Journal club 2013-03-05

8612.full

J Neurosci. 2009 Jul 1;29(26):8612-9. doi: 10.1523/JNEUROSCI.1057-09.2009.

Mrgprd enhances excitability in specific populations of cutaneous murine polymodal nociceptors.

Source

Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.

Abstract

The Mas-related G protein-coupled receptor D (Mrgprd) is selectively expressed in nonpeptidergic nociceptors that innervate the outer layers of mammalian skin. The function of Mrgprd in nociceptive neurons and the physiologically relevant somatosensory stimuli that activate Mrgprd-expressing (Mrgprd(+)) neurons are currently unknown. To address these issues, we studied three Mrgprd knock-in mouse lines using an ex vivo somatosensory preparation to examine the role of the Mrgprd receptor and Mrgprd(+) afferents in cutaneous somatosensation. In mouse hairy skin, Mrgprd, as marked by expression of green fluorescent protein reporters, was expressed predominantly in the population of nonpeptidergic, TRPV1-negative, C-polymodal nociceptors. In mice lacking Mrgprd, this population of nociceptors exhibited decreased sensitivity to cold, heat, and mechanical stimuli. Additionally, in vitro patch-clamp studies were performed on cultured dorsal root ganglion neurons from Mrgprd(-/-) and Mrgprd(+/-) mice. These studies revealed a higher rheobase in neurons from Mrgprd(-/-) mice than from Mrgprd(+/-) mice. Furthermore, the application of the Mrgprd ligand beta-alanine significantly reduced the rheobase and increased the firing rate in neurons from Mrgprd(+/-) mice but was without effect in neurons from Mrgprd(-/-) mice. Our results demonstrate that Mrgprd influences the excitability of polymodal nonpeptidergic nociceptors to mechanical and thermal stimuli.

PMID:
19571152
[PubMed – indexed for MEDLINE

Journal club 2013-01-25

1744-8069-8-75

Mol Pain. 2012 Sep 27;8:75. doi: 10.1186/1744-8069-8-75.

Prostaglandin metabolite induces inhibition of TRPA1 and channel-dependent nociception.

Source

Department of Anesthesiology, Washington University Pain Center, St, Louis, MO 63110, USA. storyg@wustl.edu.

ABSTRACT:

BACKGROUND:

The Transient Receptor Potential (TRP) ion channel TRPA1 is a key player in pain pathways. Irritant chemicals activate ion channel TRPA1 via covalent modification of N-terminal cysteines. We and others have shown that 15-Deoxy-Δ12, 14-prostaglandin J2 (15d-PGJ2) similarly activates TRPA1 and causes channel-dependent nociception. Paradoxically, 15d-PGJ2 can also be anti-nociceptive in several pain models. Here we hypothesized that activation and subsequent desensitization of TRPA1 in dorsal root ganglion (DRG) neurons underlies the anti-nociceptive property of 15d-PGJ2. To investigate this, we utilized a battery of behavioral assays and intracellular Ca2+ imaging in DRG neurons to test if pre-treatment with 15d-PGJ2 inhibited TRPA1 to subsequent stimulation.

RESULTS:

Intraplantar pre-injection of 15d-PGJ2, in contrast to mustard oil (AITC), attenuated acute nocifensive responses to subsequent injections of 15d-PGJ2 and AITC, but not capsaicin (CAP). Intraplantar 15d-PGJ2-administered after the induction of inflammation-reduced mechanical hypersensitivity in the Complete Freund’s Adjuvant (CFA) model for up to 2 h post-injection. The 15d-PGJ2-mediated reduction in mechanical hypersensitivity is dependent on TRPA1, as this effect was absent in TRPA1 knockout mice. Ca2+ imaging studies of DRG neurons demonstrated that 15d-PGJ2 pre-exposure reduced the magnitude and number of neuronal responses to AITC, but not CAP. AITC responses were not reduced when neurons were pre-exposed to 15d-PGJ2 combined with HC-030031 (TRPA1 antagonist), demonstrating that inhibitory effects of 15d-PGJ2 depend on TRPA1 activation. Single daily doses of 15d-PGJ2, administered during the course of 4 days in the CFA model, effectively reversed mechanical hypersensitivity without apparent tolerance or toxicity.

CONCLUSIONS:

Taken together, our data support the hypothesis that 15d-PGJ2 induces activation followed by persistent inhibition of TRPA1 channels in DRG sensory neurons in vitro and in vivo. Moreover, we demonstrate novel evidence that 15d-PGJ2 is analgesic in mouse models of pain via a TRPA1-dependent mechanism. Collectively, our studies support that TRPA1 agonists may be useful as pain therapeutics.

Journal club 2012-12-27

10.1177_0022034511412074

J Dent Res. 2011 Sep;90(9):1098-102. doi: 10.1177/0022034511412074. Epub 2011 Jun 10.

Cold suppresses agonist-induced activation of TRPV1.

Source

Department of Neural and Pain Sciences, University of Maryland Dental School, Program in Neuroscience, 650 W. Baltimore Street, Baltimore, MD 21201, USA. mchung@umaryland.edu

Abstract

Cold therapy is frequently used to reduce pain and edema following acute injury or surgery such as tooth extraction. However, the neurobiological mechanisms of cold therapy are not completely understood. Transient receptor potential vanilloid 1 (TRPV1) is a capsaicin- and heat-gated nociceptive ion channel implicated in thermosensation and pathological pain under conditions of inflammation or injury. Although capsaicin-induced nociception, neuropeptide release, and ionic currents are suppressed by cold, it is not known if cold suppresses agonist-induced activation of recombinant TRPV1. We demonstrate that cold strongly suppressed the activation of recombinant TRPV1 by multiple agonists and capsaicin-evoked currents in trigeminal ganglia neurons under normal and phosphorylated conditions. Cold-induced suppression was partially impaired in a TRPV1 mutant that lacked heat-mediated activation and potentiation. These results suggest that cold-induced suppression of TRPV1 may share a common molecular basis with heat-induced potentiation, and that allosteric inhibition may contribute, in part, to the cold-induced suppression. We also show that combination of cold and a specific antagonist of TRPV1 can produce an additive suppression. Our results provide a mechanistic basis for cold therapy and may enhance anti-nociceptive approaches that target TRPV1 for managing pain under inflammation and tissue injury, including that from tooth extraction.

PMID:
21666106
[PubMed – indexed for MEDLINE]
PMCID:
PMC3169882

Free PMC Article

Journal club 2012-11-30

1-s2.0-S0304395910003246-main

Pain. 2010 Aug;150(2):340-50. Epub 2010 Jun 12.

TRPM8, but not TRPA1, is required for neural and behavioral responses to acute noxious cold temperatures and cold-mimetics in vivo.

Source

Neuroscience Graduate Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.

Abstract

Somatosensory neurons detect environmental stimuli, converting external cues into neural activity that is relayed first to second-order neurons in the spinal cord. The detection of cold is proposed to be mediated by the ion channels TRPM8 and TRPA1. However, there is significant debate regarding the role of each channel in cold-evoked pain, complicating their potential as drug targets for conditions such as cold allodynia and hyperalgesia. To address this debate, we generated mice lacking functional copies of both channels and examined behaviors and neural activity in response to painful cold and noxious cooling compounds. Whereas normal mice display a robust preference for warmth over cold, both TRPM8-null (TRPM8(-/-)) and TRPM8/TRPA1 double-knockout mice (DKO) display no preference until temperatures reach the extreme noxious range. Additionally, in contrast to wildtype mice that avoid touching cold surfaces, mice lacking TRPM8 channels display no such avoidance and explore noxious cold surfaces, even at 5 degrees C. Furthermore, nocifensive behaviors to the cold-mimetic icilin are absent in TRPM8(-/-) and DKO mice, but are retained in TRPA1-nulls (TRPA1(-/-)). Finally, neural activity, measured by expression of the immediate-early gene c-fos, evoked by hindpaw stimulation with noxious cold, menthol, or icilin is reduced in TRPM8(-/-) and DKO mice, but not in TRPA1(-/-) animals. Thus our results show that noxious cold signaling is exclusive to TRPM8, mediating neural and behavioral responses to cold and cold-mimetics, and that TRPA1 is not required for acute cold pain in mammals.

Copyright (c) 2010 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved.

2012-11-26 고려인삼학회포스터

고려인삼학회포스터

A particular ginsenoside has a potential to inhibit itch transduction

Wook-Joo Lee, Dasom Choi, Won-Sik Shim*

College of Pharmacy, Gachon University, Incheon, 406-799, South Korea

Purpose:

Korean red ginseng (the steamed root of Panax ginseng C.A. Meyer, family Araliaceae) has been used as a remedy for wide variety of disorders, and recent studies have identified its beneficial use in itch-related diseases such as atopic dermatitis. Although its anti-pruritic effect was mostly revealed by animal scratching tests, its underlying molecular mechanism has never been investigated. Because the itch transduction in peripheral nervous system could be initiated via histamine-dependent (H1R/TRPV1), or -independent (MrgprA3/TRPA1) pathway, the putative inhibitory effect of various ginsenosides were investigated in the present study.

Method:

cDNA combinations of H1R/TRPV1 or MrgprA3/TRPA1 were transiently expressed in human embryonic kidney (HEK) 293T cells, and calcium influx was monitored with Fluo3-AM, a calcium-specific fluorescent dye. To test the inhibitory effect, 11 ginsenosides were pretreated for 10 min, and either histamine (for H1R/TRPV1) or chloroquine (for MrgprA3/TRPA1) was treated and changes were recorded for 1 min.

Result:

It was found that one particular ginsenoside strongly inhibited the calcium influx on both H1R/TRPV1- and MrgprA3/TRPA1-expressing cells even after histamine or chloroquine was treated, suggesting that the ginsenoside may have a potential anti-pruritic activity. Furthermore, it seems that the inhibitory effect is mediated by directly blocking the action of ion channels (TRPV1, TRPA1) rather than interfering with receptors (H1R, MrgprA3), implying other possible roles especially in nociception.

Journal club 2012-11-02

nihms225332

Enhanced scratching evoked by PAR-2 agonist and 5-HT but not histamine in a mouse model of chronic dry skin itch.

Source

Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95616, USA.

Abstract

Chronic itch is a symptom of many skin conditions and systemic disease, and it has been hypothesized that the chronic itch may result from sensitization of itch-signaling pathways. We induced experimental chronic dry skin on the rostral back of mice, and observed a significant increase in spontaneous hindlimb scratches directed to the dry skin. Spontaneous scratching was significantly attenuated by a PAR-2 antibody and 5-HT2A receptor antagonist, indicating activation of these receptors by endogenous mediators released under dry skin conditions. We also observed a significant increase in the number of scratch bouts evoked by acute intradermal injections of a protease-activated receptor (PAR)-2 agonist and serotonin (5-HT), but not histamine. We additionally investigated if pruritogen-evoked activity of dorsal root ganglion (DRG) neurons is enhanced in this model. DRG cells from dry skin mice exhibited significantly larger responses to the PAR-2 agonist and 5-HT, but not histamine. Spontaneous scratching may reflect ongoing itch, and enhanced pruritogen-evoked scratching may represent hyperknesis (enhanced itch), both potentially due to sensitization of itch-signaling neurons. The correspondence between enhanced behavioral scratching and DRG cell responses suggest that peripheral pruriceptors that respond to proteases and 5-HT, but not histamine, may be sensitized in dry skin itch.

Copyright © 2010 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved.

Journal club 2012-09-06

11330.full

TRPV1-expressing primary afferents generate behavioral responses to pruritogens via multiple mechanisms

  1. Noritaka Imamachia,1,
  2. Goon Ho Parkb,
  3. Hyosang Leec,
  4. David J. Andersonc,
  5. Melvin I. Simonc,2,
  6. Allan I. Basbaumaand
  7. Sang-Kyou Hanb,3

+ Author Affiliations


  1. aDepartment of Anatomy, University of California, San Francisco, CA 94143;

  2. bDepartment of Pharmacology, University of California at San Diego, La Jolla, CA 92093; and

  3. cDivision of Biology, California Institute of Technology, Pasadena, CA 91125
  1. Contributed by Melvin I. Simon, May 20, 2009 (received for review May 4, 2009)

Abstract

The mechanisms that generate itch are poorly understood at both the molecular and cellular levels despite its clinical importance. To explore the peripheral neuronal mechanisms underlying itch, we assessed the behavioral responses (scratching) produced by s.c. injection of various pruritogens in PLCβ3- or TRPV1-deficient mice. We provide evidence that at least 3 different molecular pathways contribute to the transduction of itch responses to different pruritogens: 1) histamine requires the function of both PLCβ3 and the TRPV1 channel; 2) serotonin, or a selective agonist, α-methyl-serotonin (α-Me-5-HT), requires the presence of PLCβ3 but not TRPV1, and 3) endothelin-1 (ET-1) does not require either PLCβ3 or TRPV1. To determine whether the activity of these molecules is represented in a particular subpopulation of sensory neurons, we examined the behavioral consequences of selectively eliminating 2 nonoverlapping subsets of nociceptors. The genetic ablation of MrgprD+ neurons that represent ≈90% of cutaneous nonpeptidergic neurons did not affect the scratching responses to a number of pruritogens. In contrast, chemical ablation of the central branch of TRPV1+ nociceptors led to a significant behavioral deficit for pruritogens, including α-Me-5-HT and ET-1, that is, the TRPV1-expressing nociceptor was required, whether or not TRPV1 itself was essential. Thus, TRPV1 neurons are equipped with multiple signaling mechanisms that respond to different pruritogens. Some of these require TRPV1 function; others use alternate signal transduction pathways.

Journal club 2012-08-22

1-s2.0-S0304394004000370-main

Histamine-induced Ca2+influx via the PLA2/lipoxygenase/TRPV1 pathway in rat sensory neurons

This paper is in honor of Manfred Zimmermann’s 70th birthday.
  • Byung Moon Kim,
  • Sang Hee Lee,
  • Won Sik Shim,
  • Uhtaek OhCorresponding author contact information, E-mail the corresponding author
  • Sensory Research Center, National Creative Research Initiatives, College of Pharmacy, Seoul National University, San 56-1, Shinlim, Kwanak-Gu, Seoul 151-742, South Korea

 

  • Available online 27 March 2004.

Abstract

Histamine is known to excite a subset of C-fibers and cause itch sensation. Despite its well-defined excitatory action on sensory neurons, intracellular signaling mechanisms are not understood. Previously, we demonstrated that bradykinin excited sensory neurons by activating TRPV1 via the phospholipase A2 (PLA2) and lipoxygenase (LO) pathway. We, thus, hypothesized that histamine excited sensory neurons via the PLA2/LO/TRPV1 pathway. Application of histamine elicited a rapid increase in intracellular Ca2+ ([Ca2+]i) that desensitized slowly in cultured dorsal root ganglion neurons. Histamine-induced [Ca2+]i was dependent on extracellular Ca2+ and inhibited by capsazepine and by SC0030, competitive antagonists of TRPV1. Quinacrine and nordihydroguaiaretic acid, a PLA2 and an LO inhibitor, respectively, blocked the histamine-induced Ca2+influx in sensory neurons, while indomethacin (a cyclooxygenase inhibitor) did not. We thus conclude that histamine activates TRPV1 after stimulating the PLA2/LO pathway, leading to the excitation of sensory neurons. These results further provide an idea for potential use of TRPV1 antagonists as anti-itch drugs.

Journal club 2012-08-08

ncomms1749-s1
Filename : ncomms1749-s1.pdf (6 MB)
Caption :

Nat Commun. 2012 Mar 20;3:746. doi: 10.1038/ncomms1749.

Transient activation of specific neurons in mice by selective expression of the capsaicin receptor.

Source

Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, 1959 NE Pacific Street, Box 357370, Seattle, Washington 98195, USA.

Abstract

The ability to control the electrical activity of a neuronal subtype is a valuable tool in deciphering the role of discreet cell populations in complex neural circuits. Recent techniques that allow remote control of neurons are either labor intensive and invasive or indirectly coupled to neural electrical potential with low temporal resolution. Here we show the rapid, reversible and direct activation of genetically identified neuronal subpopulations by generating two inducible transgenic mouse models. Confined expression of the capsaicin receptor, TRPV1, allows cell-specific activation after peripheral or oral delivery of ligand in freely moving mice. Capsaicin-induced activation of dopaminergic or serotonergic neurons reversibly alters both physiological and behavioural responses within minutes, and lasts ~10 min. These models showcase a robust and remotely controllable genetic tool that modulates a distinct cell population without the need for invasive and labour-intensive approaches.

PMID:
22434189
[PubMed – indexed for MEDLINE]
Scroll to Top