Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of...

63
Molecular Mechanisms of Itch Signaling in Keratinocytes. By Lydia The A dissertation submitted in partial satisfaction of the requirements for the degree of Doctor of Philosophy in Molecular and Cell Biology in the Graduate Division of the University of California, Berkeley Committee in charge: Professor Diana M. Bautista, Chair Professor Matthew Welch Professor Marla Feller Professor Song Li Spring 2014

Transcript of Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of...

Page 1: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

Molecular Mechanisms of Itch Signaling in Keratinocytes.

By Lydia The

A dissertation submitted in partial satisfaction of the requirements for the degree of

Doctor of Philosophy in

Molecular and Cell Biology in the

Graduate Division of the

University of California, Berkeley

Committee in charge:

Professor Diana M. Bautista, Chair Professor Matthew Welch

Professor Marla Feller Professor Song Li

Spring 2014

Page 2: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting
Page 3: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

  1

Abstract Molecular Mechanisms of Itch Signaling in Keratinocytes.

by Lydia The

Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair

There is accumulating evidence suggesting that keratinocytes, the predominant cell type in the skin, may play a key role in the transduction of several somatosensory modalities, including itch. Keratinocytes are activated by a number of itch compounds and release a variety of molecules associated with itch. Additionally, itch-transducing neurons terminate as free nerve endings in the keratinocyte layer of the skin, which would permit rapid keratinocyte-neuron signaling. Our work has focused on two areas: 1) defining the molecular mechanisms by which the atopic dermatitis cytokine TSLP is released from keratinocytes and act on neurons to cause itch and 2) identifying the distinct mechanisms by which histamine and protease signaling in keratinocytes triggers itch. Previous studies have shown that activation of the Protease-Activated Receptor 2 (PAR2) leads to upregulation of TSLP in keratinocytes and that TSLP overexpression in keratinocytes induces chronic itch in mice. We characterized the mechanisms by which PAR2 activation leads to TSLP expression and secretion, and elucidated the mechanisms by which secreted TSLP in the skin leads to itch. Understanding the molecular mechanisms underlying atopic dermatitis may identify targets that could lead to the development of drugs that specifically block atopic dermatitis itch while leaving other somatosensations intact. Previous studies have linked histamine and protease signaling to distinct clinical itch phenotypes; histamine signaling is associated with allergic itch, while protease signaling is associated with atopic dermatitis. Histamine and PAR2 agonists activate the same population of keratinocytes, thus how these pruritogens trigger distinct itch pathologies was unknown. We now demonstrate that histamine and protease signaling in keratinocytes induce distinct calcium signals that lead to divergent downstream transcriptional outputs.

Page 4: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

i

ACKNOWLEDGEMENTS For contributing to Chapter I, I would like to thank Drs. Hariharan and Heald and members of my laboratory for critical comments on the manuscript, K. Gerhold, J. Meeuwsen, T. Morita, M. Newton and M. Tsunozaki for behavior data analyses, and the Neurobiology course at the Marine Biological Laboratory for pilot experiments. For contributing to Chapter II, I would like to thank members of my laboratory for helpful discussions. I would also like to thank my committee Drs. Feller, Welch and Li for critical reading of the manuscript.

Page 5: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

ii

TABLE OF CONTENTS

• Introduction iii-xiv o Acute Versus Chronic Itch iii o The Mammalian Somatosensory System iii-v o Histamine-Dependent Versus Histamine-Independent Itch Signaling v-vi o Itch Coding vii-ix o A Role for Epidermal Cells in Itch x-xi o Mechanisms of Chronic Itch xii-xiii o Open Questions in Itch Biology xiii-xiv

• Chapter I: The Epithelial Cell-Derived Atopic Dermatitis Cytokine TSLP

Activates Neurons to Induce Itch 1-20 o Summary and Introduction 1-2 o Results 2-16 o Discussion 16-18 o Experimental Procedures 18-20

• Chapter II: Distinct Histamine and PAR2 Signaling in Keratinocytes 37-49

o Summary and Introduction 37-38 o Results 38-46 o Discussion 47-48 o Experimental Procedures 48-49

• References 50-62

Page 6: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

iii

Itch, or pruritus, is defined as the irritating sensation that elicits a desire to scratch. Itch can be categorized as acute or chronic. Like pain, acute itch serves a protective function by helping us avoid disease-carrying insects or toxic plants. However, chronic itch, which can be caused by diseases such as atopic dermatitis, is persistent and no longer serves a protective function. It is estimated that 20% of the population will experience chronic itch in their lifetime and many forms of chronic itch are refractory to available therapies. Here we introduce the cells, molecules and circuitry that mediate acute itch transduction, explore the mechanisms of chronic itch and discuss open questions in the field. ACUTE VERSUS CHRONIC ITCH Acute itch occurs in the absence of disease, following activation of primary somatosensory neurons in the skin. Sensory neuron activation may be due to direct activation by exogenous pruritogens, itch-causing compounds such as dust mite-derived proteases, or through an endogenous intermediate, such as mast cell-derived histamine. Acute itch can be triggered in experimental models by injecting an animal with either pruritogens that act directly on sensory neurons, or molecules that activate non-neuronal cells, such as 48/80, which causes mast cells to degranulate and release pruritogens. Chronic itch is associated with a variety of disorders including atopic dermatitis, psoriasis, bacterial infection, kidney disease and cutaneous T-cell lymphoma. Treatments for chronic itch include topical steroids, immunomodulators and moisturizers to prevent water loss from the skin. While acute itch models have been useful in studying itch physiology, chronic itch models have been developed to better understand clinical itch conditions. Chronic itch models include dry skin induced by organic solvents (Miyamoto et al., 2002; Nojima et al., 2003), toxic contact dermatitis induced by diphenylcyclopropenone (Seike et al., 2005; Sun et al., 2009), allergic contact dermatitis induced by 2,4-dinitro-1-fluorobenzene or oxazolone (Liu et al., 2013; Man et al., 2008; Saint-Mezard et al., 2003) and the atopic dermatitis NC/Nga genetic line (Grimstad et al., 2009; Ohsawa and Hirasawa, 2012). Importantly, most models of chronic itch reproduce itch derived from skin disorders; there is a dearth of models of itch derived from systemic disorders. CELLS AND SYSTEMS THAT PROMOTE ACUTE ITCH THE MAMMALIAN SOMATOSENSORY SYSTEM Itch and other tactile stimuli such as mechanical force and temperature are detected by the somatosensory system. The cell bodies of somatosensory neurons are located in the trigeminal ganglia, near the base of the skull, and dorsal root ganglia (DRG), outside the spinal cord. Sensory neurons in the DRG project peripheral afferent fibers to the skin and other organs in the body, where detection occurs, and central fibers to the dorsal horn of the spinal cord where they synapse onto interneurons that project to the brain stem and thalamus. Sensory neurons in the trigeminal ganglia innervate the face and send efferent projections directly to the brain stem (Figure 1).

Page 7: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

iv

Figure 1. A diverse array of somatosensory neurons transduce touch, temperature, pain and itch Unmyelinated, nociceptive C-fibers detect pain, temperature and itch. Peptidergic C-fibers terminate in the stratum spinosum (green) and non-peptidergic axons penetrate the stratum granulosum (pink). Aδ fibers terminate in the dermis as either nociceptive free nerve endings (blue) or light touch receptors associated with down hair (not shown). Myelinated Aβ fibers, which detect mechanical force, can terminate anywhere from subcutaneously to the epidermis where they associate with specialized structures, such as Merkel cells, Meissnerʼs corpuscles, Pacinian corpuscles, and Ruffini corpuscles (purple). The degree of myelination is associated with conduction velocity, axon diameter and cell body diameter in the DRG, as shown. Modified from Lumpkin and Caterina, 2007.

Page 8: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

v

Somatosensory neurons are a heterogeneous population that may be classified based on their physiological, morphological, anatomical and molecular characteristics. Physiologically, somatosensory neurons can be broadly categorized by conduction velocity: unmyelinated C-fibers, lightly myelinated Aδ-fibers and heavily myelinated Aβ-fibers (<1.3 m/s, 5 m/s and 13 m/s, respectively). These three fiber types are also specialized to detect distinct sensory stimuli. Many C-fibers respond to painful, mechanical, thermal and chemical stimuli and are thus polymodal. They terminate in the skin as free nerve endings and centrally project to superficially layers (I and II) of the dorsal horn in the spinal cord. Lighly myelinated Aδ-fibers are subdivided into Aδ-nociceptors, which detect pain and terminate in the skin as free nerve endings, and D-hair receptors, which detect mechanical force and innervate hair follicles. Heavily myelinated Aβ-fibers detect mechanical stimuli and innervate a variety of structures in the skin, including Merkel cells, hair follicles, Meissner corpuscles, Pacinian corpuscles and Ruffini endings. These mechanical touch receptors project centrally to deeper layers of the dorsal horn. C-fibers are also defined by molecular properties. For example, C-fibers can be classified as peptidergic or nonpeptidergic. Nonpeptidergic C-fibers are distinguished by their binding to the plant lectin isolectin B4 (IB4). Peptidergic C-fibers contain large dense core vesicles that release inflammatory neuropeptides, such as substance P and calcitonin gene-related peptide (CGRP), at both their central and peripheral terminals. These inflammatory peptides trigger vasodilation and immune cell infiltration into the skin. The transient receptor potential channel A1 (TRPA1) is expressed in a subset of peptidergic C-fibers (Story et al., 2003). TRPA1 detects a variety of irritants (Bandell et al., 2004; Bautista et al., 2005) and activation of TRPA1 leads to release of neuropeptides (Nakamura et al., 2012). Itch is thought to be transduced by a subpopulation of peptidergic and nonpeptidergic neurons. Genetic ablation of CGRPα-expressing peptidergic neurons reduced itch behavior in response to multiple pruritogens (McCoy et al., 2013), while ablation of MrgD-expressing nonpeptidergic neurons attenuated itch behavior in response to β-alanine (Liu et al., 2012). HISTAMINE-DEPENDENT VERSUS HISTAMINE-INDEPENDENT ITCH SIGNALING Itch can be classified as either histamine-dependent or histamine-independent. Histamine-mediated mechanisms are better understood and are important in allergic itch, while histamine-independent mechanisms are important in chronic itch. Recent work has demonstrated that histamine and histamine-independent signaling are largely distinct in the nervous system. Our work demonstrates that there are also distinct histamine and histamine-independent signaling pathways in keratinocytes. HISTAMINERGIC ITCH Histamine is a pruritogen that is secreted by a variety of cells including mast cells, basophils and keratinocytes. Histamine then acts directly on primary sensory neurons to induce itch (Jeffry et al., 2011). Four histamine receptors have been identified in mammals: GPCRs H1-4R. H1R has been most closely implicated in itch and is the

Page 9: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

vi

target of most antihistamines, which are frequently used to relieve itch from allergic reactions. H1R couples to Gq/G11 proteins and activates phospholipase Cβ3 and transient receptor potential cation channel V1 (TRPV1), resulting in increased calcium levels and action potential firing (Han et al., 2006; Shim et al., 2007). PLCβ3 and TRPV1 are required for both histamine-evoked calcium signals in DRG neurons and histamine-evoked itch behavior (Han et al., 2006; Imamachi et al., 2009; Kim et al., 2004). Recent work suggests that H4R also has a role in itch (Bell et al., 2004). H4R agonists induced dose-dependent scratching that was significantly reduced by H4R antagonists. The mechanism of H4R-mediated histamine signaling in somatosensory neurons is unknown (Liu and Ji, 2013). H4R antagonists are under investigation for their potential as superior antihistamines than H1R antagonists (Dunford et al., 2007). HISTAMINE-INDEPENDENT ITCH Many forms of chronic itch are resistant to antihistamines, and are mediated by histamine-independent mechanisms for itch. The existence of a histamine-independent itch circuit stems from studies of the cowhage plant (Mucuna pruriens). Spicules from this plant contain the cysteine protease mucunain, which causes histamine-independent itch (Johanek et al., 2007; Reddy et al., 2008). Unlike histamine, cowage induces itch without weal, flare and redness (Johanek et al., 2007). Importantly, cowage and histamine activate distinct classes of C-fibers (Johanek et al., 2008). Cowhage preferentially excited mechanically sensitive C-fibers (90% of mechano-heat-sensitive C-fibers responded to cowhage and 76% responded to histamine), while histamine preferentially excited mechano-insensitive C-fibers (20% of mechanically insensitive C-fibers responded to histamine and none responded to cowhage). The separation of histamine and cowage signaling is preserved in the spinal cord, where they activate distinct subsets of lamina I spinothalamic neurons (Davidson et al., 2007), and in the brain where they activate incompletely overlapping areas of the cortex (Papoiu et al., 2012). Mucunain activates the protease-activated receptor PAR2 (Reddy et al., 2008). PAR family members are GPCRs that are activated by cleavage of their N-terminus, revealing a new N-terminus that acts as a tethered ligand. PAR2 detects a variety of itch-inducing endogenous and exogenous agonists, including mast cell-derived tryptase, keratinocyte-derived kallikreins and dust mite-derived proteases. However, a recent study utilizing PAR2 deficient mice called the role of PAR2 in trypsin- and SLIGRL-evoked itch into question (Liu et al., 2011). Clinically, PAR2 and PAR2 agonists, such as tryptase, are upregulated in the skin of atopic dermatitis (AD) patients (Steinhoff et al., 2003). Additionally, in a dry skin model of chronic itch, the percentage of PAR2-expressing DRG neurons was increased and itch behavior was attenuated by treatment with PAR2 antibodies (Akiyama et al., 2010). Several TRP channels have been implicated in the signal transduction downstream of PAR2, including TRPA1, TRPV1 and TRPV4 (Amadesi et al., 2006; 2004; Dai et al., 2004; 2007; Grant et al., 2007; Vellani et al., 2010).

Page 10: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

vii

ITCH CODING There are multiple theories for how itch and pain are coded. Initially, itch was believed to be coded as low-intensity signals through nociceptive C-fibers. According to this intensity theory, weak activation of nociceptors triggers itch sensations, while strong activation would trigger pain sensations (Ikoma et al., 2006). This theory was supported by findings that some pruritogens can cause inflammatory pain at higher concentrations and many C-fibers are polymodal, responding to both itch and pain compounds (Sikand et al., 2009; 2011). Recently, two new hypotheses have been proposed: selectivity theory and labeled line theory. According to the selectivity theory, there are overlapping neurons that are selective for itch and pain. While most C-fibers transmit only pain, some polymodal fibers transmit both itch and pain. Itch is perceived when polymodal itch-sensitive fibers are selectively activated. Pain is perceived when a larger population of C-fibers is activated because this population inhibits itch sensation from the polymodal neurons (Garibyan et al., 2013). This theory is supported by the observation that painful scratching can suppress itch sensation. The labeled line theory postulates that itch is encoded with specific itch receptors and a dedicated neural circuit in both the periphery and spinal cord. Recent evidence supports both selectivity and labeled line theories and we believe that parts of both theories are likely to be correct. Initial evidence for itch-specific neurons in the periphery came from microneurography experiments in humans, whereby multiunit recordings of sensory neuron activity are obtained in conscious subjects. Populations of sensory neurons that respond to itch compounds but not mechanical stimuli were identified, which supported the existence of itch-specific neurons (Schmelz et al., 1997). However, they were later shown to also respond to other stimuli including heat and capsaicin, and thus are itch-selective (Schmelz et al., 2003). Genetic ablation experiments in mice offered further evidence of the existence of itch-specific primary afferent neurons (Han et al., 2013). Investigators found that MrgprA3-expressing neurons conform to three criteria for itch-specificity: they respond to pruritogens, loss of these neurons specifically reduces itch and not pain behaviors, and activation of these neurons evokes itch and not pain behaviors. They previously showed that MrgprA3 functions as a receptor for the pruritogen chloroquine (Liu et al., 2009). The authors demonstrated that ablation of MrgprA3-expressing DRG neurons reduced itch behaviors to a number of pruritogens, but had no effect on pain behavior. Additionally, when capsaicin receptor TRPV1 was ectopically expressed in MrgprA3-expressing neurons in a TRPV1 knockout background, capsaicin caused itch behavior instead of pain. Convincing evidence for itch-specific transmission in the dorsal horn comes from studies on gastrin-releasing peptide (GRP), a 27-amino acid peptide (Sun and Chen, 2007; Sun et al., 2009). GRP is expressed in unmyelinated C-fibers. Most GRP-expressing fibers also express TRPV1 and project to lamina I and II of the dorsal spinal cord. The receptor for GRP, GRPR, is expressed in a subset of lamina I spinal cord

Page 11: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

viii

neurons. Intrathecal injection of GRP causes robust scratching. Ablation of GRPR-expressing neurons attenuated itch behavior in response to a range of pruritogens, including histamine, 48/80, 5-HT, ET-1, SLIGRL and CQ, but had no effect on heat, mechanical and chemical pain behavior. This finding supports the existence of labeled lines for itch and pain at the level of the spinal cord. However, recent evidence contradicts the original hypothesis that GRP is released from primary sensory neurons. Pharmacological studies suggest that glutamate is the principle neurotransmitter between C-fibers and GRP-sensitive dorsal horn neurons (Koga et al., 2011). Additionally, a new model proposes that neuropeptide natriuretic polypeptide b (Nppb) is a neuropeptide released by a subset of TRPV1-expression neurons to signal itch upstream of GRP ((Mishra and Hoon, 2013), Figure 2). Nppb injection triggers robust scratching and mice deficient in Nppb display defective itch behavior, but normal pain behaviors. Ablation of cells expressing the natriuretic peptide receptor A (Npra) results in defective itch behavior to a variety of pruritogens, but normal itch behavior in response to GRP. In support of the selectivity theory, recent studies have identified the cellular correlates of inhibition of itch by painful stimuli. Psychophysical studies have demonstrated that painful stimuli inhibit itch sensation (Yosipovitch et al., 2007) and that inhibition of pain by opioids can cause itch (Ballantyne et al., 1988). Vesicular glutamate transporter type 2 (VGLUT2) is expressed in a subset of TRPV1-expressing neurons and is required for loading glutamate into synaptic vesicles and for synaptic glutamate release. VGLUT2 in nociceptors is required for mediating acute pain. Deletion of VGLUT2 in a subset of nociceptors resulted in deficits in pain behaviors, but also in spontaneous itch behavior and sensitization to a variety of pruritogens (Lagerström et al., 2010; Liu et al., 2010). These findings support a model where VGLUT2 is required for a constitutively inhibitory circuit whereby nociceptors block itch signals. Inhibition of itch in the dorsal horn was demonstrated through genetic deletion of atonal-related transcription factor Bhlhb5 (Ross et al., 2010). Bhlhb5 is expressed in the dorsal horn of the developing spinal cord and is required for the development of a subset of inhibitory interneurons in the dorsal horn. Mice lacking Bhlhb5 display spontaneous itch behavior. Additionally, the pain compound formalin induced itch, not nociceptive behaviors, in these mice.

Page 12: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

ix

Figure 2. Model of itch and pain coding Itch-responsive neurons, or pruriceptors, may express a range of itch receptors including MrgA3 and H1R. Activation of these primary itch afferents triggers neurotransmitter release at the central terminals in the dorsal horn. Recent evidence suggests that Nppb is required for the transmission of a variety of itch signals and lies upstream of GRP. Pain signals are detected by a population of neurons that may express VGLUT2, TRPV1 and/or TRPA1. TRPV1-expressing nociceptors are believed to exert tonic inhibition upon itch transduction, which is revealed upon deletion of VGLUT2 in nociceptors. Bhlhb5 is required for the development of a population of interneurons that inhibit itch. Further work is needed to clarify whether VGLUT2-expressing nociceptors activate Bhlhb5-dependent neurons and how Bhlhb5-dependent neurons inhibit the itch circuit.

Page 13: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

x

A ROLE FOR EPIDERMAL CELLS IN ITCH In addition to their role in releasing histamine, keratinocytes may act in concert with somatosensory neurons to detect and transduce itch stimuli. In fact, a unique feature of itch is that it is restricted to the skin, mucous membranes and the cornea (Yosipovitch and Papoiu, 2008). No other tissue experiences itch, although sensory neurons do innervate the viscera. This is tantalizing evidence that itch is unique to the activity between keratinocytes in the epidermis and the C-fibers that innervate it. There are four lines of evidence that suggest keratinocytes are key players in itch (Figure 3). First, keratinocytes are directly activated by a number of pruritogens, including histamine, SLIGRL, ET-1 and substance P (Dallos et al., 2006; Giustizieri et al., 2004; Santulli et al., 1995; Yohn et al., 1994). Keratinocytes in turn release inflammatory molecules in response to pruritogens, which might account for enhanced sensitivity under chronic itch conditions. For instance, the substance P receptor, neurokinin-1, is highly expressed in the skin of psoriasis patients (Chang et al., 2007). Substance P-treated keratinocytes respond with increased production of NGF (Dallos et al., 2006), which induces nerve sprouting and enhanced itch sensitivity. Second, the location of itch-transducing C-fibers as free nerve endings in the skin would facilitate rapid signaling between keratinocytes and neurons. Peptidergic itch-transducing C-fibers terminate in the stratum spinosum, while non-peptidergic C-fibers penetrate more superficially to the stratum granulosum (Zylka et al., 2005). Classical synapses have not been observed between keratinocytes and neurons. However, electron microscopy of the skin reveals membrane-membrane apposition, where gap-junctions or chemical synapses could exist (Hilliges et al., 1995). Third, keratinocytes secrete pruritogens and other molecules associated with itch, including histamine, endothelin-1, kallikreins, substance P, NGF, µ-opioids, interleukins (IL-31) and TSLP (Lumpkin and Caterina, 2007; Wilson et al., 2013b). Up-regulation of a number of these molecules has been detected in chronic itch patients including TSLP, NGF, substance P and IL-31 (Choi et al., 2005; Jariwala et al., 2011; Johansson et al., 2002; Sonkoly et al., 2006; Toyoda et al., 2002). Release of these molecules by keratinocytes may promote nerve sprouting, or directly activate or sensitize itch-transducing C-fibers (Urashima and Mihara, 1998). In fact, mice overexpressing TSLP or IL-31 display spontaneous scratching (Dillon et al., 2004; Li et al., 2005) and mutations affecting TSLP or IL-31 signaling have been found in chronic itch patients (Arita et al., 2008). Fourth, chronic itch is often associated with abnormalities in the epidermal barrier, which is maintained by keratinocytes. The epidermal barrier is critical for preventing both entry of exogenous objects and exit of internal components. Thus, epidermal barrier disruption may facilitate entry of pruritogens and allergens in chronic itch disorders. Disruption in the epidermal barrier is measured as transepidermal water loss (TEWL). Atopic dermatitis patients experience barrier disruption in both lesional and nonlesional skin. Additionally, TEWL is associated with itch intensity in atopic dermatitis patients (Lee et al., 2006).

Page 14: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

xi

Figure 3. Multiple lines of evidence suggest that keratinocytes act in concert with somatosensory neurons to detect and transduce itch stimuli First, keratinocytes are activated by a number of pruritogens including histamine, SLIGRL and ET-1. Second, C-fibers terminate as free nerve endings in the skin, which would enable rapid signaling between keratinocytes and itch fibers. Third, keratinocytes secrete pruritogens and other itch-mediators. Finally, chronic itch is associated with disruption of barrier function, which is maintained by keratinocytes. Barrier disruption may result from increased levels of skin proteases, such as kallikreins and tryptase, which normally regulate stratum corneum turnover.

Page 15: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

xii

MECHANISMS OF CHRONIC ITCH Many forms of chronic itch are refractory to available therapies, including antihistamines. Therefore, there is intense interest in identifying the underlying molecular mechanisms of chronic itch. Recent evidence suggests that the cells and molecules that underlie acute itch are also required for chronic itch. Mouse genetic ablation studies demonstrated that MrgprA3-expressing neurons are not only required for normal histamine and histamine-independent acute itch, but also for chronic itch (Han et al., 2013). Itch evoked by dry skin and ovalbumin-induced allergic itch models was attenuated by ablation of MrgprA3-expressing neurons. While the requirement of GRPR-expressing spinal cord neurons in chronic itch has not been directly tested, several indirect lines of evidence exist. GRP is highly expressed in cutaneous nerves of atopic dermatitis patients (Kagami et al., 2013). Additionally, upregulation of GRP has been observed in mouse models of chronic itch, including allergic contact dermatitis, dry skin itch and the NC/Nga atopic dermatitis genetic line (Tominaga et al., 2009a; Zhao et al., 2013). Clinically, PAR2 and PAR2 agonists such as tryptase are upregulated in the skin of atopic dermatitis (AD) patients (Steinhoff et al., 2003). These epidermal proteases are believed to exacerbate atopic dermatitis in three ways. First, these proteases act on keratinocytes and activate the release of inflammatory mediators. Second, these proteases are believed to act on sensory neurons to trigger itch and inflammation (Yosipovitch and Papoiu, 2008). Third, overexpression of these skin proteases, which normally regulate stratum corneum turnover, reduces barrier function (Steinhoff et al., 2003). Furthermore, skin pH is elevated in atopic dermatitis and the acidic environment enhances serine protease activity, resulting in further loss of barrier function (Hachem et al., 2005; Schmid-Wendtner and Korting, 2006). Interleukins 2 and 31 have also been implicated in atopic dermatitis. IL-2 injection induces itch in both healthy and atopic dermatitis patients. Additionally, immunosuppressants, such as cyclosporine A, are thought to produce relief in atopic dermatitis patients by inhibiting IL-2 production (Ständer and Steinhoff, 2002). Multiple lines of evidence also implicate IL-31 in atopic dermatitis (Sonkoly et al., 2006). IL-31 is overexpressed in the skin of atopic dermatitis patients and overexpression of IL-31 in mice causes spontaneous scratching. Nerve growth factor (NGF) is a neuropeptide involved in nerve growth and repair. NGF has been implicated in atopic dermatitis for two reasons. First, NGF expression is correlated with disease severity in atopic dermatitis (Toyoda et al., 2002). Second, the receptor for NGF, TrkA, is highly expressed in atopic dermatitis skin (Dou et al., 2006). Indeed, histological studies revealed increased cutaneous nerve fiber density in the skin of atopic dermatitis patients, psoriasis patients and mouse models of atopic dermatitis and dry skin (Nakamura et al., 2003; Tominaga et al., 2009a; 2007; Urashima and

Page 16: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

xiii

Mihara, 1998). The increase in sensory fibers responsive to pruritogens may be responsible for itch sensitization in chronic itch conditions. Accumulating evidence demonstrates that µ- and κ-opiod receptors have opposing effects on chronic itch (Ikoma et al., 2003a). While µ-receptor agonists induce itch, κ-receptor agonists can reduce it. In fact, atopic dermatitis patients express higher levels of endogenous µ-receptor agonists and those levels correlate with disease severity (Lee et al., 2006). Lastly, substance P expression in fibers innervating the skin is upregulated in atopic eczema (Järvikallio et al., 2003) and atopic dermatitis patients have high levels of plasma substance P (Salomon and Baran, 2008; Toyoda et al., 2002). Substance P expression may be downstream of other chronic itch mediators, as PAR2 activation of primary sensory neurons induces the release of substance P and other inflammatory peptides (Hägermark, 1995). OPEN QUESTIONS IN ITCH BIOLOGY There has been significant progress in our understanding of the cellular and molecular mechanisms of itch. The sensory neuron receptors that detect a number of pruritogens and the downstream mechanisms and ion channels have been identified. Additionally, a number of neuronal populations required for itch transmission have led to some understanding of itch circuitry. However, there are many open questions in the field. Our understanding of itch and how it interacts with pain is still in its infancy. Painful stimuli can inhibit itch, while analgesic opioids can induce itch. Many molecules implicated in pain have also been implicated in itch, including NGF, TRPA1 and TRPV1. The observations that many pruritogens can cause pain, and many pain compounds can evoke itch further demonstrate the complexity of the system. Many primary afferents are polymodal, responding to both itch and pain stimuli. Labeling of itch-specific neurons will enable a better understanding of the itch circuitry. Does processing occur at the level of the primary sensory neurons or do higher order neurons integrate signals from the primary afferents to encode itch and pain? What are the neurotransmitters for itch transmission? How do itch and pain interact in the brain? Another interesting open question concerns the use of itch models. There are well-documented species differences in itch transduction. How do findings from rodents relate to humans? Additionally, acute models of itch have been powerful in elucidating itch physiology and a number of models simulate dry skin and atopic dermatitis-evoked itch. However, new chronic itch models are needed to study systemic itch. Recent work has highlighted the importance of the interaction between keratinocytes and neurons in producing itch. Keratinocytes both respond to and release pruritogens. Additionally, disruption of the skin function result in barrier loss and dry skin, which are both associated with chronic itch. Further work is required to better understand the molecular mechanisms that cause keratinocytes to release pruritogens and other neuromodulators in chronic itch conditions.

Page 17: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

xiv

My work begins to elucidate the mechanisms of itch signaling in keratinocytes. In the following 2 chapters, I will describe our progress in: 1) defining the molecular mechanisms by which the atopic dermatitis cytokine TSLP is released from keratinocytes and act on neurons to cause itch and 2) identifying the distinct mechanisms by which histamine and protease signaling in keratinocytes triggers itch. We identify the ORAI1/NFAT calcium signaling pathway as an essential regulator of TSLP release from keratinocytes, and demonstrate that secreted TSLP in the skin acts directly on a subset of TRPA1-expressing sensory neurons to trigger itch behaviors. We also show that histamine and protease signaling in keratinocytes induce distinct calcium signals that lead to divergent downstream transcriptional outputs. An important goal in the field is to elucidate the signaling pathways responsible for chronic itch, such as atopic dermatitis. A better understanding of the underlying mechanisms will lead to itch-selective therapies that improve the quality of life for chronic itch patients.

Page 18: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

1

CHAPTER I: The Epithelial Cell-Derived Atopic Dermatitis Cytokine TSLP Activates Neurons to Induce Itch. This chapter is a reproduction of the paper by the same name published in Cell October 10, 2013. My contributions were to Figures 1A, 1C, 1D, 1F, 3A, 3B, 4A, 4D, 5A, 6A-C, 6E-G, 7B, 7C and 7E-G. For this paper I performed cellular imaging, siRNA, pharmacology, PCR and ELISA experiments and made figures and wrote the manuscript with S.R.W and D.M.B.  SUMMARY Atopic dermatitis (AD) is a chronic itch and inflammatory disorder of the skin that affects one in ten people. Patients suffering from severe AD eventually progress to develop asthma and allergic rhinitis, in a process known as the “atopic march.” Signaling between epithelial cells and innate immune cells via the cytokine Thymic Stromal Lymphopoietin (TSLP) is thought to drive AD and the atopic march. Here we report that epithelial cells directly communicate to cutaneous sensory neurons via TSLP to promote itch. We identify the ORAI1/NFAT calcium signaling pathway as an essential regulator of TSLP release from keratinocytes, the primary epithelial cells of the skin. TSLP then acts directly on a subset of TRPA1-positive sensory neurons to trigger robust itch behaviors. Our results support a new model whereby calcium-dependent TSLP release by keratinocytes activates both primary afferent neurons and immune cells to promote inflammatory responses in the skin and airways. INTRODUCTION Atopic dermatitis (AD) is a chronic itch and inflammatory disorder of the skin that affects one in ten people. AD is primarily characterized by intolerable and incurable itch. Up to 70% of AD patients go on to develop asthma in a process known as the “atopic march” (He and Geha, 2010; Locksley, 2010; Spergel and Paller, 2003; Ziegler et al., 2013). Numerous studies suggest that the cytokine Thymic Stromal Lymphopoietin (TSLP) acts as a master switch that triggers both the initiation and maintenance of AD and the atopic march (Moniaga et al., 2013; Ziegler et al., 2013). TSLP is highly expressed in human cutaneous epithelial cells in AD, and bronchial epithelial cells in asthma (Jariwala et al., 2011). Over-expression of TSLP in keratinocytes, the most prevalent cell type in the skin, triggers robust itch-evoked scratching, the development of an AD-like skin phenotype and ultimately asthma-like lung inflammation in mice (Li et al., 2005; Ying et al., 2005; Ziegler et al., 2013). However, the mechanisms by which TSLP triggers itch and AD remain enigmatic. Itch is mediated by primary afferent somatosensory neurons that have cell bodies in the dorsal root ganglia (DRG) that innervate the skin and are activated by endogenous pruritogens to drive itch behaviors (Ikoma et al., 2006; McCoy et al., 2012; Ross, 2011). Hallmarks of AD skin include robust itch sensations, increased neuronal activity and hyper-innervation (Ikoma et al., 2003b; Tobin et al., 1992; Tominaga et al., 2009b). While many studies have shown that epithelial cell-derived TSLP activates T cells,

Page 19: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

2

dendritic cells and mast cells (Ziegler et al., 2013), the role of sensory neurons in this pathway has not been studied. How does TSLP lead to sensory neuron activation to promote itch? In vitro studies suggest that keratinocytes may directly communicate with sensory neurons via neuromodulators (Ikoma et al., 2006). Indeed, many of the factors that keratinocytes secrete act on both immune cells and primary afferent sensory neurons (Andoh et al., 2001; Fitzsimons et al., 2001; Kanda et al., 2005; Ziegler et al., 2013). Thus, TSLP may evoke itch behaviors directly, by activating sensory neurons, indirectly, by activating immune cells that secrete inflammatory mediators that target sensory neurons, or both. While TSLP’s action on immune cells is well characterized, its effects on sensory neurons, and the contribution of sensory neurons to TSLP-evoked atopic disease, have not been studied. Furthermore, the mechanisms regulating TSLP release by keratinocytes are unknown. The GPCR Protease-Activated Receptor 2 (PAR2) plays a key role in keratinocyte TSLP production. Studies have shown a correlation between PAR2 activity and TSLP expression in the skin of AD patients and in mouse models of atopic disease (Briot et al., 2009; 2010; Hovnanian, 2013). In addition, PAR2 activation triggers robust TSLP expression in keratinocytes (Kouzaki et al., 2009; Moniaga et al., 2013). While there is a strong correlation between PAR2 activity and TSLP levels in the skin, virtually nothing is known about the molecular mechanisms by which PAR2 leads to TSLP expression. Here we sought to elucidate the mechanisms that regulate TSLP secretion and that promote TSLP-evoked itch. Our findings show that keratinocyte-derived TSLP activates sensory neurons directly to evoke itch behaviors. We define a new subset of sensory neurons that require both functional TSLP receptors and the ion channel, TRPA1, to promote TSLP-evoked itch behaviors, and we identify the ORAI1/NFAT signaling pathway as a key regulator of PAR2-mediated TSLP secretion by epithelial cells. RESULTS TSLP evokes robust itch behaviors in mice To identify proteins that mediate itch transduction in somatosensory neurons, we looked for biomarkers of AD (Lee and Yu, 2011) in the mouse DRG transcriptome (Gerhold et al., 2013). We were surprised to find expression of the TSLP Receptor (TSLPR) in mouse sensory ganglia. While studies have shown that TSLP acts on various immune cells, TSLP signaling in the nervous system has not been reported. TSLPR is a heterodimer, composed of the IL7 receptor alpha (IL7Ra) chain and a TSLP-specific receptor chain (TSLPR; also Crlf2; (Pandey et al., 2000). Consistent with the presence of TSLPRs in sensory neurons, we detected both TSLPR and IL7Ra transcripts in mouse and human DRG using RT-PCR (Figure 1A).

Page 20: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

3

Figure 1. TSLP triggers robust itch behaviors in mice by activating sensory neurons (A) PCR analysis of TSLPR and IL7Ra in mouse (left) and human (right) dorsal root ganglia (DRG). No product was amplified from the “no RT” control. (B) Image of itch-evoked scratching following intradermal injection of TSLP (2.5 µg/20 µl) into the cheek. (C) Quantification of scratching following TSLP injection in the cheek. TSLP (black) induced robust scratching compared to vehicle (white). n≥18 per group. (D) Itch behavior in RAG+/+, RAG-/-, NOD, and NOD/SCID mice following vehicle (PBS) or TSLP cheek injection. n≥8 per group. (E) Itch behavior in cKIT+/+ and cKIT-/- mice following vehicle (PBS) or TSLP injection. n≥8 per group. (F) TSLP-evoked scratching following neuronal ablation by RTX (red) versus control (black). n≥6 per group. *P<0.05; **P<0.01; ***P<0.001. Error bars represent s.e.m.

Page 21: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

4

Somatosensory neurons mediate itch, touch and pain. Thus, we asked if TSLP injection triggers itch and/or pain behaviors by using a mouse cheek model of itch, which permits easy distinction between these behaviors (Shimada and Lamotte, 2008). Injection of TSLP into the cheek of wild type mice evoked robust scratching that was not observed following vehicle injection (Figure 1B-C). Wiping was never observed, indicating that TSLP triggers itch, rather than pain (Shimada and Lamotte, 2008). Intradermal injection of TSLP has been previously shown to evoke inflammation of the skin and lung over the course of hours or days (Jessup et al., 2008). However, we observed robust itch behaviors within 5 minutes of TSLP injection (latency to scratch = 4.1 ± 0.3 min). While immune cells play a key role in long-term TSLP-evoked inflammation, whether immune cells are required for acute TSLP-triggered itch behaviors is unknown. The current model posits that TSLP acts on various immune cells to promote TH2 cell differentiation and inflammation. We thus compared TSLP-evoked itch behaviors of wild type mice to mouse strains lacking either T and B cells (RAG1-/-, NOD SCID) or mast cells (Kit(W-sh), Figure 1D-E). TSLP triggered robust itch behaviors in all strains, with no significant differences between transgenic and congenic wild type littermates. Together, these data indicate that acute TSLP-evoked itch does not specifically require lymphocytes or mast cells, nor does it require the cytokines or other products produced when these cells are activated, and suggest that TSLP may act directly on sensory neurons. Previous studies have shown that intradermal injection of the TRPV1 agonist, resiniferatoxin (RTX), results in ablation of primary afferent sensory neurons that express TRPV1, or TRPV1 and TRPA1, and consequently eliminates pain and itch behaviors (Imamachi et al., 2009; Mitchell et al., 2010). TSLP-evoked scratching was significantly decreased in RTX-treated mice as compared to control mice (Figure 1F). These findings show for the first time that the AD cytokine, TSLP, induces itch via sensory neurons. TSLP directly activates an uncharacterized subset of sensory neurons We next asked whether TSLPRs are expressed in sensory neurons. DRG neurons are a heterogeneous population of cells, including a subset of small-diameter, peripherin-positive neurons that transmit itch and pain signals to the CNS, and release inflammatory mediators in the skin and other target organs (Basbaum et al., 2009). We thus examined the prevalence of TSLPR-positive neurons and co-localization with known neuronal markers. In situ hybridization revealed that TSLPR and IL7Ra were expressed in a subset of small diameter DRG neurons (Figure 2A). Using antibodies against TSLPR, we observed TSLPR protein expression in 5.9% of cells in DRG sections (Figure 2B). Co-staining of TSLPR and peripherin, a marker of small-diameter DRG neurons, demonstrated that all TSLPR-positive neurons are also peripherin-positive, with an average diameter of 18.1±0.6µm (Figure 2B). Overall, the characteristics of TSLPR-positive neurons match those of sensory neurons that mediate itch and/or pain (McCoy et al., 2013).

Page 22: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

5

Figure 2. TSLP receptor components are expressed in sensory neurons (A) DIC overlay images of in situ hybridization with cDNA probes detecting TSLPR, IL7Ra and TRPV1 in mouse DRG. Scale bar = 400µm. (B) Immunostaining of DRG sections with antibodies against peripherin and TSLPR in DRG sections. White arrows (right) mark peripherin- and TSLPR-positive neurons. Scale bar = 400µm. n≥4 mice/condition. (C) Immunostaining of PGP 9.5 and TSLPR in glabrous hind paw skin. The white arrows (right) mark PGP 9.5- and TSLPR-positive neurons. Scale bar = 200µm. n≥3 mice per condition.

Page 23: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

6

If TSLPRs mediate somatosensory transduction, they should localize to primary afferent nerve terminals in the skin. We thus performed immunohistochemistry with antibodies against TSLPR and the pan-neuronal fiber marker PGP9.5 on mouse skin (Figure 2C). We observed TSLPR staining in 9% of PGP9.5-positive free nerve endings in the skin (Figure 2C). These data show that TSLPRs are localized to sensory neuronal endings that innervate the skin in close apposition to keratinocytes in the epidermis. Taken together, these data demonstrate that the TSLPR subunits are expressed in a subset of sensory neurons that innervate the skin and mediate itch and/or pain transduction. To test whether TSLPR is functional in sensory neurons, we used ratiometric Ca2+ imaging (Figures 3A-B). We found that 4.1 ± 0.6% of DRG neurons showed robust increases in intracellular Ca2+ following TSLP application (Figure 3E); this is similar to the percentage of neurons that respond to other endogenous pruritogens, like BAM8-22 (Liu et al., 2009; Wilson et al., 2011). Previous studies have shown that small diameter sensory neurons transduce itch and/or pain signals via the ion channels TRPA1 and TRPV1 (Basbaum et al., 2009; Ross, 2011). Indeed, subsequent exposure to the TRPA1 agonist, allyl isothiocyanate (AITC), or the TRPV1 agonist, capsaicin (CAP), further increased Ca2+ levels in all TSLP-positive cells (Figures 3A-B). Similarly, TSLP triggered action potential firing in a subset of CAP-sensitive neurons (Figure 3C). These data suggest that TSLP activates a subset of TRPV1- and TRPA1-positive sensory neurons. The itch compounds histamine, chloroquine (CQ) and BAM8-22 have been shown to activate 5-20% of sensory neurons (Ikoma et al., 2006; Imamachi et al., 2009; Liu et al., 2009; Wilson et al., 2011) that express TRPA1 and/or TRPV1. TSLP appears to activate an undescribed subset of itch neurons, as most TSLP-positive neurons were insensitive to other itch compounds (Figure 3A, B, D). TSLPR and TRPA1 mediate TSLP-evoked neuronal activation To ask whether TSLPRs mediate TSLP-evoked neuronal activation, we examined TSLP-evoked Ca2+ signals in neurons isolated from IL7Ra-deficient mice. TSLP-, but not AITC- or CAP-evoked Ca2+ signaling, was abolished in IL7a-deficient neurons (Figure 3E). These results are consistent with previous studies in immune cells showing that functional IL7Ra is required for TSLP signaling (Pandey et al., 2000). Here we show that functional TSLPRs are required for TSLP-evoked neuronal activation. TRPV1 and TRPA1 channels are required for acute itch signaling and behavior (Ross, 2011). We thus asked whether these channels are required for TSLP-evoked neuronal activation. TRPV1 and TRPA1 inhibition by the nonselective inhibitor, ruthenium red, significantly decreased neuronal sensitivity to TSLP (Figure 3E). We also compared neurons isolated from TRPA1- and TRPV1-deficient mice to those from wild type littermates. TSLP-evoked Ca2+ signals were significantly attenuated in TRPA1-deficient neurons, but not TRPV1-deficient neurons (Figure 3E). Our results show that TRPA1 channels mediate TSLP-evoked neuronal excitability.

Page 24: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

7

Figure 3. TSLP directly activates a subset of sensory neurons (A) Representative images of Fura-2 loaded DRG neurons treated with vehicle, TSLP (2 ng/mL), histamine (HIS, 1mM), AITC (200µM) and capsaicin (CAP, 1µM). (B) Representative trace shows a neuron that responds to TSLP, AITC and CAP, but not HIS. (C) Current-clamp recording showing TSLP- and CAP-evoked action potential firing in a DRG neuron. n≥60 cells. (D) A small percentage of the TSLP-sensitive population overlaps with the population of histamine- (HIS, 6%) or chloroquine-sensitive neurons (CQ, 6%), but not the BAM8-22 population (BAM, 0%). (E) Left: Prevalence of TSLP sensitivity in wild-type neurons (black), IL7Ra-deficient (grey) neurons, neurons treated with 20µM ruthenium red (RR; red), TRPA1-deficient neurons (blue) and TRPV1-deficient neurons (white). Right: prevalence of AITC and CAP sensitivity in wild-type (black) and IL7Ra-deficient (grey) neurons n≥1000 cells. (F) Prevalence of TSLP sensitivity in neurons pre-treated with vehicle (black), a PLC blocker, U73122 (red) and the Gbg blocker, gallein (grey) n≥600 cells. (G) Representative response to TSLP in the absence (0mM Ca2+) and presence (2mM Ca2+) of extracellular Ca2+ n≥200 cells. *P<0.05; **P<0.01; ***P<0.001. Error bars represent s.e.m.

Page 25: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

8

We next examined the mechanisms by which TSLPR activation promotes TRPA1 activity. Two signaling pathways have linked itch receptors to TRPA1 activation: Phospholipase C (PLC) signaling couples MrgprC11 to TRPA1; and, Gbg signaling couples MrgprA3 to TRPA1 (Wilson et al., 2011). Treatment of cells with the PLC inhibitor, U73122, significantly reduced the prevalence of TSLP-sensitive neurons (Fig. 3F). In contrast, gallein, a Gbg inhibitor, had no effect on TSLP-evoked Ca2+ signals (Fig. 3F). Consistent with TSLP activation of the PLC pathway, TSLP triggers both release of Ca2+ from intracellular stores, and subsequent Ca2+ influx in sensory neurons (Figure 3G). Overall, these experiments suggest that TSLPR and TRPA1 communicate via PLC signaling. TSLPR and TRPA1 mediate TSLP-evoked itch To test whether TSLP and TRPA1 receptors are required for TSLP-evoked itch behaviors, we used the cheek model of itch. TSLP-evoked scratching was significantly attenuated in IL7Ra-deficient mice, supporting a role for TSLPRs in TSLP itch signaling (Figure 4A). These mice were not generally deficient in itch behaviors, as CQ-evoked scratching, which occurs via MrgprA3 (Liu et al., 2009), was normal (Figure 4B). These data demonstrate that TSLP targets TSLPRs to trigger itch behaviors in vivo. We next asked whether TSLP-evoked itch behaviors require TRP channels. TSLP-evoked scratching was abolished in TRPA1-deficient mice, but normal in TRPV1-deficient mice (Figure 4D). These experiments show that both functional TSLPRs and TRPA1 channels are required for TSLP-evoked itch. PLC signaling is also required for the functional coupling between TSLPR and TRPA1 in vivo, as TSLP-evoked scratching was significantly attenuated by intradermal injection of U73122. Such treatment selectively silenced TSLP-evoked behaviors, as these mice displayed normal CQ-evoked scratching, which is PLC-independent (Wilson et al., 2011). Overall, these data demonstrate a new role for TSLP as a pruritogen and a robust activator of sensory neurons, and suggest that these neurons may contribute to the initiation of TSLP-evoked inflammatory responses in the skin in AD, and airways in asthma. Keratinocyte release of TSLP is Ca2+-dependent Our data establish a new cellular target for TSLP, supporting a model whereby both immune cells and sensory neurons are activated by keratinocyte-derived TSLP to drive itch and AD. What are the upstream mechanisms that govern the expression and release of TSLP by keratinocytes? Protease signaling via PAR2 plays a key role in TSLP production and AD. PAR2 activity, and levels of the endogenous PAR2 agonist, tryptase, are increased in the skin of AD patients (Steinhoff et al., 2003). Consistent with a previous study (Ui et al., 2006), injection of tryptase induced robust itch behaviors in mice (Figure 5A). Tryptase-evoked itch was significantly attenuated in both PAR2- and IL7Ra-deficient mice (Figure 5A), consistent with a pathway where PAR2 signaling promotes the release of TSLP from keratinocytes, which then acts on TSLPR-positive neurons to drive itch behaviors. We next sought to determine the signaling pathways that control PAR2-induced TSLP expression in keratinocytes.

Page 26: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

9

Figure 4. TSLP induces robust TSLPR- and TRPA1-dependent itch behaviors (A) Itch behaviors following intradermal cheek injection of vehicle (10µL PBS, white) or TSLP (2.5µg/10 µL) into wild type (WT; black) or IL7Ra-deficient (red) mice. (B) Scratching in WT (black) and IL7Ra-deficient (red) mice following chloroquine (CQ) injection in the cheek. (C) Scratching in WT (black), TRPA1-deficient (red) and TRPV1-deficient (white) mice following TSLP injection (2.5 µg/10 µL). (D) Attenuation of TSLP-evoked scratching by 30 min preinjection with the PLC blocker, U73122 (U7) compared to vehicle (VEH). (E) CQ-evoked scratching in mice preinjected with U73122 or vehicle. The time spent scratching was quantified for 20 min after injection. n≥7 mice/condition. **P<0.01; ***P<0.001. Error bars represent s.e.m.

Page 27: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

10

Figure 5. PAR2 activation promotes itch behaviors and Ca2+-dependent release of TSLP (A) Itch-evoked scratching following injection of tryptase into the cheek (100 pg/20 µL) of wild type (WT; black), PAR2-deficient (blue) or IL7Ra-deficient mice (red), or PBS (white, 20µL) injection into WT mice, n≥8 mice per condition. The time spent scratching was quantified for 1 h after injection. (B) TSLP secretion evoked by 24 h treatment with vehicle (VEH), tryptase (TRY, 100nM), tryptase in the absence of extracellular Ca2+ (TRY 0Ca), SLIGRL (100µM), SLIGRL in the absence of extracellular Ca2+ (SLIGRL 0Ca), or TG (1µM). n≥4 replicates/condition *P<0.05; **P<0.01;, ***P<0.001. Error bars represent s.e.m.

Page 28: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

11

Studies on keratinocytes have shown that the endogenous PAR2 agonist, tryptase, and the widely used PAR2 ligand mimetic, Ser-Leu-Ile-Gly-Arg-Leu (SLIGRL), elicits Ca2+ influx (Schechter et al., 1998; Zhu et al., 2009) and triggers the Ca2+-dependent release of inflammatory mediators (Halfter et al., 2005; Santulli et al., 1995; Schechter et al., 1998). For example, SLIGRL triggers a rise in intracellular Ca2+ in keratinocytes (Zhu et al., 2009) and also promotes TSLP expression (Moniaga et al., 2013). We thus asked if PAR2-evoked TSLP expression is Ca2+-dependent. ELISA measurements revealed that treatment of keratinocytes with tryptase or SLIGRL, but not vehicle, triggered the robust secretion of TSLP (Figure 5B). These data show that PAR2 stimulation of keratinocytes triggers TSLP release. TSLP secretion was highly dependent on Ca2+. First, TSLP secretion was not observed in keratinocytes treated with tryptase or SLIGRL in the absence of external Ca2+ (Figure 5B). In addition, treatment with the drug thapsigargin (TG), which promotes depletion of intracellular Ca2+ stores and subsequent Ca2+ influx, caused a significant increase in TSLP secretion (Figure 5B). These data demonstrate that Ca2+ is required and sufficient to drive TSLP secretion. A recent study has shown that some PAR2 agonists, including SLIGRL, also activate the sensory neuron-specific itch receptor, MrgprC11 (MrgprX1 in human, (Liu et al., 2011). However, this result does not impact our in vitro studies for several reasons. First, keratinocytes do not express MrgprX1 (Supplementary Figure 1A). Second, keratinocytes are insensitive to the MrgprX1-specific ligand, BAM8-22 (Supplementary Figure 1B). Third, tryptase-evoked itch is dependent on PAR2 (Figure 5A). Finally, tryptase does not activate MrgprC11 in mice (Supplementary Figure 1C-D). Overall, our findings support a model where tryptase- and SLIGRL treatment of keratinocytes promotes PAR2-evoked Ca2+ signaling and subsequent secretion of TSLP. ORAI1 and STIM1 are required for PAR2-evoked Ca2+ influx We next used ratiometric Ca2+ imaging to dissect the mechanisms underlying PAR2-evoked Ca2+ signals. Consistent with previous studies, tryptase and SLIGRL evoked a rise in intracellular Ca2+ in keratinocytes (Figure 6A-C; (Zhu et al., 2009). In some cells, PAR2 signals via PLC (Dai et al., 2007), and PLC activation leads to Ca2+-release from IP3-dependent stores and influx via the store-operated Ca2+ entry (SOCE) pathway. Indeed, PAR2 activation in keratinocytes induced both Ca2+ release from intracellular stores and Ca2+ influx, consistent with activation of SOCE (Figure 6A-B).

Page 29: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

12

Supplementary Figure 1. Human keratinocytes do not express human MRGPRX11 and the PAR2 agonist, tryptase, does not activate mouse MrgprC11, Related to Figure 6 (A) PCR analysis of the human BAM8-22 (BAM) receptor, MrgprX1, in human dorsal root ganglia (DRG) and human keratinocytes (KRT). MrgprX1 was amplified from DRG, but not keratinocytes. MrgprX1 and GAPDH were amplified from RT-treated tissue but not from “no RT” controls. (B) Representative response to BAM8-22 (BAM, 2 µM) in human keratinocytes. (C) Representative response to tryptase (TRY, 3 µM) and BAM8-22 (BAM, 2 µM) in the presence or absence of the mouse BAM8-22 receptor, MrgprC11. (D) Left: representative traces showing a neuron that is sensitive to BAM8-22 (BAM) but not tryptase (TRY, blue), and a neuron that is sensitive to tryptase but not BAM8-22 (black). Right: quantification of the prevalence of tryptase-responsive (TRY, black), BAM8-22-responsive (blue, BAM), and tryptase- and BAM8-22-responsive neurons in mouse dorsal root ganglia. n ≥ 500 cells. Data are represented as mean +/- SEM.

201510

50Pe

rcen

t neu

rons

4

3

2

1

Ratio

(F34

0/F3

80)

1251007550250Time (s)

3

2

1

Ratio

(F34

0/F3

80)

150100500Time (s)

Transfected HEK293T cells

DRG neurons

3

2

1

Ratio

(F34

0/F3

80)

16012080400Time (s)

Keratinocytes

TRY BAM TRY/BAM

TRY BAM

TRY BAM

MrgprC11+

MrgprC11-

A

D

C

Supplementary Figure 1

MrgprX1

GAPDH

DRG KRT No RT

140 K+

BAM IONOB

Page 30: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

13

Figure 6. ORAI1 and STIM1 are required for PAR2- and TG-evoked Ca2+ influx (A) Representative response to SLIGRL (100µM) following pretreatment with vehicle (black) or 2-aminoethoxydiphenyl borate (50 µM 2-APB; red). (B) Representative response to tryptase (100nM) following pretreatment with vehicle (black) or 2-APB (red). (C) Average steady state Ca2+ level following SLIGRL- or tryptase (TRY)-evoked Ca2+ influx (2 mM Ca2+), in the presence of 2-APB (red), lanthanum (50nM La3+, blue), or vehicle (CTRL, black). n≥1000 cells. (D) Representative current-voltage trace in the presence of SLIGRL (100µM) in perforated-patch, whole-cell voltage-clamp recordings. Representative baseline subtracted currents before (red) and during application of SLIGRL (black). n≥3 cells/condition. (E) siRNA-induced silencing of STIM1 (red), ORAI1 (blue), and ORAI2 (grey) mRNA in keratinocytes. Expression was normalized to scrambled-siRNA control (black). n≥1000 cells. (F) Representative traces of SLIGRL-evoked (100µM) Ca2+ signals following treatment with siRNA targeting STIM1 (red) or scrambled control (CTRL, black). (G) Average steady state Ca2+ concentration after treatment with SLIGRL (100µM) or TG (1µM) in cells treated with scrambled siRNA (black), STIM1 (red), ORAI1 (blue), or ORAI2 (grey) siRNA. n≥500 cells. *P<0.05; **P<0.01, ***P<0.001. Error bars represent s.e.m.

Page 31: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

14

What are the molecules mediating PAR2-evoked SOCE in keratinocytes? Both ORAI and TRPC channels have been implicated in SOCE (Cahalan, 2009; Ramsey et al., 2006). We next asked whether PAR2 activates SOCE via ORAI or TRPC channels, which can be distinguished by their distinct pharmacological profiles (DeHaven et al., 2008; Lis et al., 2007; Zhang et al., 2008). The drugs 2-Aminoethoxydiphenyl borate (2-APB) and lanthanum (La3+) inhibit ORAI1 and ORAI2 channels, but not ORAI3 or TRPC channels (DeHaven et al., 2008; Lis et al., 2007; Zhang et al., 2008). Tryptase and SLIGRL-evoked Ca2+ influx was significantly attenuated by treatment with 2-APB or La3+. These data show that tryptase and SLIGRL activate the same SOCE pathway and support a role for ORAI channels in PAR2-evoked SOCE (Figure 6C). ORAI and TRPC channels can also be distinguished by their distinct biophysical characteristics: ORAI1 and ORAI2 are Ca2+-selective channels that are inwardly-rectifying, while TRPC channels are outwardly-rectifying, non-selective channels (Cahalan, 2009; Owsianik et al., 2006; Yeromin et al., 2006). Thus, we measured SLIGRL-evoked currents using perforated-patch, voltage-clamp recordings on keratinocytes. Treatment with SLIGRL triggered an ORAI1/2-like current; the currents were dependent on extracellular Ca2+, displayed an inwardly-rectifying current-voltage relationship, and displayed no measurable reversal potentials below +80 mV (Figure 6D). These results implicate ORAI1 and/or ORAI2 in PAR2-evoked SOCE. qPCR demonstrated that keratinocytes express ORAI1, ORAI2 and the ORAI regulator, Stromal Interaction Molecule 1 (STIM1). We thus examined the role of ORAI1, ORAI2, and STIM1 in SOCE using siRNA-mediated knockdown. Depletion of ORAI1 transcripts by 71% or STIM1 transcripts by 84% significantly diminished Ca2+ entry in response to SLIGRL as compared to scrambled control siRNA (Figure 6E-G). ORAI1 and STIM1 knockdown also significantly attenuated tryptase-evoked Ca2+ signals (not shown). In contrast, depletion of ORAI2 transcripts by 86% had no effect on SLIGRL-evoked SOCE (Figure 6E, 6G). These data demonstrate that ORAI1 and STIM1 are required for PAR2-evoked SOCE in human keratinocytes. ORAI1 and STIM1 knockdown also attenuated TG-evoked SOCE (Figure 6G), suggesting that ORAI1 is the primary store-operated Ca2+ pathway in keratinocytes. PAR2-activation induces Ca2+-dependent NFAT translocation and TSLP secretion In immune cells, ORAI1 signaling activates NFAT, which triggers cytokine expression and secretion (Feske et al., 2006a; Gwack et al., 2006). The ORAI1/NFAT pathway may play a similar role in keratinocytes, promoting the expression and secretion of TSLP. Consistent with a regulatory role for NFAT in TSLP expression, two NFAT binding motifs (GGAAAATN) (Rao et al., 1997; Zhu et al., 2009) are present in the 5′-upstream regulatory region of the human TSLP gene. These findings imply that PAR2 may trigger NFAT-dependent expression and release of TSLP; however, the evidence is merely correlative. To directly test this hypothesis, we measured PAR2-dependent NFAT translocation and TSLP expression and release in keratinocytes.

Page 32: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

15

Figure 7. PAR2 activation promotes Ca2+-dependent NFAT translocation and TSLP secretion (A) Representative images displaying cytosolic and nuclear localization of NFAT (green) and DAPI (red) in keratinocytes after a 30 min incubation with vehicle (VEH), SLIGRL (100µM), SLIGRL + 2APB (50µM) or SLIGRL + CsA (1µM). Pretreatment with 2APB or CsA prevented SLIGRL-induced NFAT nuclear translocation. n≥300 cells. (B) Fraction of HaCaT keratinocytes displaying nuclear localization of NFAT-GFP following treatment with SLIGRL (100µM; black), SLIGRL and 2APB (50 µM; red), SLIGRL + CsA (1µM; blue) or vehicle (VEH; white). n≥1000 cells. (C) TSLP expression in human keratinocytes following a 3h treatment with vehicle (VEH, black) or SLIGRL (100µM, red). n≥3. (D) SLIGRL-evoked TSLP release in cells treated with scrambled (black), STIM1 (red) or ORAI1 siRNA (blue). Secretion was normalized to vehicle-treated cells (white). n≥3. (E) TSLP release in response to treatment with vehicle (VEH, black), SLIGRL (100 µM, red) or SLIGRL + CsA (1µM, blue). (F) Western blot of skin lysates from mice following intradermal injection with vehicle (VEH), SLIGRL, or SLIGRL+CsA. Samples were probed with antibodies against TSLP and calnexin (loading control). n≥3 mice. (G) Western blot of skin lysates isolated from mice following intradermal injection with vehicle (VEH), tryptase (TRY; 100pg/20µL), or tryptase+CsA (TRY + CsA). Samples were probed with antibodies against TSLP, and actin (loading control). n≥3 mice. *P<0.05; **P<0.01, ***P<0.001. Error bars represent s.e.m. (H) Schematic diagram depicting the ORAI1 signaling pathway in keratinocytes that links PAR2 to TSLP secretion and activation of itch neurons. Activation of PAR2 triggers release of Ca2+ from the ER and activation of STIM1, which opens ORAI1 channels to promote Ca2+ influx. Ca2+ activates the phosphatase calcineurin, which dephosphorylates NFAT and causes nuclear translocation, thus inducing transcription of TSLP. Secreted TSLP depolarizes a subset of C-fibers to evoke itch, in a TSLPR- and TRPA1-dependent manner. Activation of TRPA1-expressing sensory neurons can then lead to release of neuropeptides in the skin in a process known as neurogenic inflammation.

Page 33: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

16

Following a rise in Ca2+, NFAT is dephosphorylated by the Ca2+-dependent phosphatase calcineurin and translocates from the cytosol to the nucleus to promote transcription of target genes (Rao et al., 1997). Immunostaining demonstrated that treatment of keratinocytes with SLIGRL for 30 minutes induced robust NFAT translocation to the nucleus (Figure 7A). This translocation was attenuated by blocking ORAI channels with 2-APB, or by inhibiting NFAT activity with cyclosporine A (CsA), an inhibitor of calcineurin (Figure 7A); similar results were observed using live cell imaging of a human keratinocyte cell line, HaCat, that expressed NFAT-GFP (Figure 7B). These results show that PAR2 activation induces Ca2+-dependent NFAT translocation, which may lead to NFAT-dependent changes in gene expression. In support of this model, PAR2-evoked SOCE robustly increased expression of TSLP transcripts in keratinocytes (Figure 7C). We next addressed whether ORAI1/NFAT signaling mediates PAR2-evoked TSLP release. We found that siRNA-mediated knockdown of ORAI1 or STIM1 significantly attenuated SLIGRL-evoked TSLP release by keratinocytes, suggesting that ORAI1 is required for PAR2-evoked TSLP secretion (Figure 7D). Likewise, inhibition of NFAT-mediated transcription with CsA also attenuated TSLP release (Figure 7E), but had no effect on SOCE-evoked Ca2+ signals (not shown). In addition to cutaneous epithelial cells, airway epithelial cells of patients with allergic rhinitis, AD and asthma also display high TSLP expression (Ziegler et al., 2013). Previous studies have shown that TG induces ORAI1-dependent Ca2+ signals in human airway epithelial cells (Gusarova et al., 2011). Interestingly, we found that, like keratinocytes, SOCE triggers robust TSLP expression in human airway epithelial cells, which can be blocked by CsA (not shown). These data identify ORAI1-dependent NFAT activation as a regulator of TSLP expression and release in both cutaneous and airway epithelial cells. We next tested the hypothesis that NFAT promotes TSLP expression in vivo. Mice were treated with SLIGRL, SLIGRL and CSA, or vehicle via intradermal injection into the back. TSLP protein levels in treated skin were measured three hours after injection. Co-injection of CsA significantly attenuated SLIGRL-evoked TSLP protein expression in skin (Figure 7F). Similar results were also observed with the endogenous PAR2 agonist, tryptase (Figure 7G), demonstrating that PAR2 triggers TSLP expression via the Ca2+-calmodulin/NFAT pathway in vivo. DISCUSSION TSLP plays a key role in the triad of atopic diseases: asthma, allergic rhinitis and atopic dermatitis (Ziegler et al., 2013). Recent studies have also implicated TSLP in a number of disorders, including cancer, gastrointestinal diseases, and autoimmunity (Ziegler et al., 2013). As such, there is much interest in understanding the mechanisms of TSLP expression and downstream effects of TSLP secretion. Here we present molecular, cellular and behavioral data showing that ORAI1/NFAT signaling regulates TSLP release by keratinocytes, and that TRPA1 is required for TSLP-evoked activation of sensory neurons and subsequent itch behaviors. Our data support a new model

Page 34: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

17

whereby TSLP released from keratinocytes acts directly on sensory neurons to trigger robust itch-evoked scratching (Figure 7H). Sensory neurons mediate TSLP-evoked itch Studies on the role of TSLP in promoting atopic disease have focused solely on its effects on immune cells. A variety of immune cells are activated by TSLP, including dendritic cells, T cells, B cells, natural killer cells, mast cells, basophils and eosinophils, which together promote allergic inflammation (Ziegler et al., 2013). The inflammatory cytokines produced by these immune cells can activate sensory neurons (Cevikbas et al., 2007). TSLP expression in keratinocytes leads to robust scratching in mice, which was previously assumed to occur solely downstream of immune cell cytokine release (Bogiatzi et al., 2012; Yoo et al., 2005). The current model is that sensory neurons are activated downstream of TSLP-activated immune cells to induce itch. Our data support the direct activation of sensory neurons by TSLP. First, we show that mast cell release of histamine, or other pruritogens, is not required for TSLP-evoked itch behaviors. In addition, histamine-dependent itch requires TRPV1 (Imamachi et al., 2009), and our data show that TRPV1-deficient mice display normal TSLP-evoked itch behaviors. Finally, we show that acute TSLP-evoked itch does not require lymphocytes. These results were surprising given the well-known role of immune cells in TSLP-evoked atopic disease. However, until now, studies have focused on the long-term, rather than the acute effects of TSLP. These data suggest that the acute versus chronic phases of TSLP-evoked inflammation may be mediated by distinct mechanisms. In addition, because activation of primary afferent neurons triggers the release of inflammatory agents that promote immune cell chemoattraction and activation (Basbaum et al., 2009), neuron-to-immune cell communication may also play a key role in the development of AD. Thus far, all published studies have focused on global knockouts of TSLPR. Future studies using tissue specific TSLPR knockout mice are required to determine the relative contributions of sensory neurons and immune cells to both the acute and chronic phases of AD. TRPA1 is required for TSLP-evoked itch TSLP activates a subset of sensory neurons that express TSLPRs and the irritant receptor TRPA1. TRPA1-positive sensory neurons are required for the transmission of itch and pain stimuli to the CNS (Basbaum et al., 2009; Ross, 2011). Recent studies have shown that TRPA1 is also required for dry skin- and allergen-evoked chronic itch (Liu et al., 2013; Wilson et al., 2013a), but the endogenous signaling molecules that promote TRPA1 activation in these itch models are unknown. We now show that the endogenous AD cytokine, TSLP, leads to TRPA1 activation, downstream of TSLPR. Inhibition of PLC significantly attenuates such coupling both in vitro and in vivo. Despite the extensive literature on TSLP in immune cells, little is known about the signaling pathways activated downstream of TSLPR. The JAK/STAT pathway has been implicated in TSLP signaling but thus far, neither TSLPR nor IL7R have been linked to PLC (Ziegler et al., 2013).

Page 35: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

18

TRPA1 afferents are not restricted to the skin, but also densely innervate the airways and gastrointestinal tract (Bautista et al., 2013). Indeed, TRPA1 activation promotes lung inflammation in mouse models of airway inflammation and asthma and triggers inflammation in mouse models of inflammatory bowel disease (Bautista et al., 2013). Interestingly, we found that like keratinocytes, Ca2+ signaling through ORAI1 triggers robust TSLP expression in human airway epithelial cells (data not shown). Thus, crosstalk between sensory neurons and epithelial cells via TSLP and TRPA1 may not be restricted to the skin, but may also occur in the airways and gut. The “atopic march” has been largely attributed to the actions of epithelial and immune cells (Holgate, 2007). Future studies using tissue specific TSLPR-deficient animals are required to resolve the contributions of neuronal and immune cell TSLP signaling to atopic disease. Nonetheless, our findings highlight a potential new role for TRPA1 and sensory neurons in promoting the atopic march. ORAI1/NFAT regulates TSLP release in keratinocytes ORAI1 was first identified as the channel that mediates store-operated Ca2+ influx required for NFAT-dependent cytokine expression during immune cell activation; loss of function mutations in ORAI1 and STIM1 lead to severe combined immunodeficiencies in patients (Feske, 2010; Feske et al., 2006b; Prakriya et al., 2006; Yeromin et al., 2006). Our work shows that in addition to lymphocytes, epithelial cells also utilize ORAI1-mediated Ca2+ influx to regulate cytokine expression and release, suggesting that ORAI1 plays a more general role in the pathogenesis of inflammatory disease. Thus, ORAI1 may represent a new therapeutic target for atopic disease. A role for the ORAI1/NFAT pathway in AD is consistent with a number of disparate clinical findings. First, SNPs in the ORAI1 gene have been linked to susceptibility to atopic disease in humans (Chang et al., 2012) but the role of ORAI1 in AD had not been studied. Second, NFAT displays an abnormally high degree of nuclear localization in the keratinocytes of chronic itch patients (Al-Daraji et al., 2002), but the consequences of NFAT activation on AD was unknown. Finally, CsA, an inhibitor of NFAT-mediated transcription, is a potent immunosuppressant drug and is often prescribed for itchy inflammatory skin diseases, such as psoriasis and AD (Madan and Griffiths, 2007). While its effects have been mainly attributed to immune cell inhibition, our work suggests that the effectiveness of CsA in treating chronic itch may, in part, be due to its effects on keratinocyte-mediated TSLP release. EXPERIMENTAL PROCEDURES Cell culture Primary human epidermal keratinocytes (PromoCell) and HaCaT cells were cultured in PromoCell Keratinocyte Medium 2 and DMEM, respectively. siRNA directed against ORAI1, ORAI2, and STIM1 (Qiagen; 100ng) were transfected using HiPerFect (Qiagen). HaCaT cells were transiently transfected with Lipofectamine 2000 (Invitrogen)

Page 36: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

19

using 1µg HA-NFAT1(1-460)-GFP plasmid (Addgene 11107). DRG neurons were isolated from P18-30 mice and cultured as previously described (Wilson et al., 2011). All media and cell culture supplements were purchased from the UCSF Cell Culture Facility. Ca2+ imaging Ca2+ imaging was carried out as previously described (Wilson et al., 2011). Physiological Ringer solution: 140mM NaCl, 5mM KCl, 10mM HEPES, 2mM CaCl2, 2mM MgCl2, 10mM D-(+)-glucose, pH 7.4 with NaOH. Images were collected and analyzed using MetaFluor (Molecular Devices). [Ca2+]i was determined from background-corrected F340/F380 ratio images using the relation [Ca2+]I = K*(R−Rmin)/(Rmax−R) (Almers 1985), with the following parameters measured in keratinocytes: Rmin=0.3; Rmax=2.2; and K*=3µM. Cells were classified as responders if [Ca2+]i increased 15% above baseline. Electrophysiology Recordings were collected at 5 kHz and filtered at 2 kHz using an Axopatch 200B and PClamp software. Electrode resistances were 2-6 MΩ. Perforated patch internal solution: 140mM CsCl, 5mM EGTA ,10mM HEPES, pH 7.4 with CsOH, 0.24 mg ml−1 Amphotericin B (Rae et al., 1991). Stimulation protocol: 10ms step to -80 mV, 150ms ramp from -80mV to +80mV. Current clamp internal solution: 140mM KCl, 5mM EGTA and 10mM HEPES (pH 7.4 with KOH). Series resistance of all cells were <30 MΩ and liquid junction potentials were < 5mV (no correction). RT-PCR RNA was extracted using RNeasy (Qiagen) and reverse transcription was performed using Superscript III. RT-PCR was carried out using SYBR Green (Invitrogen) on a StepOnePlus ABI machine. Threshold cycles for each transcript (Bogiatzi et al., 2012) were normalized to GAPDH (ΔCt). Calibrations and normalizations used the 2-ΔΔCt method where ΔΔCt = [(Ct (target gene) - Ct (reference gene)] - [Ct (calibrator) - Ct (reference gene)]; GAPDH=reference gene; scrambled siRNA=calibrator. Experiments were performed in triplicate. Histology Histology was carried out as previously described (Gerhold et al., 2013). Antibodies: rabbit anti-PGP9.5 and rabbit anti-peripherin (Millipore) 1:1000; goat anti-TSLPR and mouse anti-NFATc1 (Santa Cruz Biotechnology) 1:100. IL7Ra and TSLPR probes (Panomics) were used for in situ hybridization following the Quantigene protocol (Panomics). Protein detection

Page 37: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

20

TSLP protein levels were measured using the DuoSet ELISA kit (R&D Systems) on media collected 24h after stimulation. TSLP release was normalized to vehicle. For western blots, 50µg of cleared tissue lysate was resolved by SDS-PAGE, transferred to nitrocellulose membranes and probed with Anti-TSLP (1:250, Genetex), Anti-Calnexin (1:2,000, Abcam) and Anti-Actin (1:2,000). Mice and Behavior Mice (20-35g) were housed in 12h light-dark cycle at 21ºC. Behavioral measurements were performed as previously described (Wilson et al., 2011). Compounds injected: 2.5µg TSLP, 200µg CQ, 100pg tryptase dissolved in PBS, or RTX 1µg/mL in 0.05% ascorbic acid and 7% Tween 80 (two days prior to pruritogen injection). For AITC behavior, 5µL 10% AITC in mineral oil was applied to the right hind paw. Behavioral scoring was performed while blind to treatment and genotype. All experiments were performed under the policies and recommendations of the International Association for the Study of Pain and approved by the University of California, Berkeley Animal Care and Use Committee. Data analysis Data are shown as mean ± s.e.m. Statistical significance was evaluated using a one-way ANOVA followed by a Tukey-Kramer post hoc test or unpaired two-tailed Student’s t-test for comparing difference between two samples. *p < 0.05, **p < 0.01, ***p < 0.001.

Page 38: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

37

CHAPTER II: Distinct Histamine and PAR2 Signaling in Keratinocytes. This chapter is not yet in print. My experimental contributions were to Figures 1, 2, 3A-C and 4-6. For this paper I performed cellular imaging, siRNA, pharmacology, PCR and ELISA experiments and made figures and wrote the manuscript with D.M.B. Sarah R. Wilson contributed to Figures 3D-F. SUMMARY While histamine signaling mediates some forms of allergic itch, histamine-independent signaling has been implicated in chronic itch. Antihistamines are frequently used to treat allergic itch reactions, but are generally ineffective in treating chronic itch disorders. Additionally, histamine-dependent and -independent itch are mediated by distinct cells and molecules in the nervous system. Previous studies have shown that histamine and the PAR2 agonist SLIGRL both evoke a rise in intracellular calcium in keratinocytes. We show that SLIGRL and histamine trigger release of calcium from distinct intracellular stores. In addition, while histamine triggers a rise in intracellular calcium that peaks within 5 s and decays to a steady state plateau, SLIGRL triggers oscillations with a period of 233 s. Such calcium dynamics have significant consequences on calcium-dependent gene expression. SLIGRL, but not histamine, activates NFAT and NFAT-dependent transcription. We suggest that distinct SLIGRL- and histamine-evoked calcium signaling dynamics in keratinocytes are responsible for the differing transcriptional outputs. INTRODUCTION Itch can be classified as either histamine-dependent or histamine-independent. Histamine-mediated mechanisms are better understood and are important in acute allergic itch, while histamine-independent mechanisms are important in chronic itch. Recent work has demonstrated that histamine and histamine-independent signaling are largely distinct in the nervous system. The first indication that there is separate histamine and histamine-independent circuitry came from work on the pruritogen cowhage. Spicules from the cowhage plant (Mucuna pruriens) contain the cysteine protease mucunain, which causes histamine-independent itch through activation of the Protease Activated Receptor 2 (PAR2; (Johanek et al., 2007; Reddy et al., 2008). Cowage and histamine activate distinct classes of primary afferent C-fibers (Johanek et al., 2008), distinct subsets of lamina I spinothalamic neurons in the spinal cord (Davidson et al., 2007), and incompletely overlapping areas of the cortex (Papoiu et al., 2012). Whether distinct histamine and histamine-independent signaling pathways exist in non-neuronal cells, such as keratinocytes, is not known. Keratinocytes play a key role in itch. Keratinocytes are directly activated by a number of pruritogens, including histamine, the PAR2 agonist SLIGRL, ET-1 and substance P (Dallos et al., 2006; Giustizieri et al., 2004; Santulli et al., 1995; Yohn et al., 1994). Keratinocytes in turn release inflammatory molecules in response to pruritogens, which can directly activate the itch-transducing C-fibers that terminate as free nerve endings in the skin (Zylka et al., 2005). For instance, SLIGRL triggers release of the atopic dermatitis pruritogen TSLP (Wilson et al., 2013b) and cathepsin E triggers release of

Page 39: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

38

pruritogen endothelin-1 (Andoh et al., 2012). However, the mechanisms by which activation of keratinocytes by pruritogens leads to release of inflammatory molecules remains enigmatic. T cells display calcium oscillations, which depend on both external and ER calcium stores, in response to T-cell receptor engagement. Different agonists can generate unique oscillation patterns that differ in amplitude, duration and frequency (Kar et al., 2012). Calcium-dependent transcription factors are tuned to different calcium patterns (Dolmetsch and Lewis, 1994; Dolmetsch et al., 1997; 1998). JNK and NK-κB are tuned to respond to transient calcium spikes, while NFAT is tuned to sustained low increases in calcium (Dolmetsch et al., 1997), however, oscillations reduce the calcium threshold required to activate these transcription factors (Dolmetsch and Lewis, 1994). While keratinocytes are known to oscillate in response to numerous stimuli, it is not known whether pruritogens can induce oscillations or what occurs downstream of such oscillations. Here we show that histamine and histamine-independent pruritogens evoked distinct calcium signaling pathways in keratinocytes. While histamine evokes a sustained rise in intracellular calcium, the PAR2 agonist SLIGRL evokes calcium oscillations, which lead to distinct transcriptional outputs. These findings demonstrate that like in the nervous system, histamine-dependent and histamine-independent itch signaling is distinct in keratinocytes. RESULTS Histamine and SLIGRL Evoke Distinct Calcium Dynamics in Keratinocytes We used ratiometric calcium imaging to characterize pruritogen-induced signaling pathways in keratinocytes (Figure 1). Similar to previous findings, histamine and the PAR2 agonist SLIGRL evoked a rise in intracellular calcium (Figure 1 A-D) in a dose-dependent manner (Osawa et al., 1991; Steinhoff et al., 2003). Higher doses recruited a larger percentage of responsive cells, and elicited greater response amplitudes in individual cells (Figure 1C-D). Histamine-evoked calcium signals were attenuated by the histamine H1 receptor (H1R) antagonist diphenhydramine, consistent with reports that histamine acts on keratinocytes via H1R (Figure 1E-F, (Gschwandtner et al., 2013). While histamine and SLIGRL-evoked calcium dynamics were similar over a short time scale, they evoked vastly different signals over 30 minutes. Histamine evoked a transient rise in calcium concentration that peaked before decaying to a steady state plateau (Figure 1G). In contrast, SLIGRL evoked calcium oscillations in 19% of cells (Figure 1H-I). The remaining SLIGRL-treated cells displayed responses that peaked before decaying to a steady state plateau, like histamine-treated cells. SLIGRL-evoked oscillations displayed average amplitudes of 295 ± 11 nM and periods of 233 ± 3 s, while histamine evoked a calcium concentration of 241 ± 5 nM at the steady state plateau (average calcium concentration 342-1878 s after histamine addition). We next sought to determine the mechanism by which histamine and SLIGRL evoke differing calcium dynamics.

Page 40: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

39

Figure 1. Histamine and SLIGRL trigger distinct Ca2+ dynamics in primary human keratinocytes (A) Ratiometric calcium images of keratinocytes in vehicle (VEH) or 100 µM histamine (HIS, scale bar = 100 µm). (B) Representative trace showing HIS- (100 µM) and SLIGRL-evoked (100 µM) Ca2+ responses in keratinocytes. (C) Dose response curves displaying the fraction of HIS- and SLIGRL-responsive cells (HIS EC50 = 0.31 ± 0.08 µM; SLIGRL EC50 = 2.84 ± 0.28 µM; n > 600 cells per point). (D) Dose response curves displaying the response amplitude evoked by HIS and SLIGRL (HIS EC50 = 0.46 ± 0.18 µM; SLIGRL EC50 = 55.2 ± 4.6 µM; n > 600 cells per point). (E) 10uM diphenhydramine (DPH) attenuates HIS-evoked calcium signals. Representative traces showing histamine response in the presence of DPH (red) or vehicle (black). (F) Average amplitude of HIS-evoked response in the presence of DPH (red) or vehicle (black, n>300 cells). (G) Representative trace showing sustained increase in intracellular calcium in response to histamine. (H) Representative trace showing calcium oscillation in response to SLIGRL. (I) Fraction of cells that respond to HIS and SLIGRL with oscillations. n ≥ 300 cells. ***P < 0.001. Error bars represent s.e.m.

Page 41: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

40

ORAI1 and STIM1 Are Required for Histamine and SLIGRL-Evoked Calcium Signals We first investigated whether the mechanisms that mediate histamine and SLIGRL calcium responses differ. We demonstrated previously that SLIGRL evokes calcium release from stores and store-operated calcium entry (Wilson et al., 2013b). Consistent with previous studies showing that histamine activates phospholipase C-coupled GPCRs, histamine also induced calcium release from intracellular stores in the absence of extracellular calcium and calcium influx upon subsequent addition of extracellular calcium, suggesting that histamine triggers the store-operated calcium entry (SOCE) pathway (Figure 2A, (Dai et al., 2007; Imamachi et al., 2009). Histamine and SLIGRL-evoked calcium responses were significantly attenuated by treatment with the SOCE blockers, 2-aminoethoxydiphenyl borate (2-APB; 50µM) and lanthanum (Figure 2A-C), suggesting that these agonists evoke SOCE through a common mechanism. To directly measure pruritogen-evoked currents, we performed cell-attached voltage clamp recordings on primary human keratinocytes (Figure 2D-F). Both histamine and SLIGRL evoked currents displaying inward rectification and no measurable reversal potential below +80 mV (Figure 2D). Additionally, histamine-evoked current densities demonstrate robust increases as extracellular calcium increases from 2 to 20 mM (Figure 2E). Histamine-evoked currents were significantly attenuated by preincubation with 2-APB (Figure 2F). As, both histamine and SLIGRL trigger store operated calcium entry, we next asked whether histamine and SLIGRL utilize the same SOCE molecular players. We previously demonstrated that SLIGRL evokes store-operated calcium entry in a STIM1- and ORAI1-dependent manner (Wilson et al., 2013b). We examined the role of ORAI1, ORAI2, ORAI3, STIM1 and STIM2 in histamine-evoked responses using siRNA-mediated knockdown. Depletion of ORAI1 transcripts by 75% or STIM1 transcripts by 78% significantly diminished Ca2+ entry in response to histamine and SLIGRL as compared to scrambled control siRNA (Figure 3A-D). In contrast, depletion of ORAI2 transcripts by 82%, ORAI3 transcripts by 53% or STIM2 transcripts by 65% had no effect on histamine- or SLIGRL-evoked SOCE (Figure 3C-D). These data demonstrate that histamine and SLIGRL-evoked calcium responses are mediated by the same ER calcium sensor and SOCE channel. Histamine and SLIGRL Evoke Distinct Calcium Signals We asked whether the initial kinetics of pruritogen-evoked calcium signals differ. We found that histamine and SLIGRL signals differ in time to peak. Histamine signals peak 4.7 ± 0.5s after application and SLIGRL signals peak 16.5 ± 1.3 s after application (Figure 4B). For comparison, thapsigargin signals peak 75.7 ± 5.6 s after application (Figure 4 A-B). As the majority of the early pruritogen-evoked calcium response is mediated by release from stores, we accessed the kinetics of histamine and SLIGRL signals in the absence of extracellular calcium. Histamine signals peak 11.8 ± 1.3s after application and SLIGRL signals peak 24.8 ± 3.1 s after application (Figure 4C). We next asked whether histamine and SLIGRL access different internal calcium stores.

Page 42: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

41

Figure 2. Histamine induces store-operated Ca2+ influx in primary human keratinocytes (A-B) Histamine and SLIGRL trigger Ca2+ release from stores in the absence of external Ca2+, and Ca2+ influx upon readdition of 2 mM extracellular calcium. Pretreatment with SOCE inhibitor 2-Aminoethoxydiphenyl borate (2-APB, 50 µM) attenuates influx. Representative responses to histamine (HIS, 100 µM, A) and SLIGRL (100 µM, B) in the presence (2-APB, red) or absence (CTRL, black) of 2-APB. (C) Average amplitudes of HIS- and SLIGRL-evoked calcium influx in the presence of 2-APB (red), lanthanum (50 nM La3+, blue), or vehicle (CTRL, black). HIS- and SLIGRL-evoked influx was reduced by 2-APB and La3+ (n=300-600 cells). (D) Representative current traces during voltage ramps in the presence of 100 µM HIS (top) and 1 µM SLIGRL (bottom) in cell-attached voltage clamp recordings. Representative baseline (black) and evoked currents (red; n ≥ 6). (E) Average HIS-evoked (100 µM) current densities at -120 mV in the presence of 2 mM or 20 mM external Ca2+ (n ≥ 6). (F) Average HIS-evoked (100 µM) current densities at -120 mV in the presence of 2-APB (50 µM) or vehicle (VEH), currents normalized to vehicle control (n ≥ 6). *P < 0.05; **P < 0.01, ***P < 0.001. Error bars represent s.e.m.

Page 43: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

42

Figure 3. ORAI1 and STIM1 are required for histamine-evoked calcium influx (A) siRNA-induced silencing of ORAI1-3 and STIM1-2 mRNA in primary human keratinocytes. Expression was normalized to scrambled siRNA control. (B) Representative traces showing 100 µM HIS-evoked calcium signals following treatment with siRNA targeting STIM1 (red) or scrambled control (CTRL, black). (C) Average calcium plateau at steady state after treatment with HIS or SLIGRL in cells treated with scrambled control (CTRL, black), ORAI1 (blue), ORAI2 (dark gray) or ORAI3 (light gray). (D) Average calcium plateau at steady state after treatment with HIS or SLIGRL in cells treated with scrambled control (CTRL, black), STIM1 (red) or STIM2 (dark gray). n ≥ 500 cells. NS = Not Significant, *P < 0.05; **P < 0.01, ***P < 0.001. Error bars represent s.e.m.

Page 44: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

43

Figure 4. Histamine and SLIGRL access distinct stores (A) Thapsigargin (TG, 1 µM) triggers a slow rise in intracellular calcium. (B) Average time to peak after treatment with HIS (100 µM), SLIGRL (100 µM) and TG (1 µM). (C) Average time to peak after treatment with HIS (100 µM) and SLIGRL (100 µM) in the absence of extracellular calcium. (D) HIS and SLIGRL trigger calcium release from incompletely overlapping subsets of internal calcium stores. SLIGRL triggers store release that is not accessed by histamine. Representative trace showing store release in response to consecutive treatments with HIS (100 µM), SLIGRL (100 µM) and TG (1 µM) in the absence of extracellular calcium. (E) HIS triggers store release that is not accessed by SLIGRL. Representative trace showing store release in response to consecutive treatments with SLIGRL (100 µM), HIS (100 µM) and TG (1 µM) in the absence of extracellular calcium. (F) Store release triggered by HIS and SLIGRL are not significantly different. Average calcium entry, calculated as the integral over stimulation duration, triggered by treatment of histamine and SLIGRL in the absence of extracellular calcium. (G) Stores exclusively accessed by HIS and SLIGRL are not significantly different. Average calcium entry triggered by histamine following SLIGRL treatment, and average calcium entry triggered by SLIGRL following HIS treatment in the absence of extracellular calcium. (H) Representative trace showing store release in response to consecutive treatments with TG (1 µM), HIS (100 µM) and SLIGRL (100 µM) in the absence of extracellular calcium. (I) Venn diagram showing that HIS, SLIGRL and TG trigger distinct intracellular calcium stores. HIS and SLIGRL trigger incompletely overlapping stores, which are all SERCA-dependent stores. n ≥ 100 cells. NS = Not Significant, *P < 0.05, ***P < 0.001. Error bars represent s.e.m.

Page 45: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

44

We found that histamine and SLIGRL trigger calcium release from incompletely overlapping subsets of internal calcium stores. In the absence of extracellular calcium, histamine evoked a large response due to release from stores, and subsequent SLIGRL treatment evoked a smaller calcium response (Figure 4D). Thus, SLIGRL triggers release from a subset of stores that histamine does not access. Similarly, initial treatment with SLIGRL evoked a large response and subsequent histamine treatment evoked a smaller calcium response (Figure 4E). These data demonstrate that histamine also triggers release from a subset of stores that SLIGRL does not access. Thus, histamine and SLIGRL access incompletely overlapping stores. We next asked whether the size of the stores accessed by histamine and SLIGRL differ. Calcium entry, calculated as the integral over the stimulation duration, in response to treatments with histamine and SLIGRL were not significantly different, demonstrating that histamine and SLIGRL access similarly sized stores (Figure 4F). Additionally, the stores accessed by only SLIGRL and the stores accessed by only histamine are also similarly sized, as the calcium entry triggered by histamine following SLIGRL treatment and the calcium entry triggered by SLIGRL following histamine treatment were not significantly different (Figure 4G). Addition of the SERCA inhibitor, thapsigargin, after histamine and SLIGRL treatment demonstrates that histamine and SLIGRL do not access all SERCA-dependent stores (Figure 4D-E). However, all histamine and SLIGRL-evoked calcium release from stores is SERCA-dependent as initial treatment with thapsigargin prevents subsequent histamine and SLIGRL responses (Figure 4H-I). Histamine and SLIGRL Evoke Distinct Transcriptional Outputs As calcium dynamics have significant consequences on calcium-dependent gene expression, we asked whether histamine- and SLIGRL-evoked calcium signals result in differing transcriptional outputs. We previously demonstrated that PAR2 activation by SLIGRL induces calcium-dependent NFAT translocation and TSLP secretion (Wilson et al., 2013b). In contrast to SLIGRL, histamine does not induce NFAT translocation, TSLP expression or secretion (Figure 5A-D). Published work suggests that histamine and SLIGRL regulate a distinct complement of genes (Table 1). For example, histamine was found to positively regulate SEMA3A, but PAR2 activation had no effect (Fukamachi et al., 2011). Other genes are regulated by both histamine and PAR2 antagonists, such as IL-8, which was found to be upregulated, and NGF, which was found to be down-regulated (Fukamachi et al., 2011). The transcription of many genes have been studied with one but not both pruritogens. A comprehensive comparison has not been performed. To ascertain the complete transcriptional consequence of differing histamine- and SLIGLR-evoked calcium dynamics, we are currently performing RNAseq on keratinocytes treated with histamine, SLIGRL or vehicle for 30 minutes. Such analyses will shed light on the role of keratinocytes in histamine and protease signaling and the mechanism by which keratinocyte itch signaling feeds into neural circuits for itch. Additionally, analysis of promoter sequences of the regulated genes will enable us to identify common motifs that may implicate transcription factors responsible for pruritogen gene regulation.

Page 46: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

45

Figure 5. Histamine does not induce NFAT translocation or TSLP expression (A) Representative images displaying cytosolic and nuclear localization of NFAT (green) in HaCaT keratinocytes transfected with NFAT-GFP before (baseline) and following incubation with HIS (100 µM, left) or SLIGRL (100 µM, right). (B) Fraction of cells displaying nuclear localization of NFAT-GFP following treatment with vehicle (VEH; white), HIS (100 µM; red), or SLIGRL (100 µM; black). n ≥ 100 cells. (C) TSLP expression in human keratinocytes in response to 3 h treatment with vehicle (white), HIS (100 µM ) or SLIGRL (100 µM). (D) Vehicle-, 100 µM HIS-, and 100 µM SLIGRL-evoked TSLP release 18 h after stimulation as measured by ELISA (n ≥ 6 experiments). NS = Not Significant, *P < 0.05; **P < 0.01, ***P < 0.001. Error bars represent s.e.m.

Page 47: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

46

Table 1. Histamine and SLIGRL have distinct transcriptional outputs

Gene Regulation by histamine Regulation by PAR2 References

TSLP Unchanged Upregulated Present study and Wilson et al., 2013b

IL-8 Upregulated Upregulated Fukamachi et al., 2011

SEMA3A Upregulated Unchanged Fukamachi et al., 2011

NGF Downregulated Downregulated Fukamachi et al., 2011

GM-CSF Upregulated Unknown Fukamachi et al., 2011 Kanda and Watanabe, 2004

Leukotriene B(4) Unknown Upregulated Zhu et al., 2009

Prostaglandin E(2) Unknown Upregulated Zhu et al., 2009

Keratin 5, Keratin 14 Not changed Unknown Gschwandtner et al., 2013

Filaggrin, Loricrin, Downregulated Unknown Gschwandtner et al., 2013

Keratin 1, Keratin 10 Downregulated Unknown Gschwandtner et al., 2013

Page 48: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

47

DISCUSSION Itch is a common clinical complaint associated with many diseases, including atopic dermatitis, celiac disease and some cancers (Garibyan et al., 2013). While histamine-mediated itch is important for allergic itch, most forms of chronic itch are mediated by histamine-independent mechanisms. Additionally, the neurons responsible for mediating histaminergic and non-histaminergic itch are largely distinct (Jeffry et al., 2011). Here we provide evidence for an extension of this division in non-neuronal itch signaling. While both histamine and the histamine-independent pruritogen SLIGRL activate keratinocytes in a dose-dependent manner, we show that they differ in their transcriptional outputs. SLIGRL, unlike histamine, induces NFAT activation and NFAT-dependent transcription. We show that SLIGRL- and histamine-evoked calcium signals are similar in that they both require STIM1 and ORAI1, but mobilize calcium from both shared and unique calcium stores. In addition, while SLIGRL triggers oscillations, histamine triggers a biphasic calcium response that decays to a low level plateau. Calcium oscillations have been observed in many cell types, arising from diverse mechanisms and having diverse physiological consequences (Lewis, 2003). Many studies on calcium oscillations use T-cells as model systems. Stimulation of the T-cell antigen receptor can trigger oscillations that require ORAI1 and result in NFAT activation (Ong et al., 2012). Prolonged signals of ten minutes or longer are required to trigger NFAT activation and promote departure from T-cell quiescence (Negulescu et al., 1994). Similarly, we observe that SLIGRL-evoked oscillations initiate after ~5 minutes. It is not known whether there is segregation in histamine and histamine-independent signaling pathways on a shorter time scale. We did not observe differences in histamine- and SLIGRL-evoked calcium signaling during the early phase of the response, however this does not rule out signaling via other secondary messengers. For instance, histamine receptor H4, which is expressed in keratinocytes, couples to Gi/o and leads to the inhibition of cAMP formation (Yamaura et al., 2009). SLIGRL induces oscillations and NFAT activation, while histamine does not induce oscillations or NFAT activation, suggesting that oscillations may drive NFAT activation in keratinocytes, as well. However, this evidence is merely correlative. Further evidence may come from experiments confirming single-cell correlation between oscillations and NFAT activation in SLIGRL-treated cells. As SLIGRL evokes oscillations in 19% of cells, our hypothesis would predict that the 19% of cells that oscillate will display NFAT activation, while the 81% of cells that maintain steady calcium concentrations will not display NFAT activation. To directly test whether oscillations are sufficient to induce NFAT activation in keratinocytes, we could reproduce calcium clamp experiments done in T-cells, which were used to determine the optimal frequency at which oscillations activate NFAT (Dolmetsch et al., 1998). Further work is required to elucidate the physiological role of histamine and histamine-independent signaling separation in keratinocytes. We have previously demonstrated that PAR2 evoked induces TSLP expression (Wilson et al., 2013b). Other investigators have demonstrated that PAR2 activation induces expression of IL-8, GM-CSF, leukotriene B(4) and prostaglandin E(2) (Fukamachi et al., 2011; Zhu et al., 2009), while

Page 49: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

48

histamine evokes expression of IL-8, GM-CSF, SEMA3A, NGF and chemokines (Fukamachi et al., 2011; Giustizieri et al., 2004; Kanda and Watanabe, 2003), but downregulation of differentiation markers (Gschwandtner et al., 2013). While the transcriptional pathway mediating pruritogen-evoked expression of many of these genes is unresolved, initial work has implicated a number of transcription factors. For example, we showed that SLIGRL-evoked TSLP expression has been linked to NFAT activity (Wilson et al., 2013b). Sequences homologous to activator protein 1, nuclear factor kB and NFAT cis-elements have been found in the promoter of GM-CSF (Kanda and Watanabe, 2004). Additional questions remain: Are there other downstream effects of histamine and histamine-independent signaling in keratinocytes? How do histamine and histamine-independent signaling in keratinocytes feed into the neural circuits for itch transduction? A better understanding of the role of keratinocytes in the itch circuitry may lead to valuable therapies for itch patients. EXPERIMENTAL PROCEDURES Epithelial cell culture Normal human epidermal keratinocytes (PromoCell) were cultured in PromoCell Keratinocyte Growth Medium 2. Cells passaged fewer than 5 times were plated on coverslips and imaged 12-48 h later. For siRNA experiments, 100 ng of siRNA constructs in Opti-MEM (Invitrogen) were transfected using HiPerFect (Qiagen) according to manufacturer’s instruction. FlexiTube GeneSolution siRNA directed against ORAI1, ORAI2, ORAI3, STIM1 and STIM2 were purchased from Qiagen. Calcium imaging Cells were loaded for 1 h with 10 µM Fura-2AM (Invitrogen), supplemented with 0.01% w/v Pluronic F-127 (w/vol, Invitrogen), in a physiological Ringer solution containing 140 mM NaCl, 5 mM KCl, 10 mM HEPES, 2 mM CaCl2, 2 mM MgCl2 and 10mM D-(+)-glucose, pH 7.4 with KOH. All chemicals were purchased from Sigma. Images were collected using MetaFluor (Molecular Devices) and analyzed using MetaMorph software. [Ca2+]i was determined from background-corrected F340/F380 ratio images using the relation [Ca2+]i=K*(R−Rmin)/(Rmax−R) 48, with the following parameters measured in keratinocytes: Rmin (fluorescence ratio measured in 10 mM 1,2-Bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid (BAPTA), Rmax (fluorescence ratio in the presence of 20 mM Ca2+) and K* (determined from the fluorescence ratio measured in 200 nM Ca2+ ). Acquired images were displayed as the ratio of 340 nm to 380 nm. For the identification of oscillations, the built-in fast Fourier transform function in Python was used. Oscillations were defined as having a frequency component between 0.003 and 0.01 Hz of amplitude greater than 0.025 µM over baseline. Electrophysiology Voltage-clamp recordings were collected at 5 kHz and filtered at 2 kHz using an Axopatch 200B and PClamp software. For cell-attached recordings, electrode resistance ranged between 4-6 MΩ. Solutions are described above. The following voltage protocol was applied to cell-attached patches: 10 ms voltage step to -120 mV, followed by a ramp from -120 mV to +100 mV in 150 ms. Current densities were calculated as the mean current over 10 ms at +120 mV normalized by the patch

Page 50: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

49

capacitance. In all experiments, liquid junction potentials were < 5 mV in all experiments and were not corrected. Real time qPCR Total RNA from keratinocytes was extracted using RNeasy (Qiagen). 500 ng total RNA was used to generate 1st strand cDNA using SuperScript III Reverse Transcriptase (Invitrogen). Real time PCR was carried out using SYBR GreenER qPCR Supermix for ABI PRISM (Invitrogen) on a StepOnePlus ABI machine. To determine gene expression in keratinocytes, threshold cycles for each transcript (CT) were normalized to GAPDH (ΔCt). Gene expression is shown as 2ΔCt. For the analysis of siRNA-induced down-regulation of mRNA expression, GAPDH was used as the reference gene and scrambled siRNA treated keratinocytes was the calibrator. Calibrations and normalizations were done using the 2-ΔΔCt method where ΔΔCt = [(Ct (target gene) – Ct (reference gene)] – [Ct (calibrator) – Ct (reference gene)]. Real-time PCR were performed in triplicates. We used primers for ORAI1 (forward, 5'-ATG GTG GCA ATG GTG GAG-3'; reverse, 5'-CGA TGA CGG TGG AGA AGG-3'), ORAI2 (forward, 5'-GCA ACA TCC ACA ACC TGA AC-3'; reverse, 5'-AGC AGA GCA GCA CCA CCT-3'), ORAI3 (forward, 5'-tca aaa gat gct ccc acc ac-3'; reverse, 5'-ccc aaa atg cca acc ata aa-3'), STIM1 (forward, 5'-gga atg ggt tgg gag agg-3'; reverse, 5'-ggt gtg ggt ggg agt aga ga-3'), and GAPDH (forward, 5'- CCA CTC CTC CAC CTT TGA C-3'; reverse, 5'- ACC CTG TTG CTG TAG CC-3'). ELISA Concentration of TSLP protein levels was measured with ELISA kits (DuoSet; R&D Systems) using culture supernatant collected 18h after stimulation. TSLP release is displayed normalized to vehicle. A one-way analysis of variance (ANOVA) with Tukey’s multiple comparison test was used to establish significance. Data analysis Data are shown as mean ± s.e.m. Statistical significance was evaluated using a one-way ANOVA followed by a Tukey-Kramer post hoc test or unpaired two-tailed Student’s t-test for comparing difference between two samples.

Page 51: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

50

REFERENCES: Akiyama, T., Carstens, M.I., and Carstens, E. (2010). Enhanced scratching evoked by PAR-2 agonist and 5-HT but not histamine in a mouse model of chronic dry skin itch. Pain 151, 378–383.

Al-Daraji, W.I., Grant, K.R., Ryan, K., Saxton, A., and Reynolds, N.J. (2002). Localization of calcineurin/NFAT in human skin and psoriasis and inhibition of calcineurin/NFAT activation in human keratinocytes by cyclosporin A. J Invest Dermatol 118, 779–788.

Amadesi, S., Cottrell, G.S., Divino, L., Chapman, K., Grady, E.F., Bautista, F., Karanjia, R., Barajas-Lopez, C., Vanner, S., Vergnolle, N., et al. (2006). Protease-activated receptor 2 sensitizes TRPV1 by protein kinase Cepsilon- and A-dependent mechanisms in rats and mice. J Physiol (Lond) 575, 555–571.

Amadesi, S., Nie, J., Vergnolle, N., Cottrell, G.S., Grady, E.F., Trevisani, M., Manni, C., Geppetti, P., McRoberts, J.A., Ennes, H., et al. (2004). Protease-activated receptor 2 sensitizes the capsaicin receptor transient receptor potential vanilloid receptor 1 to induce hyperalgesia. J Neurosci 24, 4300–4312.

Andoh, T., Katsube, N., Maruyama, M., and Kuraishi, Y. (2001). Involvement of leukotriene B(4) in substance P-induced itch-associated response in mice. J Invest Dermatol 117, 1621–1626.

Andoh, T., Yoshida, T., Lee, J.-B., and Kuraishi, Y. (2012). Cathepsin E induces itch-related response through the production of endothelin-1 in mice. Eur J Pharmacol 686, 16–21.

Arita, K., South, A.P., Hans-Filho, G., Sakuma, T.H., Lai-Cheong, J., Clements, S., Odashiro, M., Odashiro, D.N., Hans-Neto, G., Hans, N.R., et al. (2008). Oncostatin M receptor-beta mutations underlie familial primary localized cutaneous amyloidosis. Am J Hum Genet 82, 73–80.

Ballantyne, J.C., Loach, A.B., and Carr, D.B. (1988). Itching after epidural and spinal opiates. Pain 33, 149–160.

Bandell, M., Story, G.M., Hwang, S.W., Viswanath, V., Eid, S.R., Petrus, M.J., Earley, T.J., and Patapoutian, A. (2004). Noxious cold ion channel TRPA1 is activated by pungent compounds and bradykinin. Neuron 41, 849–857.

Basbaum, A.I., Bautista, D.M., Scherrer, G., and Julius, D. (2009). Cellular and molecular mechanisms of pain. Cell 139, 267–284.

Bautista, D.M., Movahed, P., Hinman, A., Axelsson, H.E., Sterner, O., Högestätt, E.D., Julius, D., Jordt, S.-E., and Zygmunt, P.M. (2005). Pungent products from garlic activate the sensory ion channel TRPA1. Proc Natl Acad Sci USA 102, 12248–12252.

Page 52: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

51

Bautista, D.M., Pellegrino, M., and Tsunozaki, M. (2013). TRPA1: A gatekeeper for inflammation. Annu Rev Physiol 75, 181–200.

Bell, J.K., McQueen, D.S., and Rees, J.L. (2004). Involvement of histamine H4 and H1 receptors in scratching induced by histamine receptor agonists in Balb C mice. Br J Pharmacol 142, 374–380.

Bogiatzi, S.I., Guillot-Delost, M., Cappuccio, A., Bichet, J.-C., Chouchane-Mlik, O., Donnadieu, M.-H., Barillot, E., Hupé, P., Chlichlia, K., Efremidou, E.I., et al. (2012). Multiple-checkpoint inhibition of thymic stromal lymphopoietin-induced TH2 response by TH17-related cytokines. J Allergy Clin Immunol 130, 233–40.e235.

Briot, A., Deraison, C., Lacroix, M., Bonnart, C., Robin, A., Besson, C., Dubus, P., and Hovnanian, A. (2009). Kallikrein 5 induces atopic dermatitis-like lesions through PAR2-mediated thymic stromal lymphopoietin expression in Netherton syndrome. J. Exp. Med. 206, 1135–1147.

Briot, A., Lacroix, M., Robin, A., Steinhoff, M., Deraison, C., and Hovnanian, A. (2010). Par2 inactivation inhibits early production of TSLP, but not cutaneous inflammation, in Netherton syndrome adult mouse model. J Invest Dermatol 130, 2736–2742.

Cahalan, M.D. (2009). STIMulating store-operated Ca(2+) entry. Nat Cell Biol 11, 669–677.

Cevikbas, F., Steinhoff, A., Homey, B., and Steinhoff, M. (2007). Neuroimmune interactions in allergic skin diseases. Curr Opin Allergy Clin Immunol 7, 365–373.

Chang, S.-E., Han, S.-S., Jung, H.-J., and Choi, J.-H. (2007). Neuropeptides and their receptors in psoriatic skin in relation to pruritus. Br J Dermatol 156, 1272–1277.

Chang, W.-C., Lee, C.-H., Hirota, T., Wang, L.-F., Doi, S., Miyatake, A., Enomoto, T., Tomita, K., Sakashita, M., Yamada, T., et al. (2012). ORAI1 genetic polymorphisms associated with the susceptibility of atopic dermatitis in Japanese and Taiwanese populations. PLoS ONE 7, e29387.

Choi, J.C., Yang, J.H., Chang, S.-E., and Choi, J.-H. (2005). Pruritus and nerve growth factor in psoriasis. Korean Journal of Dermatology.

Dai, Y., Moriyama, T., Higashi, T., Togashi, K., Kobayashi, K., Yamanaka, H., Tominaga, M., and Noguchi, K. (2004). Proteinase-activated receptor 2-mediated potentiation of transient receptor potential vanilloid subfamily 1 activity reveals a mechanism for proteinase-induced inflammatory pain. J Neurosci 24, 4293–4299.

Dai, Y., Wang, S., Tominaga, M., Yamamoto, S., Fukuoka, T., Higashi, T., Kobayashi, K., Obata, K., Yamanaka, H., and Noguchi, K. (2007). Sensitization of TRPA1 by PAR2 contributes to the sensation of inflammatory pain. J Clin Invest 117, 1979–1987.

Dallos, A., Kiss, M., Polyánka, H., Dobozy, A., Kemény, L., and Husz, S. (2006). Effects

Page 53: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

52

of the neuropeptides substance P, calcitonin gene-related peptide, vasoactive intestinal polypeptide and galanin on the production of nerve growth factor and inflammatory cytokines in cultured human keratinocytes. Neuropeptides 40, 251–263.

Davidson, S., Zhang, X., Yoon, C.H., Khasabov, S.G., Simone, D.A., and Giesler, G.J. (2007). The itch-producing agents histamine and cowhage activate separate populations of primate spinothalamic tract neurons. J Neurosci 27, 10007–10014.

DeHaven, W.I., Smyth, J.T., Boyles, R.R., Bird, G.S., and Putney, J.W. (2008). Complex actions of 2-aminoethyldiphenyl borate on store-operated calcium entry. J Biol Chem 283, 19265–19273.

Dillon, S.R., Sprecher, C., Hammond, A., Bilsborough, J., Rosenfeld-Franklin, M., Presnell, S.R., Haugen, H.S., Maurer, M., Harder, B., Johnston, J., et al. (2004). Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice. Nat Immunol 5, 752–760.

Dolmetsch, R.E., and Lewis, R.S. (1994). Signaling between intracellular Ca2+ stores and depletion-activated Ca2+ channels generates [Ca2+]i oscillations in T lymphocytes. J Gen Physiol 103, 365–388.

Dolmetsch, R.E., Lewis, R.S., Goodnow, C.C., and Healy, J.I. (1997). Differential activation of transcription factors induced by Ca2+ response amplitude and duration. Nature 386, 855–858.

Dolmetsch, R.E., Xu, K., and Lewis, R.S. (1998). Calcium oscillations increase the efficiency and specificity of gene expression. Nature 392, 933–936.

Dou, Y.-C., Hagströmer, L., Emtestam, L., and Johansson, O. (2006). Increased nerve growth factor and its receptors in atopic dermatitis: an immunohistochemical study. Arch Dermatol Res 298, 31–37.

Dunford, P.J., Williams, K.N., Desai, P.J., Karlsson, L., McQueen, D., and Thurmond, R.L. (2007). Histamine H4 receptor antagonists are superior to traditional antihistamines in the attenuation of experimental pruritus. J Allergy Clin Immunol 119, 176–183.

Feske, S. (2010). CRAC channelopathies. Pflugers Arch 460, 417–435.

Feske, S., Gwack, Y., Prakriya, M., Srikanth, S., Puppel, S.-H., Tanasa, B., Hogan, P.G., Lewis, R.S., Daly, M., and Rao, A. (2006a). A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature 441, 179–185.

Feske, S., Gwack, Y., Prakriya, M., Srikanth, S., Puppel, S.-H., Tanasa, B., Hogan, P.G., Lewis, R.S., Daly, M., and Rao, A. (2006b). A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature 441, 179–185.

Fitzsimons, C., Engel, N., Durán, H., Policastro, L., Cricco, G., Martín, G., Molinari, B., and Rivera, E. (2001). Histamine production in mouse epidermal keratinocytes is

Page 54: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

53

regulated during cellular differentiation. Inflamm. Res. 50 Suppl 2, S100–S101.

Fukamachi, S., Bito, T., Shiraishi, N., Kobayashi, M., Kabashima, K., Nakamura, M., and Tokura, Y. (2011). Modulation of semaphorin 3A expression by calcium concentration and histamine in human keratinocytes and fibroblasts. J Dermatol Sci 61, 118–123.

Garibyan, L., Rheingold, C.G., and Lerner, E.A. (2013). Understanding the pathophysiology of itch. Dermatol Ther 26, 84–91.

Gerhold, K.A., Pellegrino, M., Tsunozaki, M., Morita, T., Leitch, D.B., Tsuruda, P.R., Brem, R.B., Catania, K.C., and Bautista, D.M. (2013). The star-nosed mole reveals clues to the molecular basis of mammalian touch. PLoS ONE 8, e55001.

Giustizieri, M.L., Albanesi, C., Fluhr, J., Gisondi, P., Norgauer, J., and Girolomoni, G. (2004). H1 histamine receptor mediates inflammatory responses in human keratinocytes. J Allergy Clin Immunol 114, 1176–1182.

Grant, A.D., Cottrell, G.S., Amadesi, S., Trevisani, M., Nicoletti, P., Materazzi, S., Altier, C., Cenac, N., Zamponi, G.W., Bautista-Cruz, F., et al. (2007). Protease-activated receptor 2 sensitizes the transient receptor potential vanilloid 4 ion channel to cause mechanical hyperalgesia in mice. J Physiol (Lond) 578, 715–733.

Grimstad, O., Sawanobori, Y., Vestergaard, C., Bilsborough, J., Olsen, U.B., Grønhøj-Larsen, C., and Matsushima, K. (2009). Anti-interleukin-31-antibodies ameliorate scratching behaviour in NC/Nga mice: a model of atopic dermatitis. Exp Dermatol 18, 35–43.

Gschwandtner, M., Mildner, M., Mlitz, V., Gruber, F., Eckhart, L., Werfel, T., Gutzmer, R., Elias, P.M., and Tschachler, E. (2013). Histamine suppresses epidermal keratinocyte differentiation and impairs skin barrier function in a human skin model. Allergy 68, 37–47.

Gusarova, G.A., Trejo, H.E., Dada, L.A., Briva, A., Welch, L.C., Hamanaka, R.B., Mutlu, G.M., Chandel, N.S., Prakriya, M., and Sznajder, J.I. (2011). Hypoxia leads to Na,K-ATPase downregulation via Ca(2+) release-activated Ca(2+) channels and AMPK activation. Mol. Cell. Biol. 31, 3546–3556.

Gwack, Y., Sharma, S., Nardone, J., Tanasa, B., Iuga, A., Srikanth, S., Okamura, H., Bolton, D., Feske, S., Hogan, P.G., et al. (2006). A genome-wide Drosophila RNAi screen identifies DYRK-family kinases as regulators of NFAT. Nature 441, 646–650.

Hachem, J.-P., Man, M.-Q., Crumrine, D., Uchida, Y., Brown, B.E., Rogiers, V., Roseeuw, D., Feingold, K.R., and Elias, P.M. (2005). Sustained serine proteases activity by prolonged increase in pH leads to degradation of lipid processing enzymes and profound alterations of barrier function and stratum corneum integrity. J Invest Dermatol 125, 510–520.

Page 55: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

54

Halfter, U.M., Derbyshire, Z.E., and Vaillancourt, R.R. (2005). Interferon-gamma-dependent tyrosine phosphorylation of MEKK4 via Pyk2 is regulated by annexin II and SHP2 in keratinocytes. Biochem J 388, 17–28.

Han, L., Ma, C., Liu, Q., Weng, H.-J., Cui, Y., Tang, Z., Kim, Y., Nie, H., Qu, L., Patel, K.N., et al. (2013). A subpopulation of nociceptors specifically linked to itch. Nat Neurosci 16, 174–182.

Han, S.-K., Mancino, V., and Simon, M.I. (2006). Phospholipase Cbeta 3 mediates the scratching response activated by the histamine H1 receptor on C-fiber nociceptive neurons. Neuron 52, 691–703.

Hägermark, O. (1995). Itch mediators. Semin Dermatol 14, 271–276.

He, R., and Geha, R.S. (2010). Thymic stromal lymphopoietin. Ann N Y Acad Sci 1183, 13–24.

Hilliges, M., Wang, L., and Johansson, O. (1995). Ultrastructural evidence for nerve fibers within all vital layers of the human epidermis. J Invest Dermatol 104, 134–137.

Holgate, S.T. (2007). The epithelium takes centre stage in asthma and atopic dermatitis. Trends Immunol. 28, 248–251.

Hovnanian, A. (2013). Netherton syndrome: skin inflammation and allergy by loss of protease inhibition. Cell Tissue Res 351, 289–300.

Ikoma, A., Rukwied, R., Ständer, S., Steinhoff, M., Miyachi, Y., and Schmelz, M. (2003a). Neurophysiology of pruritus: interaction of itch and pain. Arch Dermatol 139, 1475–1478.

Ikoma, A., Rukwied, R., Ständer, S., Steinhoff, M., Miyachi, Y., and Schmelz, M. (2003b). Neuronal sensitization for histamine-induced itch in lesional skin of patients with atopic dermatitis. Arch Dermatol 139, 1455–1458.

Ikoma, A., Steinhoff, M., Ständer, S., Yosipovitch, G., and Schmelz, M. (2006). The neurobiology of itch. Nat Rev Neurosci 7, 535–547.

Imamachi, N., Park, G.H., Lee, H., Anderson, D.J., Simon, M.I., Basbaum, A.I., and Han, S.-K. (2009). TRPV1-expressing primary afferents generate behavioral responses to pruritogens via multiple mechanisms. Proc Natl Acad Sci USA 106, 11330–11335.

Jariwala, S.P., Abrams, E., Benson, A., Fodeman, J., and Zheng, T. (2011). The role of thymic stromal lymphopoietin in the immunopathogenesis of atopic dermatitis. Clin Exp Allergy 41, 1515–1520.

Järvikallio, A., Harvima, I.T., and Naukkarinen, A. (2003). Mast cells, nerves and neuropeptides in atopic dermatitis and nummular eczema. Arch Dermatol Res 295, 2–7.

Page 56: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

55

Jeffry, J., Kim, S., and Chen, Z.-F. (2011). Itch signaling in the nervous system. Physiology 26, 286–292.

Jessup, H.K., Brewer, A.W., Omori, M., Rickel, E.A., Budelsky, A.L., Yoon, B.-R.P., Ziegler, S.F., and Comeau, M.R. (2008). Intradermal administration of thymic stromal lymphopoietin induces a T cell- and eosinophil-dependent systemic Th2 inflammatory response. J. Immunol. 181, 4311–4319.

Johanek, L.M., Meyer, R.A., Friedman, R.M., Greenquist, K.W., Shim, B., Borzan, J., Hartke, T., Lamotte, R.H., and Ringkamp, M. (2008). A role for polymodal C-fiber afferents in nonhistaminergic itch. J Neurosci 28, 7659–7669.

Johanek, L.M., Meyer, R.A., Hartke, T., Hobelmann, J.G., Maine, D.N., Lamotte, R.H., and Ringkamp, M. (2007). Psychophysical and physiological evidence for parallel afferent pathways mediating the sensation of itch. J Neurosci 27, 7490–7497.

Johansson, O., Liang, Y., and Emtestam, L. (2002). Increased nerve growth factor- and tyrosine kinase A-like immunoreactivities in prurigo nodularis skin -- an exploration of the cause of neurohyperplasia. Arch Dermatol Res 293, 614–619.

Kagami, S., Sugaya, M., Suga, H., Morimura, S., Kai, H., Ohmatsu, H., Fujita, H., Tsunemi, Y., and Sato, S. (2013). Serum gastrin-releasing Peptide levels correlate with pruritus in patients with atopic dermatitis. J Invest Dermatol 133, 1673–1675.

Kanda, N., and Watanabe, S. (2003). Histamine enhances the production of nerve growth factor in human keratinocytes. J Invest Dermatol 121, 570–577.

Kanda, N., and Watanabe, S. (2004). Histamine enhances the production of granulocyte-macrophage colony-stimulating factor via protein kinase C alpha and extracellular signal-regulated kinase in human keratinocytes. J Invest Dermatol 122, 863-872.

Kanda, N., Koike, S., and Watanabe, S. (2005). Prostaglandin E2 enhances neurotrophin-4 production via EP3 receptor in human keratinocytes. J Pharmacol Exp Ther 315, 796–804.

Kar, P., Bakowski, D., Di Capite, J., Nelson, C., and Parekh, A.B. (2012). Different agonists recruit different stromal interaction molecule proteins to support cytoplasmic Ca2+ oscillations and gene expression. Proc Natl Acad Sci USA 109, 6969–6974.

Kim, B.M., Lee, S.H., Shim, W.-S., and Oh, U. (2004). Histamine-induced Ca(2+) influx via the PLA(2)/lipoxygenase/TRPV1 pathway in rat sensory neurons. Neurosci Lett 361, 159–162.

Koga, K., Chen, T., Li, X.-Y., Descalzi, G., Ling, J., Gu, J., and Zhuo, M. (2011). Glutamate acts as a neurotransmitter for gastrin releasing peptide-sensitive and insensitive itch-related synaptic transmission in mammalian spinal cord. Mol Pain 7, 47.

Page 57: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

56

Kouzaki, H., O'Grady, S.M., Lawrence, C.B., and Kita, H. (2009). Proteases induce production of thymic stromal lymphopoietin by airway epithelial cells through protease-activated receptor-2. J. Immunol. 183, 1427–1434.

Lagerström, M.C., Rogoz, K., Abrahamsen, B., Persson, E., Reinius, B., Nordenankar, K., Olund, C., Smith, C., Mendez, J.A., Chen, Z.-F., et al. (2010). VGLUT2-dependent sensory neurons in the TRPV1 population regulate pain and itch. Neuron 68, 529–542.

Lee, C.-H., Chuang, H.-Y., Shih, C.-C., Jong, S.-B., Chang, C.-H., and Yu, H.-S. (2006). Transepidermal water loss, serum IgE and beta-endorphin as important and independent biological markers for development of itch intensity in atopic dermatitis. Br J Dermatol 154, 1100–1107.

Lee, C.-H., and Yu, H.-S. (2011). Biomarkers for itch and disease severity in atopic dermatitis. Curr. Probl. Dermatol. 41, 136–148.

Lewis, R.S. (2003). Calcium oscillations in T-cells: mechanisms and consequences for gene expression. Biochem. Soc. Trans. 31, 925–929.

Li, M., Messaddeq, N., Teletin, M., Pasquali, J.-L., Metzger, D., and Chambon, P. (2005). Retinoid X receptor ablation in adult mouse keratinocytes generates an atopic dermatitis triggered by thymic stromal lymphopoietin. Proc Natl Acad Sci USA 102, 14795–14800.

Lis, A., Peinelt, C., Beck, A., Parvez, S., Monteilh-Zoller, M., Fleig, A., and Penner, R. (2007). CRACM1, CRACM2, and CRACM3 are store-operated Ca2+ channels with distinct functional properties. Curr Biol 17, 794–800.

Liu, B., Escalera, J., Balakrishna, S., Fan, L., Caceres, A.I., Robinson, E., Sui, A., McKay, M.C., McAlexander, M.A., Herrick, C.A., et al. (2013). TRPA1 controls inflammation and pruritogen responses in allergic contact dermatitis. Faseb J 27, 3549–3563.

Liu, Q., Sikand, P., Ma, C., Tang, Z., Han, L., Li, Z., Sun, S., Lamotte, R.H., and Dong, X. (2012). Mechanisms of itch evoked by β-alanine. J Neurosci 32, 14532–14537.

Liu, Q., Tang, Z., Surdenikova, L., Kim, S., Patel, K.N., Kim, A., Ru, F., Guan, Y., Weng, H.-J., Geng, Y., et al. (2009). Sensory neuron-specific GPCR Mrgprs are itch receptors mediating chloroquine-induced pruritus. Cell 139, 1353–1365.

Liu, Q., Weng, H.-J., Patel, K.N., Tang, Z., Bai, H., Steinhoff, M., and Dong, X. (2011). The distinct roles of two GPCRs, MrgprC11 and PAR2, in itch and hyperalgesia. Sci Signal 4, ra45.

Liu, T., and Ji, R-R. (2013). New insights into the mechanisms of itch: are pain and itch controlled by distinct mechanisms? Eur J Physiol 465, 1671–1685.

Liu, Y., Samad, O.A., Zhang, L., Duan, B., Tong, Q., Lopes, C., Ji, R.-R., Lowell, B.B.,

Page 58: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

57

and Ma, Q. (2010). VGLUT2-dependent glutamate release from nociceptors is required to sense pain and suppress itch. Neuron 68, 543–556.

Locksley, R.M. (2010). Asthma and allergic inflammation. Cell 140, 777–783.

Lumpkin, E.A., and Caterina, M.J. (2007). Mechanisms of sensory transduction in the skin. Nature 445, 858–865.

Madan, V., and Griffiths, C.E.M. (2007). Systemic ciclosporin and tacrolimus in dermatology. Dermatol Ther 20, 239–250.

Man, M.-Q., Hatano, Y., Lee, S.H., Man, M., Chang, S., Feingold, K.R., Leung, D.Y.M., Holleran, W., Uchida, Y., and Elias, P.M. (2008). Characterization of a hapten-induced, murine model with multiple features of atopic dermatitis: structural, immunologic, and biochemical changes following single versus multiple oxazolone challenges. J Invest Dermatol 128, 79–86.

McCoy, E.S., Taylor-Blake, B., and Zylka, M.J. (2012). CGRPα-expressing sensory neurons respond to stimuli that evoke sensations of pain and itch. PLoS ONE 7, e36355.

McCoy, E.S., Taylor-Blake, B., Street, S.E., Pribisko, A.L., Zheng, J., and Zylka, M.J. (2013). Peptidergic CGRPα primary sensory neurons encode heat and itch and tonically suppress sensitivity to cold. Neuron 78, 138–151.

Mishra, S.K., and Hoon, M.A. (2013). The cells and circuitry for itch responses in mice. Science 340, 968–971.

Mitchell, K., Bates, B.D., Keller, J.M., Lopez, M., Scholl, L., Navarro, J., Madian, N., Haspel, G., Nemenov, M.I., and Iadarola, M.J. (2010). Ablation of rat TRPV1-expressing Adelta/C-fibers with resiniferatoxin: analysis of withdrawal behaviors, recovery of function and molecular correlates. Mol Pain 6, 94.

Miyamoto, T., Nojima, H., Shinkado, T., Nakahashi, T., and Kuraishi, Y. (2002). Itch-associated response induced by experimental dry skin in mice. Jpn J Pharmacol 88, 285–292.

Moniaga, C.S., Jeong, S.K., Egawa, G., Nakajima, S., Hara-Chikuma, M., Jeon, J.E., Lee, S.H., Hibino, T., Miyachi, Y., and Kabashima, K. (2013). Protease activity enhances production of thymic stromal lymphopoietin and basophil accumulation in flaky tail mice. Am. J. Pathol. 182, 841–851.

Nakamura, M., Toyoda, M., and Morohashi, M. (2003). Pruritogenic mediators in psoriasis vulgaris: comparative evaluation of itch-associated cutaneous factors. Br J Dermatol 149, 718–730.

Nakamura, Y., Une, Y., Miyano, K., Abe, H., Hisaoka, K., Morioka, N., and Nakata, Y. (2012). Activation of transient receptor potential ankyrin 1 evokes nociception through

Page 59: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

58

substance P release from primary sensory neurons. J Neurochem 120, 1036–1047.

Negulescu, P.A., Shastri, N., and Cahalan, M.D. (1994). Intracellular calcium dependence of gene expression in single T lymphocytes. Proc Natl Acad Sci USA 91, 2873–2877.

Nojima, H., Carstens, M.I., and Carstens, E. (2003). c-fos expression in superficial dorsal horn of cervical spinal cord associated with spontaneous scratching in rats with dry skin. Neurosci Lett 347, 62–64.

Ohsawa, Y., and Hirasawa, N. (2012). The antagonism of histamine H1 and H4 receptors ameliorates chronic allergic dermatitis via anti-pruritic and anti-inflammatory effects in NC/Nga mice. Allergy 67, 1014–1022.

Ong, H.L., Jang, S.-I., and Ambudkar, I.S. (2012). Distinct contributions of Orai1 and TRPC1 to agonist-induced [Ca(2+)](i) signals determine specificity of Ca(2+)-dependent gene expression. PLoS ONE 7, e47146.

Osawa, Y., Koizumi, H., Fukaya, T., Yasui, C., Ohkawara, A., and Ueda, T. (1991). Adenylate cyclase induces intracellular Ca2+ increase in single human epidermal keratinocytes of the epidermal sheet as measured by digital imaging microscopy using Fura 2-AM. Arch Dermatol Res 283, 91–95.

Owsianik, G., Talavera, K., Voets, T., and Nilius, B. (2006). Permeation and selectivity of TRP channels. Annu Rev Physiol 68, 685–717.

Pandey, A., Ozaki, K., Baumann, H., Levin, S.D., Puel, A., Farr, A.G., Ziegler, S.F., Leonard, W.J., and Lodish, H.F. (2000). Cloning of a receptor subunit required for signaling by thymic stromal lymphopoietin. Nat Immunol 1, 59–64.

Papoiu, A.D.P., Coghill, R.C., Kraft, R.A., Wang, H., and Yosipovitch, G. (2012). A tale of two itches. Common features and notable differences in brain activation evoked by cowhage and histamine induced itch. Neuroimage 59, 3611–3623.

Prakriya, M., Feske, S., Gwack, Y., Srikanth, S., Rao, A., and Hogan, P.G. (2006). Orai1 is an essential pore subunit of the CRAC channel. Nature 443, 230–233.

Rae, J., Cooper, K., Gates, P., and Watsky, M. (1991). Low access resistance perforated patch recordings using amphotericin B. J Neurosci Methods 37, 15–26.

Ramsey, I.S., Delling, M., and Clapham, D.E. (2006). An introduction to TRP channels. Annu Rev Physiol 68, 619–647.

Rao, A., Luo, C., and Hogan, P.G. (1997). Transcription factors of the NFAT family: regulation and function. Annu. Rev. Immunol. 15, 707–747.

Reddy, V.B., Iuga, A.O., Shimada, S.G., Lamotte, R.H., and Lerner, E.A. (2008). Cowhage-evoked itch is mediated by a novel cysteine protease: a ligand of protease-

Page 60: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

59

activated receptors. J Neurosci 28, 4331–4335.

Ross, S.E. (2011). Pain and itch: insights into the neural circuits of aversive somatosensation in health and disease. Current Opinion in Neurobiology 21, 880–887.

Ross, S.E., Mardinly, A.R., Mccord, A.E., Zurawski, J., Cohen, S., Jung, C., Hu, L., Mok, S.I., Shah, A., Savner, E.M., et al. (2010). Loss of inhibitory interneurons in the dorsal spinal cord and elevated itch in Bhlhb5 mutant mice. Neuron 65, 886–898.

Saint-Mezard, P., Krasteva, M., Chavagnac, C., Bosset, S., Akiba, H., Kehren, J., Kanitakis, J., Kaiserlian, D., Nicolas, J.F., and Berard, F. (2003). Afferent and efferent phases of allergic contact dermatitis (ACD) can be induced after a single skin contact with haptens: evidence using a mouse model of primary ACD. J Invest Dermatol 120, 641–647.

Salomon, J., and Baran, E. (2008). The role of selected neuropeptides in pathogenesis of atopic dermatitis. J Eur Acad Dermatol Venereol 22, 223–228.

Santulli, R.J., Derian, C.K., Darrow, A.L., Tomko, K.A., Eckardt, A.J., Seiberg, M., Scarborough, R.M., and Andrade-Gordon, P. (1995). Evidence for the presence of a protease-activated receptor distinct from the thrombin receptor in human keratinocytes. Proc Natl Acad Sci USA 92, 9151–9155.

Schechter, N.M., Brass, L.F., Lavker, R.M., and Jensen, P.J. (1998). Reaction of mast cell proteases tryptase and chymase with protease activated receptors (PARs) on keratinocytes and fibroblasts. J Cell Physiol 176, 365–373.

Schmelz, M., Schmidt, R., Bickel, A., Handwerker, H.O., and Torebjörk, H.E. (1997). Specific C-receptors for itch in human skin. J Neurosci 17, 8003–8008.

Schmelz, M., Schmidt, R., Weidner, C., Hilliges, M., Torebjörk, H.E., and Handwerker, H.O. (2003). Chemical response pattern of different classes of C-nociceptors to pruritogens and algogens. J Neurophysiol 89, 2441–2448.

Schmid-Wendtner, M.-H., and Korting, H.C. (2006). The pH of the skin surface and its impact on the barrier function. Skin Pharmacol Physiol 19, 296–302.

Seike, M., Ikeda, M., Kodama, H., Terui, T., and Ohtsu, H. (2005). Inhibition of scratching behaviour caused by contact dermatitis in histidine decarboxylase gene knockout mice. Exp Dermatol 14, 169–175.

Shim, W.-S., Tak, M.-H., Lee, M.-H., Kim, M., Kim, M., Koo, J.-Y., Lee, C.-H., Kim, M., and Oh, U. (2007). TRPV1 mediates histamine-induced itching via the activation of phospholipase A2 and 12-lipoxygenase. J Neurosci 27, 2331–2337.

Shimada, S.G., and Lamotte, R.H. (2008). Behavioral differentiation between itch and pain in mouse. Pain 139, 681–687.

Page 61: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

60

Sikand, P., Shimada, S.G., Green, B.G., and Lamotte, R.H. (2009). Similar itch and nociceptive sensations evoked by punctate cutaneous application of capsaicin, histamine and cowhage. Pain 144, 66–75.

Sikand, P., Shimada, S.G., Green, B.G., and Lamotte, R.H. (2011). Sensory responses to injection and punctate application of capsaicin and histamine to the skin. Pain 152, 2485–2494.

Sonkoly, E., Muller, A., Lauerma, A.I., Pivarcsi, A., Soto, H., Kemény, L., Alenius, H., Dieu-Nosjean, M.-C., Meller, S., Rieker, J., et al. (2006). IL-31: a new link between T cells and pruritus in atopic skin inflammation. J Allergy Clin Immunol 117, 411–417.

Spergel, J.M., and Paller, A.S. (2003). Atopic dermatitis and the atopic march. J Allergy Clin Immunol 112, S118–S127.

Ständer, S., and Steinhoff, M. (2002). Pathophysiology of pruritus in atopic dermatitis: an overview. Exp Dermatol 11, 12–24.

Ständer, S., Weisshaar, E., Mettang, T., Szepietowski, J.C., Carstens, E., Ikoma, A., Bergasa, N.V., Gieler, U., Misery, L., Wallengren, J., et al. (2007). Clinical classification of itch: a position paper of the International Forum for the Study of Itch. Acta Derm Venereol 87, 291–294.

Steinhoff, M., Neisius, U., Ikoma, A., Fartasch, M., Heyer, G., Skov, P.S., Luger, T.A., and Schmelz, M. (2003). Proteinase-activated receptor-2 mediates itch: a novel pathway for pruritus in human skin. J Neurosci 23, 6176–6180.

Story, G.M., Peier, A.M., Reeve, A.J., Eid, S.R., Mosbacher, J., Hricik, T.R., Earley, T.J., Hergarden, A.C., Andersson, D.A., Hwang, S.W., et al. (2003). ANKTM1, a TRP-like channel expressed in nociceptive neurons, is activated by cold temperatures. Cell 112, 819–829.

Sun, Y.-G., and Chen, Z.-F. (2007). A gastrin-releasing peptide receptor mediates the itch sensation in the spinal cord. Nature 448, 700–703.

Sun, Y.-G., Zhao, Z.-Q., Meng, X.-L., Yin, J., Liu, X.-Y., and Chen, Z.-F. (2009). Cellular basis of itch sensation. Science 325, 1531–1534.

Tobin, D., Nabarro, G., Baart de la Faille, H., van Vloten, W.A., van der Putte, S.C., and Schuurman, H.J. (1992). Increased number of immunoreactive nerve fibers in atopic dermatitis. J Allergy Clin Immunol 90, 613–622.

Tominaga, M.M., Ogawa, H.H., and Takamori, K.K. (2009a). Histological characterization of cutaneous nerve fibers containing gastrin-releasing peptide in NC/Nga mice: an atopic dermatitis model. J Invest Dermatol 129, 2901–2905.

Tominaga, M., Ozawa, S., Tengara, S., Ogawa, H., and Takamori, K. (2007). Intraepidermal nerve fibers increase in dry skin of acetone-treated mice. J Dermatol Sci

Page 62: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

61

48, 103–111.

Tominaga, M., Tengara, S., Kamo, A., Ogawa, H., and Takamori, K. (2009b). Psoralen-ultraviolet A therapy alters epidermal Sema3A and NGF levels and modulates epidermal innervation in atopic dermatitis. J Dermatol Sci 55, 40–46.

Toyoda, M., Nakamura, M., Makino, T., Hino, T., Kagoura, M., and Morohashi, M. (2002). Nerve growth factor and substance P are useful plasma markers of disease activity in atopic dermatitis. Br J Dermatol 147, 71–79.

Ui, H., Andoh, T., Lee, J.-B., Nojima, H., and Kuraishi, Y. (2006). Potent pruritogenic action of tryptase mediated by PAR-2 receptor and its involvement in anti-pruritic effect of nafamostat mesilate in mice. Eur J Pharmacol 530, 172–178.

Urashima, R., and Mihara, M. (1998). Cutaneous nerves in atopic dermatitis. A histological, immunohistochemical and electron microscopic study. Virchows Arch 432, 363–370.

Vellani, V., Kinsey, A.M., Prandini, M., Hechtfischer, S.C., Reeh, P., Magherini, P.C., Giacomoni, C., and McNaughton, P.A. (2010). Protease activated receptors 1 and 4 sensitize TRPV1 in nociceptive neurones. Mol Pain 6, 61.

Wilson, S.R., Gerhold, K.A., Bifolck-Fisher, A., Liu, Q., Patel, K.N., Dong, X., and Bautista, D.M. (2011). TRPA1 is required for histamine-independent, Mas-related G protein-coupled receptor-mediated itch. Nat Neurosci 14, 595–602.

Wilson, S.R., Nelson, A.M., Batia, L., Morita, T., Estandian, D., Owens, D.M., Lumpkin, E.A., and Bautista, D.M. (2013a). The ion channel TRPA1 is required for chronic itch. J Neurosci 33, 9283–9294.

Wilson, S.R., The, L., Batia, L.M., Beattie, K., Katibah, G.E., McClain, S.P., Pellegrino, M., Estandian, D.M., and Bautista, D.M. (2013b). The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell 155, 285–295.

Yamaura, K., Oda, M., Suwa, E., Suzuki, M., Sato, H., and Ueno, K. (2009). Expression of histamine H4 receptor in human epidermal tissues and attenuation of experimental pruritus using H4 receptor antagonist. J Toxicol Sci. 34, 427-31.

Yeromin, A.V., Zhang, S.L., Jiang, W., Yu, Y., Safrina, O., and Cahalan, M.D. (2006). Molecular identification of the CRAC channel by altered ion selectivity in a mutant of Orai. Nature 443, 226–229.

Ying, S., O'Connor, B., Ratoff, J., Meng, Q., Mallett, K., Cousins, D., Robinson, D., Zhang, G., Zhao, J., Lee, T.H., et al. (2005). Thymic stromal lymphopoietin expression is increased in asthmatic airways and correlates with expression of Th2-attracting chemokines and disease severity. J. Immunol. 174, 8183–8190.

Yohn, J.J., Smith, C., Stevens, T., Morelli, J.G., Shurnas, L.R., Walchak, S.J., Hoffman,

Page 63: Lydia The - thesis · Lydia The Doctor of Philosophy Molecular and Cell Biology University of California, Berkeley Professor Diana Bautista, Chair There is accumulating evidence suggesting

 

62

T.A., Kelley, K.K., Escobedo-Morse, A., and Yanagisawa, M. (1994). Autoregulation of endothelin-1 secretion by cultured human keratinocytes via the endothelin B receptor. Biochim Biophys Acta 1224, 454–458.

Yoo, J., Omori, M., Gyarmati, D., Zhou, B., Aye, T., Brewer, A., Comeau, M.R., Campbell, D.J., and Ziegler, S.F. (2005). Spontaneous atopic dermatitis in mice expressing an inducible thymic stromal lymphopoietin transgene specifically in the skin. J. Exp. Med. 202, 541–549.

Yosipovitch, G., Duque, M.I., Fast, K., Dawn, A.G., and Coghill, R.C. (2007). Scratching and noxious heat stimuli inhibit itch in humans: a psychophysical study. Br J Dermatol 156, 629–634.

Yosipovitch, G., and Papoiu, A.D.P. (2008). What causes itch in atopic dermatitis? Curr Allergy Asthma Rep 8, 306–311.

Zhang, X.-F., Chen, J., Faltynek, C.R., Moreland, R.B., and Neelands, T.R. (2008). Transient receptor potential A1 mediates an osmotically activated ion channel. Eur J Neurosci 27, 605–611.

Zhao, Z.-Q., Huo, F.-Q., Jeffry, J., Hampton, L., Demehri, S., Kim, S., Liu, X.-Y., Barry, D.M., Wan, L., Liu, Z.-C., et al. (2013). Chronic itch development in sensory neurons requires BRAF signaling pathways. J Clin Invest.

Zhu, Y., Wang, X.R., Peng, C., Xu, J.G., Liu, Y.X., Wu, L., Zhu, Q.G., Liu, J.Y., Li, F.Q., Pan, Y.H., et al. (2009). Induction of leukotriene B(4) and prostaglandin E(2) release from keratinocytes by protease-activated receptor-2-activating peptide in ICR mice. Int Immunopharmacol 9, 1332–1336.

Ziegler, S.F., Roan, F., Bell, B.D., Stoklasek, T.A., Kitajima, M., and Han, H. (2013). The biology of thymic stromal lymphopoietin (TSLP). Adv. Pharmacol. 66, 129–155.

Zylka, M.J., Rice, F.L., and Anderson, D.J. (2005). Topographically distinct epidermal nociceptive circuits revealed by axonal tracers targeted to Mrgprd. Neuron 45, 17–25.