18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      WNK1/HSN2 kinase inhibition ameliorates neuropathic pain by reducing maladaptive KCC2 inhibitory phosphorylation and restoring GABA inhibition

      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

          WNK1/HSN2 kinase, mutated in a Mendelian form of congenital pain insensitivity, contributes to a maladaptive decrease in KCC2 cotransporter activity and a loss of GABA inhibition in the spared nerve injury (SNI) model of neuropathic pain by increasing KCC2 inhibitory phosphorylation at Thr906/Thr1007. Antagonizing WNK1/HSN2 signaling reduces SNI-induced cold allodynia and mechanical hyperalgesia, decreases up-regulated KCC2 Thr906/Thr1007 phosphorylation, and normalizes pathological GABA depolarizations of injured spinal cord lamina II neurons. These data collectively provide novel mechanistic insight into, and a compelling therapeutic target for, neuropathic pain after nerve injury.

          Related collections

          Most cited references25

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

          Harnessing homologous recombination in vitro to generate recombinant DNA via SLIC.

          We describe a new cloning method, sequence and ligation-independent cloning (SLIC), which allows the assembly of multiple DNA fragments in a single reaction using in vitro homologous recombination and single-strand annealing. SLIC mimics in vivo homologous recombination by relying on exonuclease-generated ssDNA overhangs in insert and vector fragments, and the assembly of these fragments by recombination in vitro. SLIC inserts can also be prepared by incomplete PCR (iPCR) or mixed PCR. SLIC allows efficient and reproducible assembly of recombinant DNA with as many as 5 and 10 fragments simultaneously. SLIC circumvents the sequence requirements of traditional methods and functions much more efficiently at very low DNA concentrations when combined with RecA to catalyze homologous recombination. This flexibility allows much greater versatility in the generation of recombinant DNA for the purposes of synthetic biology.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Human hypertension caused by mutations in WNK kinases.

            Hypertension is a major public health problem of largely unknown cause. Here, we identify two genes causing pseudohypoaldosteronism type II, a Mendelian trait featuring hypertension, increased renal salt reabsorption, and impaired K+ and H+ excretion. Both genes encode members of the WNK family of serine-threonine kinases. Disease-causing mutations in WNK1 are large intronic deletions that increase WNK1 expression. The mutations in WNK4 are missense, which cluster in a short, highly conserved segment of the encoded protein. Both proteins localize to the distal nephron, a kidney segment involved in salt, K+, and pH homeostasis. WNK1 is cytoplasmic, whereas WNK4 localizes to tight junctions. The WNK kinases and their associated signaling pathway(s) may offer new targets for the development of antihypertensive drugs.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain.

              Modern pain-control theory predicts that a loss of inhibition (disinhibition) in the dorsal horn of the spinal cord is a crucial substrate for chronic pain syndromes. However, the nature of the mechanisms that underlie such disinhibition has remained controversial. Here we present evidence for a novel mechanism of disinhibition following peripheral nerve injury. It involves a trans-synaptic reduction in the expression of the potassium-chloride exporter KCC2, and the consequent disruption of anion homeostasis in neurons of lamina I of the superficial dorsal horn, one of the main spinal nociceptive output pathways. In our experiments, the resulting shift in the transmembrane anion gradient caused normally inhibitory anionic synaptic currents to be excitatory, substantially driving up the net excitability of lamina I neurons. Local blockade or knock-down of the spinal KCC2 exporter in intact rats markedly reduced the nociceptive threshold, confirming that the reported disruption of anion homeostasis in lamina I neurons was sufficient to cause neuropathic pain.
                Bookmark

                Author and article information

                Journal
                101465400
                34171
                Sci Signal
                Sci Signal
                Science signaling
                1945-0877
                1937-9145
                8 April 2017
                29 March 2016
                29 March 2016
                09 December 2017
                : 9
                : 421
                : ra32
                Affiliations
                [1 ]Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston (MA) 02115, USA
                [2 ]Department of Neurosurgery, Boston Children’s Hospital, Boston (MA), 02124 USA
                [3 ]Montreal Neurological Institute and Hospital, McGill University, Montréal (QC), H3A 2B4, Canada
                [4 ]Department of Neurology and Neurosurgery, McGill University, Montréal (QC), Canada
                [5 ]F.M. Kirby Neurobiology Center, Boston Children’s Hospital and Department of Neurobiology, Harvard Medical School, Boston (MA), 02115, USA
                [6 ]MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
                [7 ]Department of Psychology and Centre for Research on Pain, McGill University, Montréal (QC), H3A 0G1, Canada
                [8 ]Institut des Neurosciences Cellulaires et Integratives, UPR 3212 Centre National de la Recherche Scientifique, Strasbourg, France; Universite de Strasbourg, 5 rue Blaise Pascal, F-67084, Strasbourg, France
                Author notes
                [* ]To whom correspondence should be addressed: Guy A. Rouleau M.D., Ph.D., F.R.C.P.C., Phone: +1-514-398-2690, Fax: +1-514-398-8248, guy.rouleau@ 123456mcgill.ca ; Kristopher T. Kahle M.D., Ph.D., Phone: +1-203-737-2096, Fax: +1-203-785-2044, kristopher.kahle@ 123456yale.edu
                [†]

                These authors contributed equally to this work.

                [#]

                Current affiliation: Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology, Yale School of Medicine, New Haven (CT) 06511 USA

                Article
                PMC5723157 PMC5723157 5723157 nihpa862166
                10.1126/scisignal.aad0163
                5723157
                27025876
                be71eac0-e024-473d-83eb-cde685cc2271
                History
                Categories
                Article

                Comments

                Comment on this article