66
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      A subpopulation of nociceptors specifically linked to itch

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Itch-specific neurons have been sought for decades. The existence of such neurons is in doubt recently due to the observation that itch-mediating neurons also respond to painful stimuli. Here, we genetically labeled and manipulated MrgprA3 + neurons in dorsal root ganglion (DRG) and found that they exclusively innervate the epidermis of the skin and respond to multiple pruritogens. Ablation of MrgprA3 + neurons led to significant reductions in scratching evoked by multiple pruritogens and occurring spontaneously under chronic itch conditions whereas pain sensitivity remained intact. Importantly, mice with TRPV1 exclusively expressed in MrgprA3 + neurons exhibited only itch- and not pain behavior in response to capsaicin. Although MrgprA3 + neurons are sensitive to noxious heat, activation of TRPV1 in these neurons by noxious heat did not alter pain behavior. These data suggest that MrgprA3 defines a specific subpopulation of DRG neurons mediating itch. Our study opens new avenues for studying itch and developing anti-pruritic therapies.

          Related collections

          Most cited references39

          • Record: found
          • Abstract: found
          • Article: not found

          A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration.

          A new system for lineage ablation is based on transgenic expression of a diphtheria toxin receptor (DTR) in mouse cells and application of diphtheria toxin (DT). To streamline this approach, we generated Cre-inducible DTR transgenic mice (iDTR) in which Cre-mediated excision of a STOP cassette renders cells sensitive to DT. We tested the iDTR strain by crossing to the T cell- and B cell-specific CD4-Cre and CD19-Cre strains, respectively, and observed efficient ablation of T and B cells after exposure to DT. In MOGi-Cre/iDTR double transgenic mice expressing Cre recombinase in oligodendrocytes, we observed myelin loss after intraperitoneal DT injections. Thus, DT crosses the blood-brain barrier and promotes cell ablation in the central nervous system. Notably, we show that the developing DT-specific antibody response is weak and not neutralizing, and thus does not impede the efficacy of DT. Our results validate the use of iDTR mice as a tool for cell ablation in vivo.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            TRPA1 is required for histamine-independent, Mas-related G protein-coupled receptor-mediated itch

            SUMMARY Itch, the unpleasant sensation that evokes a desire to scratch, accompanies numerous skin and nervous system disorders. In many cases, pathological itch is insensitive to antihistamine treatment. Recent studies have identified members of the Mas-related GPCR (Mrgpr) family that are activated by mast cell mediators and promote histamine-independent itch. MrgprA3 and MrgprC11 act as receptors for the pruritogens chloroquine and BAM8–22, respectively. However, the signaling pathways and transduction channels activated downstream of these pruritogens are largely unknown. We found that TRPA1 is the downstream target of both MrgprA3 and MrgprC11, in cultured sensory neurons and heterologous cells. TRPA1 is required for Mrgpr-mediated signaling, as sensory neurons from TRPA1-deficient mice exhibited profoundly diminished responses to chloroquine and BAM8–22. Likewise, TRPA1-deficient mice displayed little to no scratching in response to these pruritogens. Our findings demonstrate that TRPA1 is an essential component of the signaling pathways that promote histamine-independent itch.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A diverse family of GPCRs expressed in specific subsets of nociceptive sensory neurons.

              In vertebrates, peripheral chemosensory neurons express large families of G protein-coupled receptors (GPCRs), reflecting the diversity and specificity of stimuli they detect. However, somatosensory neurons, which respond to chemical, thermal, or mechanical stimuli, are more broadly tuned. Here we describe a family of approximately 50 GPCRs related to Mas1, called mrgs, a subset of which is expressed in specific subpopulations of sensory neurons that detect painful stimuli. The expression patterns of mrgs thus reveal an unexpected degree of molecular diversity among nociceptive neurons. Some of these receptors can be specifically activated in heterologous cells by RFamide neuropeptides such as NPFF and NPAF, which are analgesic in vivo. Thus, mrgs may regulate nociceptor function and/or development, including the sensation or modulation of pain.
                Bookmark

                Author and article information

                Journal
                9809671
                21092
                Nat Neurosci
                Nat. Neurosci.
                Nature neuroscience
                1097-6256
                1546-1726
                30 November 2012
                23 December 2012
                February 2013
                01 August 2013
                : 16
                : 2
                : 174-182
                Affiliations
                [1 ]The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology
                [2 ]Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205
                [3 ]Department of Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China, 100005
                [4 ]Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06520
                [5 ]The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
                [6 ]Department of Anesthesiology & Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205
                Author notes
                [* ]Correspondence: Xinzhong Dong, The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N Wolfe Street, Baltimore, MD 21205, Phone: 410-502-2993, Fax: 410-614-6249, xdong2@ 123456jhmi.edu , Robert H. LaMotte, Department of Anesthesiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, Robert.lamotte@ 123456yale.edu
                Article
                NIHMS425086
                10.1038/nn.3289
                3557753
                23263443
                9858cd0d-5d71-4e58-8b55-c04b7c098f30

                Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Funding
                Funded by: National Institute of Neurological Disorders and Stroke : NINDS
                Award ID: R01 NS054791 || NS
                Funded by: National Institute of Neurological Disorders and Stroke : NINDS
                Award ID: R01 NS014624 || NS
                Funded by: National Institute of General Medical Sciences : NIGMS
                Award ID: R01 GM087369 || GM
                Categories
                Article

                Neurosciences
                Neurosciences

                Comments

                Comment on this article