You are on page 1of 12

SECTION 2 PRURITUS

5 Cutaneous Neurophysiology
Gil Yosipovitch and Tasuku Akiyama

junctions (e.g. conjunctivae). Interestingly, nerves in the deep reticular


Chapter Contents dermis and subcutaneous fat do not transmit itch.
Pruritus pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .100
Mediators of pruritus . . . . . . . . . . . . . . . . . . . . . . . . . . . . .102
PRURITUS PATHWAYS
Chronic itch . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .105
Itch neurophysiology includes specific itch mediators and nerve fibers
Treatment of pruritus . . . . . . . . . . . . . . . . . . . . . . . . . . . . .108
that transmit itch peripherally and centrally4–6. While the concept of
pruritus as a sensory modality entirely separate from pain was not
generally appreciated until the mid-twentieth century, studies have
clearly identified individual histaminergic and non-histaminergic C
fibers that transmit itch7,8. C fibers that transmit itch have exception-
Key features ally slow conduction velocities (0.3–1.0 m/second) and innervate
■ Two subsets of pruritoceptive C neurons that respond to histamine unusually wide territories.
versus cowhage and then activate distinct spinothalamic tract Histamine-sensitive C fibers are sensitive to heat as well as prurito-
neurons genic stimuli but not to mechanical stimuli; in contrast, the vast
majority of C fibers are sensitive to mechanical and heat stimuli but
■ Peripheral itch mediators include histamine, proteases, and
have little or no response to histamine9. The co-responsiveness of itch-
interleukin-31, while central itch mediators include opioids,
transmitting C fibers to temperature explains aggravation of pruritus
gastrin-releasing peptide, and B-type natriuretic peptide
in a warm environment. However, the ineffectiveness of oral antihis-
■ There is an overlap between chronic itch and chronic pain, tamines for most types of pruritus suggests that other fibers have
including activation of multiple brain areas and associated important roles in itch sensation10.
neuromediators and receptors, e.g. nerve growth factor, Indeed, a distinct parallel pathway of non-histaminergic C fibers that
neurotrophin 4, transient receptor potential (TRP) channels transmit itch has been identified in the peripheral nervous system of
■ Cross-talk between cutaneous nerve fibers and the stratum humans and the spinothalamic tract of other primates10,11. These fibers
corneum is a possible mechanism for the pruritus associated with are activated by spicules of the tropical legume cowhage (Mucuna pru-
impaired barrier function (e.g. xerosis, atopic dermatitis) riens), which induces an intense sensation of itch when rubbed,
■ Therapies with topical and systemic drugs that reduce itch inserted, or injected into the skin, without producing a histaminergic
sensitization by counteracting the responsible mediators represent axon reflex12. Cowhage spicules induce itch via release of the protease
promising treatment strategies mucunain, which activates proteinase-activated receptor (PAR)-2 and
PAR-413. Cowhage-sensitive fibers transmit a burning sensation
together with itch and are also sensitive to mechanical and other
stimuli. PAR-2 receptors have a major role in mediating itch in patients
with atopic dermatitis14 (see Ch. 12). The non-histaminergic, polymo-
INTRODUCTION dal C fibers stimulated by mucunain may also have clinical relevance
in chronic itch. In addition to C fibers, A-delta fibers contribute to
The skin is a sensory organ with a dense network of highly specialized cowhage-evoked itch with a more rapid onset15.
afferent sensory nerves that convey sensations such as pain, itch, touch, Other non-histaminergic C fibers respond to β-alanine and activate
temperature, vibration, and pressure (Table 5.1); efferent autonomic distinct populations of primate primary sensory neurons16,17. Intrader-
nerve branches are also present. Neuropeptides such as substance P, mal injection of β-alanine elicits itch but not a wheal and flare response.
calcitonin gene related peptide (CGRP), nerve growth factor (NGF), and MAS-related G protein-coupled receptor D (MrgprD) is a β-alanine
other neurotrophins are secreted from nerve fibers, with multiple effects receptor that is exclusively expressed by these C fibers. In mice, toll-like
that include immune modulation. Itch (pruritus) is the dominant receptor 7 (TLR7) is expressed in C fibers and thought to have a role in
symptom of many cutaneous diseases. Almost all inflammatory skin itch (especially that elicited by non-histaminergic pruritogens) but not
disorders can result in pruritus, which patients often perceive as their in pain sensations; however, its role in human itch is questionable18.
most unendurable symptom. Pruritus can also occur in association A number of other neuromediators and receptors have been identified
with systemic disease (e.g. renal failure, cholestasis; see Ch. 6), psychi- in animal models, but their role in itch pathophysiology in humans
atric conditions (see Ch. 7), and damage to nerve fibers1,2. Itch is a remains to be determined19,20. Gastrin-releasing peptide receptor
multidimensional phenomenon with sensory discriminative, cognitive, (GRPR)-positive neurons in the spinal cord of mice transmit itch, but
evaluative, and motivational components. In most instances, itch not pain, via VGLUT2-mediated signaling21. Overexpression of gastrin-
results from interactions that involve the brain–skin axis. releasing peptide in cutaneous nerve fibers and GRPR in the spinal cord
Pruritus has many similarities to pain. Both are unpleasant sensory has also been observed in mice and primates with chronic itch19,20,22.
experiences that can impair quality of life in affected individuals. B-type natriuretic peptide is an itch-selective neuropeptide found in
However, the behavioral reaction patterns differ – pain elicits a reflex dorsal root ganglia that is expressed at higher levels in mice than in
withdrawal, whereas itch leads to a scratching response3. Despite being rats and humans23,24. Preprotachykinin A, a substance P precursor, is
an extremely common complaint and a sensation so rudimentary that also expressed by a distinct population of murine dorsal horn neurons
almost every two- or four-footed creature experiences it, medical science that respond to pruritic and noxious stimuli25.
is still struggling to understand the mechanisms of itch and how it can The perceived sensation of pruritus can vary greatly in quality26–30.
be inhibited. Patients may describe burning, pricking, “insects crawling” on the skin,
The connection between itch and scratching is so close that in some or even a tickle, but the neurophysiologic and psychologic correlates of
100 languages the same word refers to both itch and scratch. Itch is restricted these qualitative differences have not yet been elucidated. However,
to the skin, tracheal mucous membrane, and several mucocutaneous information obtained from itch questionnaires has enabled a better
non-print metadata
ABSTRACT KEYWORDS:
Cutaneous neurophysiology encompasses specific itch mediators and pruritus,
nerve fibers that transmit itch peripherally and centrally. This chapter primary sensory afferent neurons,
CHAPTER
provides an overview of fundamental mechanisms of itch, from the skin C nerve fibers,
to the brain. There are two subsets of pruritoceptive C neurons which
respond to histamine versus cowhage and then activate distinct spino-
pruritoceptive C neurons,
nerve growth factor,
5
thalamic tract neurons. Peripheral itch mediators include histamine, neurotrophins,

Cutaneous Neurophysiology
proteases, and interleukin-31, while central itch mediators include cowhage,
opioids, gastrin-releasing peptide, and B-type natriuretic peptide. There histamine,
is an overlap between chronic itch and chronic pain, including activa- opioids,
tion of multiple brain areas and associated neuromediators and recep- substance P,
tors such as nerve growth factor and transient receptor potential (TRP) gastrin-releasing peptide,
channels. Therapy with topical and systemic drugs that reduce itch B-type natriuretic peptide,
sensitization by counteracting the responsible mediators represent transient receptor potential channels,
promising treatment strategies. interleukin-31,
cathepsin S,
MAS-related G protein coupled receptors,
alloknesis,
hyperknesis

100.e1
PRIMARY AFFERENT NEURONS THAT INNERVATE THE SKIN NEUROANATOMY OF ITCH

Conduction
Fiber Diameter Myelination velocity Responds to CHAPTER
Cerebral cortex:
A-beta
(Aβ)
Large + >30 m/s Light touch
Moving stimuli Anterior cingulate
5

Cutaneous Neurophysiology
A-delta Small + 2–30 m/s Pain (nociceptors) Somatosensory I
(Aδ) Itch, cowhage-
Somatosensory II
sensitive‡
Thermal
Mechanical Thalamus:
C Small − <2 m/s Pain (nociceptors)
Medial dorsal
Itch*, histamine-
sensitive† Ventral posterior
Itch*, cowhage-
sensitive‡
Thermal†
Mechanical

*‡ ~5% of total C fibers transmit itch.


Transmit pruritus accompanied by a burning sensation; also sensitive to mechanical stimuli.
†Separate C fibers carry both pruritogenic and thermal stimuli, but not mechanical stimuli.
Cerebral
Table 5.1 Primary afferent neurons that innervate the skin. aqueduct
Midbrain:
Cowhage-
sensitive Periaqueductal
understanding of the different characteristics of itch and its features in C fiber gray matter
various skin diseases31. transmitting
itch
Ascending lamina I
Central Pathways to Higher Nervous Histamine- spinothalamic fibers
System Centers sensitive
C fiber Descending
In the spinal cord, pruriceptive C fibers synapse with secondary sensory transmitting inhibitory fibers
neurons in the gray matter of the dorsal horn (Fig. 5.1). These neurons itch Dorsal root
then cross over and ascend in the lateral spinothalamic tract to the ganglion
thalamus. Studies using in vivo extracellular recordings from cats iden- Spinal cord:
tified a subclass of lamina I spinothalamic tract neurons that are excited
Dorsal horn
by iontophoretically administered histamine32. Neurophysiology exper-
iments in primates then found that cowhage-induced itch stimulates a C fiber Lateral
transmitting spinothalamic
distinct population of spinothalamic tract neurons that are not involved pain
in the transmission of histamine-mediated itch11. Studies in human tract
subjects utilizing functional MRI or positron emission tomography
(PET) have shown that histamine and cowhage activate different brain
Fig. 5.1 Neuroanatomy of itch. Itch and pain transmission occur via
areas, providing insight into the supraspinal processing of itch and the unmyelinated C nerve fibers that excite lamina I neurons in the dorsal horn of
corresponding scratch response33–37. the spinal cord. Two subsets of pruritoceptive C-fiber neurons (which respond
In healthy individuals, induction of itch by histamine or cowhage can to histamine and cowhage, respectively) are conducted through distinct lateral
elicit activation of the anterior and posterior cingulate cortex, precu- spinothalamic pathways, with projections to the thalamus. Processing of itch
neus, somatosensory areas I and II, supramarginal gyrus, inferior pari- through either pathway activates several regions of the brain, which are similar
etal lobe, and insula-claustrum complex. In addition to most of the to those involved in pain (see Table 5.4). Concomitant painful stimuli can
previous areas, cowhage evokes more extensive activation of the insular reduce the sensation of itch, possibly by a descending inhibitory mechanism
cortex, claustrum, basal ganglia, putamen, and thalamic nuclei on the resulting from activation of the periaqueductal gray matter. In mice, gastrin-
contralateral side of stimuli33. In one study, atopic dermatitis patients 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.
exhibited significantly more activation of the posterior cingulate cortex
Adapted from Yosipovitch G. Pruritus: an update. Curr Probl Dermatol. 2003;15:137–64.
and precuneus, a cortical area involved in integrative tasks such as
visuospatial imagery, episodic memory retrieval and self-awareness,
than did healthy controls38. This highlights the emotional and affective
Seeing other people scratch can induce sensations of itch and an urge
processing of itch experience in atopic individuals, in whom the degree
to scratch45. Psychophysical studies have shown that the perception of
of brain activation correlated with itch intensity and the severity of the
itch intensifies in atopic dermatitis patients when they are exposed to
atopic disease38.
visual cues of itch45. Recent studies utilizing functional MRI showed
In healthy subjects, scratching has been found to inhibit the activa-
that “contagious itch” activates many of the neural regions linked to
tion of the cingulate cortex and to activate the prefrontal cortex and
the physical perception of itch, including anterior insular, primary
cerebellum. It is possible that the cerebellum plays a role in coordina-
somatosensory, prefrontal, and premotor cortices as well as the
tion of the itch–scratch cycle39,40. Scratching also inhibits histamine-
striatum46,46a. However, more studies are needed to verify the hypoth-
evoked activity of spinothalamic neurons in primates, but not
esis that itch “contagion” involves processing in associative cortical
spontaneous activity or that stimulated by pain41. Self-scratching also
networks such as “mirror neurons” of the prefrontal cortex.
activates areas of the brain involved in reward processing, including the
striatum and substantia nigra. While these responses correlate with the
pleasure of scratching, activation of other areas is associated with itch Pruritus Receptor Units
relief42. Removal of the epidermis abolishes the perception of itch, suggesting
Activation of multiple brain areas argues against a single “itch that putative pruritus receptor units are located predominantly within
center”, emphasizing the multidimensionality of the itch sensation. this layer. Light microscopic and ultrastructural studies of human skin
Pain demonstrates a similar pattern of brain activation involving many have shown the existence of intraepidermal nerve fibers with “free” 101
of the same cortical regions43,44 (see Fig. 5.1). non-specialized nerve endings extending to the stratum granulosum47.
complement C5a. In IgE-mediated acute urticaria, histamine is released
MAJOR MEDIATORS OF PRURITUS: RELATIVE POTENCIES WITH when a specific antigen/allergen cross-links adjacent receptor-bound
REGARD TO PRURITUS AND PAIN specific IgE antibodies. In autoimmune chronic urticaria, similar cross-
linking occurs via functional circulating IgG that react with epitopes
Mediator Pruritus Pain
SECTION
expressed on the α-chain of adjacent FcεRIs51 or less commonly anti-
2 Primary mediators
Histamine +++* +*
IgE autoantibodies (see Fig. 18.3). Histologically, dermal mast cells and
unmyelinated neurons are closely juxtaposed (see Fig. 5.2B, C, E),
raising the possibility of a close (“synapsis-like”) functional relationship
PRURITUS

Tryptase (protease) +++ +


between the immune and nervous systems.
Cathepsin S (protease) +++ ? Histamine’s pruritic action can be potentiated by prostaglandin E1
Interleukin-31 +++ ? and E2. Evidence for histamine as the main mediator of pruritus is
limited to a few skin diseases, including acute and chronic urticaria
Secondary mediators and mastocytosis (e.g. urticaria pigmentosa). H1 antihistamines are
Prostaglandin E1,2 +† + usually effective in these disorders.
Substance P‡ + + Recognition of the histamine H4 receptor has expanded our under-
standing of the physiologic actions of histamine. The H4 receptor is
μ-Opioid receptor agonists ++ − expressed by neurons and bone marrow-derived cells such as eosino-
Nerve growth factor (NGF) + ++ phils, mast cells, dendritic cells, monocytes, and CD8+ T cells. It medi-
ates chemotaxis in the latter group and is thought to have a role in the
Interleukin-2 +++ +/−
inflammation and pruritus of atopic dermatitis. H4 antagonists are
*Superficial (intraepidermal) injection of histamine causes pruritus; deep dermal injection under development and have been shown to alleviate experimental
causes pain. pruritus. In animal models of itch, the effects of H4 antagonists are
†Lowers threshold to pruritus induced by other mediators.
‡Actions partly due to histamine release from mast cells. synergistic with those of centrally acting H1 antihistamines (e.g.
diphenhydramine)52,53.
Table 5.2 Major mediators of pruritus: relative potencies with regard to
pruritus and pain. +/−, little or no activity; +, weak activity; ++, moderately Gastrin-Releasing Peptide
active; +++, highly active; −, no activity.
As noted above, itch-specific GRPR-positive neurons in the dorsal horn
of the spinal cord have been identified in mice22. Although named for
its role in regulating gastrointestinal functions, the GRP ligand of
A subclass of C fibers in the epidermis that express Mrgprs, in particu- GRPRs is widely expressed in the CNS. Whether peripheral sensory
lar MrgprX1, are involved in chloroquine-induced pruritus (see Fig. neurons express GRP is still debated54–56.
5.2F)47. Cathepsin S, an endogenous protease, can cleave and thereby
activate both MrgprC11 in mice, which leads to scratching behavior,
and MrgprX2 in humans48; this is in addition to its ability to activate
B-Type Natriuretic Peptide
PAR-2/4 receptors (Fig. 5.2). In mice, neurons that express MrgprA3 B-type natriuretic peptide (BNP; natriuretic polypeptide B) is a 32-amino
relay itch-specific information to the spinal cord49. acid polypeptide that is secreted by the cardiac ventricles to regulate
Keratinocytes express a variety of neural mediators and receptors blood pressure and fluid balance. BNP is also expressed by a subset of
involved in the sensation of itch (Table 5.2). These include opioids, C fibers and may function in itch transmission54,55, likely driving an
proteases, substance P, NGF, and neurotrophin 4 as well as their respec- itch circuit that includes GRPR-expressing neurons in the spinal cord.
tive receptors, including μ- and κ-opioid receptors, PAR-2, neurotrophic
tyrosine kinase receptor type 1 (NTRK1 [TRKA]), and transient receptor Proteases
potential vanilloid ion channels (particularly TRPV1 and TRPV3). Human dermal mast cells produce two proteases, tryptase and
Keratinocytes also have ATP-gated ion channels and adenosine receptor chymase14. Tryptase released by activated mast cells cleaves PAR-2, a
ligands similar to those observed in C fibers involved in pain transmis- G protein-coupled receptor present on C-fiber terminals (see Fig. 5.2B,
sion. These structural similarities to nerve fibers suggest that keratin- D); this exposes a tethered ligand domain and thereby “self-activates”
ocytes may be involved in the transduction and generation of itch. PAR-2, leading to itch transmission. PAR-2 activation results in local
PAR-2 receptors are thought to be involved in the itch of atopic derma- release of neuropeptides, including substance P and calcitonin gene-
titis and mediate cowhage-induced itch (see above). Cathepsin S also related peptide, which induce neurogenic inflammation57. Kallikrein
induces itch via PAR-2/4 receptors and as mentioned previously, and cathepsins in the skin can also activate PAR-2 in a similar manner.
Mrgprs50. In addition, cleavage of murine MrgprC11 or human MrgprX2 by
cathepsin S activates these receptors and evokes itch48 (see above).
Tryptase levels are elevated fourfold in non-lesional forearm skin of
atopic dermatitis patients14, while expression of PAR-2 is significantly
MEDIATORS OF PRURITUS increased in the epidermis and cutaneous nerve fibers of eczematous
Various mediators that act centrally and/or peripherally are involved in lesions and to a lesser degree in non-lesional skin. Proteases such as
pruritus, including histamine, proteases, substance P, opiates, NGF, and cathepsin B can also be found in common allergens (e.g. grass pollen,
prostaglandins. In inflammatory skin diseases, proinflammatory medi- house dust mites)58, and Staphylococcus aureus can induce secretion
ators produce pruritus and other signs of inflammation, in particular of proteases; both of these exogenous factors are known to aggravate
erythema due to vasodilation and edema from increased vascular per- atopic dermatitis and itch.
meability. The relative potencies of the major mediators with regard to Transgenic mice overexpressing a serine protease exhibit severe itch
these responses (including pruritus) are listed in Table 5.2. Some of and scratching (Table 5.3). In Netherton syndrome, serine protease
these mediators cause pruritus indirectly by evoking release of hista- inhibitor deficiency leads to excess epidermal protease activity, resulting
mine and tryptase from mast cells or by potentiating the actions of in pruritus and atopic manifestations (see Ch. 57). These observations
mediators such as prostaglandins E1 and E2. suggest that interactions between proteases and receptors on C fibers
play important roles in itch and cutaneous inflammation.
Histamine
Histamine is the archetypal mediator of signs and symptoms of inflam- Opioid Peptides
mation, including pruritus. In the skin, histamine is contained primar- Central pruritus, which involves pruritic mediators within the CNS,
ily within the granules of dermal mast cells. Histamine can be released can occur in both cutaneous and systemic diseases. Endogenous opiates
from mast cells upon activation of a range of receptors, including the modify the perception of pruritus via central and peripheral opioid
102 high-affinity IgE receptor (FcεRI), the KIT receptor for stem cell factor, receptors, and generalized pruritus may be induced by an imbalance
and receptors for neuropeptides (e.g. substance P, NGF) and between the μ- and κ-opioid systems. Activation of μ- and κ-opioid
TYPICAL CUTANEOUS NERVE AND ITCH TRANSMISSION VIA ACTIVATION OF C NERVE FIBERS

Peripheral itch mediators and their receptors on C fibers


CHAPTER
Mast cells Neutrophils Keratinocytes Macrophages T cells Liver
ACh, acetylcholine
!-End, !-endorphin
5

Cutaneous Neurophysiology
ET-1, endothelin-1
5-HT, serotonin
IL-7R", interleukin 7 receptor "
M3, muscarinic M3 receptor
Histamine 5-HT Tryptase Elastase Cathepsin ACh ET-1 TSLP !- ProEnkA TNF ** IL-31 Bile acid TSLP, thymic stromal lymphopoietin
S * End
Enk
BAM8-22

H1 + 5-HT3 PAR-2 M3 ETA µ- Mrgpr TNFR Gpbar1 Receptors


H4 IL-7R" TSLPR opioid IL-31RA Oncostatin M
C-fiber terminal *Can also activate Mrgprs.
**Also released from mast cells.
Augmentation by tryptase and substance P Augmentation by NGF Cowhage-induced itch

Mast cell Chemotaxis and PAR-2 Keratinocyte


NK1R Mast cell survival of mast cells Cathepsin S
NGF
Tryptase H TRPV1
Cowhage
(mucunain)
More substance P
released More tryptase
released
TRPA1
More substance P
released
Itch
Cowhage-sensitive,
PAR-2 receptor nonhistaminergic,
H1 polymodal C fibers
receptor NTRK1
Itch

Transduction of itch by the TRPA1 ion channel


Transduction of itch by the TRPV1 ion
channel Keratinocyte
Epidermal Sensitized
Mast cell Capsaicin, noxious heat, hyperplasia C fibers Sprouting BAM8-22
lower pH, eicosanoids
(e.g. prostaglandins),
neurotrophins (e.g. NGF) Chloroquine Mrgpr

TRPA1
Early
release of H1 + H4
puritogenic Itch receptors Itch
mediators IL-31R
TRPV1 IL-31 TSLPR
IL-31R ETA
Keratinocyte Gpbar1

Fig. 5.2 Typical cutaneous nerve and itch transmission via activation of C nerve fibers. There are two categories of axons in a typical cutaneous nerve: (1)
primary afferent Aβ, Aδ, and C fibers with cell bodies in dorsal root ganglia; and (2) sympathetic postganglionic fibers with cell bodies in sympathetic ganglia.
Separate C fibers (~5% of the total) carry pruritogenic stimuli via two pathways: (1) histamine-induced itch that is transmitted by mechanically insensitive, capsaicin-
sensitive fibers via the transient receptor potential vanilloid receptor 1 (TRPV1); and (2) cowhage (mucunain)-induced itch accompanied by a burning sensation that
is transmitted by polymodal fibers sensitive to both capsaicin and mechanical stimuli. A Peripheral itch mediators released by immune cells, keratinocytes, and the
liver activate pruriceptors on C-fiber terminals in the skin. For example, activated mast cells release histamine, which causes transmission of itch via histamine H1
and H4 receptors on histamine-sensitive C fibers. Proteases such as tryptase, elastase, and cathepsin S can cleave proteinase-activated receptor-2 (PAR-2), exposing
a tethered ligand and thereby resulting in “self-activation.” Proenkephalin A (ProEnkA) may be cleaved to form enkephalin (ENK) or bovine adrenal medulla 8-22
peptide (BAM8-22). B Self-activation of PAR-2 following tryptase cleavage results in transmission of itch by the C fibers as well as release of additional substance P,
which primes mast cells via neurokinin-1 receptors (NK1Rs). C Nerve growth factor (NGF) released by mast cells and keratinocytes (with NGF production stimulated
by histamine [H]) activates neurotrophic tyrosine kinase receptor type 1 (NTRK1 [TRKA]) on C fibers, mast cells, and keratinocytes. This induces C fiber sprouting,
sensitization to cowhage-induced itch, and increased substance P release; mast cell chemotaxis, survival, and increased tryptase release; and epidermal
hyperplasia. D Cowhage-induced itch occurs through the release of mucunain, a protease that activates PAR-2 and PAR-4 receptors (the latter not yet identified in
the skin); these receptors can also be activated by endogenous proteases such as cathepsin S as well as tryptase. Activation of PAR-2/4 sensitizes TRPV1 and TRP
ankyrin 1 (TRPA1) channels, resulting in cross-talk and itch transmission. E Activation of H1/H4 receptors and the IL-31 receptor (IL-31R) heterodimer leads to
membrane depolarization of C fibers via TRPV1, which is required for transduction of histamine-induced itch. The TRPV1 ion channel on C fibers, keratinocytes, and
mast cells can also be activated by capsaicin, heat, low pH, eicosanoids, and neurotrophins; although this initially stimulates transmission of itch and release of
pruritogenic mediators, it may eventually lead to desensitization, neuropeptide depletion, and attenuation of itch. F C fibers (including those responsive to
histamine) in the epidermis express MAS-related G protein-coupled receptors (Mrgprs), which can be activated by chloroquine, BAM8-22, and cathepsin S. TRPA1 is 103
involved in transduction of itch by Mrgprs as well as other pruriceptors, including the IL-31R, thymic stromal lymphopoietin receptor (TSLPR) heterodimer,
endothelin type A receptor (ETA), and G protein-coupled bile acid receptor-1 (Gpbar1 [TGR5]).
MURINE MODELS OF ITCH

Model Clinical and laboratory features Pathogenesis/comments


SECTION
Transgenic mice overexpressing PAR-2 Epidermal hyperplasia and scaling as well as
2 Transgenic mice overexpressing a serine
pruritus and scratching
Pruritus and scratching
PRURITUS

protease
TRPV1-deficient mice and Pirt-deficient Lack of response to pruritogens Pirt normally plays an important role in sensing itch
mice (histaminergic and non-histaminergic) as well as pain via
TRPV1 channels (and also TRPV1-independent itch)
Mrgpr-deficient mice and TRPA1- Lack of scratching response to chloroquine and Mrgprs are normally activated by chloroquine and
deficient mice 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 Lack 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
μ-Opioid receptor-deficient mice Thinner epidermis, higher density of epidermal
nerve endings, and less scratching after
induction of dry skin
Transgenic mice overexpressing Pruritus, scratching, excoriations and alopecia
interleukin-31
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; PAR-2, protease-activated receptor 2; Pirt, phosphoinositide-interacting regulator of transient receptor potential channels; TRPV1, transient receptor
potential vanilloid 1.

receptors stimulates and inhibits itch perception, respectively59,60. mast cells, leading to sensitization of these cells and increased produc-
κ-opioid receptor agonists can act within the skin, spinal cord (e.g. tion of tumor necrosis factor (TNF; see Fig. 5.2B)77. In turn, TNF
interneurons), and brain to reduce itch61,62. sensitizes nociceptive nerve endings, producing a self-amplifying loop
Morphine as well as other exogenous and endogenous μ-opioid recep- between neurons and mast cells. Substance P also plays an important
tor agonists may cause generalized pruritus1,2,63,64. Morphine also pro- role in spinal itch transmission78,79.
duces local pruritus and erythema when injected intradermally; this
response is only partially inhibited by the μ-opioid receptor antagonist Neurotrophins
naloxone but is substantially inhibited by topical pretreatment with the
H1 antihistamine doxepin65. In contrast to morphine, the highly potent Neurotrophins are factors that regulate the growth and function of
μ-opioid agonist fentanyl does not induce mast cell degranulation, even nerve cells. Members of this family include the prototypic nerve growth
when applied in high doses. Therefore, two possible mechanisms for factor (NGF) as well as brain-derived neurotrophic factor (BDNF) and
opioid-induced itch are: (1) degranulation of cutaneous mast cells66; neurotrophins 3, 4, and 580. Increased levels of epidermal NGF corre-
and (2) activation of μ-opioid receptors with direct central and periph- late with proliferation of terminal cutaneous nerves and upregulated
eral pruritogenic effects67–69. Specifically, morphine-induced itch can expression of neuropeptides such as substance P81. NGF can also
result from activation of a heterodimeric μ-opioid and gastrin-releasing induce sprouting of nerve fibers, sensitization of nerve endings, and
peptide receptor; inhibition of the latter receptor component blocked axonal transport in dorsal root ganglia cells (see Fig. 5.2C)82.
opioid-related itch but not analgesia, an observation that may have Keratinocytes express high levels of NGF, which is not only required
therapeutic relevance70. for survival and regeneration of sensory neurons but also controls the
Nociceptin, the endogenous peptide ligand for the opioid receptor-like responsiveness of such neurons to external stimuli83,84. Patients with
1 (ORL1) receptor, has also been implicated in cutaneous inflamma- atopic dermatitis and psoriasis have increased expression of NGF in
tion, pain, and pruritus71. In a murine model, nociceptin interaction cutaneous mast cells, keratinocytes, and fibroblasts85,86. NGF is thought
with ORL1 receptors on keratinocytes led to production of leukotriene to act as a signaling molecule between mast cells and keratinocytes in
B4, which induced scratching that was inhibited by systemic adminis- allergic skin diseases. Mast cell-derived histamine induces keratin-
tration of naloxone72. ocytes to increase NGF production, and the latter may promote infiltra-
tion of mast cells into inflamed skin87,88. Upregulation of other
neurotrophins, such as neurotrophin 4, has also been observed in
Substance P keratinocytes from atopic dermatitis patients89.
Substance P, a neuropeptide with a widespread distribution in periph-
eral nerves and the CNS, intensifies itch perception. Levels of substance
P in the serum of patients with atopic dermatitis are elevated and cor- Prostanoids
relate with disease severity73. Intradermal injection of substance P In the skin, prostaglandins enhance histamine-induced itch90,91. Pros-
provokes itch as well as elements of neurogenic inflammation such as taglandins are the products of the transformation of the essential fatty
erythema and the wheal-and-flare reaction. Substance P is synthesized acid arachidonic acid by cyclooxygenase-1 (COX-1) or cyclooxygenase-2
in the cell bodies of C neurons, transported towards the peripheral (COX-2). When injected into the dermis, prostaglandin E1 (PGE1) is
nerve terminals, and released by antidromic depolarization to cause not itself pruritogenic but enhances pruritus due to histamine subse-
vasodilation and increased vascular permeability. quently injected into the same site92. It appears that only itch-mediating
Although endogenously released substance P does not degranulate neurons that display lasting activation following exposure to histamine
mast cells in healthy human skin or cause any sensation at physiologic are excited by PGE2, and mechanosensitive fibers are unresponsive to
concentrations74,75, direct communication between nerve fibers and both histamine and PGE2.
mast cells via substance P has been verified76. High concentrations of PGE2 has also been shown to have a direct, low-level pruritogenic
104 substance P can cause immediate mast cell degranulation, whereas low effect in both atopic dermatitis patients and unaffected individuals
concentrations specifically activate neurokinin-1 (NK-1) receptors on without inducing protein extravasation. This suggests that prostanoids’
peripheral action is not solely via histamine and that prostanoids may Immune Cells as Itch Mediators and Modulators
potentiate pruritus via other effects on nerve fibers.
Interactions between the nervous and immune systems in the skin
Although oral administration of aspirin, a cyclooxygenase inhibitor,
have important roles in itch induction108. Neuropeptides such as sub-
does not generally ameliorate pruritus93, topical application of aspirin
stance P, CGRP and vasointestinal peptide, which are released by cuta-
may reduce chronic localized itch94. The role of other eicosanoids, CHAPTER

including leukotrienes and 12-hydroxyeicosatetraenoic acid (12-HETE),


in the pathogenesis of pruritus is unclear. In mice, leukotriene B4 can
neous sensory nerves, can activate transcription factors and regulate
the expression of adhesion molecules and proinflammatory cytokines, 5
thereby modulating immune and inflammatory responses57. These
provoke scratching and may be involved in skin disease-related

Cutaneous Neurophysiology
neuropeptides also influence cellular proliferation and differentiation,
itch72,95–97.
tissue repair, and antigen presentation involving keratinocytes, mast
cells, dermal microvascular endothelial cells, and Langerhans cells.
This interaction is bidirectional, as cytokines and chemokines are also
Mediators That Activate Transient Receptor 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 intradermally109. 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 located on C fibers, dermal mast intense generalized pruritus. Moreover, treatment with topical calcineu-
cells, dendritic cells, and keratinocytes (see Fig. 5.2E)98. This receptor rin inhibitors, which block the production of IL-2, can result in
is activated by capsaicin, endogenous substances such as cannabinoids decreased itch. Neither antihistamines nor NSAIDs reduce IL-2-in-
(e.g. anandamide), prostaglandins and various neurotrophins, as well as duced pruritus, and whether the latter is directly receptor-mediated or
by acidosis and temperatures >43°C (109°F); as a result, it can mediate an indirect effect via mast cells or endothelial cells remains to be
heat pain. Mechano-insensitive pruritoceptive C-nerve pathways can determined.
be activated by capsaicin, indicating that there is expression of TRPV1. IL-31 is produced by T helper 2 (Th2) cells and belongs to the IL-6
In addition, the experimental induction of histamine-mediated itch family. It induces pruritus by modulating the function of sensory
requires cooperation of TRPV1 ion channels99. Stimulation of TRPV1- neurons, with the itch developing after a mean delay of ~2 hours110.
positive nerve fibers also leads to the release of multiple pruritoceptive IL-31 may exert its pruritogenic effect via activation of the IL-31 recep-
mediators such as interleukins (ILs) and neuropeptides. tor (IL-31R) on keratinocytes, which could subsequently stimulate C
TRPV3 is a thermosensor of warmth (>33°C, 91°F) that is expressed fibers in the skin. The IL-31R is a heterodimer composed of the
in keratinocytes and dorsal root ganglion neurons in humans. In mice, oncostatin M receptor (OSMR) β protein plus the IL-31 receptor A and
a gain-of-function missense mutation in TRPV3 results in chronic itch, it is also found on TRPV1+/ TRPA1+ cutaneous C fibers and in dorsal
scratching, and an atopic-like dermatitis100. In contrast, TRP mela- root ganglia111.
statin 8 (TRPM8) on C nerve fibers functions as a thermosensor of cool Signaling via either the IL-31R or OSMR, a heterodimer composed
temperatures (<28°C, 82°F) and is activated by menthol and icilin, of the OSMR β protein and a gp130 subunit, can result in cutaneous
which provide a “cooling” sensation that may relieve itch. inflammation as well as keratinocyte proliferation, differentiation, and
TRP ankyrin 1 (TRPA1), a polymodal nociceptor, functions as a apoptosis112. Mutations in the gene encoding the OSMR β protein
downstream mediator of histamine-independent itch stimulated by underlie familial primary localized cutaneous amyloidosis, an autoso-
Mrgprs present in a subset of epidermal C fibers101. TRPA1 can be mal dominant disorder characterized by chronic localized itching and
activated directly by menthol as well as via pruriceptors such as the scratching that results in deposition of keratin-derived amyloid in the
heterodimeric receptor for thymic stromal lymphopoietin (TSLP), a dermis113. Higher levels of IL-31 expression are found in the skin of
cytokine released by keratinocytes that plays a role in the pruritus of patients with atopic dermatitis, prurigo nodularis, and cutaneous T-cell
atopic dermatitis102 (see Fig. 5.2F). lymphoma (CTCL)114–116. Serum levels of IL-31 have been found to
correlate with pruritus severity in patients with advanced CTCL but
not in those with atopic dermatitis117,118.
Other Peripheral Mediators of Itch Thymic stromal lymphopoietin (TSLP) is produced by keratinocytes
and promotes Th2-type responses. TSLP acts directly on TRPA1-
Other neurotransmitters expressing neurons to elicit itch in mice102, and TSLP expression is
Intradermal injection of acetylcholine, an important neurotransmitter increased within lesions of atopic dermatitis119. IL-4 and IL-13 pro-
in the autonomic nervous system, typically induces pain; however, in duced by Th2 cells contribute to the itch of atopic dermatitis via activa-
patients with chronic itch, it induces itch. In murine models of itch, tion of JAK (Janus kinase)/STAT (signal transducer and activator of
activation of the muscarinic receptor 3 triggers pruritus103. Norepi- transcription) signaling cascades.
nephrine, a catecholamine neurotransmitter, exerts tonic inhibition of TNF binding to its receptors is known to sensitize nociceptive nerve
itch signaling in the spinal cord104. Currently there are no data regard- endings, but its role in itch is unclear. Although targeted TNF inhibitors
ing the role of epinephrine or dopamine in itch transmission. do not directly decrease pruritus, thalidomide has anti-TNF effects and
can be effective in treating the itch associated with prurigo nodularis120.
Peptidases
Mast cells express both neuropeptides and neuropeptide-degrading pep-
tidases including angiotensin-converting enzyme (ACE) and neural CHRONIC ITCH
endopeptidases. Medications such as ACE inhibitors, which can induce
Chronic itch may occur in the setting of pruritoceptive itch originating
pruritus without a rash, may induce itch by inhibiting the activity of
from skin disease, neuropathic itch due to pathology in the nervous
these degrading enzymes. Likewise, the neural peptidase endothelin-
system, and systemic or psychiatric disorders121 (see Ch. 6). It often
converting enzyme 1 (ECE-1) negatively regulates endothelin 1-induced
has a significant effect on patients’ quality of life. Chronic itch and
itch, which is largely histamine-independent105.
chronic pain share several features, with both potentially involving
Other mediators with potential roles in itch peripheral and central sensitization122 (Table 5.4; Fig. 5.3). Interest-
ingly, familial chronic itch was recently associated with heterozygous
Serotonin has been implicated as an inducer of itch in murine models; variants in the gene encoding the collagen type VI α5 chain, which is
however, in humans it is a very mild pruritogen. Nitric oxide is another expressed in the dermis123.
factor that may induce itch via neurogenic inflammation106. The bovine
adrenal medulla 8-22 (BAM8-22) peptide, a proteolytically cleaved
product of proenkephalin A, is a potent activator of Mrgprs and can Peripheral Sensitization in Chronic Itch
stimulate itch, usually accompanied by a stinging or burning sensa- An increased density of cutaneous nerve endings is observed in some
tion (see Fig. 5.2F)107. Chloroquine can also induce itch by activating forms of chronic itch. Patients with chronic itch in the setting of atopic
Mrgprs, which are expressed by a subset of C fibers in the epidermis dermatitis have increased neurotrophin levels in involved skin, includ- 105
(see above). ing NGF and neurotrophin 489. Chronic localized pain is associated
with elevated levels of the same neurotrophins, which are known to amplified itching via lipocalin-2 signaling126. Spinal TLR4 signaling
sensitize nociceptive neurons80. In humans, intradermally injected also plays a role in the spinal astrocyte activation and astrogliosis that
NGF sensitizes non-histaminergic itch124, while selective sensitization underlies chronic itch127.
of non-histaminergic itch was observed in a murine model of chronic There are two forms of increased sensitivity to itch, alloknesis and
SECTION
dry skin125. hyperknesis. In alloknesis, stimuli that normally do not induce itch
2 Central Sensitization in Chronic Itch
such as touch or gentle warming do so in the skin that surrounds a
pruritic area128. This phenomenon is analogous to allodynia, in which
gentle mechanical stimuli give rise to a perception of pain. Like allo-
PRURITUS

Chronic itch leads to sensitization of second-order neurons within the


dorsal horn of the spinal cord (see Fig. 5.1). In a mouse model of chronic dynia, alloknesis requires ongoing activity in primary afferent C fibers
pruritus, STAT3-dependent reactive astrogliosis within the dorsal horn and is probably mediated by low-threshold myelinated mechanorecep-
tor Aβ fibers (see Fig. 5.2). Alloknesis is common and represents a
prominent feature of atopic dermatitis, explaining pruritus associated
with dressing and undressing. Hyperknesis is characterized by more
COMPARISON OF CHARACTERISTICS OF CHRONIC ITCH AND intense itch induced by a stimulus that usually produces slight itch and
CHRONIC PAIN occurs within the skin surrounding an area of inflammation128a. It is
similar to the phenomenon in chronic pain termed hyperalgesia. In
Chronic itch Chronic pain mice, neurokinin-1 receptor-expressing spinal neurons play a major
Peripheral Sensitized C nerve fibers Sensitized C nerve fibers role in chronic itch, whereas gastrin-releasing peptide receptor-
sensitization expressing spinal neurons contribute to hyperknesis, but not alloknesis
or ongoing itch129.
Central Alloknesis Allodynia
In patients with chronic itch, painful electrical and heat stimuli may
sensitization Hyperknesis Hyperalgesia
be perceived as itch130–132. An analogous phenomenon can occur in
Neuromediators Nerve growth factor Nerve growth factor patients with chronic pain, in whom histamine iontophoresis may be
Neurotrophin 4 Neurotrophin 4 perceived as painful133. These findings indicate that pain-induced inhi-
Chemical Histamine • Bradykinin bition of pruritus may be compromised in patients with chronic itch.
mediators Cowhage (mucunain) • Most opioids reduce This may also explain why scratching aggravates itch in patients with
Most opioids induce itch pain chronic itch, thereby inducing a vicious itch–scratch cycle.
CNS areas • Anterior and posterior • Anterior cingulate
activated cingulate cortex cortex Itch Related to Impaired Skin Barrier Function
• Premotor area • Premotor area Itch is a common symptom of xerotic skin and is aggravated during
• Precuneus • Somatosensory cortex the winter in cold climates when the relative humidity falls indoors.
• Supramarginal gyrus I, II Damage to the stratum corneum and the impaired barrier function that
• Dorsolateral prefrontal • Dorsolateral prefrontal results can induce itch even without inflammation. Environmental
cortex cortex changes in pH, temperature, and humidity may activate C fibers to
• Insula–claustrum • Insula–claustrum transmit the sensation of itch. Cross-talk between the stratum corneum
complex complex and nerve fibers may explain the pruritus associated with impaired
• Somatosensory cortex
barrier function. Keratinocytes release neuromediators upon damage to
I, II
the stratum corneum barrier, and nerve fibers sprout in the epidermis
Table 5.4 Comparison of characteristics of chronic itch and chronic pain. in response to this damage. In a murine model of dry skin, enhanced

Fig. 5.3 Mechanisms of itch sensitization


MECHANISMS OF ITCH SENSITIZATION UNDERLYING CHRONIC PRURITUS underlying chronic pruritus. Three main
mechanisms of itch sensitization have important
roles in chronic pruritus. A Peripheral sensitization
of C-fiber pruriceptors occurs in the setting of
A Peripheral Pruritogens Scratch, heat chronic pruritus, which may be associated with
sensitization increased levels of neurotrophins such as nerve
↑ Itch growth factor (NGF; see Fig. 5.2C). PAR-2 activation
in C fibers can also play a role in itch sensitization
by enhancing responses to MAS-related G
protein-coupled receptor (Mrgpr) agonists.
B Spinal itch-transmitting neurons are sensitized in
the setting of chronic pruritus. There may be
increased synaptic efficacy and signaling via
additional synapses. C Itch-inhibitory circuits are
B Central normally driven by vesicular glutamate transporter
sensitization 2 (VGLUT2)-dependent glutamate release from
nociceptors as well as by descending pathways;
this results in activation of itch-inhibitory Bhlhb5
interneurons, which release GABA, glycine, and/or
dynorphins. Decreased function of these
+ interneurons can result in disinhibition of itch. H1R,
+ histamine H1 receptor; TRPA1, transient receptor
potential ankyrin receptor 1; TRPV1, transient
– C Dysfunction of receptor potential vanilloid receptor 1.
+ itch-inhibitory circuits

H1R+ or Mrgpr+ pruriceptors


TRPV1+, TRPA1+ nociceptors
Descending
Bhlhb5 interneuron
pathways
GABA, Glycine
Glutamine released from VGLUT2
106
DRUG TREATMENT OF PRURITUS

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


CHAPTER
Topical agents
Capsaicin (active
component in hot chili
• Activates and subsequently desensitizes
TRPV1, a vanilloid TRP receptor
• Used primarily for localized, chronic
pruritic disorders, especially those of
• Transient burning sensation,
which usually resolves after
5

Cutaneous Neurophysiology
peppers) • Causes release from C neurons and neuropathic origin several days of consistent use;
(eventually) depleted stores of • Controlled studies support use for: application of topical lidocaine
neuropeptides (e.g. substance P, CGRP, - Notalgia paresthetica or EMLA may be helpful
somatostatin) - Brachioradial pruritus
• Prolonged use leads to neuronal - Hemodialysis-associated pruritus
degeneration at sites of application, - Postherpetic neuralgia, with a
which can result in a reversible decrease transdermal patch FDA-approved for this
in epidermal nerve fiber density indication
Doxepin • Tricyclic compound with potent • Shown to relieve pruritus in patients with • Drowsiness in ~25% of patients
antihistamine effects (H1 and H2); also atopic dermatitis due to percutaneous absorption
anticholinergic properties • Allergic contact dermatitis
Menthol • Activates TRPM8 and TRPA1, producing • Inconsistent results in controlled studies • Skin irritation
a cool sensation • 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 disease (uncommon)
Tacrolimus, • Calcineurin inhibitors that block • Can decrease itch related to the • Transient burning sensation
pimecrolimus production of multiple proinflammatory inflammation of atopic dermatitis
cytokines (see Ch. 128)
• May initially activate and subsequently
desensitize TRPV1
“Barrier repair” creams, • Diminish transepidermal water loss and • Decrease itch in patients with atopic None
other emollients and decrease flux between a distended and dermatitis
humectants desiccated state
• 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
Strontium • Calcimimetic thought to inhibit ion • Strontium 4% hydrogel was shown to None identified
channels in nerve fibers relieve localized cowhage-induced itch
Ketamine-amitriptyline- • Anesthetic effect; targets ion channels in • May decreases neuropathic itch and other • Mild burning sensation
lidocaine (10%-5%-5% peripheral nerves and NMDA receptors forms of pruritus, including the itch of • Potential systemic absorption
compounded cream) prurigo nodularis
Systemic agents
Antihistamines • Doxepin is a tricyclic compound with • Decrease wheal formation as well as • Sedation
(especially doxepin) potent antihistamine effects (H1 and H2) pruritus in patients with urticaria • Anticholinergic effects
and anticholinergic properties; first- • Interaction with MAO inhibitors
generation, sedating antihistamine (for doxepin)
Naloxone, naltrexone • μ-Opioid receptor antagonists* • Controlled trials show efficacy for • Hepatotoxicity
cholestatic and renal pruritus • Nausea/vomiting
• May be of benefit for severe pruritus of • Insomnia
other etiologies • Reversal of analgesia
Nalfurafine† • κ-Opioid receptor agonist* • Controlled trials show efficacy for renal • Insomnia
pruritus
Butorphanol • κ-Opioid receptor agonist and μ-opioid • Relieves pruritus associated with morphine • Nausea/vomiting
receptor antagonist* with epidural administration of both agents • Sedation
• May reduce severe pruritus associated • Dependence (rare)
with systemic disease or inflammatory skin
conditions‡
Mirtazapine • Serotonin and norepinephrine inverse • May reduce nocturnal pruritus (at low • Sedation
agonist that reduces central itch doses, e.g. 15 mg nightly) • Weight gain
perception
Paroxetine • Selective serotonin reuptake inhibitor • A controlled study showed benefit for • Sexual dysfunction
(SSRI) pruritus related to systemic disease • Sedation
• Insomnia
• Weight gain
Table 5.5 Drug treatment of pruritus. Topical naltrexone, aspirin, and cannabinoids may also have antipruritic effects, and injections of botulinum toxin type A
have been used to treat neuropathic itch. In the future, topical anti-itch medications may target 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. CTCL, cutaneous T-cell lymphoma; EGFR, epidermal growth factor 107
receptor; GABA, γ-aminobutyric acid; NMDA, N-methyl-D-aspartate; TNF, tumor necrosis factor.
DRUG TREATMENT OF PRURITUS

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


SECTION
Thalidomide • Blocks neuropathic afferent pathways • May be helpful for prurigo nodularis, • Teratogenicity
2 •
(peripherally and centrally)
Inhibits TNF synthesis
actinic prurigo and pruritus of advanced
age
• Peripheral neuropathy
PRURITUS

Gabapentin, pregabalin • Structural analogues of the • Beneficial for neuropathic itch (e.g. • Peripheral edema
neurotransmitter GABA brachioradial pruritus), post-burn pruritus • Sedation
• Thought to inhibit central itch pathways and postherpetic neuralgia • Abdominal pain
Aprepitant • Antagonist of the neurokinin-1 receptor, • Primarily used as an antiemetic in • Fatigue
which is activated by substance P oncology patients; reports of reduced • Hiccups
pruritus in patients with atopic dermatitis,
Sézary syndrome, and other forms of
CTCL as well as decreases in pruritus
induced by anti-EGFR antibodies or
tyrosine kinase inhibitors

*† Inhibit itch transmission within the central nervous system.


Currently not commercially available in the US.
‡An intranasal spray is currently available.

Table 5.5 Drug treatment of pruritus. (cont’d)

c-fos expression in the CNS was observed, reflecting activation of chronic pruritus can have sequelae such as depression and decreased
axons134,135. sexual desire or function140.
Serine proteases that activate PAR-2 and thereby stimulate itch are
secreted in response to an increasing (alkaline) stratum corneum pH, Pharmacologic Treatments
which is commonly noted during barrier damage. This suggests that
Unlike pain, for which a host of effective medications are avail-
environmental factors that increase stratum corneum pH may increase
able, there are no general-purpose, consistently beneficial antipruritic
itch perception136. In contrast, a low-pH extracellular environment
drugs. However, recent discoveries of specific neural networks that
causes C nociceptors to induce pain.
are involved in itch transmission have led to advances in this rela-
tively unexplored area of medicine140a. The mechanisms, uses, and
Senescent Skin and Itch side effects of currently available antipruritic drug treatments are out-
Itch is particularly frequent in individuals over 65 years of age137,138. lined in Table 5.5. Of note, although capable of relieving pruritus by
Although dry skin is probably the most common trigger, elderly patients treating inflammatory skin disease, corticosteroids are not intrinsically
can have idiopathic itch without xerosis. Other possible explanations antipruritic.
include age-related changes in nerve fibers and central disinhibition of
itch due to loss of input from pain fibers. Additional cutaneous changes Behavioral Therapy
that may contribute to pruritus as well as xerosis in elderly patients
Stress and other psychogenic factors are important in itch141. Patients
include decreased skin surface lipids and diminished barrier repair.
with pruritus experience higher levels of psychological stress than those
without this symptom142. An impaired response of the autonomic
Cholestatic Pruritus nervous system to itch, scratching, and emotional stress has also been
The enzyme autotaxin and its product lysophosphatidic acid (LPA), a identified in atopic dermatitis patients143. Several studies have shown
neuronal activator, have a pathogenic role in cholestatic pruritus (see that behavioral therapy reduces the intensity of itch that is
Ch. 6). Serum autotaxin levels correlate with itch intensity in patients perceived144.
with cholestasis139. In addition, the G protein-coupled bile acid recep-
tor-1 (Gpbar1; TGR5) has been found to have a role in cholestatic
pruritus in mouse models.
FUTURE DIRECTIONS
TREATMENT OF PRURITUS A critical issue is identification of the specific mechanisms and media-
tors responsible for particular “itch states”. Hopefully, our emerging
Pruritus can significantly impair quality of life in affected individuals. understanding of the pathways of itch transmission will soon result in
Acute itch may lead to agitation and difficulty in concentrating, while the development of effective targeted treatments.

REFERENCES
1. Yosipovitch G, Bernhard JD. Clinical practice. Chronic 7. Schmelz M. Itch and pain. Neurosci Biobehav Rev 12. Shelley WB, Arthur RP. Mucunain, the active
pruritus. N Engl J Med 2013;368:1625–34. 2010;34:171–6. pruritogenic proteinase of cowhage. Science
2. Yosipovitch G, Greaves MW, Fleischer AB Jr, McGlone F, 8. Handwerker HO, Schmelz M. Pain: itch without pain 1955;122:469–70.
editors. Itch: basic mechanisms and therapy. New York: – a labeled line for itch sensation? Nat Rev Neurol 13. Reddy VB, Iuga AO, Shimada SG, et al. Cowhage-
Marcel Dekker; 2004. 2009;5:640–1. evoked itch is mediated by a novel cysteine protease:
3. Yosipovitch G, Carstens E, McGlone F. Chronic itch and 9. Johanek L, Meyer R, Hartke T, et al. Psychophysical and a ligand of protease-activated receptors. J Neurosci
chronic pain: analogous mechanisms. Pain physiological evidence for parallel afferent pathways 2008;28:4331–5.
2007;131:4–7. mediating the sensation of itch. J Neurosci 14. Steinhoff M, Neisius U, Ikoma A, et al. Proteinase-
4. Akiyama T, Carstens E. Neural processing of itch. 2007;27:7490–7. activated receptor-2 mediates itch: a novel pathway
Neuroscience 2013;10:697–714. 10. Namer B, Carr R, Johanek LM, et al. Separate for pruritus in human skin. J Neurosci
5. Bautista DM, Wilson SR, Hoon MA. Why we scratch an peripheral pathways for pruritus in man. J 2003;23:6176–80.
itch: the molecules, cells and circuits of itch. Nat Neurophysiol 2008;100:2062–9. 15. Ringkamp M, Schepers RJ, Shimada SG, et al. A role for
Neurosci 2014;17:175–82. 11. Davidson S, Zhang X, Yoon CH, et al. The itch- nociceptive, myelinated nerve fibers in itch sensation.
6. Davidson S, Giesler G Jr. The multiple pathways for producing agents histamine and cowhage activate J Neurosci 2011;31:14841–9.
108 itch and their interactions with pain. Trends Neurosci separate populations of primate spinothalamic tract 16. Liu Q, Sikand P, Ma C, et al. Mechanisms of itch evoked
2010;33:550–8. neurons. J Neurosci 2007;27:10007–14. by β-alanine. J Neurosci 2012;32:14532–7.
17. Wooten M, Weng HJ, Hartke TV, et al. Three 41. Davidson S, Zhang X, Khasabov SG, et al. Relief of itch 65. Heyer G, Dotzer M, Diepgen TL, Handwerker HO.
functionally distinct classes of C-fibre nociceptors in by scratching: state-dependent inhibition of primate Opiate and H1 antagonist effects on histamine
primates. Nat Commun 2014;5:4122. spinothalamic tract neurons. Nat Neurosci induced pruritus and alloknesis. Pain 1997;73:239–43.
18. Liu T, Xu ZZ, Park CK, et al. Toll-like receptor 7 2009;12:544–6. 66. Fjellner B, Hagermark O. Potentiation of histamine-
mediates pruritus. Nat Neurosci 2010;13:1460–2. 42. Papoiu AD, Nattkemper LA, Sanders KM, et al. Brain’s induced itch and flare responses in human skin by the
CHAPTER
19. Sun YG, Zhao ZQ, Meng XL, et al. Cellular basis of itch reward circuits mediate itch relief. a functional MRI enkephalin analogue FK-33-824, beta-endorphin and
sensation. Science 2009;325:1531–4.
20. Nattkemper LA, Zhao ZQ, Nichols AJ, et al.
study of active scratching. PLoS ONE 2013;8:e82389.
43. Mochizuki H, Papoiu AD, Yosipovitch G. Brain
morphine. Arch Dermatol Res 1982;274:29–37.
67. Fjellner B, Hagermark O. The influence of the opiate 5
Overexpression of the gastrin-releasing peptide in processing of itch and scratching. In: Carstens E, antagonist naloxone on experimental pruritus. Acta

Cutaneous Neurophysiology
cutaneous nerve fibers and its receptor in the spinal Akiyama T, editors. Itch: mechanisms and treatment. Derm Venereol 1984;64:73–5.
cord in primates with chronic itch. J Invest Dermatol Boca Raton (FL): CRC Press; 2014. p. 391–408. 68. Hagermark O. Peripheral and central mediators of itch.
2013;133:2489–92. 44. Mochizuki H, Sadato N, Saito DN, et al. Neural Skin Pharmacol 1992;5:1–8.
21. Aresh B, Freitag FB, Perry S, et al. Spinal cord correlates of perceptual difference between itching 69. Bernstein JE, Swift R. Relief of intractable pruritus with
interneurons expressing the gastrin releasing peptide and pain: a human fMRI study. Neuroimage naloxone. Arch Dermatol 1979;115:1366–7.
receptor convey itch through VGLUT2-mediated 2007;36:706–17. Erratum in: Neuroimage. 70. Liu XY, Liu ZC, Sun YG, et al. Unidirectional cross-
signaling. Pain 2017;158:945–61. 2008;39:911–12. activation of GRPR by MOR1D uncouples itch and
22. Sun YG, Chen ZF. A gastrin-releasing peptide receptor 45. Papoiu AD, Wang H, Coghill RC, et al. Contagious itch analgesia induced by opioids. Cell 2011;147:447–58.
mediates the itch sensation in the spinal cord. Nature in humans: a study of visual ‘transmission’ of itch in 71. Meunier JC. Nociceptin/orphanin FQ and the opioid
2007;448:700–3. atopic dermatitis and healthy subjects. Br J Dermatol receptor-like ORL1 receptor. Eur J Pharmacol
23. Mishra SK, Hoon MA. The cells and circuitry for itch 2011;164:1299–303. 1997;340:1–15.
responses in mice. Science 2013;340:968–71. 46. Holle H, Warne K, Seth AK, et al. Neural basis of 72. Andoh T, Yageta Y, Takeshima H, Kuraishi Y. Intradermal
24. Goswami SC, Thierry-Mieg D, Thierry-Mieg J, et al. contagious itch and why some people are more prone nociceptin elicits itch-associated responses through
Itch-associated peptides: RNA-Seq and bioinformatic to it. Proc Natl Acad Sci USA 2012;109:19816–21. leukotriene B(4) in mice. J Invest Dermatol
analysis of natriuretic precursor peptide B and gastrin 46a. Schut C, Mochizuki H, Grossman SK, et al. Brain 2004;123:196–201.
releasing peptide in dorsal root and trigeminal processing of contagious itch in patients with atopic 73. Toyoda M, Nakamura M, Makino T, et al. Nerve growth
ganglia, and the spinal cord. Mol Pain 2014;10:44. dermatitis. Front Psychol 2017;8:1267. factor and substance P are useful plasma markers of
25. Gutierrez-Mecinas M, Bell AM, Marin A, et al. 47. Liu Q, Tang Z, Surdenikova L, et al. Sensory neuron- disease activity in atopic dermatitis. Br J Dermatol
Preprotachykinin A (PPTA) is expressed by a distinct specific GPCR Mrgprs are itch receptors mediating 2002;147:71–9.
population of excitatory neurons in the mouse chloroquine-induced pruritus. Cell 2009;139:1353–65. 74. Schmelz M, Zeck S, Raithel M, Rukwied R. Mast cell
superficial spinal dorsal horn including cells that 48. Reddy VB, Sun S, Azimi E, et al. Redefining the concept tryptase in dermal neurogenic inflammation. Clin Exp
respond to noxious and pruritic stimuli. Pain of protease-activated receptors: cathepsin S evokes Allergy 1999;29:695–702.
2017;158:440–56. itch via activation of Mrgprs. Nat Commun 75. Weidner C, Klede M, Rukwied R, et al. Acute effects of
26. Dawn A, Papoiu AD, Chan YH, et al. Itch characteristics 2015;6:7864. substance P and calcitonin gene-related peptide in
in atopic dermatitis: results of a web-based 49. Han L, Ma C, Liu Q, et al. A subpopulation of human skin – a microdialysis study. J Invest Dermatol
questionnaire. Br J Dermatol 2009;160:642–4. nociceptors specifically linked to itch. Nat Neurosci 2000;115:1015–20.
27. Yosipovitch G, Ansari N, Goon A, et al. Clinical 2013;16:174–82. 76. Suzuki R, Furuno T, McKay DM, et al. Direct neurite-
characteristics of pruritus in chronic idiopathic 50. Reddy VB, Shimada SG, Sikand P, et al. Cathepsin S mast cell communication in vitro occurs via the
urticaria. Br J Dermatol 2002;147:32–6. elicits itch and signals via protease-activated neuropeptide substance P. J Immunol
28. Yosipovitch G, Goon AT, Wee J, et al. Itch receptors. J Invest Dermatol 2010;130:468–70. 1999;163:2410–15.
characteristics in Chinese patients with atopic 51. Hide M, Francis DM, Grattan CE, et al. Autoantibodies 77. Cocchiara R, Lampiasi N, Albeggiani G, et al. Mast cell
dermatitis using a new questionnaire for the against the high-affinity IgE receptor as a cause of production of TNF-alpha induced by substance P
assessment of pruritus. Int J Dermatol 2002;41: histamine release in chronic urticaria. N Engl J Med evidence for a modulatory role of substance
212–16. 1993;328:1599–604. P-antagonists. J Neuroimmunol 1999;101:128–36.
29. Darsow U, Scharein E, Simon D, et al. New aspects of 52. Engelhardt H, Smits RA, Leurs R, et al. A new 78. Akiyama T, Tominaga M, Takamori K, et al. Roles of
itch pathophysiology: component analysis of atopic generation of anti-histamines: histamine H4 receptor glutamate, substance P, and gastrin-releasing peptide
itch using the ‘Eppendorf Itch Questionnaire. Int Arch antagonists on their way to the clinic. Curr Opin Drug as spinal neurotransmitters of histaminergic and
Allergy Immunol 2001;124:326–31. Discov Devel 2009;12:628–43. nonhistaminergic itch. Pain 2014;155:80–92.
30. Yosipovitch G. Itch questionnaires as tools for itch 53. Huang JF, Thurmond RL. The new biology of histamine 79. Akiyama T, Tominaga M, Davoodi A, et al. Roles for
evaluation. In: Yosipovitch G, Greaves MW, Fleischer receptors. Curr Allergy Asthma Rep 2008;8:21–7. substance P and gastrin-releasing peptide as
AB Jr, McGlone F, editors. Itch: basic mechanisms and 54. Goswami SC, Thierry-Mieg D, Thierry-Mieg J, et al. neurotransmitters released by primary afferent
therapy. New York: Marcel Dekker; 2004. p. 169–82. Itch-associated peptides: RNA-Seq and bioinformatic pruriceptors. J Neurophysiol 2013;109:742–8.
31. O’Neill JL, Chan YH, Rapp SR, et al. Differences in itch analysis of natriuretic precursor peptide B and gastrin 80. Mendell LM, Albers KM, Davis BM. Neurotrophins,
characteristics between psoriasis and atopic dermatitis releasing peptide in dorsal root and trigeminal nociceptors, and pain. Microsc Res Tech
patients: results of a web-based questionnaire. Acta ganglia, and the spinal cord. Mol Pain 2014;10:44. 1999;45:252–61.
Derm Venereol 2011;91:537–40. 55. Liu XY, Wan L, Huo FQ, et al. B-type natriuretic peptide 81. Nockher WA. Neurotrophins in allergic diseases: from
32. Andrew D, Craig AD. Spinothalamic lamina I neurons is neither itch-specific nor functions upstream of the neuronal growth factors to intercellular signaling
selectively sensitive to histamine: a central neural GRP-GRPR signaling pathway. Mol Pain 2014;10:4. molecules. J Allergy Clin Immunol 2006;117:583–9.
pathway for itch. Nat Neurosci 2001;4:72–7. 56. Fleming MS, Ramos D, Han SB, et al. The majority of 82. Donnerer J, Schuligoi R, Amann R. Increased content
33. Papoiu AD, Coghill RC, Kraft RA, et al. A tale of two dorsal spinal cord gastrin releasing peptide is and transport of substance P and calcitonin
itches. Common features and notable differences in synthesized locally whereas neuromedin B is highly gene-related peptide in sensory nerves innervating
brain activation evoked by cowhage and histamine expressed in pain- and itch-sensing somatosensory inflamed tissue: evidence for a regulatory function of
induced itch. Neuroimage 2012;59:3611–23. neurons. Mol Pain 2012;8:52. nerve growth factor in vivo. Neuroscience
34. Hsieh JC, Hagermark O, Stahle-Backdahl M, et al. Urge 57. Luger TA. Neuromediators – a crucial component of 1992;49:693–8.
to scratch represented in the human cerebral cortex the skin immune system. J Dermatol Sci 83. Nakamura M, Toyoda M, Morohashi M. Pruritogenic
during itch. J Neurophysiol 1994;72:3004–8. 2002;30:87–93. mediators in psoriasis vulgaris: comparative evaluation
35. Drzezga A, Darsow U, Treede RD, et al. Central 58. Grobe K, Poppelmann M, Becker WM, Petersen A. of itch-associated cutaneous factors. Br J Dermatol
activation by histamine-induced itch: analogies to Properties of group I allergens from grass pollen and 2003;149:718–30.
pain processing: a correlational analysis of O-15 H2O their relation to cathepsin B, a member of the C1 84. Raychaudhuri SP, Raychaudhuri SK. Role of NGF and
positron emission tomography studies. Pain family of cysteine proteinases. Eur J Biochem neurogenic inflammation in the pathogenesis of
2001;92:295–305. 2002;269:2083–92. psoriasis. Prog Brain Res 2004;146:433–7.
36. McGlone F, Rukweid R, Howard M, Hitchcock D. 59. Pan ZZ. mu-Opposing actions of the kappa-opioid 85. Groneberg DA, Serowka F, Peckenschneider N, et al.
Histamine-induced discriminative and affective receptor. Trends Pharmacol Sci 1998;19:94–8. Gene expression and regulation of nerve growth
responses revealed by functional MRI. In: Yosipovitch 60. Umeuchi H, Togashi Y, Honda T, et al. Involvement of factor in atopic dermatitis mast cells and the human
G, Greaves MW, Fleischer AB Jr, McGlone F, editors. central mu-opioid system in the scratching behavior mast cell line-1. J Neuroimmunol 2005;161:87–92.
Itch: basic mechanisms and therapy. New York: Marcel in mice, and the suppression of it by the activation of 86. Yamaguchi J, Aihara M, Kobayashi Y, et al. Quantitative
Decker; 2004. p. 51–61. kappa-opioid system. Eur J Pharmacol analysis of nerve growth factor (NGF) in the atopic
37. Darsow U, Drzezga A, Frisch M, et al. Processing of 2003;477:29–35. dermatitis and psoriasis horny layer and effect of
histamine-induced itch in the human cerebral cortex: 61. Papoiu AD, Kraft RA, Coghill RC, et al. Butorphanol treatment on NGF in atopic dermatitis. J Dermatol Sci
a correlation analysis with dermal reactions. J Invest suppression of histamine itch is mediated by nucleus 2009;53:48–54.
Dermatol 2000;115:1029–33. accumbens and septal nuclei. A pharmacological fMRI 87. Kanda N, Watanabe S. Histamine enhances the
38. Ishiuji Y, Coghill RC, Patel TS, et al. Distinct patterns of study. J Invest Dermatol 2015;135:560–8. production of granulocyte-macrophage colony-
brain activity evoked by histamine-induced itch reveal 62. Kardon AP, Polgár E, Hachisuka J, et al. Dynorphin acts stimulating factor via protein kinase C(alpha) and
an association with itch intensity and disease severity as a neuromodulator to inhibit itch in the dorsal horn extracellular signal-regulated kinase in human
in atopic dermatitis. Br J Dermatol 2009;161:1072–80. of the spinal cord. Neuron 2014;82:573–86. keratinocytes. J Invest Dermatol 2004;122:863–72.
39. Yosipovitch G, Ishiuji Y, Patel TS, et al. The brain 63. Togashi Y, Umeuchi H, Okano K, et al. Antipruritic 88. Dou YC, Hagstromer L, Emtestam L, Johansson O.
processing of scratching. J Invest Dermatol activity of the kappa-opioid receptor agonist, TRK-820. Increased nerve growth factor and its receptors in
2008;128:1806–11. Eur J Pharmacol 2002;435:259–64. atopic dermatitis: an immunohistochemical study.
40. Vierow V, Fukuoka M, Ikoma A, et al. Cerebral 64. Bigliardi PL, Tobin DJ, Gaveriaux-Ruff C, Bigliardi-Qi M. Arch Dermatol Res 2006;298:31–7.
representation of the relief of itch by scratching. J Opioids and the skin—where do we stand? Exp 89. Grewe M, Vogelsang K, Ruzicka T, et al. Neurotrophin-4 109
Neurophysiol 2009;102:3216–24. Dermatol 2009;18:424–30. production by human epidermal keratinocytes:
increased expression in atopic dermatitis. J Invest neuroendocrine basis of pruritus. J Invest Dermatol 127. Liu T, Han Q, Chen G, et al. Toll-like receptor 4
Dermatol 2000;114:1108–12. 2006;126:1705–18. contributes to chronic itch, alloknesis, and spinal
90. Hagermark O, Strandberg K, Hamberg M. Potentiation 109. Wahlgren CF, Tengvall LM, Hagermark O, Scheynius A. astrocyte activation in male mice. Pain
of itch and flare responses in human skin by Itch and inflammation induced by intradermally 2016;157:806–17.
prostaglandins E2 and H2 and a prostaglandin injected interleukin-2 in atopic dermatitis patients and 128. Schmelz M. Itch and pain. Dermatol Ther
SECTION
endoperoxide analog. J Invest Dermatol healthy subjects. Arch Dermatol Res 1995;287:572–80. 2005;18:304–7.

2 91.
1977;69:527–30.
Hagermark O, Strandberg K. Pruritogenic activity of
110. Hawro T, Saluja R, Weller K, et al. Interleukin-31 does
not induce immediate itch in atopic dermatitis
128a. Andersen HH, Elberling J, Sølvsten H, et al.
Nonhistaminergic and mechanical itch sensitization in
prostaglandin E2. Acta Derm Venereol 1977;57:37–43. patients and healthy controls after skin challenge. atopic dermatitis. Pain 2017;158:1780–91.
PRURITUS

92. Greaves MW, McDonald-Gibson W. Itch: role of Allergy 2014;69:113–17. 129. Akiyama T, Nguyen T, Curtis E, et al. A central role for
prostaglandins. Br Med J 1973;3:608–9. 111. Cevikbas F, Wang X, Akiyama T, et al. A sensory spinal dorsal horn neurons that express neurokinin-1
93. Daly BM, Shuster S. Effect of aspirin on pruritus. Br neuron-expressed IL-31 receptor mediates T helper receptors in chronic itch. Pain 2015;156:1240–6.
Med J (Clin Res Ed) 1986;293:907. cell-dependent itch: Involvement of TRPV1 and TRPA1. 130. Ikoma A, Handwerker H, Miyachi Y, Schmelz M.
94. Yosipovitch G, Sugeng MW, Chan YH, et al. The effect J Allergy Clin Immunol 2014;133:448–60. Electrically evoked itch in humans. Pain
of topically applied aspirin on localized circumscribed 112. Zhang Q, Putheti P, Zhou Q, et al. Structures and 2005;113:148–54.
neurodermatitis. J Am Acad Dermatol 2001;45:910–13. biological functions of IL-31 and IL-31 receptors. 131. Ikoma A, Fartasch M, Heyer G, et al. Painful stimuli
95. Andoh T, Haza S, Saito A, et al. Involvement of Cytokine Growth Factor Rev 2008;19:347–56. evoke itch in patients with chronic pruritus: central
leukotriene B4 in spontaneous itch-related behaviour 113. Arita K, South AP, Hans-Filho G, et al. Oncostatin M sensitization for itch. Neurology 2004;62:212–17.
in NC mice with atopic dermatitis-like skin lesions. Exp receptor-beta mutations underlie familial primary 132. Ishiuji Y, Coghill RC, Patel TS, et al. Repetitive
Dermatol 2011;20:894–8. localized cutaneous amyloidosis. Am J Hum Genet scratching and noxious heat do not inhibit histamine-
96. Andoh T, Takayama Y, Kuraishi Y. Involvement of 2008;82:73–80. induced itch in atopic dermatitis. Br J Dermatol
leukotriene B4 in dermatophyte-related itch in mice. 114. Sonkoly E, Muller A, Lauerma AI, et al. IL-31: a new link 2008;158:78–83.
Pharmacol Rep 2014;66:699–703. between T cells and pruritus in atopic skin 133. Binder A, Koroschetz J, Baron R. Disease mechanisms
97. Andoh T, Kuraishi Y. Lipid mediators and itch. In: inflammation. J Allergy Clin Immunol in neuropathic itch. Nat Clin Pract Neurol
Carstens E, Akiyama T, editors. Itch: mechanisms and 2006;117:411–17. 2008;4:329–37.
treatment. Boca Raton (FL): CRC Press; 2014. p. 115. Neis MM, Peters B, Dreuw A, et al. Enhanced 134. Akiyama T, Merrill AW, Zanotto K, et al. Scratching
271–80. expression levels of IL-31 correlate with IL-4 and IL-13 behavior and Fos expression in superficial dorsal horn
98. Stander S, Moormann C, Schumacher M, et al. in atopic and allergic contact dermatitis. J Allergy Clin elicited by protease-activated receptor agonists and
Expression of vanilloid receptor subtype 1 in Immunol 2006;118:930–7. other itch mediators in mice. J Pharmacol Exp Ther
cutaneous sensory nerve fibers, mast cells, and 116. Nattkemper LA, Martinez-Escala ME, Gelman AB, et al. 2009;329:945–51.
epithelial cells of appendage structures. Exp Dermatol Cutaneous T-cell lymphoma and pruritus: the 135. Nojima H, Carstens MI, Carstens E. c-fos expression in
2004;13:129–39. expression of IL-31 and its receptors in the skin. Acta superficial dorsal horn of cervical spinal cord
99. Imamachi N, Park GH, Lee H, et al. TRPV1-expressing Derm Venereol 2016;96:894–8. associated with spontaneous scratching in rats with
primary afferents generate behavioral responses to 117. Singer EM, Shin DB, Nattkemper LA, et al. IL-31 is dry skin. Neurosci Lett 2003;347:62–4.
pruritogens via multiple mechanisms. Proc Natl Acad produced by the malignant T-cell population in 136. Ali SM, Yosipovitch G. Skin pH: from basic science to
Sci USA 2009;106:11330–5. cutaneous T-Cell lymphoma and correlates with CTCL basic skin care. Acta Derm Venereol 2013;93:261–7.
100. Yoshioka T, Imura K, Asakawa M, et al. Impact of the pruritus. J Invest Dermatol 2013;133:2783–5. 137. Valdes-Rodriguez R, Mollanazar NK, González-Muro J,
Gly573Ser substitution in TRPV3 on the development 118. Sokołowska-Wojdyło M, Gleń J, Zabłotna M, et al. et al. Itch prevalence and characteristics in a hispanic
of allergic and pruritic dermatitis in mice. J Invest Association of distinct IL-31 polymorphisms with geriatric population: a comprehensive study using a
Dermatol 2009;129:714–22. pruritus and severity of atopic dermatitis. J Eur Acad standardized itch questionnaire. Acta Derm Venereol
101. Wilson SR, Gerhold KA, Bifolck-Fisher A, et al. TRPA1 is Dermatol Venereol 2013;27:662–4. 2015;95:417–21.
required for histamine-independent, Mas-related G 119. Luo Y, Zhou B, Zhao M, et al. Promoter demethylation 138. Berger TG, Shive M, Harper GM. Pruritus in the older
protein-coupled receptor-mediated itch. Nat Neurosci contributes to TSLP overexpression in skin lesions of patient: a clinical review. JAMA 2013;310:2443–50.
2011;14:595–602. patients with atopic dermatitis. Clin Exp Dermatol 139. Kremer AE, Bolier R, van Dijk R, et al. Advances in
102. Wilson SR, Thé L, Batia LM, et al. The epithelial 2014;39:48–53. pathogenesis and management of pruritus in
cell-derived atopic dermatitis cytokine TSLP activates 120. Maurer T, Poncelet A, Berger T. Thalidomide treatment cholestasis. Dig Dis 2014;32:637–45.
neurons to induce itch. Cell 2013;155:285–95. for prurigo nodularis in human immunodeficiency 140. Yosipovitch G. Pruritus: an update. Curr Probl
103. Miyamoto T, Nojima H, Kuraishi Y. Intradermal virus-infected subjects: efficacy and risk of Dermatol 2003;15:137–64.
cholinergic agonists induce itch-associated response neuropathy. Arch Dermatol 2004;140:845–9. 140a. Stull C, Lavery MJ, Yosipovitch G. Advances in
via M3 muscarinic acetylcholine receptors in mice. Jpn 121. Twycross R, Greaves MW, Handwerker H, et al. Itch: therapeutic strategies for the treatment of pruritus.
J Pharmacol 2002;88:351–4. scratching more than the surface. QJM 2003;96:7–26. Expert Opin Pharmacother 2016;17:671–87.
104. Gotoh Y, Andoh T, Kuraishi Y. Noradrenergic regulation 122. Yosipovitch G, Carstens E, McGlone F. Chronic itch and 141. Tey HL, Wallengren J, Yosipovitch G. Psychosomatic
of itch transmission in the spinal cord mediated by chronic pain: Analogous mechanisms. Pain factors in pruritus. Clin Dermatol 2013;31:31–40.
α-adrenoceptors. Neuropharmacology 2007;131:4–7. 142. Yamamoto Y, Yamazaki S, Hayashino Y, et al.
2011;61:825–31. 123. Martinelli-Boneschi F, Colombi M, Castori M, et al. Association between frequency of pruritic symptoms
105. Kido-Nakahara M, Buddenkotte J, Kempkes C, et al. COL6A5 variants in familial neuropathic chronic itch. and perceived psychological stress: a Japanese
Neural peptidase endothelin-converting enzyme 1 Brain 2017;140:155–67. population-based study. Arch Dermatol
regulates endothelin 1-induced pruritus. J Clin Invest 124. Rukwied RR, Main M, Weinkauf B, et al. NGF sensitizes 2009;145:1384–8.
2014;124:2683–95. nociceptors for cowhage- but not histamine-induced 143. Tran BW, Papoiu ADP, Russoniello C, et al. The effect of
106. Andoh T, Kuraishi Y. Nitric oxide enhances substance itch in human skin. J Invest Dermatol 2013;133:268–70. itch, scratching and mental stress on autonomic
P-induced itch-associated responses in mice. Br J 125. Akiyama T, Carstens MI, Carstens E. Enhanced nervous system function in atopic dermatitis. Acta
Pharmacol 2003;138:202–8. scratching evoked by PAR-2 agonist and 5-HT but not Derm Venereol 2010;90:354–61.
107. Sikand P, Dong X, Lamotte RH. BAM8-22 peptide histamine in a mouse model of chronic dry skin itch. 144. Kupfer J, Gieler U, Yosipovitch G. Psychological
produces itch and nociceptive sensations in humans Pain 2010;151:378–83. interventions in the treatment of chronic itch. Acta
independent of histamine release. J Neurosci 126. Shiratori-Hayashi M, Koga K, Tozaki-Saitoh H, et al. Derm Venereol 2016;96:157–61.
2011;31:7563–7. STAT3-dependent reactive astrogliosis in the spinal
108. Steinhoff M, Bienenstock J, Schmelz M, et al. dorsal horn underlies chronic itch. Nat Med
Neurophysiological, neuroimmunological, and 2015;21:927–31.

110

You might also like