogous to allodynia, over 65 years of age100,101. Although dry skin is probably the most
in which gentle mechanical stimuli give rise to a perception of pain. common trigger, elderly patients can have idiopathic itch without
Like allodynia, alloknesis requires ongoing activity in primary afferent xerosis. Other possible explanations include age-related changes in
C fibers and is mediated by low-threshold myelinated mechanoreceptor nerve fibers and central disinhibition of itch due to loss of input from
A fibers (see Fig. 5.2). Alloknesis is common and represents a promi- pain fibers. Additional cutaneous changes that may contribute to pru-
nent feature of atopic dermatitis; it explains patients complaints of ritus (as well as xerosis) in elderly patients include decreased skin
severe pruritus associated with sweating, sudden changes in tempera- surface lipids, decreased sweat and sebum production, and diminished
ture, dressing and undressing. Punctate hyperknesis is characterized by barrier repair102.
more intense prick-induced itch sensations in the zone surrounding an

TREATMENT OF PRURITUS
Pruritus can significantly impair the quality of life in affected individu-
als. Acute itch may lead to agitation and difficulty concentrating, while
COMPARISON OF CHARACTERISTICS OF CHRONIC ITCH AND
chronic pruritus can have sequelae such as depression and decreased
CHRONIC PAIN
sexual desire or function103.
Chronic itch Chronic pain

Peripheral Peripheral sensitization of C Peripheral sensitization


Pharmacologic Treatments
sensitization nerve fibers of C nerve fibers Unlike pain, for which a host of effective medications are available,
there are no general-purpose, consistently beneficial antipruritic drugs.
Central Alloknesis Allodynia
However, recent discoveries of specific neural networks that are involved
sensitization Punctate hyperknesis Punctate hyperalgesia
in itch transmission have led to advances in this relatively unexplored
Neuromediators Nerve growth factor Nerve growth factor area of medicine. The mechanisms, uses and side effects of currently
Neurotrophin 4 Neurotrophin 4 available antipruritic drug treatments are outlined in Table 5.5. Of note,
Chemical Histamine Bradykinin although capable of relieving pruritus due to inflammatory skin disease,
mediators Cowhage (mucunain) Most opioids reduce pain corticosteroids are not intrinsically antipruritic.
Most opioids induce itch
CNS areas Anterior and posterior cingulate Anterior cingulate cortex Non-Pharmacologic Treatments
activated cortex Premotor area
Premotor area Somatosensory cortex I, II Cutaneous field stimulation
Precuneus Dorsolateral prefrontal Cutaneous field stimulation (CFS) is a technique that electrically stim-
Supramarginal gyrus cortex ulates afferent fibers, including nociceptive C fibers. CFS is similar to
Dorsolateral prefrontal cortex Insula-claustrum complex transcutaneous electrical nerve stimulation (TENS), but the latter acti-
Insula-claustrum complex
vates large myelinated nerve fibers while CFS specifically targets unmy-
Somatosensory cortex I, II
elinated C nerve fibers. In a study of patients with localized pruritus, 105
Table 5.4 Comparison of characteristics of chronic itch and chronic pain. CFS significantly reduced patient-reported itch and caused degeneration
DRUG TREATMENT OF PRURITUS

Medication Mechanism(s) of action Uses and efficacy Major adverse effects


SECTION
TOPICAL AGENTS
2 Capsaicin (active
component in
Activates and subsequently desensitizes
TRPV1, a vanilloid TRP receptor107
Used primarily for localized, chronic
pruritic disorders, especially those of
Transient burning sensation, which
usually resolves after several days
hot chili Causes release from C neurons and neuropathic origin109 of consistent use; application of
Pruritus

peppers) (eventually) depleted stores of neuropeptides Controlled studies support use for: topical lidocaine or EMLA may be
(e.g. substance P, CGRP, somatostatin) Notalgia paresthetica helpful
Prolonged use leads to neuronal degeneration Brachioradial pruritus
at sites of application, which can result in a Hemodialysis-associated pruritus
reversible decrease in epidermal nerve fiber Postherpetic neuralgia (transdermal
density108 patch FDA-approved for this
indication)110
Doxepin Tricyclic compound with potent antihistamine Shown to relieve pruritus in patients with Drowsiness in 25% of patients due
effects (H1 and H2); also anticholinergic atopic dermatitis111 to percutaneous absorption
properties Allergic contact dermatitis
Menthol Activates TRPM8 and TRPA1, producing a cool Inconsistent results in controlled Skin irritation
sensation112 studies113,114
May be especially beneficial for patients
whose itch is relieved by cold showers
Pramoxine Anesthetic (blocks transmission of nerve Beneficial for histamine-induced itch and Allergic contact dermatitis
impulses) uremic pruritus in end-stage renal (uncommon)
disease115
Tacrolimus, Calcineurin inhibitors that block production of Can decrease itch related to the Transient burning sensation
pimecrolimus multiple proinflammatory cytokines (see Ch. inflammation of atopic dermatitis
128)
May initially activate and subsequently
desensitize TRPV1116
Barrier repair Diminish transepidermal water loss and Decrease itch in patients with atopic None
creams, other decrease flux between a distended and dermatitis
emollients and desiccated state
humectants Minimize microfissures that expose C nerve
fibers
Agents that acidify the stratum corneum may
reduce itch, as alkaline agents (e.g. bar soaps)
can activate proteases and increase lipid
rigidity
SYSTEMIC AGENTS
Antihistamines Doxepin is a tricyclic compound with potent Decrease wheal formation as well as Sedation
(especially antihistamine effects (H1 and H2) and pruritus in patients with urticaria Anticholinergic effects
doxepin) anticholinergic properties; it represents a Interaction with MAO inhibitors
first-generation (classic), sedating (for doxepin)
antihistamine
Naloxone, -Opioid receptor antagonists* Controlled trials show efficacy for Hepatotoxicity
naltrexone cholestatic and renal pruritus117121 Nausea/vomiting
May be of benefit for severe pruritus of Insomnia
other etiologies122 Reversal of analgesia
Nalfurafine -Opioid receptor agonist* Controlled trials show efficacy for renal Insomnia
(TRK-820) pruritus123
Butorphanol -Opioid receptor agonist and -opioid Relieves pruritus associated with morphine Nausea/vomiting
receptor antagonist* (with epidural administration of both Dependence (rare)
agents)124
May reduce severe pruritus associated with
systemic disease or inflammatory skin
conditions125
Mirtazapine Serotonin and norepinephrine inverse agonist May reduce nocturnal pruritus (at low Sedation
that reduces central itch perception doses, e.g. 15mg nightly)126 Weight gain
Paroxetine Selective serotonin reuptake inhibitor (SSRI) A controlled study showed benefit for Sexual dysfunction
pruritus related to systemic disease127 Sedation
Insomnia
Weight gain
*Inhibit itch transmission within the central nervous system.

Currently not commercially available in the US.

An intranasal spray is currently available, and transdermal formulations are being developed.

Table 5.5 Drug treatment of pruritus. Topical naltrexone104, aspirin and cannabinoids may also have antipruritic effects, and injections of botulinum toxin type A
have been used to treat neuropathic itch105. In the future, topical drugs for the relief of itch may include prostaglandin DP1 receptor agonists106, and agents that
inhibit serine proteases (e.g. cathepsin S), nerve growth factor or neurotrophin 4. CGRP, calcitonin gene-related polypeptide; EMLA, eutectic mixture of local
anesthetics (lidocaine + prilocaine); FDA, US Food and Drug Administration; MAO, monoamine oxidase; TRP, transient receptor potential ion channel family.
Continued

106
DRUG TREATMENT OF PRURITUS

Medication Mechanism(s) of action Uses and efficacy Major adverse effects


CHAPTER
Thalidomide Blocks neuropathic afferent pathways May be helpful for prurigo nodularis, Teratogenicity
(peripherally and centrally)
Inhibits TNF- synthesis
actinic prurigo and pruritus of advanced
age
Peripheral neuropathy 5

Cutaneous Neurophysiology
Gabapentin, Structural analogues of the neurotransmitter Beneficial for neuropathic itch (e.g. Peripheral edema
pregabalin GABA brachioradial pruritus), post-burn pruritus Sedation
Thought to inhibit central itch pathways and postherpetic neuralgia128130a Abdominal pain
Aprepitant Antagonist of the neurokinin-1 receptor, Primarily used as an antiemetic in Fatigue
which is activated by substance P oncology patients, with anecdotal reports Hiccups
of reduced pruritus (e.g. in Szary
syndrome or erlotinib-induced pruritus)
Table 5.5 Drug treatment of pruritus (contd). Topical naltrexone104, aspirin and cannabinoids may also have antipruritic effects, and injections of botulinum toxin
type A have been used to treat neuropathic itch105. In the future, topical drugs for the relief of itch may include prostaglandin DP1 receptor agonists106, and agents
that inhibit serine proteases (e.g. cathepsin S), nerve growth factor or neurotrophin 4. GABA, -aminobutyric acid; TNF, tumor necrosis factor.

of epidermal nerve fibers131. However, CFS is practical only for localized shown that behavioral therapy reduces the intensity of itch that is
disease. perceived135.

Behavioral therapy targeting the CNS


FUTURE DIRECTIONS
Stress and other psychogenic factors are important in itch132. Patients
with pruritus experience higher levels of psychological stress than One of the most critical issues is to identify the specific mechanisms
those without this symptom133. An impaired response of the autonomic and mediators responsible for particular itch states. Hopefully, our
nervous system to itch, scratching and emotional stress has also emerging understanding of the pathways of itch transmission will soon
been identified in atopic dermatitis patients134. Several studies have result in the development of effective targeted treatments.

REFERENCES
1. Yosipovitch G, Greaves MW, Schmelz M. Itch. Lancet. 16. Sun YG, Zhao ZQ, Meng XL, et al. Cellular basis of itch induced itch. Neuroimage. 2011 doi: 10.1016/
2003;361:6904. sensation. Science. 2009;325:15314. j.neuroimage.2011.10.099.
2. Yosipovitch G, Greaves MW, Fleischer AB Jr, McGlone F 17. Liu T, Xu ZZ, Park CK, et al. Toll-like receptor 7 mediates 29. Ishiuji Y, Coghill RC, Patel TS, et al. Distinct patterns of
(eds). Itch: Basic Mechanisms and Therapy. New York: pruritus. Nat Neurosci. 2010;13:14602. brain activity evoked by histamine-induced itch reveal
Marcel Dekker, 2004. 18. Dawn A, Papoiu AD, Chan YH, et al. Itch characteristics an association with itch intensity and disease severity
3. Rea JN, Newhouse ML, Halil T. Skin disease in Lambeth. in atopic dermatitis: results of a web-based in atopic dermatitis. Br J Dermatol. 2009;161:107280.
A community study of prevalence and use of medical questionnaire. Br J Dermatol. 2009;160:6424. 30. Yosipovitch G, Ishiuji Y, Patel TS, et al. The brain
care. Br J Prev Soc Med. 1976;30:10714. 19. Yosipovitch G, Ansari N, Goon A, et al. Clinical processing of scratching. J Invest Dermatol.
4. Davidson S, Giesler G Jr. The multiple pathways for itch characteristics of pruritus in chronic idiopathic urticaria. 2008;128:180611.
and their interactions with pain. Trends Neurosci. Br J Dermatol. 2002;147:326. 31. Vierow V, Fukuoka M, Ikoma A, et al. Cerebral
2010;33:5508. 20. Yosipovitch G, Goon AT, Wee J, et al. Itch characteristics representation of the relief of itch by scratching.
5. Schmelz M. Itch and pain. Neurosci Biobehav Rev. in Chinese patients with atopic dermatitis using a new J Neurophysiol. 2009;102:321624.
2010;34:1716. questionnaire for the assessment of pruritus. Int J 32. Davidson S, Zhang X, Khasabov SG, et al. Relief of itch
6. Handwerker HO, Schmelz M. Pain: itch without pain a Dermatol. 2002;41:2126. by scratching: state-dependent inhibition of primate
labeled line for itch sensation? Nat Rev Neurol. 21. Darsow U, Scharein E, Simon D, et al. New aspects of spinothalamic tract neurons. Nat Neurosci.
2009;5:6401. itch pathophysiology: component analysis of atopic 2009;12:5446.
7. Schmelz M, Schmidt R, Weidner C, et al. Chemical itch using the Eppendorf Itch Questionnaire. Int Arch 33. Mochizuki H, Sadato N, Saito DN, et al. Neural correlates
response pattern of different classes of C-nociceptors to Allergy Immunol. 2001;124:32631. of perceptual difference between itching and pain:
pruritogens and algogens. J Neurophysiol. 22. Yosipovitch G. Itch questionnaires as tools for itch a human fMRI study. Neuroimage. 2007;36:70617.
2003;89:24418. evaluation. In: Yosipovitch G, Greaves MW, Erratum in: Neuroimage. 2008;39:91112.
8. Fruhstorfer H, Hermanns M, Latzke L. The effects of Fleischer AB Jr, McGlone F (eds). Itch: Basic 34. Papoiu AD, Wang H, Coghill RC, et al. Contagious itch in
thermal stimulation on clinical and experimental itch. Mechanisms and Therapy. New York: Marcel Dekker, humans: a study of visual transmission of itch in atopic
Pain. 1986;24:25969. 2004:16982. dermatitis and healthy subjects. Br J Dermatol.
9. Johanek L, Meyer R, Hartke T, et al. Psychophysical and 23. Andrew D, Craig AD. Spinothalamic lamina I neurons 2011;164:1299303.
physiological evidence for parallel afferent pathways selectively sensitive to histamine: a central neural 35. Hilliges M, Wang L, Johansson O. Ultrastructural
mediating the sensation of itch. J Neurosci. pathway for itch. Nat Neurosci. 2001;4:727. evidence for nerve fibers within all vital layers of
2007;27:74907. 24. Hsieh JC, Hagermark O, Stahle-Backdahl M, et al. Urge the human epidermis. J Invest Dermatol. 1995;104
10. Namer B, Carr R, Johanek LM, et al. Separate peripheral to scratch represented in the human cerebral cortex :1347.
pathways for pruritus in man. J Neurophysiol. during itch. J Neurophysiol. 1994;72:30048. 36. Liu Q, Tang Z, Surdenikova L, et al. Sensory neuron-
2008;100:20629. 25. Drzezga A, Darsow U, Treede RD, et al. Central specific GPCR Mrgprs are itch receptors mediating
11. Davidson S, Zhang X, Yoon CH, et al. The itch-producing activation by histamine-induced itch: analogies to chloroquine-induced pruritus. Cell. 2009;139:
agents histamine and cowhage activate separate pain processing: a correlational analysis of O-15 H2O 135365.
populations of primate spinothalamic tract neurons. positron emission tomography studies. Pain. 2001;92: 37. VB Reddy, SG Shimada, P Sikand, et al. Cathepsin S
J Neurosci. 2007;27:1000714. 295305. elicits itch and signals via protease-activated receptors.
12. Shelley WB, Arthur RP. Mucunain, the active 26. McGlone F, Rukweid R, Howard M, Hitchcock D. J Invest Dermatol. 2010;130:46870.
pruritogenic proteinase of cowhage. Science. Histamine-induced discriminative and affective 38. Hide M, Francis DM, Grattan CE, et al. Autoantibodies
1955;122:46970. responses revealed by functional MRI. In: Yosipovitch G, against the high-affinity IgE receptor as a cause of
13. Reddy VB, Iuga AO, Shimada SG, et al. Cowhage-evoked Greaves MW, Fleischer AB Jr, McGlone F (eds). Itch: Basic histamine release in chronic urticaria. N Engl J Med.
itch is mediated by a novel cysteine protease: a ligand Mechanisms and Therapy. New York: Marcel Decker, 1993;328:1599604.
of protease-activated receptors. J Neurosci. 2004:5161. 39. Engelhardt H, Smits RA, Leurs R, et al. A new generation
2008;28:43315. 27. Darsow U, Drzezga A, Frisch M, et al. Processing of of anti-histamines: histamine H4 receptor antagonists
14. Steinhoff M, Neisius U, Ikoma A, et al. Proteinase- histamine-induced itch in the human cerebral cortex: on their way to the clinic. Curr Opin Drug Discov Devel.
activated receptor-2 mediates itch: a novel pathway for a correlation analysis with dermal reactions. J Invest 2009;12:62843
pruritus in human skin. J Neurosci. 2003;23:617680. Dermatol. 2000;115:102933. 40. Huang JF, Thurmond RL. The new biology of histamine
15. Sun YG, Chen ZF. A gastrin-releasing peptide receptor 28. Papoiu ADP, Coghill RC, Kraft RA, et al. A Tale of two receptors. Curr Allergy Asthma Rep. 2008;8:217.
mediates the itch sensation in the spinal cord. Nature. itches. Common features and notable differences in 41. Luger TA. Neuromediators a crucial component of the 107
2007;448:7003. brain activation evoked by cowhage and histamine skin immune system. J Dermatol Sci. 2002;30:8793.
42. Grobe K, Poppelmann M, Becker WM, Petersen A. atopic dermatitis: an immunohistochemical study. Arch 90. Twycross R, Greaves MW, Handwerker H, et al. Itch:
Properties of group I allergens from grass pollen and Dermatol Res. 2006;298:317. scratching more than the surface. QJM. 2003;96:726.
their relation to cathepsin B, a member of the C1 family 67. Grewe M, Vogelsang K, Ruzicka T, et al. Neurotrophin-4 91. Yosipovitch G, Carstens E, McGlone F. Chronic itch and
of cysteine proteinases. Eur J Biochem. production by human epidermal keratinocytes: chronic pain: Analogous mechanisms. Pain.
2002;269:208392. increased expression in atopic dermatitis. J Invest 2007;131:47.
SECTION 43. Pan ZZ. mu-Opposing actions of the kappa-opioid Dermatol. 2000;114:110812. 92. Schmelz M. Itch and pain. Dermatol Ther.

2 receptor. Trends Pharmacol Sci. 1998;19:948.


44. Umeuchi H, Togashi Y, Honda T, et al. Involvement of
68. Hagermark O, Strandberg K, Hamberg M. Potentiation
of itch and flare responses in human skin by
2005;18:3047.
93. Ikoma A, Handwerker H, Miyachi Y, Schmelz M.
central mu-opioid system in the scratching behavior in prostaglandins E2 and H2 and a prostaglandin Electrically evoked itch in humans. Pain.
Pruritus

mice, and the suppression of it by the activation of endoperoxide analog. J Invest Dermatol. 2005;113:14854.
kappa-opioid system. Eur J Pharmacol. 2003;477: 1977;69:52730. 94. Ikoma A, Fartasch M, Heyer G, et al. Painful stimuli
2935. 69. Hagermark O, Strandberg K. Pruritogenic activity of evoke itch in patients with chronic pruritus: central
45. Togashi Y, Umeuchi H, Okano K, et al. Antipruritic prostaglandin E2. Acta Derm Venereol. 1977;57:3743. sensitization for itch. Neurology. 2004;62:2127.
activity of the kappa-opioid receptor agonist, TRK-820. 70. Greaves MW, McDonald-Gibson W. Itch: role of 95. Ishiuji Y, Coghill RC, Patel TS, et al. Repetitive scratching
Eur J Pharmacol. 2002;435:25964. prostaglandins. Br Med J. 1973;3:6089. and noxious heat do not inhibit histamine-induced itch
46. Bigliardi PL, Tobin DJ, Gaveriaux-Ruff C, Bigliardi-Qi M. 71. Daly BM, Shuster S. Effect of aspirin on pruritus. Br Med in atopic dermatitis. Br J Dermatol. 2008;158:7883.
Opioids and the skinwhere do we stand? Exp J (Clin Res Ed). 1986;293:907. 96. Binder A, Koroschetz J, Baron R Disease mechanisms
Dermatol. 2009;18:42430. 72. Yosipovitch G, Sugeng MW, Chan YH, et al. The in neuropathic itch. Nat Clin Pract Neurol. 2008;4:
47. Heyer G, Dotzer M, Diepgen TL, Handwerker HO. Opiate effect of topically applied aspirin on localized 32937.
and H1 antagonist effects on histamine induced circumscribed neurodermatitis. J Am Acad Dermatol. 97. Akiyama T, Merrill AW, Zanotto K, et al. Scratching
pruritus and alloknesis. Pain. 1997;73:23943. 2001;45:9103. behavior and Fos expression in superficial dorsal horn
48. Fjellner B, Hagermark O. Potentiation of histamine- 73. Stander S, Moormann C, Schumacher M, et al. elicited by protease-activated receptor agonists and
induced itch and flare responses in human skin by the Expression of vanilloid receptor subtype 1 in cutaneous other itch mediators in mice. J Pharmacol Exp Ther.
enkephalin analogue FK-33-824, beta-endorphin and sensory nerve fibers, mast cells, and epithelial cells of 2009;329:94551.
morphine. Arch Dermatol Res. 1982;274:2937. appendage structures. Exp Dermatol. 2004;13:12939. 98. Nojima H, Carstens MI,Carstens E. c-fos expression in
49. Fjellner B, Hagermark O. The influence of the opiate 74. Imamachi N, Park GH, Lee H, et al. TRPV1-expressing superficial dorsal horn of cervical spinal cord associated
antagonist naloxone on experimental pruritus. Acta primary afferents generate behavioral responses to with spontaneous scratching in rats with dry skin.
Derm Venereol. 1984;64:735. pruritogens via multiple mechanisms. Proc Natl Acad Neurosci Lett. 2003;347:624.
50. Hagermark O. Peripheral and central mediators of itch. Sci U S A. 2009;106:113305. 99. Miyamoto T, Nojima H, Shinkado T, et al. Itch-associated
Skin Pharmacol. 1992;5:18. 75. Rukweid R, Dvorak M, Watkinson A, McGlone F. Putative response induced by experimental dry skin in mice. Jpn
51. Bernstein JE, Swift R. Relief of intractable pruritus with role of cannabinoids in experimentally induced itch J Pharmacol. 2002;88:28592.
naloxone. Arch Dermatol. 1979;115:13667. and inflammation in human skin. In: Yosipovitch G, 100. Beauregard S, Gilchrest BA. A survey of skin problems
51a. Liu XY, Liu ZC, Sun YG, et al. Unidirectional cross- Greaves MW, Fleischer AB Jr, McGlone F (eds). Itch: and skin care regimens in the elderly. Arch Dermatol.
activation of GRPR by MOR1D uncouples itch and Basic Mechanisms and Therapy. New York: Marcel 1987;123:163843.
analgesia induced by opioids. Cell. 2011;147:44758. Dekker, 2004:11530. 101. Norman RA. Xerosis and pruritus in the elderly:
52. Meunier JC. Nociceptin/orphanin FQ and the opioid 76. Yoshioka T, Imura K, Asakawa M, et al. Impact of the recognition and management. Dermatol Ther.
receptor-like ORL1 receptor. Eur J Pharmacol. Gly573Ser substitution in TRPV3 on the development of 2003;16:2549.
1997;340:115. allergic and pruritic dermatitis in mice. J Invest 102. Ward JR, Bernhard JD. Willans itch and other causes of
53. Andoh T, Yageta Y, Takeshima H, Kuraishi Y. Intradermal Dermatol. 2009;129:71422. pruritus in the elderly. Int Dermatol. 2005;44:26773.
nociceptin elicits itch-associated responses through 77. Wilson SR, Gerhold KA, Bifolck-Fisher A, et al. TRPA1 is 103. Yosipovitch G. Pruritus: an update. Curr Probl Dermatol.
leukotriene B(4) in mice. J Invest Dermatol. required for histamine-independent, Mas-related G 2003;15:13764.
2004;123:196201. protein-coupled receptor-mediated itch. Nat Neurosci. 104. Bigliardi PL, Stammer H, Jost G, et al. Treatment of
54. Toyoda M, Nakamura M, Makino T, et al. Nerve growth 2011;14:595602. pruritus with topically applied opiate receptor
factor and substance P are useful plasma markers of 78. Miyamoto T, Nojima H, Kuraishi Y. Intradermal antagonist. J Am Acad Dermatol. 2007;56:97988.
disease activity in atopic dermatitis. Br J Dermatol. cholinergic agonists induce itch-associated response 105. Wallengren J, Bartosik J. Botulinum toxin type A for
2002;147:719. via M3 muscarinic acetylcholine receptors in mice. Jpn J neuropathic itch. Br J Dermatol. 2010;163:4246.
55. Schmelz M, Zeck S, Raithel M, Rukwied R. Mast cell Pharmacol. 2002;88:3514. 106. Arai I, Takaoka A, Hashimoto Y, et al. Effects of TS-022,
tryptase in dermal neurogenic inflammation. Clin Exp 79. Andoh T, Kuraishi Y. Nitric oxide enhances substance a newly developed prostanoid DP1 receptor agonist,
Allergy. 1999;29:695702. P-induced itch-associated responses in mice. Br J on experimental pruritus, cutaneous barrier
56. Weidner C, Klede M, Rukwied R, et al. Acute effects of Pharmacol. 2003;138:2028. disruptions and atopic dermatitis in mice. Eur J
substance P and calcitonin gene-related peptide in 80. Sikand P, Dong X, Lamotte RH. BAM8-22 peptide Pharmacol. 2007;556:20714.
human skin a microdialysis study. J Invest Dermatol. produces itch and nociceptive sensations in humans 107. Inoue K, Koizumi S, Fuziwara S, et al. Functional
2000;115:101520. independent of histamine release. J Neurosci. vanilloid receptors in cultured normal human
57. Suzuki R, Furuno T, McKay DM, et al. Direct neurite- 2011;31:75637. epidermal keratinocytes. Biochem Biophys Res
mast cell communication in vitro occurs via the 81. Steinhoff M, Bienenstock J, Schmelz M, et al. Commun. 2002;291:1249.
neuropeptide substance P. J Immunol. Neurophysiological, neuroimmunological, and 108. Simone DA, Nolano M, Johnson T, et al. Intradermal
1999;163:241015. neuroendocrine basis of pruritus. J Invest Dermatol. injection of capsaicin in humans produces
58. Cocchiara R, Lampiasi N, Albeggiani G, et al. Mast cell 2006;126:170518. degeneration and subsequent reinnervation of
production of TNF-alpha induced by substance P 82. Wahlgren CF, Tengvall LM, Hagermark O, Scheynius A. epidermal nerve fibers: correlation with sensory
evidence for a modulatory role of substance Itch and inflammation induced by intradermally function. J Neurosci. 1998;18:894759.
P-antagonists. J Neuroimmunol. 1999;101:12836. injected interleukin-2 in atopic dermatitis patients 109. Papoiu ADP, Yosipovitch G. Topical capsaicin. The fire of
59. Mendell LM, Albers KM, Davis BM. Neurotrophins, and healthy subjects. Arch Dermatol Res. 1995;287: a hot medicine is reignited. Expert Opin
nociceptors, and pain. Microsc Res Tech. 1999;45: 57280. Pharmacother. 2010;11:135971.
25261. 83. Zhang Q, Putheti P, Zhou Q, et al. Structures and 110. Backonja MM, Malan TP, Vanhove GF, Tobias JK.
60. Nockher WA. Neurotrophins in allergic diseases: from biological functions of IL-31 and IL-31 receptors. NGX-4010, a high-concentration capsaicin patch, for
neuronal growth factors to intercellular signaling Cytokine Growth Factor Rev. 2008;19:34756. the treatment of postherpetic neuralgia: a randomized,
molecules. J Allergy Clin Immunol. 2006;117:5839. 84. Arita K, South AP, Hans-Filho G, et al. Oncostatin M double-blind, controlled study with an open label
61. Donnerer J, Schuligoi R, Amann R. Increased content receptor-beta mutations underlie familial primary extension. Pain Med. 2011;12:99109.
and transport of substance P and calcitonin gene- localized cutaneous amyloidosis. Am J Hum Genet. 111. Drake LA, Fallon JD, Sober A. Relief of pruritus in
related peptide in sensory nerves innervating inflamed 2008;82:7380. patients with atopic dermatitis after treatment with
tissue: evidence for a regulatory function of nerve 85. Sonkoly E, Muller A, Lauerma AI, et al. IL-31: a new link topical doxepin cream. The Doxepin Study Group. J Am
growth factor in vivo. Neuroscience. 1992;49:6938. between T cells and pruritus in atopic skin Acad Dermatol. 1994;31:6136.
62. Nakamura M, Toyoda M, Morohashi M. Pruritogenic inflammation. J Allergy Clin Immunol. 2006;117:4117. 112. Peier AM, Moqrich A, Hergarden AC, et al. A TRP
mediators in psoriasis vulgaris: comparative evaluation 86. Neis MM, Peters B, Dreuw A, et al. Enhanced expression channel that senses cold stimuli and menthol. Cell.
of itch-associated cutaneous factors. Br J Dermatol. levels of IL-31 correlate with IL-4 and IL-13 in atopic and 2002;108:70515.
2003;149:71830. allergic contact dermatitis. J Allergy Clin Immunol. 113. Bromm B, Scharein E, Darsow U, Ring J. Effects of
63. Raychaudhuri SP, Raychaudhuri SK. Role of NGF and 2006;118:9307. menthol and cold on histamine-induced itch and skin
neurogenic inflammation in the pathogenesis of 87. Maurer T, Poncelet A, Berger T. Thalidomide treatment reactions in man. Neurosci Lett. 1995;187:15760.
psoriasis. Prog Brain Res. 2004;146:4337. for prurigo nodularis in human immunodeficiency 114. Yosipovitch G, Szolar C, Hui XY, Maibach H. Effect of
64. Groneberg DA, Serowka F, Peckenschneider N, et al. virus-infected subjects: efficacy and risk of neuropathy. topically applied menthol on thermal, pain and itch
Gene expression and regulation of nerve growth factor Arch Dermatol. 2004;140:8459. sensations and biophysical properties of the skin. Arch
in atopic dermatitis mast cells and the human mast cell 88. Grothe C, Heese K, Meisinger C, et al. Expression of Dermatol Res. 1996;288:2458.
line-1. J Neuroimmunol. 2005;161:8792. interleukin-6 and its receptor in the sciatic nerve and 115. Young TA, Patel TS, Camacho F, et al. A pramoxine-
65. Kanda N, Watanabe S. Histamine enhances the cultured Schwann cells: relation to 18-kD fibroblast based anti-itch lotion is more effective than a control
production of granulocyte-macrophage colony- growth factor-2. Brain Res. 2000;885:17281. lotion for the treatment of uremic pruritus in adult
stimulating factor via protein kinase C(alpha) and 89. Nordlind K, Chin LB, Ahmed AA, et al. hemodialysis patients. J Dermatol Treat. 2009;20:
extracellular signal-regulated kinase in human Immunohistochemical localization of interleukin-6-like 7681.
keratinocytes. J Invest Dermatol. 2004;122:86372. immunoreactivity to peripheral nerve-like structures in 116. Pereira U, Boulais N, Lebonvallet N, et al. Mechanisms of
108 66. Dou YC, Hagstromer L, Emtestam L, Johansson O. normal and inflamed human skin. Arch Dermatol Res. sensory effects of tacrolimus on the skin. Br J Dermatol.
Increased nerve growth factor and its receptors in 1996;288:4315. 2010;163:707.
117. Peer G, Kivity S, Agami O, et al. Randomised cross-over randomized, double-blind, placebo-controlled clinical combination in the relief of post-burn pruritus. Burns.
trial of naltrexone in uraemic pruritus. Lancet. studies. J Am Soc Nephrol. 2005;16:37427. 2011;37:2037.
1996;348:15524. 124. Gunter JB, McAuliffe J, Gregg T, et al. Continuous 131. Wallengren J, Sundler F. Cutaneous field stimulation in
118. Wolfhagen FH, Sternieri E, Hop WC, et al. Oral epidural butorphanol relieves pruritus associated with the treatment of severe itch. Arch Dermatol.
naltrexone treatment for cholestatic pruritus: a epidural morphine infusions in children. Paediatr 2001;137:13235.
CHAPTER
double-blind, placebo-controlled study. Anaesth. 2000;10:16772. 132. Gieler U, Niemeier V, Brosig B, Kupfer J. Psychosomatic
Gastroenterology. 1997;113:12649.
119. Terg R, Coronel E, Sorda J, et al. Efficacy and safety of
125. Dawn AG, Yosipovitch G. Butorphanol for treatment of
intractable pruritus. J Am Acad Dermatol.
aspects of pruritus. In: Yosipovitch G, Greaves MW,
Fleischer AB Jr, McGlone F (eds). Itch: Basic Mechanisms 5
oral naltrexone treatment for pruritus of cholestasis, 2006;54:52731. and Therapy. New York: Marcel Dekker, 2004:34350.

Cutaneous Neurophysiology
a crossover, double blind, placebo-controlled study. 126. Hundley JL, Yosipovitch G. Mirtazapine for reducing 133. Yamamoto Y, Yamazaki S, Hayashino Y, et al. Association
J Hepatol. 2002;37:71722. nocturnal itch in patients with chronic pruritus: a pilot between frequency of pruritic symptoms and perceived
120. Bergasa NV, Talbot TL, Alling DW, et al. A controlled trial study. J Am Acad Dermatol. 2004;50:88991. psychological stress: a Japanese population-based
of naloxone infusions for the pruritus of chronic 127. Zylicz Z, Krajnik M, Sorge AA, Costantini M. Paroxetine study. Arch Dermatol. 2009;145:13848.
cholestasis. Gastroenterology. 1992;102:5449. in the treatment of severe non-dermatological pruritus: 134. Tran BW, Papoiu ADP, Russoniello C, et al. The effect of
121. Bergasa NV, Alling DW, Talbot TL, et al. Effects of a randomized, controlled trial. J Pain Symptom Manage. itch, scratching and mental stress on autonomic
naloxone infusions in patients with the pruritus of 2003;26:110512. nervous system function in atopic dermatitis. Acta
cholestasis. A double-blind, randomized, controlled 128. Ehrchen J, Stnder S. Pregabalin in the treatment of Derm Venereol. 2010;90:35461.
trial. Ann Intern Med. 1995;123:1617. chronic pruritus. J Am Acad Dermatol. 2008;58:S36S37. 135. Gieler U. Psychosomatic aspects of pruritus. In:
122. Metze D, Reimann S, Beissert S, Luger T. Efficacy and 129. Yosipovitch G, Samuel LS. Neuropathic and Yosipovitch G, Greaves MW, Fleischer AB, McGlone F
safety of naltrexone, an oral opiate receptor antagonist, psychogenic itch. Dermatol Ther. 2008;21:3241. (eds): Itch: Basic Mechanisms and Therapy. New York:
in the treatment of pruritus in internal and 130. Porzio G, Aielli F, Verna L, et al. Efficacy of pregabalin in Marcel Dekker, 2004:3439.
dermatological diseases. J Am Acad Dermatol. the management of cetuximab-related itch. J Pain
1999;41:5339. Symptom Manage. 2006;32:3978.
123. Wikstrom B, Gellert R, Ladefoged SD, et al. Kappa- 130a. Ahuja RB, Gupta R, Gupta G, Shrivastava P. A
opioid system in uremic pruritus: multicenter, comparative analysis of cetirizine, gabapentin and their

109

Você também pode gostar