Deletion of interleukin-6 signal transducer gp130 in small sensory neurons attenuates mechanonociception and down-regulates TRPA1 expression

Philipp Malsch, Manfred Andratsch, Christian Vogl, Andrea S Link, Christian Alzheimer, Stuart M Brierley, Patrick A Hughes, Michaela Kress, Philipp Malsch, Manfred Andratsch, Christian Vogl, Andrea S Link, Christian Alzheimer, Stuart M Brierley, Patrick A Hughes, Michaela Kress

Abstract

Glycoprotein 130 (gp130) is the signal transducing receptor subunit for cytokines of the interleukin-6 (IL-6) family, and it is expressed in a multitude of cell types of the immune and nervous system. IL-6-like cytokines are not only key regulators of innate immunity and inflammation but are also essential factors for the differentiation and development of the somatosensory system. Mice with a null mutation of gp130 in primary nociceptive afferents (SNS-gp130(-/-)) are largely protected from hypersensitivity to mechanical stimuli in mouse models of pathological pain. Therefore, we set out to investigate how neuronal gp130 regulates mechanonociception. SNS-gp130(-/-) mice revealed reduced mechanosensitivity to high mechanical forces in the von Frey assay in vivo, and this was associated with a reduced sensitivity of nociceptive primary afferents in vitro. Together with these findings, transient receptor potential ankyrin 1 (TRPA1) mRNA expression was significantly reduced in DRG from SNS-gp130(-/-) mice. This was also reflected by a reduced number of neurons responding with calcium transients to TRPA1 agonists in primary DRG cultures. Downregulation of Trpa1 expression was predominantly discovered in nonpeptidergic neurons, with the deficit becoming evident during stages of early postnatal development. Regulation of Trpa1 mRNA expression levels downstream of gp130 involved the classical Janus kinase family-signal transducer and activator of transcription pathway. Our results closely link proinflammatory cytokines to the expression of TRPA1, both of which have been shown to contribute to hypersensitive pain states. We suggest that gp130 has an essential role in mechanonociception and in the regulation of TRPA1 expression.

Keywords: Il-6 signal transducer; JAK/STAT signaling; cytokine; mechanonociceptor; mechanotransduction; pain.

Copyright © 2014 the authors 0270-6474/14/349845-12$15.00/0.

Figures

Figure 1.
Figure 1.
SNS-gp130−/− mice were less sensitive to mechanical stimulation. A, SNS-gp130−/− mice responded with significantly lower frequencies to mechanical stimulation with von Frey hairs than control gp130fl/fl mice (N = 9, two-way ANOVA, genotype: p < 0.001, forces: p < 0.001, genotype × forces: p = 0.084). B, Stimulus–response relationship of SNS-gp130−/− mice in the von Frey test was shifted toward stronger forces as SNS-gp130−/− mice exhibited significantly higher mechanical thresholds than gp130fl/fl mice. Boxes represent median as well as lower and upper quartile; whiskers represent the 10th and 90th percentiles. C, In vitro, the distribution of mechanical thresholds was significantly shifted toward stronger stimuli for unmyelinated mechanosensitive afferents obtained from SNS-gp130−/− (black columns) compared with those from gp130fl/fl mice (white columns; gp130fl/fl: median: 32 mN, n = 63, N = 30; black columns: SNS-gp130−/−: median: 128 mN, n = 35, N = 23; Mann–Whitney U test, p < 0.05). D, The cumulative distribution showed that a significantly lower percentage of fibers responded to medium to high mechanical forces in SNS-gp130−/− mice. *p < 0.05. **p < 0.01.
Figure 2.
Figure 2.
Genome-wide mRNA expression profiling assay. A, Average ion channel expression intensities in SNS-gp130−/− compared with gp130fl/fl DRG explants (N = 5). Depicted are the means of the quantile-normalized signals. The linear values are shown in a log2 scaling. FDR-adjusted p values for deregulated genes: Scn5a, p = 0.162797384; Kcnh1, p = 0.309818818; Chrnb4, p = 0.112616368; Kcnj11, p = 0.36698117; Scn3b, p = 0.396426446; Trpc7, p = 0.378056322; Cacna1f, p = 0.5246788; Trpa1, p = 0.363034703; LOC637107, p = 0.31986923. B, Heat map showing ion channel gene expression in SNS-gp130−/− versus gp130fl/fl DRG explants. Genes, which are upregulated (green, left) or downregulated (magenta, right), are sorted by their unadjusted p value. Only genes with p < 0.05 are shown. C, Expressed ion channels with a putative function in mechanotransduction. Expression levels are shown as log2 quantile-normalized signal values for each candidate and genotype (open circles represent gp130fl/fl; filled circles represent SNS-gp130−/−), and variation between individuals is minimal. Among the 14 genes, Trpa1 is the only deregulated ion channel. For fold change in expression levels, see Table 1.
Figure 3.
Figure 3.
Neuronal gp130 was essential for Trpa1 mRNA expression in DRG. A, TaqMan mRNA analysis revealed significantly lower Trpa1 mRNA levels in DRG explants and primary neuronal cultures of SNS-gp130−/− mice (DRG from thoracic and lumbar levels). B, Trpa1 mRNA expression was significantly reduced in lumbar DRG explants from SNS-gp130−/− mice compared with gp130fl/fl and SNS-Cre, which were not significantly different. Furthermore, Trpv1 and Trpv4 mRNA levels were similar between gp130fl/fl and SNS-gp130−/− mice. C, Trpa1 mRNA expression of lumbar DRG explants within the first 14 postnatal days. In SNS-gp130−/− mice, Trpa1 mRNA expression was already reduced at P1–P14. Expression levels are normalized to adult gp130fl/fl mice. *p < 0.05. **p < 0.01. ***p < 0.001.
Figure 4.
Figure 4.
Reduced Trpa1 mRNA levels in DRG sections of SNS-gp130−/− mice. A, ISH with Trpa1 antisense riboprobe produced a selective staining of Trpa1-expressing neurons, whereas sense probes did not show specific staining in the lumbar DRG sections. Scale bar, 50 μm. B, Samples of gp130fl/fl and SNS-gp130−/− lumbar DRG sections with Trpa1 antisense riboprobe display reduced Trpa1 mRNA labeling in SNS-gp130−/− sections. Scale bar, 50 μm. C, Quantification of the ISH labeling in lumbar gp130fl/fl and SNS-gp130−/− sections. SNS-gp130−/− sections reveal significantly lower numbers of Trpa1 expressing neurons. *p < 0.05.
Figure 5.
Figure 5.
Trpa1 deficiency was predominantly observed in IB4-binding nociceptive neurons. A, B, Staining for IB4+ neurons in gp130fl/fl and SNS-gp130−/− DRG sections. Scale bar, 50 μm. C, The percentage of IB4-binding neurons was similar in both genotypes. D, E, FISH for Trpa1 mRNA in gp130fl/fl and SNS-gp130−/− DRG sections. Scale bar, 50 μm. F, Quantification of FISH shows that the number of Trpa1 mRNA-expressing neurons (Trpa1+) was significantly reduced in SNS-gp130−/− mice. G, H, Colabeling of Trpa1 mRNA expressing (magenta) and IB4-binding (green) neurons in both genotypes. Scale bar, 50 μm. I, In control mice, Trpa1 mRNA was predominantly expressed in IB4-positive neurons. In SNS-gp130−/− mice, reduction of Trpa1 was most noticeable in the IB4-positive population. n.s., Not significant (p > 0.05). *p < 0.05. **p < 0.01.
Figure 6.
Figure 6.
Expression of Trpa1 mRNA was controlled by the JAK-STAT pathway. TaqMan mRNA analysis revealed significantly reduced Trpa1 expression levels in DRG neurons incubated with a selective JAK inhibitor I (JAKI, 1 μm) or a STAT inhibitor, with selectivity for STAT3 (Stattic, 1.5 μm). *p < 0.05. **p < 0.01.
Figure 7.
Figure 7.
Functional TRPA1 expression was reduced in cultured primary DRG neurons of SNS-gp130−/− mice. A, Calcium imaging sample traces of primary neurons stimulated with the TRPA1-agonist CA (200 μm), high potassium chloride (KCl, 25 mm) and TRPV1-agonist capsaicin (0.3 μm). B, The percentage of neurons responding to CA was significantly decreased in SNS-gp130−/− cultures. In contrast, the responsiveness to capsaicin (Caps) was unchanged. C, Calcium imaging sample traces of primary neurons stimulated with NPPB (100 μm) and KCl (25 mm). Responses to NPPB were greatly diminished in TRPA1−/− neurons. Magnitude of ratio increase in response to stimulation with 100 μm NPPB in TRPA1+/+ (n = 53, N = 2) and TRPA1−/− (n = 101, N = 3) neuronal cultures. The highest ratio increase in TRPA1−/− cultures was considered as threshold for TRPA1-mediated responses to NPPB. TRPA1+/+ and TRPA1−/−: boxes represent median as well as lower and upper quartile; whiskers represent the 10th and 90th percentiles. D, Percentage and ratio increase of neurons responding to NPPB with a suprathreshold ratio increase in gp130fl/fl and SNS-gp130−/− neuron cultures. In SNS-gp130−/− cultures, the number of neurons responding to NPPB was significantly decreased, and NPPB-responsive cells showed a significantly lower ratio increase to the stimulus. *p < 0.05. **p < 0.01. ***p < 0.001.
Figure 8.
Figure 8.
Attenuated CA-induced nocifensive behavior in SNS-gp130−/− mice. SNS-gp130−/− mice displayed a significantly shorter licking duration and reduced number of licks of their hindpaws in response to an injection of 20 μl 1% CA during the 5 min observation period. *p < 0.05. **p < 0.01.

References

    1. Abdel Samad O, Liu Y, Yang FC, Kramer I, Arber S, Ma Q. Characterization of two Runx1-dependent nociceptor differentiation programs necessary for inflammatory versus neuropathic pain. Mol Pain. 2010;6:45. doi: 10.1186/1744-8069-6-45.
    1. Agarwal N, Offermanns S, Kuner R. Conditional gene deletion in primary nociceptive neurons of trigeminal ganglia and dorsal root ganglia. Genesis. 2004;38:122–129. doi: 10.1002/gene.20010.
    1. Andratsch M, Mair N, Constantin CE, Scherbakov N, Benetti C, Quarta S, Vogl C, Sailer CA, Uceyler N, Brockhaus J, Martini R, Sommer C, Zeilhofer HU, Müller W, Kuner R, Davis JB, Rose-John S, Kress M. A key role for gp130 expressed on peripheral sensory nerves in pathological pain. J Neurosci. 2009;29:13473–13483. doi: 10.1523/JNEUROSCI.1822-09.2009.
    1. Bandell M, Story GM, Hwang SW, Viswanath V, Eid SR, Petrus MJ, Earley TJ, Patapoutian A. Noxious cold ion channel TRPA1 is activated by pungent compounds and bradykinin. Neuron. 2004;41:849–857. doi: 10.1016/S0896-6273(04)00150-3.
    1. Banner LR, Patterson PH, Allchorne A, Poole S, Woolf CJ. Leukemia inhibitory factor is an anti-inflammatory and analgesic cytokine. J Neurosci. 1998;18:5456–5462.
    1. Barabas ME, Kossyreva EA, Stucky CL. TRPA1 is functionally expressed primarily by IB4-binding, non-peptidergic mouse and rat sensory neurons. PLoS One. 2012;7:e47988. doi: 10.1371/journal.pone.0047988.
    1. Bauer S, Kerr BJ, Patterson PH. The neuropoietic cytokine family in development, plasticity, disease and injury. Nat Rev Neurosci. 2007;8:221–232. doi: 10.1038/nrn2054.
    1. Bautista DM, Movahed P, Hinman A, Axelsson HE, Sterner O, Högestätt ED, Julius D, Jordt SE, Zygmunt PM. Pungent products from garlic activate the sensory ion channel TRPA1. Proc Natl Acad Sci U S A. 2005;102:12248–12252. doi: 10.1073/pnas.0505356102.
    1. Bautista DM, Jordt SE, Nikai T, Tsuruda PR, Read AJ, Poblete J, Yamoah EN, Basbaum AI, Julius D. TRPA1 mediates the inflammatory actions of environmental irritants and proalgesic agents. Cell. 2006;124:1269–1282. doi: 10.1016/j.cell.2006.02.023.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300.
    1. Brenn D, Richter F, Schaible HG. Sensitization of unmyelinated sensory fibers of the joint nerve to mechanical stimuli by interleukin-6 in the rat: an inflammatory mechanism of joint pain. Arthritis Rheum. 2007;56:351–359. doi: 10.1002/art.22282.
    1. Brierley SM, Castro J, Harrington AM, Hughes PA, Page AJ, Rychkov GY, Blackshaw LA. TRPA1 contributes to specific mechanically activated currents and sensory neuron mechanical hypersensitivity. J Physiol. 2011;589:3575–3593. doi: 10.1113/jphysiol.2011.206789.
    1. Camprubí-Robles M, Mair N, Andratsch M, Benetti C, Beroukas D, Rukwied R, Langeslag M, Proia RL, Schmelz M, Ferrer Montiel AV, Haberberger RV, Kress M. Sphingosine-1-phosphate-induced nociceptor excitation and ongoing pain behavior in mice and humans is largely mediated by S1P3 receptor. J Neurosci. 2013;33:2582–2592. doi: 10.1523/JNEUROSCI.4479-12.2013.
    1. Caspani O, Zurborg S, Labuz D, Heppenstall PA. The contribution of TRPM8 and TRPA1 channels to cold allodynia and neuropathic pain. PLoS One. 2009;4:e7383. doi: 10.1371/journal.pone.0007383.
    1. Dai Y, Wang S, Tominaga M, Yamamoto S, Fukuoka T, Higashi T, Kobayashi K, Obata K, Yamanaka H, Noguchi K. Sensitization of TRPA1 by PAR2 contributes to the sensation of inflammatory pain. J Clin Invest. 2007;117:1979–1987. doi: 10.1172/JCI30951.
    1. Delmas P, Coste B. Mechano-gated ion channels in sensory systems. Cell. 2013;155:278–284. doi: 10.1016/j.cell.2013.09.026.
    1. Delmas P, Hao J, Rodat-Despoix L. Molecular mechanisms of mechanotransduction in mammalian sensory neurons. Nat Rev Neurosci. 2011;12:139–153. doi: 10.1038/nrn2993.
    1. Diogenes A, Akopian AN, Hargreaves KM. NGF up-regulates TRPA1: implications for orofacial pain. J Dent Res. 2007;86:550–555.
    1. Dittert I, Vlachová V, Knotková H, Vitásková Z, Vyklicky L, Kress M, Reeh PW. A technique for fast application of heated solutions of different composition to cultured neurones. J Neurosci Methods. 1998;82:195–201. doi: 10.1016/S0165-0270(98)00051-X.
    1. Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. J Clin Invest. 2010;120:3760–3772. doi: 10.1172/JCI42843.
    1. Eid SR, Crown ED, Moore EL, Liang HA, Choong KC, Dima S, Henze DA, Kane S, Urban MO. HC-030031, a TRPA1 selective antagonist, attenuates inflammatory- and neuropathy-induced mechanical hypersensitivity. Mol Pain. 2008;4:48. doi: 10.1186/1744-8069-4-48.
    1. Elitt CM, McIlwrath SL, Lawson JJ, Malin SA, Molliver DC, Cornuet PK, Koerber HR, Davis BM, Albers KM. Artemin overexpression in skin enhances expression of TRPV1 and TRPA1 in cutaneous sensory neurons and leads to behavioral sensitivity to heat and cold. J Neurosci. 2006;26:8578–8587. doi: 10.1523/JNEUROSCI.2185-06.2006.
    1. Franck MC, Stenqvist A, Li L, Hao J, Usoskin D, Xu X, Wiesenfeld-Hallin Z, Ernfors P. Essential role of Ret for defining non-peptidergic nociceptor phenotypes and functions in the adult mouse. Eur J Neurosci. 2011;33:1385–1400. doi: 10.1111/j.1460-9568.2011.07634.x.
    1. Gascon E, Gaillard S, Malapert P, Liu Y, Rodat-Despoix L, Samokhvalov IM, Delmas P, Helmbacher F, Maina F, Moqrich A. Hepatocyte growth factor-Met signaling is required for Runx1 extinction and peptidergic differentiation in primary nociceptive neurons. J Neurosci. 2010;30:12414–12423. doi: 10.1523/JNEUROSCI.3135-10.2010.
    1. Hatano N, Itoh Y, Suzuki H, Muraki Y, Hayashi H, Onozaki K, Wood IC, Beech DJ, Muraki K. Hypoxia-inducible factor-1α (HIF1α) switches on transient receptor potential ankyrin repeat 1 (TRPA1) gene expression via a hypoxia response element-like motif to modulate cytokine release. J Biol Chem. 2012;287:31962–31972. doi: 10.1074/jbc.M112.361139.
    1. Heinrich PC, Behrmann I, Haan S, Hermanns HM, Müller-Newen G, Schaper F. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J. 2003;374:1–20. doi: 10.1042/BJ20030407.
    1. Hjerling-Leffler J, Alqatari M, Ernfors P, Koltzenburg M. Emergence of functional sensory subtypes as defined by transient receptor potential channel expression. J Neurosci. 2007;27:2435–2443. doi: 10.1523/JNEUROSCI.5614-06.2007.
    1. Jordt SE, Bautista DM, Chuang HH, McKemy DD, Zygmunt PM, Högestätt ED, Meng ID, Julius D. Mustard oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature. 2004;427:260–265. doi: 10.1038/nature02282.
    1. Kim YS, Son JY, Kim TH, Paik SK, Dai Y, Noguchi K, Ahn DK, Bae YC. Expression of transient receptor potential ankyrin 1 (TRPA1) in the rat trigeminal sensory afferents and spinal dorsal horn. J Comp Neurol. 2010;518:687–698. doi: 10.1002/cne.22238.
    1. Kwan KY, Allchorne AJ, Vollrath MA, Christensen AP, Zhang DS, Woolf CJ, Corey DP. TRPA1 contributes to cold, mechanical, and chemical nociception but is not essential for hair-cell transduction. Neuron. 2006;50:277–289. doi: 10.1016/j.neuron.2006.03.042.
    1. Kwan KY, Glazer JM, Corey DP, Rice FL, Stucky CL. TRPA1 modulates mechanotransduction in cutaneous sensory neurons. J Neurosci. 2009;29:4808–4819. doi: 10.1523/JNEUROSCI.5380-08.2009.
    1. Langeslag M, Constantin CE, Andratsch M, Quarta S, Mair N, Kress M. Oncostatin M induces heat hypersensitivity by gp130-dependent sensitization of TRPV1 in sensory neurons. Mol Pain. 2011;7:102. doi: 10.1186/1744-8069-7-102.
    1. Langeslag M, Malsch P, Welling A, Kress M. Reduced excitability of gp130-deficient nociceptors is associated with increased voltage-gated potassium currents and Kcna4 channel upregulation. Pflugers Arch. 2014 doi: 10.1007/s00424-014-1443-0. doi: 10.1007/s00424-014-1443-0. Advance online publication. Retrieved Jan. 25, 2014.
    1. Liu K, Samuel M, Ho M, Harrison RK, Paslay JW. NPPB structure-specifically activates TRPA1 channels. Biochem Pharmacol. 2010;80:113–121. doi: 10.1016/j.bcp.2010.03.005.
    1. Loy B, Apostolova G, Dorn R, McGuire VA, Arthur JS, Dechant G. P38|gA and P38|gB mitogen-activated protein kinases determine cholinergic transdifferentiation of sympathetic neurons. J Neurosci. 2011;31:12059–12067. doi: 10.1523/JNEUROSCI.0448-11.2011.
    1. Lumpkin EA, Caterina MJ. Mechanisms of sensory transduction in the skin. Nature. 2007;445:858–865. doi: 10.1038/nature05662.
    1. Luo W, Wickramasinghe SR, Savitt JM, Griffin JW, Dawson TM, Ginty DD. A hierarchical NGF signaling cascade controls Ret-dependent and Ret-independent events during development of nonpeptidergic DRG neurons. Neuron. 2007;54:739–754. doi: 10.1016/j.neuron.2007.04.027.
    1. Macpherson LJ, Dubin AE, Evans MJ, Marr F, Schultz PG, Cravatt BF, Patapoutian A. Noxious compounds activate TRPA1 ion channels through covalent modification of cysteines. Nature. 2007;445:541–545. doi: 10.1038/nature05544.
    1. McNamara CR, Mandel-Brehm J, Bautista DM, Siemens J, Deranian KL, Zhao M, Hayward NJ, Chong JA, Julius D, Moran MM, Fanger CM. TRPA1 mediates formalin-induced pain. Proc Natl Acad Sci U S A. 2007;104:13525–13530. doi: 10.1073/pnas.0705924104.
    1. Melemedjian OK, Asiedu MN, Tillu DV, Peebles KA, Yan J, Ertz N, Dussor GO, Price TJ. IL-6- and NGF-induced rapid control of protein synthesis and nociceptive plasticity via convergent signaling to the eIF4F complex. J Neurosci. 2010;30:15113–15123. doi: 10.1523/JNEUROSCI.3947-10.2010.
    1. Merrill L, Girard BM, May V, Vizzard MA. Transcriptional and translational plasticity in rodent urinary bladder TRP channels with urinary bladder inflammation, bladder dysfunction, or postnatal maturation. J Mol Neurosci. 2012;48:744–756. doi: 10.1007/s12031-012-9867-5.
    1. Morikawa Y, Tamura S, Minehata K, Donovan PJ, Miyajima A, Senba E. Essential function of oncostatin m in nociceptive neurons of dorsal root ganglia. J Neurosci. 2004;24:1941–1947. doi: 10.1523/JNEUROSCI.4975-03.2004.
    1. Murphy M, Reid K, Hilton DJ, Bartlett PF. Generation of sensory neurons is stimulated by leukemia inhibitory factor. Proc Natl Acad Sci U S A. 1991;88:3498–3501. doi: 10.1073/pnas.88.8.3498.
    1. Murphy M, Reid K, Brown MA, Bartlett PF. Involvement of leukemia inhibitory factor and nerve growth factor in the development of dorsal root ganglion neurons. Development. 1993;117:1173–1182.
    1. Murphy M, Dutton R, Koblar S, Cheema S, Bartlett P. Cytokines which signal through the LIF receptor and their actions in the nervous system. Prog Neurobiol. 1997;52:355–378. doi: 10.1016/S0301-0082(97)00020-8.
    1. Nakashima K, Wiese S, Yanagisawa M, Arakawa H, Kimura N, Hisatsune T, Yoshida K, Kishimoto T, Sendtner M, Taga T. Developmental requirement of gp130 signaling in neuronal survival and astrocyte differentiation. J Neurosci. 1999;19:5429–5434.
    1. Nassini R, Gees M, Harrison S, De Siena G, Materazzi S, Moretto N, Failli P, Preti D, Marchetti N, Cavazzini A, Mancini F, Pedretti P, Nilius B, Patacchini R, Geppetti P. Oxaliplatin elicits mechanical and cold allodynia in rodents via TRPA1 receptor stimulation. Pain. 2011;152:1621–1631. doi: 10.1016/j.pain.2011.02.051.
    1. Nilius B, Appendino G, Owsianik G. The transient receptor potential channel TRPA1: from gene to pathophysiology. Pflugers Arch. 2012;464:425–458. doi: 10.1007/s00424-012-1158-z.
    1. Obata K, Katsura H, Mizushima T, Yamanaka H, Kobayashi K, Dai Y, Fukuoka T, Tokunaga A, Tominaga M, Noguchi K. TRPA1 induced in sensory neurons contributes to cold hyperalgesia after inflammation and nerve injury. J Clin Invest. 2005;115:2393–2401. doi: 10.1172/JCI25437.
    1. Obreja O, Schmelz M, Poole S, Kress M. Interleukin-6 in combination with its soluble IL-6 receptor sensitises rat skin nociceptors to heat, in vivo. Pain. 2002;96:57–62. doi: 10.1016/S0304-3959(01)00420-1.
    1. Obreja O, Biasio W, Andratsch M, Lips KS, Rathee PK, Ludwig A, Rose-John S, Kress M. Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons. Brain. 2005;128:1634–1641. doi: 10.1093/brain/awh490.
    1. Oh MH, Oh SY, Lu J, Lou H, Myers AC, Zhu Z, Zheng T. TRPA1-dependent pruritus in IL-13-induced chronic atopic dermatitis. J Immunol. 2013;191:5371–5382. doi: 10.4049/jimmunol.1300300.
    1. Pellegrino MJ, Habecker BA. STAT3 integrates cytokine and neurotrophin signals to promote sympathetic axon regeneration. Mol Cell Neurosci. 2013;56:272–282. doi: 10.1016/j.mcn.2013.06.005.
    1. Petrus M, Peier AM, Bandell M, Hwang SW, Huynh T, Olney N, Jegla T, Patapoutian A. A role of TRPA1 in mechanical hyperalgesia is revealed by pharmacological inhibition. Mol Pain. 2007;3:40. doi: 10.1186/1744-8069-3-40.
    1. Quarta S, Vogl C, Constantin CE, Üçeyler N, Sommer C, Kress M. Genetic evidence for an essential role of neuronally expressed IL-6 signal transducer gp130 in the induction and maintenance of experimentally induced mechanical hypersensitivity in vivo and in vitro. Mol Pain. 2011;7:73. doi: 10.1186/1744-8069-7-73.
    1. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–675. doi: 10.1038/nmeth.2089.
    1. Schwartz ES, Christianson JA, Chen X, La JH, Davis BM, Albers KM, Gebhart GF. Synergistic role of TRPV1 and TRPA1 in pancreatic pain and inflammation. Gastroenterology. 2011;140 doi: 10.1053/j.gastro.2010.12.033. 1283–1291.e1–2.
    1. Story GM, Peier AM, Reeve AJ, Eid SR, Mosbacher J, Hricik TR, Earley TJ, Hergarden AC, Andersson DA, Hwang SW, McIntyre P, Jegla T, Bevan S, Patapoutian A. ANKTM1, a TRP-like channel expressed in nociceptive neurons, is activated by cold temperatures. Cell. 2003;112:819–829. doi: 10.1016/S0092-8674(03)00158-2.
    1. Tamura S, Morikawa Y, Miyajima A, Senba E. Expression of oncostatin M receptor beta in a specific subset of nociceptive sensory neurons. Eur J Neurosci. 2003;17:2287–2298. doi: 10.1046/j.1460-9568.2003.02681.x.
    1. Thompson SW, Dray A, Urban L. Leukemia inhibitory factor induces mechanical allodynia but not thermal hyperalgesia in the juvenile rat. Neuroscience. 1996;71:1091–1094. doi: 10.1016/0306-4522(95)00537-4.
    1. Vilceanu D, Stucky CL. TRPA1 mediates mechanical currents in the plasma membrane of mouse sensory neurons. PLoS One. 2010;5:e12177. doi: 10.1371/journal.pone.0012177.
    1. Zhong J, Dietzel ID, Wahle P, Kopf M, Heumann R. Sensory impairments and delayed regeneration of sensory axons in interleukin-6-deficient mice. J Neurosci. 1999;19:4305–4313.

Source: PubMed

Подписаться