intrathecal administration of proteinase-activated receptor-2 … · 2019. 5. 8. · intrathecal...

10
Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by Exciting the Cell Bodies of Primary Sensory Neurons Kwai A. Alier, Jaclyn A. Endicott, Patrick L. Stemkowski, Nicolas Cenac, Laurie Cellars, Kevin Chapman, Patricia Andrade-Gordon, Nathalie Vergnolle, and Peter A. Smith Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (K.A.A., J.A.E., P.L.S., P.A.S.); Department of Pharmacology and Therapeutics, University of Calgary, Calgary, Alberta, Canada (N.C., L.C., K.C., N.V.); and Johnson & Johnson Pharmaceutical Research and Development, Spring House, Pennsylvania (P.A.-G.) Received July 24, 2007; accepted October 3, 2007 ABSTRACT Proteinase-activated receptors (PARs) are a family of G-pro- tein-coupled receptors that are activated by endogenous serine proteinases that cleave the N-terminal domain of the receptor unmasking a “tethered ligand” sequence. Trypsin and other agonists at PAR 2 act on peripheral nerves to augment the transfer of nociceptive information. We tested whether PAR 2 agonists also exert a spinal pronociceptive effect by i.t. admin- istering the selective ligand, Ser-Leu-Ile-Gly-Arg-Leu-NH 2 (SLI- GRL). This produced thermal and mechanical hyperalgesia in rats and mice and augmented mechanical and thermal hyper- algesia seen in the formalin inflammatory pain test. Effects of SLIGRL were abrogated in PAR 2 -deficient mice and were not seen with the inactive control peptide, Leu-Arg-Gly-Ile- Leu-Ser-NH 2 . Surprisingly, electrophysiological studies, using whole-cell recording from rat substantia gelatinosa neurons, failed to demonstrate an increase in excitatory transmission or neuronal excitability following treatment with SLIGRL or trypsin. In fact, the actions of trypsin were consistent with a decrease in dorsal horn excitability. SLIGRL and trypsin did, however, de- polarize and increase the excitability of large, medium and small primary afferent, dorsal root ganglion neurons. The ef- fects were associated with an increase in conductance at hy- perpolarized potentials and a decrease in conductance at de- polarized potentials. PAR 2 -like immunoreactivity was found in DRG but not in spinal dorsal horn. These results suggest that activation of DRG neuron cell bodies may account for the pronociceptive actions of i.t. applied PAR 2 agonists. They also imply that pathophysiological release of PAR 2 -activating pro- teases in the vicinity of DRG neurons may produce profound effects on nociceptive processing in vivo. Proteinase-activated receptors (PARs) are a novel family of G-protein-coupled receptors (Hoogerwerf et al., 2001; Stein- hoff et al., 2005) that are found in various tissues throughout the body, including the central and peripheral nervous sys- tems (Noorbakhsh et al., 2003; Vergnolle et al., 2003; Stein- hoff et al., 2005). They have been implicated in the activation and/or modulation of nociceptive pathways (Vergnolle et al., 2001; Fiorucci and Distrutti, 2002; Cenac and Vergnolle, 2005). PARs are activated by endogenous serine proteinases that cleave the N-terminal domain of the receptor unmasking a “tethered ligand” sequence (Hoogerwerf et al., 2001; Vergnolle et al., 2003; Steinhoff et al., 2005). This tethered ligand subsequently binds to an active site within the recep- tor leading to activation and a downstream response (Hoogerwerf et al., 2001; Steinhoff et al., 2005). Activating enzymes are derived from cellular sources (trypsin and mast cell tryptase) as well as humoral sources (thrombin and fac- tor Xa) and are released or generated during trauma and/or inflammation (Hoogerwerf et al., 2001). Trypsin may also be released from microglia following traumatic damage to the central nervous system (Noorbakhsh et al., 2003). So far, four types of PAR receptors, PAR 1 , PAR 2 , PAR 3 , and PAR 4 , have been identified (Hollenberg, 1999; Steinhoff et al., 2005). These can be distinguished functionally on the basis of the agonist effectiveness of various serine proteases. For exam- ple, thrombin selectively activates PAR 1 but not PAR 2 , whereas PAR 1 is less sensitive to trypsin than PAR 2 (for review, see Vergnolle et al., 2003). Moreover, synthetic pep- This work was supported by the Canadian Institutes of Health Research. Article, publication date, and citation information can be found at http://jpet.aspetjournals.org. doi:10.1124/jpet.107.129171. ABBREVIATIONS: PAR, proteinase-activated receptor; SLIGRL, PAR 2 ligand (SLIGRL-NH 2 ; Ser-Leu-Ile-Gly-Arg-Leu-NH 2 ); SP, substance P; LRGILS, reverse sequence (control) PAR 2 ligand (LRGILS-NH 2 ; Leu-Arg-Gly-Ile-Leu-Ser-NH 2 ); DRG, dorsal root ganglion; DMEM, Dulbecco’s modified Eagle’s medium; AP, action potential; EPSC, excitatory postsynaptic current; sEPSC, spontaneous EPSC; PBS, phosphate-buffered saline; TRPV, transient receptor potential vanilloid; I-V, current-voltage. 0022-3565/08/3241-224–233$20.00 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 324, No. 1 Copyright © 2008 by The American Society for Pharmacology and Experimental Therapeutics 129171/3285095 JPET 324:224–233, 2008 Printed in U.S.A. 224 at ASPET Journals on May 8, 2019 jpet.aspetjournals.org Downloaded from

Upload: others

Post on 15-Mar-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

Intrathecal Administration of Proteinase-Activated Receptor-2Agonists Produces Hyperalgesia by Exciting the Cell Bodiesof Primary Sensory Neurons

Kwai A. Alier, Jaclyn A. Endicott, Patrick L. Stemkowski, Nicolas Cenac, Laurie Cellars,Kevin Chapman, Patricia Andrade-Gordon, Nathalie Vergnolle, and Peter A. SmithDepartment of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (K.A.A., J.A.E., P.L.S., P.A.S.); Department ofPharmacology and Therapeutics, University of Calgary, Calgary, Alberta, Canada (N.C., L.C., K.C., N.V.); andJohnson & Johnson Pharmaceutical Research and Development, Spring House, Pennsylvania (P.A.-G.)

Received July 24, 2007; accepted October 3, 2007

ABSTRACTProteinase-activated receptors (PARs) are a family of G-pro-tein-coupled receptors that are activated by endogenous serineproteinases that cleave the N-terminal domain of the receptorunmasking a “tethered ligand” sequence. Trypsin and otheragonists at PAR2 act on peripheral nerves to augment thetransfer of nociceptive information. We tested whether PAR2agonists also exert a spinal pronociceptive effect by i.t. admin-istering the selective ligand, Ser-Leu-Ile-Gly-Arg-Leu-NH2 (SLI-GRL). This produced thermal and mechanical hyperalgesia inrats and mice and augmented mechanical and thermal hyper-algesia seen in the formalin inflammatory pain test. Effectsof SLIGRL were abrogated in PAR2-deficient mice and werenot seen with the inactive control peptide, Leu-Arg-Gly-Ile-Leu-Ser-NH2. Surprisingly, electrophysiological studies, usingwhole-cell recording from rat substantia gelatinosa neurons,

failed to demonstrate an increase in excitatory transmission orneuronal excitability following treatment with SLIGRL or trypsin.In fact, the actions of trypsin were consistent with a decrease indorsal horn excitability. SLIGRL and trypsin did, however, de-polarize and increase the excitability of large, medium andsmall primary afferent, dorsal root ganglion neurons. The ef-fects were associated with an increase in conductance at hy-perpolarized potentials and a decrease in conductance at de-polarized potentials. PAR2-like immunoreactivity was found inDRG but not in spinal dorsal horn. These results suggest thatactivation of DRG neuron cell bodies may account for thepronociceptive actions of i.t. applied PAR2 agonists. They alsoimply that pathophysiological release of PAR2-activating pro-teases in the vicinity of DRG neurons may produce profoundeffects on nociceptive processing in vivo.

Proteinase-activated receptors (PARs) are a novel family ofG-protein-coupled receptors (Hoogerwerf et al., 2001; Stein-hoff et al., 2005) that are found in various tissues throughoutthe body, including the central and peripheral nervous sys-tems (Noorbakhsh et al., 2003; Vergnolle et al., 2003; Stein-hoff et al., 2005). They have been implicated in the activationand/or modulation of nociceptive pathways (Vergnolle et al.,2001; Fiorucci and Distrutti, 2002; Cenac and Vergnolle,2005). PARs are activated by endogenous serine proteinasesthat cleave the N-terminal domain of the receptor unmaskinga “tethered ligand” sequence (Hoogerwerf et al., 2001;Vergnolle et al., 2003; Steinhoff et al., 2005). This tethered

ligand subsequently binds to an active site within the recep-tor leading to activation and a downstream response(Hoogerwerf et al., 2001; Steinhoff et al., 2005). Activatingenzymes are derived from cellular sources (trypsin and mastcell tryptase) as well as humoral sources (thrombin and fac-tor Xa) and are released or generated during trauma and/orinflammation (Hoogerwerf et al., 2001). Trypsin may also bereleased from microglia following traumatic damage to thecentral nervous system (Noorbakhsh et al., 2003). So far, fourtypes of PAR receptors, PAR1, PAR2, PAR3, and PAR4, havebeen identified (Hollenberg, 1999; Steinhoff et al., 2005).These can be distinguished functionally on the basis of theagonist effectiveness of various serine proteases. For exam-ple, thrombin selectively activates PAR1 but not PAR2,whereas PAR1 is less sensitive to trypsin than PAR2 (forreview, see Vergnolle et al., 2003). Moreover, synthetic pep-

This work was supported by the Canadian Institutes of Health Research.Article, publication date, and citation information can be found at

http://jpet.aspetjournals.org.doi:10.1124/jpet.107.129171.

ABBREVIATIONS: PAR, proteinase-activated receptor; SLIGRL, PAR2 ligand (SLIGRL-NH2; Ser-Leu-Ile-Gly-Arg-Leu-NH2); SP, substance P;LRGILS, reverse sequence (control) PAR2 ligand (LRGILS-NH2; Leu-Arg-Gly-Ile-Leu-Ser-NH2); DRG, dorsal root ganglion; DMEM, Dulbecco’smodified Eagle’s medium; AP, action potential; EPSC, excitatory postsynaptic current; sEPSC, spontaneous EPSC; PBS, phosphate-bufferedsaline; TRPV, transient receptor potential vanilloid; I-V, current-voltage.

0022-3565/08/3241-224–233$20.00THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 324, No. 1Copyright © 2008 by The American Society for Pharmacology and Experimental Therapeutics 129171/3285095JPET 324:224–233, 2008 Printed in U.S.A.

224

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 2: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

tides that mimic the structure of the tethered ligand alsospecifically activate various PAR subtypes. For example, Ser-Leu-Ile-Gly-Arg-Leu-NH2 (SLIGRL) selectively activatesPAR2 (Vergnolle et al., 2001).

Intraplantar injection of PAR2 agonists in rats causesmarked and sustained hyperalgesia. This and other workstrongly implicates PAR2 in peripheral inflammatory pain(Hoogerwerf et al., 2001; Vergnolle et al., 2001; Fiorucci andDistrutti, 2002; Dai et al., 2004). Although PAR2 agonistshave also been reported to release substance P (SP) andcalcitonin gene-related peptide from peripheral and spinalcord tissues (Vergnolle et al., 2001) and to augment capsa-icin-induced neuropeptide release in spinal cord (Amadesi etal., 2004), the possibility that PAR2 agonists act directlywithin the spinal cord to produce pronociceptive effects re-mains to be established. We have therefore directly appliedPAR2 agonists to the spinal cord via the i.t. route in vivo andexamined their effects in various pain models. We have usedwhole-cell recording from DRG and dorsal horn (substantiagelatinosa) neurons to elucidate the underlying mechanismof the effects that we observed.

Materials and MethodsAll experimental protocols were approved by the Health Sciences

Animal Welfare Committee of the University of Alberta and theAnimal Care Committee of the University of Calgary. They wereperformed in accordance with the guidelines established by the Ca-nadian Council on Animal Care and the Committee for Research andEthical Issues and are in concordance with the guidelines of theInternational Association for the Study of Pain.

Behavioral Experiments: Intrathecal and Intraplantar In-jections. Male Wistar rats (175–200 g) were obtained from CharlesRiver Laboratories (Montreal, Quebec, Canada). The rats had freeaccess to food and water and were housed under constant tempera-ture and light cycle. PAR2-deficient mice and wild-type littermatesused in the control experiments were originally obtained from John-son and Johnson Pharmaceutical (Springhouse, PA). Both were bredat the University of Calgary Animal Care Facility and maintained asstable lines. Genotyping of animals was undertaken on a regularbasis (three to four times per year) to verify their genetic profile.C57BL6 mice were purchased from Charles River Laboratories forthe formalin experiments.

The PAR2-activating peptide, SLIGRL, or the control peptide,Leu-Arg-Gly-Ile-Leu-Ser-NH2 (LRGILS), were administered i.t. asdescribed previously (Vergnolle et al., 2001) at the L4 to L5 region, ina total volume of 5 �l. For i.t. injections, the animals were held by thepelvic girdle, and a 25-gauge needle connected to a 25-�l Hamiltonsyringe was inserted between L4 and L5, into the subarachnoidalspace. The placement of the needle was verified by the elicitation ofa tail-flick movement. The syringe was kept in place for few secondsafter the injection.

In animals treated with formalin, the i.t. injections of peptides orsaline were performed 10 min before the intraplantar injection offormalin. Formalin (5%) was diluted in saline and injected into themouse hindpaw in a total volume of 10 �l as described previously(Vergnolle et al., 2001).

Behavioral Experiments: Measures of Nociception. Nocicep-tive responses to a thermal stimulus were evaluated in rats andmice, using a plantar test apparatus (Stoelting, Chicago, IL), mea-suring paw withdrawal latency in response to a radiant heat stim-ulus. Nociceptive responses to a mechanical stimulus were evaluatedin rats hindpaws, using a Ugo Basile (Milan, Italy) analgesy meter,measuring nociceptive flexion reflex in response to increasing weight(Asfaha et al., 2002). In mice, mechanical nociception was evaluatedusing three von Frey filaments of different size and bending forces as

described previously (Amadesi et al., 2004). The filaments wereapplied for 1 to 2 s, a minimum of three times randomly among thetested animals. A score was assigned based on the animal’s response:0, no movement; 1, removal of the paw; and 2, removal of the paw andvocalization, licking or holding of the paw. Mechanical nociceptivescore was expressed as a percentage of the maximal score that wouldbe obtained for the number of tests at each time-point. Before allexperiments, baseline measures of nociception were recorded (time0) for all assays, and nociception was then measured again at differ-ent time-points after treatments.

Thermal and mechanical hyperalgesia were defined as a signifi-cant increase in withdrawal latency and nociceptive threshold orscore, respectively. Mechanical allodynia was observed in mice whennociceptive score values � 0 were recorded using the smallest sizevon Frey filament (3.61), which under basal conditions did not pro-voke any nociceptive response.

Electrophysiological Experiments on Dorsal Root Gan-glion Neurons. Sprague-Dawley rats, aged 18 to 26 days old, wereanesthetized with 1.5 g/kg i.p. urethane. Laminectomy was per-formed under sterile conditions with carbogen-saturated dissectionsolution. The fourth, fifth, and sixth lumbar dorsal root ganglions(DRGs) were harvested with the aid of a dissection microscope.Dissection solution used throughout the DRG removal process con-tained 118 mM NaCl, 2.5 mM KCl, 1.3 mM MgSO4, 1.2 mMNaH2PO4, 5 mM MgCl2, 25 mM D-glucose, 26 mM NaHCO3, and 1.5mM CaCl2. DRGs were then enzymatically dissociated while shakingin a water bath at 35°C for 40 min. Digestion media was preparedin 6 ml of Dulbecco’s modified Eagle’s medium (DMEM) withGlutaMAX (Invitrogen, Carlsbad, CA) containing 0.5 mg/ml trypsin(Sigma), 1 mg/ml collagenase (Sigma), and 0.1 mg/ml deoxyribonu-clease I (Sigma). Soybean trypsin inhibitor (Worthington Biochemi-cals, Lakewood, NJ), at a concentration high enough to neutralize 2times the amount of trypsin present in the digestion medium, wasdissolved in 1.5 ml of DMEM and added to the digestion mediumafter the 40 min of incubation. After trituration, the dissociated cellswere plated in sterile 3.5-cm polystyrene culture dishes (Nalge NuncInternational, Rochester, NY). Cells were allowed to adhere to cul-ture dishes for 2 min before 2 ml of culture medium was added toeach dish. The culture medium used was made up in DMEM, includ-ing: 10% fetal bovine serum (Invitrogen), 10% horse serum (Invitro-gen), 100 units/ml penicillin (Invitrogen), 100 �g/ml streptomycin(Invitrogen), and 5 ng/ml mouse nerve growth factor 2.5S (AlomoneLabs, Jerusalem, Israel). Culture medium was changed every 48 h.Experiments were done at room temperature (20°C) on neurons thathad been maintained for 2 to 4 days in a tissue culture incubator at37.0°C with 6% CO2. Standard whole-cell recordings were madeusing an Axoclamp 2A amplifier in current-clamp mode or an Axo-patch 1B amplifier in voltage-clamp mode. Borosilicate glass patchelectrodes had a DC resistance of 4 to 6 M� for action potential (AP)recordings. Data were acquired and analyzed using pClamp 5.5software, and final figures were produced using Origin 7.0 (Origin-Lab Corporation, Northampton, MA). The cells were continuouslysuperfused with carbogen-saturated extracellular solution. For allrecordings, the external solution contained the following: 127 mMNaCl, 2.5 mM KCl, 1.2 mM NaH2PO4, 1.3 mM MgSO4, 26 mMNaHCO3, 25 mM D-glucose, and 2.5 mM CaCl2. Internal pipettesolution contained: 130 mM D-gluconic acid, 10 mM EGTA, 4 mMMg-ATP, 0.3 mM Na-GTP, 10 mM HEPES, and 2 mM CaCl2. The pHwas adjusted to 7.2 with 1 M KOH. Single action potentials weregenerated using 20-ms depolarizing current pulses. In current-clamprecordings, changes in conductance were measured by means of200-ms, 0.1-nA hyperpolarizing current pulses applied through therecording electrode at 2 Hz. Conductance changes in voltage clampwere measured from I-V relationships derived from slow ramp com-mands from �110 to �40mV. Excitability was monitored by injectingdepolarizing current ramps and observing the latency to discharge ofthe first action potential on the ramp. Drugs were applied by a rapidsuperfusion system constructed from 0.8-mm-diameter polyamide

PAR2 and Pain 225

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 3: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

tubes, and flow was controlled by solenoid valves. The applicator tipwas positioned within 100 �m of the neuronal cell under study. Eachdrug was applied for 30 s.

Electrophysiological Experiments on Spinal Cord Slices.These were carried out according to our previously published meth-ods (Moran et al., 2004; Balasubramanyan et al., 2006). In brief,male Sprague-Dawley rats (15–42 days old) were deeply anesthe-tized with urethane (1.5 g/kg i.p.), and a dorsal incision was madefrom the base of the tail to the skull. The incision site was immedi-ately irrigated with �2 ml of ice-cold oxygenated (95% O2-5% CO2)dissection solution containing 118 mM NaCl, 2.5 mM KCl, 26 mMNaHCO3, 1.3 mM MgSO4, 1.2 mM NaH2PO4, 1.5 mM CaCl2, 5 mMMgCl2, 25 mM D-glucose, and 1 mM kynurenic acid. A laminectomywas performed extending from the thoracic to the sacral portions ofthe spinal cord. The spinal cord was removed from the vertebralcolumn, and the ventral roots and all dorsal roots except L4 to L5were cut. Transverse slices (300–500 �m) were cut using a Vi-bratome (TPI, St. Louis, MO). Slices were incubated in a holdingchamber (Sakmann and Stuart, 1995) at 36°C for 1 h and subse-quently stored at room temperature (�22°C) in oxygenated dissec-tion solution (see above, without 1 mM kynurenic acid).

Slices were placed in a circular glass-bottomed recording chamberand held in place with a U-shaped platinum wire frame with at-tached parallel nylon fibers. They were superfused at room temper-ature (�22°C) with 95% O2-5% CO2 saturated artificial cerebrospi-nal fluid, which contained 127 mM NaCl, 2.5 mM KCl, 1.2 mMNaH2PO4, 26 mM NaHCO3, 1.3 mM MgSO4, 2.5 mM CaCl2, and 25mM D-glucose, pH 7.4. Spinal cord slices were viewed with a ZeissAxioskop FS upright microscope (Carl Zeiss GmbH, Jena, Germany)equipped with infrared differential interference contrast optics.Whole-cell recordings from visually identified substantia gelatinosaneurons were made with an NPI SEC 05L amplifier (NPI Electronic,Tamm, Germany) in discontinuous single-electrode voltage-clamp orbridge-balance current-clamp mode. Switching frequencies were typ-ically between 30 and 40 kHz. Signals were digitized between 5 and10 kHz and filtered at 1 to 2 kHz. Patch pipettes were pulled fromthin-walled borosilicate glass (TW-150F-4; WPI, Sarasota, FL). Pi-pettes for recording APs had resistances of 5 to 10 M� when filledwith an internal solution containing 130 mM potassium gluconate, 1mM MgCl2, 2 mM CaCl2, 10 mM HEPES, 10 mM EGTA, 4 mMMg-ATP, and 0.3 mM Na-GTP, pH 7.2 (290–300 mOsm). For someexperiments recording synaptic currents, a Cs�-based internal solu-tion containing 140 mM CsCl, 5 mM HEPES, 10 mM EGTA, 2 mMCaCl2, 2 mM Mg-ATP, and 0.3 mM Na-GTP, pH 7.2 (290–300mOsm) was used.

Data for I-V plots were obtained in voltage-clamp mode usingincremental 10-mV, 800-ms commands. Current values were mea-sured from the close-to-steady-state values attained at the end ofthese commands. As in DRG experiments, the excitability of sub-stantia gelatinosa neurons was monitored by injecting depolarizingcurrent ramps (60 pA/s) and observing the latency to discharge of thefirst action potential on the ramp (see also Balasubramanyan et al.,2006).

Spontaneous EPSCs (sEPSCs) were recorded at �70 mV, digitizedat 5 kHz, and filtered at 1 kHz. Data were acquired for 3 min before,and 5 or 10 min after, superfusion of trypsin or peptides. MiniAnalysis Program software (Synaptosoft, Decatur, GA) was used todistinguish sEPSCs and spontaneous inhibitory postsynaptic cur-rents from the baseline noise and to prepare cumulative probabilityplots or frequency histograms. Data were only included in the anal-ysis if the series resistance was below 25 M� and did not change by�20% during the course of an experiment. Spontaneous postsynapticcurrents were detected automatically using an amplitude thresholdof 10 pA and an area threshold of 15 femtocoloumbs. All detectedevents were then re-examined and visually accepted or rejectedbased on subjective visual examination. The Mini Analysis Programwas used to further analyze the data and to generate figures. Ap-proximately 50 events were used for analysis before and after agonist

superfusion. Whenever possible, the first 50 events after 1 min ofacquisition were used. In some cells, however, spontaneous activitywas limited, so the whole 3-min acquisition period was used toprovide data.

Immunohistochemistry. To localize PAR2 in DRG and spinalcord sections, rats were anesthetized with sodium pentobarbital (200mg/kg i.p.) and transcardially perfused with 4% paraformaldehyde in100 mM PBS, pH 7.4. Spinal cord and DRG (T10-L5) were fixedovernight at 4°C, placed in 25% sucrose in PBS for 24 h at 4°C,embedded in ornithine carbamyl transferase compound (Miles,Elkhart, IN), and sectioned at 25 �m. A section of DRG neurons andspinal cord was adjacently fixed on each slide. Sections were washedin PBS containing 1 to 10% normal growth serum, 1% bovine serumalbumin, and 0.3 to 0.5% Triton X-100 and incubated with primaryantibodies to PAR2 (goat; Santa Cruz Biotechnology, Santa Cruz,CA, 1:200; or B5 antibody, 1:200, a generous gift from Dr. M.D.Hollenberg, University of Calgary) overnight at 4°C. Sections werewashed and incubated with secondary antibodies conjugated to Al-exa Fluor 488 (Molecular Probes, Invitrogen Canada Inc., Burling-ton, ON, Canada; 1:1000, room temperature, 2 h). Sections werewashed and mounted in Prolong (Molecular Probes, Eugene, OR). Asa control for PAR2 specificity, the primary antiserum was preincu-bated with the peptide used for immunization (10 �M) for 24 h at 4°Cbefore staining. Confocal images were acquired using a Zeiss LSM-510 META confocal microscope using 10� and 20� objectives in theinverted configuration. For all confocal images, a regular phasetransmission image was obtained. Images of stained and control cellswere collected and processed identically.

Drugs and Chemicals. The PAR2-activating peptide, SLIGRL,and the inactive control peptide, LRGILS, were obtained from thePeptide Synthesis Facility of the University of Calgary (Calgary,Alberta, Canada). The composition and purity of peptides wereconfirmed by HPLC analysis. Formalin was purchased from VWR(Mississauga, ON, Canada). All other chemicals were from Sigma(Oakville, ON, Canada). SLIGRL and LRGILS were dissolved inexternal solution that contained 0.1% bovine albumin (Sigma). Tryp-sin was also dissolved in external solution and molarities calculatedassuming a molecular mass of 24 kDa.

Statistics. All data are presented as mean � S.E.M. Statisticalsignificance in electrophysiological experiments was assessed by theStudent’s unpaired t test after passing a normality test. The Kol-mogorov-Smirnov two-sample test was used to compare distributionsof amplitudes and interevent intervals in neurons in acute slices.The withdrawal latency, nociceptive threshold, and nociceptive scoredata were analyzed using a one-way analysis of variance followed bya Dunnett’s test. With all statistical analyses, distributions wereconsidered statistically significant if p � 0.05

ResultsIntrathecal Injection of PAR2 Agonists Causes Allo-

dynia and Hyperalgesia. Intrathecal injection of the PAR2

agonist SLIGRL, but not the control peptide LRGILS, causeda decrease in withdrawal latency in response to a thermalstimulus, characteristic of thermal hyperalgesia both in rats(Fig. 1A) and in mice (Fig. 2A). Likewise, i.t. injection ofSLIGRL, but not LRGILS, caused a significant decrease innociceptive threshold in rats (Fig. 1B) and a significant in-crease in nociceptive score in mice (Fig. 2, B and C). Thiseffect of the PAR2 agonist is characteristic of mechanicalhyperalgesia. Although mice did not respond to the smallestsize von Frey filament (3.61) in basal conditions, the PAR2

agonist SLIGRL, but not the control peptide LRGILS, in-jected i.t., caused a significant increase in nociceptive scorecharacteristic of mechanical allodynia (Fig. 2B). The effectsof SLIGRL on withdrawal latency and nociceptive score were

226 Alier et al.

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 4: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

completely abolished in PAR2-deficient mice, demonstratingthe specificity of SLIGRL as a PAR2 agonist (Fig. 2, A–C).

Intrathecal Injection of PAR2 Agonists Enhances Pe-ripherally Induced Inflammatory Allodynia and Hy-peralgesia. Intraplantar injection of formalin caused ther-mal and mechanical hyperalgesia characterized by adecrease in withdrawal latency in response to a thermalstimulus compared with basal measurements (Fig. 3A) andan increased nociceptive score in response to von Frey fila-ment application (Fig. 3C). In addition, formalin intraplantarinjection caused mechanical allodynia, as measured by anociceptive response to the smallest size von Frey filament(3.61) (Fig. 3B). Both thermal and mechanical hyperalgesiaas well as mechanical allodynia induced by formalin wereenhanced by i.t. injection of the PAR2 agonist SLIGRL com-pared with the effect of the control peptide LRGILS, as ob-served by a larger decrease in withdrawal latency and en-hanced nociceptive scores to von Frey filament exposure(Fig. 3).

Effects of Trypsin and PAR2 Agonists on SubstantiaGelatinosa Neurons. To understand the mechanismwhereby i.t. applied PAR2 agonists facilitate nociception, weexamined the effects of trypsin, SLIGRL, and LRGILS on theelectrophysiological properties of second order sensory neu-rons in the substantia gelatinosa. There were no obviouseffects of SLIGRL on the I-V relationship of substantia ge-latinosa neurons. Because PAR2 resides on DRG cell bodiesand on the peripheral sensory nerve endings (Steinhoff et al.,2000), it has been suggested that it also exists on primaryafferent terminals (Fiorucci and Distrutti, 2002). We there-fore examined the actions of PAR2 agonists on sEPSCs. Thefirst series of experiments was done with Cs�-filled pipettesso as to maximize sEPSC amplitude (Moran et al., 2004).Trypsin (21 �M) affected 11 of 15 cells examined (Table 1),but its dominant effect was to decrease both sEPSC ampli-tude and frequency (Fig. 4, A–C). These effects, which weresustained for at least 10 min, could not be attributed todeterioration of recording conditions because sEPSC fre-quency and amplitude were sustained in the absence of tryp-sin (Fig. 4, D and E). SLIGRL (10 �M) also reduced sEPSCamplitude in two of five cells examined but had less effect onsEPSC frequency (Fig. 4, F and G). However, the effect ofSLIGRL on sEPSC amplitude was also seen with the controlcompound, LRGILS, which also decreased SEPSC frequency(Fig. 4, H and I).

We also considered the possibility that trypsin and SLI-GRL may exert different effects on different types of substan-tia gelatinosa neuron. Substantia gelatinosa neurons may beclassified according to their discharge pattern in responseto depolarizing current pulses as “tonic,” “phasic,” “delay,”“transient,” and “irregular” neurons (Balasubramanyan etal., 2006). The need to classify neurons on the basis of theirAP firing pattern precluded the use of Cs�-filled electrodesfor this series of experiments. Many tonic neurons (Fig. 4J)may be GABAergic and inhibitory, whereas delay neurons(Fig. 4K) may be excitatory (Lu and Perl, 2003, 2005). How-

Fig. 1. Withdrawal latency (A) and nociceptive threshold (B) measured inrat hindpaws, before (time 0) and after i.t. injection of 10 �g of SLIGRLor LRGILS. Data are expressed as mean � S.E.M. �, significantly differ-ent from basal latency and threshold measured at time 0, for p � 0.05.

Fig. 2. Withdrawal latency (A) and nociceptive score (B and C) measuredin PAR2-deficient (PAR2

�/�) or wild-type (PAR2�/�) mouse hindpaws,

before (time 0) and after i.t. injection of 1 �g of SLIGRL or LRGILS. Dataare expressed as mean � S.E.M. �, significantly different from LRGILS,for p � 0.05.

PAR2 and Pain 227

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 5: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

ever, SLIGRL or trypsin had little effect on spontaneoussynaptic activity in identified tonic neurons (Table 1) butreduced sEPSC frequency and amplitude in �50% of delaycells. Trypsin failed to alter excitability of delay or tonic cellsbecause first spike latency on a 60 pA/s ramp in the presenceof trypsin was unchanged in both cell types (p � 0.4, n 4 fordelay cells and p � 0.2, n 3 for tonic cells). Typical exper-iments illustrating spike latency experiments are shown inFig. 4, L and M.

Thus, the tendency for trypsin and SLIGRL to depressexcitatory synaptic transmission in substantia gelatinosa aswell as the tendency of trypsin to reduce membrane excit-

ability seem at odds with the observation that i.t. appliedPAR2 agonists have pronociceptive actions (Figs. 1–3). How-ever, i.t. applied drugs likely have access to the DRG (Abramet al., 2006), so we next examined possible excitatory effectsof trypsin, SLIGRL, and LRGILS on the cell bodies of pri-mary afferent neurons.

Classification of DRG Cells. Because there is a reason-able correlation between AP shape and cell size of DRGneurons (Abdulla and Smith, 1997), “large” cells were definedas those with an AP duration �3 ms, “medium” cells hadan AP duration of 3 to 5 ms, and “small” cells had an APduration �5 ms (Fig. 5, A–C). Spike width was measured at50% of the maximum amplitude. Both medium and smallcells displayed a “hump” on the falling phase of their AP,whereas large cells did not.

Effects of Trypsin and SLIGRL on the MembranePotential of DRG Cells. Twenty-eight of 43 (65%) of DRGneurons studied were depolarized in response to 30-s expo-sure to 42 �M trypsin. Responses to trypsin were not con-fined to any one cell type because it affected 16 of 23 largecells, 5 of 10 medium cells, and 7 of 10 small cells. Responseamplitude was 6.8 � 1.2 mV for large cells, 4.7 � 1.3 mV formedium cells, and 5.2 � 1.9 mV for small cells. It is likelythat these responses were supramaximal because similaramplitude depolarizations were seen with much lower con-centrations of trypsin (2.1 �M). A typical response to 42 �Mtrypsin is illustrated in Fig. 5D. In this experiment, hy-perpolarizing current commands were passed through therecording electrode to monitor membrane conductance. Be-cause the voltage responses observed in the presence of tryp-sin were reduced in amplitude, membrane conductance ap-peared to increase in this cell. The depolarizing response totrypsin following a second application to the same cell wasrarely present and, if seen, was generally substantially lowerin amplitude and shorter in duration. Application of trypsinoccasionally produced a rapid and transient hyperpolariza-tion before the onset of depolarization (Fig. 5D, arrow).

LRGILS

SLIGRL

Saline

Fig. 3. Withdrawal latency (A) and nociceptive score (B and C) measuredin mouse hindpaws, before (time 0) and after intraplantar injection offormalin in mice that have received (10 min before formalin) an i.t.injection of 1 �g of SLIGRL, LRGILS, or saline. Data are expressed asmean � S.E.M. �, significantly different from LRGILS; #, significantlydifferent from basal latency/score measured at time 0, for p � 0.05.

TABLE 1Effects of PAR2 agonists and the control compound LRGILS onfrequency and amplitude of spontaneous EPSCs in substantiagelatinosa neuronsEffectiveness of drugs was assessed using the Komogorov-Smirnoff test on cumula-tive probability plots of event amplitude and duration.

Increase insEPSC

Amplitude

Decrease insEPSC

Amplitude

Increase insEPSC

Frequency

Decrease insEPSC

Frequency

Cs� internal 2/15 9/15 2/10 5/15Trypsin, 5 min

Cs� internal 0/5 2/5 1/5 0/5SLIGRL, 10 min

Cs� internal 0/5 2/5 0/5 1/5LRGILS, 5 min

Delay cells 0/8 4/8 1/8 3/8Trypsin, 10 min

Delay cells 1/11 4/11 0/11 3/11SLIGRL, 5 min

Delay cells 0/11 6/11 1/11 4/11SLIGRL, 10 min

Tonic cells 1/4 0/4 1/4 0/4Trypsin, 10 min

Tonic cells 1/11 1/11 1/11 2/11SLIGRL, 5 min

Tonic cells 3/11 1/11 2/11 1/11SLIGRL, 10 min

228 Alier et al.

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 6: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

50pA

2s

Fig. 4. Effects of trypsin and PAR2 agonists on rat substantia gelatinosa neurons. A, effect of trypsin on sEPSCs. Note decrease in frequency andamplitude. B to I, cumulative probability plots to illustrate the effect of trypsin (21 �M), time, SLIGRL (10 �M), and LRGILS (10 �M) on SEPSCamplitude and interevent interval (reciprocal frequency). Note that trypsin, SLIGRL, and LRGILS decrease sEPSC amplitude, and trypsin andLRGILS decrease sEPSC frequency. Data in B to I obtained from approximately 50 events during 3-min recording in two to seven neurons thatresponded to agonists. J, characteristic firing pattern of a tonic neuron in response to depolarizing current command. K, characteristic firing patternof a delay neuron in response to depolarizing current command. L and M, experiments to illustrate the lack of effect of 21 �M trypsin on spike latencyin response to current ramps in tonic and delay cells, respectively. All data records in traces J to M are on the same time scale.

PAR2 and Pain 229

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 7: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

Like trypsin, the synthetic activating peptide SLIGRL(10 �M) was capable of depolarizing all types of DRG cells,and coincidentally, 65% of cells (13 of 20) were affected.Responses to SLIGRL were seen in 7 of 12 large cells, threeof five medium cells, and three of three small cells. Re-sponse amplitudes were 6.6 � 2.1 mV for large cells, 2.7 �0.4 mV for medium cells, and 1.9 � 0.2 mV for small cells.A typical response is illustrated in Fig. 5E. In this cell,

SLIGRL reduced the amplitude of voltage responses tohyperpolarizing current pulses, an observation consistentwith an increase in membrane conductance. The responseto SLIGRL following a second application to the same cellwas generally similar in amplitude and duration to thefirst. Unlike trypsin, SLIGRL never produced transienthyperpolarization of DRG neurons. The synthetic peptideLRGILS, the reverse sequence of SLIGRL, was used as a

-100 -80 -60 -40 -20 0 20 40-3

-2

-1

0

1

2

3

4

Cur

rent

(nA

)

Voltage (mV)

I

0

1

2

3

4

5

6

Res

pons

e D

urat

ion

(min

)

Trypsin SLIGRL

2nA

100ms

20mV

100ms

H

20mV

2min

LRGILS

20mV

2min

SLIGRL

20mV

2min

Trypsin

20mV

5ms

20mV

5ms20 mV

5 ms

A B

D E F

G

SmallMediumLarge

Control

Trypsin

Control

SLIGRL

I

C

Fig. 5. Effects of PAR2 agonists on ratDRG neurons. Rat DRG cells wereclassified according to cell size and APshape. A, “large” cells had an AP du-ration of �3 ms. B, “medium” cellshad an AP duration of 3 to 5 ms. C,“small” cells had an AP duration of�5 ms. Both medium and small cellsexhibit a hump on the falling phaseof their AP. D to F, effects of trypsinSLIGRL on resting potential and lackof effect of LRGILS. Repetitive hyper-polarizing current pulses were passedthrough the recording electrode, pro-ducing a voltage change across themembrane, and thickness of the re-cording thus represents the voltagechange. Because current is constant,and voltage is decreasing in the pres-ence of SLIGRL and trypsin, both ago-nists appear to increase membraneconductance. G, comparison of the du-ration of trypsin responses (5.20 �0.93 min; n 9) and SLIGRL re-sponses (1.95 � 0.56 min; n 7; p �0.02). H, trypsin-induced increase inexcitability of a medium DRG neuron.Three depolarizing current ramps ofincreasing amplitude were injected intothe neuron before (upper trace) and af-ter (lower trace) application of trypsin.Note increased discharge of action po-tentials in the presence of trypsin. I,voltage-clamp experiment to examineconductance change produced by SLI-GRL. Traces are current responses toramp commands from �110 to �40 mVbefore and after application of SLIGRL.Note that agonist increases conduc-tance at potentials more negative than�40 mV but decreases conductance atpotentials more positive than �20 mV.

230 Alier et al.

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 8: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

control. LRGILS (10 �M) failed to affect the membranepotential of seven of seven large cells and one of onemedium cells (Fig. 5F).

Data from all cell types were pooled to compare the dura-tion of depolarizing response between 42 �M trypsin (n 9),5.20 � 0.93 min, and 10 �M SLIGRL (n 7), 1.95 � 0.56 min.The trypsin response was significantly longer than that pro-duced by SLIGRL (p � 0.05; Fig. 5G).

After a 30-s exposure to 10 �M SLIGRL, some neurons alsoexhibited spontaneous action potential discharge at restingpotential. To examine this phenomenon in more detail, neu-ronal excitability was examined in response to depolarizingcurrent steps or ramps. Four of eight medium or large cellsexamined exhibited an increase in excitability in the pres-ence of 42 �M trypsin. A typical experiment is illustrated inFig. 5H. Before the application of trypsin, a series of depo-larizing current ramps of increasing amplitude produces amodest discharge of action potentials. The most shallow cur-rent ramp fails to generate action potentials, and even withthe steepest ramp, the latency to the first action potential is�300 ms. In contrast, in the presence of trypsin, an actionpotential is seen on the shallowest ramp, and the latency tothe first action potential on the steepest ramp is reduced to�130 ms.

Conductance Changes in DRG Neurons in Responseto PAR2 Agonists. When studied under current clamp, de-polarizing responses to trypsin were associated with an in-crease in conductance in two of two small cells and four of sixlarge cells. Conductance appeared unchanged in the remain-ing two large cells and in three of three medium cells. In-creased conductances were produced by SLIGRL in five offive large cells, one medium cell, and two of two small cells.Application of the reverse sequence peptide, LRGILS, did notchange conductance in four of four large cells.

To examine conductance changes in more detail, cells

were studied under voltage clamp. Voltage-ramp com-mands from �110 to �40mV were used to establish cur-rent-voltage relationships before and during application oftrypsin or SLIGRL. A typical voltage-clamp experiment isillustrated in Fig. 5I. Multiple downward deflections atpositive voltages reflect unclamped action potentials. Whenstudied under voltage clamp, SLIGRL responses usually in-volved an increase in conductance at voltages more negativethan �40 mV and a decrease in conductance at voltagespositive to �30 mV. Presumably, the latter effect, which wasalso seen with trypsin, reflects suppression of voltage-depen-dent K� currents, which may in turn contribute to the in-creased excitability seen in the presence of trypsin (Fig. 5H).Careful examination of the voltage responses to currentramps in Fig. 5H is consistent with this possibility. Voltageresponses to current ramps rise more rapidly in the presenceof trypsin, indicating decreased conductance at depolarizedvoltages. The increase in conductance seen at hyperpolarizedpotentials under voltage clamp (Fig. 5I) would explain theincrease in conductance often seen under current-clamp con-ditions (Fig. 5, D and E) because the hyperpolarizing currentpulses passed throughout the response would assess conduc-tance changes that are dominant at more negative voltages.

Localization of PAR2 Immunoreactivity. The aboveexperiments suggest that although PAR2 receptors are ab-sent from substantia gelatinosa, they are present on the cellbodies of DRG neurons. To confirm this, we investigated PAR2

expression by immunohistochemistry on rat spinal cord slidesand DRGs, using a Santa Cruz Biotechnology anti-PAR2 anti-body. Although PAR2 was detected in rat DRG neurons (Fig.6B), no specific staining for PAR2 was detected from rat spinalcord slices (Fig. 6D). Additional immunohistochemistry stain-ings using another anti-PAR2 antibody (B5 antibody) were per-formed in spinal cord slices and confirmed the absence of PAR2

staining in those tissues (data not shown).

Fig. 6. PAR2 immunoreactivity in rat DRG (A and B) andspinal cord (C and D). DRG photographs without fluores-cence (A) or with a fluorescent lamp exposure (B); typicaldistribution of PAR2 on DRG neurons can be seen on thecell membrane of 62% of DRG neurons (arrows). Spinalcord photographs without fluorescence (C) or with exposureto a fluorescent lamp (D); no specific fluorescent staining isobserved on spinal cord section, demonstrating a lack ofPAR2 expression at the spinal cord level.

PAR2 and Pain 231

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 9: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

DiscussionWe have found that i.t. applied PAR2 agonists produce

mechanical and thermal hyperalgesia and augment the ther-mal and mechanical hyperalgesia produced in an inflamma-tory pain model (Figs. 1–3). The lack of effect of the controlpeptide, LRGILS, and the absence of SLIGRL effects inknockout animals strongly supports the notion that activa-tion of PAR2 augments the transfer of nociceptive informa-tion. Although neither SLIGRL nor trypsin exert excitatoryactions within the substantia gelatinosa per se (Fig. 4), thechanges produced in DRG neurons (Fig. 5) are consistentwith an increased excitation, which may in turn contribute tothe pronociceptive action. This implies that i.t. applied drugsmust have access to the cell bodies of sensory neurons withinthe DRG. At least two studies suggest this is likely to be thecase. Abram et al. (2006) showed that fluorescein, which doesnot cross the blood-brain barrier, can be detected in rat DRGafter i.t. injection. Earlier work had shown that even fluores-cein-labeled proteins accessed the DRG when administeredby this route (Klatzo et al., 1964).

PAR2-induced mechanical allodynia may reflect activationof A� fibers, whereas thermal and mechanical hyperalgesiamay reflect activation of C-fibers. The ability of PAR2 ago-nists to augment nociception in a variety of pain models mayreflect their ability to increase the excitability of all types ofDRG neurons (Fig. 5).

The presence of PAR2 receptors in DRG neurons is con-firmed and their absence from the spinal cord is suggested byour immunohistochemical studies (Fig. 6). To the best of ourknowledge, there are no immunohistochemistry, immuno-blot, or binding studies that demonstrate the presence ofPAR2 in the dorsal horn. This correlates with the inability ofSLIGRL and trypsin to increase spontaneous synaptic eventsin our electrophysiological experiments (Fig. 4). These obser-vations appear to contradict data from other functional stud-ies that suggest that PAR2 resides on the central terminals ofprimary afferent nerves (Fiorucci and Distrutti, 2002). Theobservation that SLIGRL releases SP from DRG neurons inculture (Steinhoff et al., 2000) may either reflect release fromDRG cell bodies (Huang and Neher, 1996) or from neurites.In the latter case, it does not necessarily follow that PAR2

receptors exist on the central terminals of primary afferent invivo because the release in vitro (Steinhoff et al., 2000) maycome from neurites analogous to peripheral nerve endings. Itis already established that PAR2 agonists release SP fromperipheral nerve endings in vivo (Steinhoff et al., 2000). Ithas been shown, however, that PAR2 agonists augmentTRPV1-induced increase of SP from spinal cord slices thatcontain neither DRG neuron bodies nor peripheral nerveendings (Amadesi et al., 2004). One possible explanation forthis effect is that activation of TRPV1 receptors promotes arapid appearance of PAR2 within the dorsal horn. We havepreliminary electrophysiological and histochemical data tosuggest this may be the case. One might therefore speculatethat the appearance of PAR2 is dependent upon the prior orconstitutive activation of TRPV1 receptors. If this is so, itmay explain why PAR2 is found on peripheral nerve termi-nals and DRG cell bodies but not usually on the centralterminals of primary afferents.

Because our electrophysiological studies were confined tothe substantia gelatinosa, it could be argued that spinal

pronociceptive actions of PAR2 agonists reflect actions withindeeper sensory lamina, such as laminae III and IV, whichalso receive sensory information from pain fibers. This isunlikely to be the case because these areas, like lamina II,fail to display PAR2-like immunoreactivity (Fig. 6D).

Unlike PAR2 agonists, PAR1 and PAR4 agonists attenuatenociception (Steinhoff et al., 2000; Vergnolle et al., 2003;Asfaha et al., 2007), and these effects may be mediated bothcentrally and peripherally (Asfaha et al., 2002). The occa-sional transient hyperpolarization of DRG neurons seen withtrypsin, but not with the PAR2 agonist SLIGRL, may reflectactivation of PAR1 or PAR4. The depressant actions oftrypsin on sEPSC amplitude and frequency in putativeexcitatory delay cells in substantia gelatinosa may be me-diated by PAR1 or PAR4 (Vergnolle et al., 2003), whereasdepressant effects of SLIGRL may reflect nonspecific ac-tions because these were also seen with the control peptideinactive on PAR2, LRGILS (Fig. 4).

Effects on DRG Cells. Even at supramaximal concentra-tions, the membrane depolarizations produced by trypsinand SLIGRL were of quite small amplitude and unlikely todepolarize DRG neurons to the threshold for action potentialgeneration, although spontaneous action potentials did ap-pear in a few of the cells studied. It is probably more relevantthat the agonists affected voltage-gated channels that controlaction potential discharge when neurons are subject to otherdepolarizing stimuli. Thus, in a pathological situation, whereDRG neurons may be depolarized by a host of pronociceptivesubstances such as neuropeptides, cytokines, or even low pH,the concomitant activation of PAR2 receptors would facilitateaction potential generation in response to depolarization. This,in turn, would lead to increased firing of fibers that synapse inthe dorsal horn. A variety of voltage-gated K� currents areactivated by depolarization of DRG neurons (Gold et al., 1996).During the ramp commands used in our voltage-clamp experi-ments (Fig. 5I), most of the outward current at positive voltagesprobably flows through delayed rectifier and voltage-dependentCa2�-sensitive K� channels (BK channels) (Abdulla and Smith,2001). Suppression of conductance at these voltages is morelikely to reflect an action on delayed rectifier currents than onBK channels because PAR2 agonists are known to elevate in-tracellular Ca2� in DRG cells (Steinhoff et al., 2000). More-over, PAR2-induced depolarization was not blocked by Ca2�

channel blockers such as Mg2� or Ni2� (J. A. Endicott andP. A. Smith, unpublished data). Decreased K� conductance islikely to contribute to the PAR2-mediated increase in DRGexcitability (Fig. 5H). Because PAR2 receptors are known tomodulate TRPV1 channel activity (Amadesi et al., 2004; Daiet al., 2004), it is possible that the inward current seen atnegative voltages reflects activation of these channels. Thiswould seem unlikely because TRPV1 channels are confinedto a subset of small DRG neurons, yet PAR2 agonists pro-duced increased conductance in “large” and “medium” DRGneurons that do not express TRPV1 channels. Moreover,TRPV1 channels exhibit outward rather than inward rectifi-cation so that would be more available at positive rather thannegative voltages (Raisinghani et al., 2005). Another possi-bility is that PAR2 agonists affected H-currents. These non-selective cation currents that are activated by hyperpolariza-tion are found in some DRG cells (Abdulla and Smith, 2001;Chaplan et al., 2003). However, PAR2 agonists failed to affectH-currents activated by standard hyperpolarizing voltage

232 Alier et al.

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from

Page 10: Intrathecal Administration of Proteinase-Activated Receptor-2 … · 2019. 5. 8. · Intrathecal Administration of Proteinase-Activated Receptor-2 Agonists Produces Hyperalgesia by

protocols (P. Stemkowski, unpublished data). Thus, the chan-nels underlying PAR2-induced changes in conductance re-main to be identified.

The observed overall 65% response rate of DRG neurons toPAR2 agonists fits well with the report that approximately60% of them display PAR2 (Steinhoff et al., 2000). In trigem-inal ganglion neurons, PAR2 receptors are found on neuronsthat do not express tachykinins (Dinh et al., 2005). This andthe present observation that PAR2 depolarized large cellssuggest that it is capable of affecting neurons that do notexpress tachykinins.

Significance of Findings. Our findings illustrate howselective stimulation of DRG neurons can produce hyperal-gesia and underline the importance of DRG cell bodies as asite of action of i.t. applied drugs (Abram et al., 2006). More-over, pathological or pharmacological excitation of DRG cellbodies is able to produce pain-like behavior in animal models.Although trypsin is regarded as the principal PAR2-activat-ing proteinase, other activators include mast cell tryptase,trypsinogen IV, and a trypsin-like proteinase known as P22that was identified in brain slices (Sawada et al., 2000).Because mast cells are found in perivascular areas of thenervous system, including the sensory ganglia (Undem et al.,1993), mast cell tryptase is perhaps the most likely endoge-nous activator of PAR2 in DRG (Vergnolle et al., 2001). Al-though the actions of endogenous PAR2 agonists may beconfined to DRG neurons, our findings suggest that sucheffects play a major role in nociceptive processing.

ReferencesAbdulla FA and Smith PA (1997) Ectopic �2-adrenoceptors couple to N-type Ca2�

channels in axotomized rat sensory neurons. J Neurosci 17:1633–1641.Abdulla FA and Smith PA (2001) Axotomy- and autotomy-induced changes in Ca2�

and K� channel currents of rat dorsal root ganglion neurons. J Neurophysiol85:644–658.

Abram SE, Yi J, Fuchs A, and Hogan QH (2006) Permeability of injured and intactperipheral nerves and dorsal root ganglia. Anesthesiology 105:146–153.

Amadesi S, Nie J, Vergnolle N, Cottrell GS, Grady EF, Trevisani M, Manni C,Geppetti P, McRoberts JA, Ennes H, et al. (2004) Protease-activated receptor 2sensitizes the capsaicin receptor transient receptor potential vanilloid receptor 1 toinduce hyperalgesia. J Neurosci 24:4300–4312.

Asfaha S, Brussee V, Chapman K, Zochodne DW, and Vergnolle N (2002) Proteinase-activated receptor-1 agonists attenuate nociception in response to noxious stimuli.Br J Pharmacol 135:1101–1106.

Asfaha S, Cenac N, Houle S, Altier C, Papez MD, Nguyen C, Steinhoff M, ChapmanK, Zamponi GW, and Vergnolle N (2007) Protease-activated receptor-4: a novelmechanism of inflammatory pain modulation. Br J Pharmacol 150:176–185.

Balasubramanyan S, Stemkowski PL, Stebbing MJ, and Smith PA (2006) Sciaticchronic constriction injury produces cell-type specific changes in the electrophys-iological properties of rat substantia gelatinosa neurons. J Neurophysiol 96:579–590.

Cenac N and Vergnolle N (2005) Proteases and protease-activated receptors (PARs):novel signals for pain. Curr Top Med Chem 5:569–576.

Chaplan SR, Guo HQ, Lee DH, Luo L, Liu C, Kuei C, Velumian AA, Butler MP,Brown SM, and Dubin AE (2003) Neuronal hyperpolarization-activated pace-maker channels drive neuropathic pain. J Neurosci 23:1169–1178.

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

Dinh QT, Cryer A, Dinh S, Trevisani M, Georgiewa P, Chung F, Geppetti P, HepptW, Klapp BF, and Fischer A (2005) Protease-activated receptor 2 expression intrigeminal neurons innervating the rat nasal mucosa. Neuropeptides 39:461–466.

Fiorucci S and Distrutti E (2002) Role of PAR2 in pain and inflammation. TrendsPharmacol Sci 23:153–155.

Gold MS, Shuster MJ, and Levine JD (1996) Characterization of six voltage-gatedK� currents in adult rat sensory neurons. J Neurophysiol 75:2629–2646.

Hollenberg MD (1999) Protease-activated receptors: PAR4 and counting: how long isthe course? Trends Pharmacol Sci 20:271–273.

Hoogerwerf WA, Zou L, Shenoy M, Sun D, Micci MA, Lee-Hellmich H, Xiao SY,Winston JH, and Pasricha PJ (2001) The proteinase-activated receptor 2 is in-volved in nociception. J Neurosci 21:9036–9042.

Huang LY and Neher E (1996) Ca2�-dependent exocytosis in the somata of dorsalroot ganglion neurons. Neuron 17:135–145.

Klatzo I, Miguel J, Ferris PJ, Prokop JD, and Smith FM (1964) Observations on thepassage of fluorescein labeled serum proteins (FSLP) from the cerebrospinal fluid.J Neuropathol Exp Neurol 23:18–24.

Lu Y and Perl ER (2003) A specific inhibitory pathway between substantia gelati-nosa neurons receiving direct C-fiber input. J Neurosci 23:8752–8758.

Lu Y and Perl ER (2005) Modular organization of excitatory circuits between neu-rons of the spinal superficial dorsal horn (laminae I and II). J Neurosci 25:3900–3907.

Moran TD, Colmers WF, and Smith PA (2004) Opioid-like actions of neuropeptide Yin rat substantia gelatinosa: Y1 suppression of inhibition and Y2 suppression ofexcitation. J Neurophysiol 92:3266–3275.

Noorbakhsh F, Vergnolle N, Hollenberg MD, and Power C (2003) Proteinase-activated receptors in the nervous system. Nat Rev Neurosci 4:981–990.

Raisinghani M, Pabbidi RM, and Premkumar LS (2005) Activation of transientreceptor potential vanilloid 1 (TRPV1) by resiniferatoxin. J Physiol (Lond) 567:771–786.

Sakmann B and Stuart GJ (1995) Patch clamp recordings from the soma, dendritesand axons of neurons in brain slices, in Single-Channel Recordings (Sakmann Band Neher E eds) pp 199–211, Plenum, New York.

Sawada K, Nishibori M, Nakaya N, Wang Z, and Saeki K (2000) Purification andcharacterization of a trypsin-like serine proteinase from rat brain slices thatdegrades laminin and type IV collagen and stimulates protease-activated recep-tor-2. J Neurochem 74:1731–1738.

Steinhoff M, Buddenkotte J, Shpacovitch V, Rattenholl A, Moormann C, Vergnolle N,Luger TA, and Hollenberg MD (2005) Proteinase-activated receptors: transducersof proteinase-mediated signaling in inflammation and immune response. EndocrRev 26:1–43.

Steinhoff M, Vergnolle N, Young SH, Tognetto M, Amadesi S, Ennes HS, TrevisaniM, Hollenberg MD, Wallace JL, Caughey GH, et al. (2000) Agonists of proteinase-activated receptor 2 induce inflammation by a neurogenic mechanism. Nat Med6:151–158.

Undem BJ, Hubbard W, and Weinreich D (1993) Immunologically induced neuro-modulation of guinea pig nodose ganglion neurons. J Auton Nerv Syst 44:35–44.

Vergnolle N, Bunnett NW, Sharkey KA, Brussee V, Compton SJ, Grady EF, CirinoG, Gerard N, Basbaum AI, Andrade-Gordon P, et al. (2001) Proteinase-activatedreceptor-2 and hyperalgesia: a novel pain pathway. Nat Med 7:821–826.

Vergnolle N, Ferazzini M, D’Andrea MR, Buddenkotte J, and Steinhoff M (2003)Proteinase-activated receptors: novel signals for peripheral nerves. Trends Neu-rosci 26:496–500.

Address correspondence to: Dr. Peter Smith, Department of Pharmacology,University of Alberta, 9.75 Medical Sciences Building, Edmonton, AB T6G2H7, Canada. E-mail: [email protected]

PAR2 and Pain 233

at ASPE

T Journals on M

ay 8, 2019jpet.aspetjournals.org

Dow

nloaded from