10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      PICK1 inhibits the E3 ubiquitin ligase activity of Parkin and reduces its neuronal protective effect

      research-article

      Read this article at

      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.

          Significance

          Parkinson’s disease (PD) is the second most common neurodegenerative disorder. It is characterized by progressive deterioration of motor function caused by loss of dopamine-producing neurons. Currently, there is no treatment that could stop the progress of the disease. Loss-of-function mutations in Parkin account for ∼50% of early onset PD. Parkin functions as an E3 ubiquitin ligase that exhibits multiple protective roles especially in dopaminergic neurons. Here, we demonstrate that PICK1 directly binds to Parkin. PICK1 is a potent endogenous inhibitor of Parkin’s E3 ubiquitin ligase activity and blocks Parkin’s protective functions. Conversely, knockout of PICK1 enhances the neuroprotective effect of Parkin. Thus, the reduction of PICK1 may provide a therapeutic target for the treatment of PD.

          Abstract

          Parkin functions as a multipurpose E3 ubiquitin ligase, and Parkin loss of function is associated with both sporadic and familial Parkinson’s disease (PD). We report that the Bin/Amphiphysin/Rvs (BAR) domain of protein interacting with PRKCA1 (PICK1) bound to the really interesting new gene 1 (RING1) domain of Parkin and potently inhibited the E3 ligase activity of Parkin by disrupting its interaction with UbcH7. Parkin translocated to damaged mitochondria and led to their degradation in neurons, whereas PICK1 robustly inhibited this process. PICK1 also impaired the protective function of Parkin against stresses in SH-SY5Y cells and neurons. The protein levels of several Parkin substrates were reduced in young PICK1-knockout mice, and these mice were resistant to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated toxicity. Taken together, the results indicate that PICK1 is a potent inhibitor of Parkin, and the reduction of PICK1 enhances the protective effect of Parkin.

          Related collections

          Most cited references34

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

          Association between early-onset Parkinson's disease and mutations in the parkin gene.

          Mutations in the parkin gene have recently been identified in patients with early-onset Parkinson's disease, but the frequency of the mutations and the associated phenotype have not been assessed in a large series of patients. We studied 73 families in which at least one of the affected family members was affected at or before the age of 45 years and had parents who were not affected, as well as 100 patients with isolated Parkinson's disease that began at or before the age of 45 years. All subjects were screened for mutations in the parkin gene with use of a semiquantitative polymerase-chain-reaction assay that simultaneously amplified several exons. We sequenced the coding exons in a subgroup of patients. We also compared the clinical features of patients with parkin mutations and those without mutations. Among the families with early-onset Parkinson's disease, 36 (49 percent) had parkin mutations. The age at onset ranged from 7 to 58 years. Among the patients with isolated Parkinson's disease, mutations were detected in 10 of 13 patients (77 percent) with an age at onset of 20 years or younger, but in only 2 of 64 patients (3 percent) with an age at onset of more than 30 years. The mean (+/-SD) age at onset in the patients with parkin mutations was younger than that in those without mutations (32+/-11 vs. 42+/-11 years, P<0.001), and they were more likely to have symmetric involvement and dystonia at onset, to have hyperreflexia at onset or later, to have a good response to levodopa therapy, and to have levodopa-induced dyskinesias during treatment. Nineteen different rearrangements of exons (deletions and multiplications) and 16 different point mutations were detected. Mutations in the parkin gene are a major cause of early-onset autosomal recessive familial Parkinson's disease and isolated juvenile-onset Parkinson's disease (at or before the age of 20 years). Accurate diagnosis of these cases cannot be based only on the clinical manifestations of the disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Selective autophagy: ubiquitin-mediated recognition and beyond.

            Eukaryotic cells use autophagy and the ubiquitin-proteasome system as their major protein degradation pathways. Whereas the ubiquitin-proteasome system is involved in the rapid degradation of proteins, autophagy pathways can selectively remove protein aggregates and damaged or excess organelles. Proteasome-mediated degradation requires previous ubiquitylation of the cargo, which is then recognized by ubiquitin receptors directing it to 26S proteasomes. Although autophagy has long been viewed as a random cytoplasmic degradation system, the involvement of ubiquitin as a specificity factor for selective autophagy is rapidly emerging. Recent evidence also suggests active crosstalk between proteasome-mediated degradation and selective autophagy. Here, we discuss the molecular mechanisms that link autophagy and the proteasome system, as well as the emerging roles of ubiquitin and ubiquitin-binding proteins in selective autophagy. On the basis of the evolutionary history of autophagic ubiquitin receptors, we propose a common origin for metazoan ubiquitin-dependent autophagy and the cytoplasm-to-vacuole targeting pathway of yeast.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Structure of parkin reveals mechanisms for ubiquitin ligase activation.

              Mutations in the PARK2 (parkin) gene are responsible for an autosomal recessive form of Parkinson's disease. The parkin protein is a RING-in-between-RING E3 ubiquitin ligase that exhibits low basal activity. We describe the crystal structure of full-length rat parkin. The structure shows parkin in an autoinhibited state and provides insight into how it is activated. RING0 occludes the ubiquitin acceptor site Cys(431) in RING2, whereas a repressor element of parkin binds RING1 and blocks its E2-binding site. Mutations that disrupted these inhibitory interactions activated parkin both in vitro and in cells. Parkin is neuroprotective, and these findings may provide a structural and mechanistic framework for enhancing parkin activity.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                24 July 2018
                9 July 2018
                9 July 2018
                : 115
                : 30
                : E7193-E7201
                Affiliations
                [1] aDivision of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology , Clear Water Bay, Kowloon, Hong Kong, China;
                [2] bSchool of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong, China;
                [3] cState Key Laboratory of Biopharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong , Pokfulam, Hong Kong, China
                Author notes
                3To whom correspondence should be addressed. Email: jxia@ 123456ust.hk .

                Edited by Solomon H. Snyder, Johns Hopkins University School of Medicine, Baltimore, MD, and approved June 21, 2018 (received for review September 19, 2017)

                Author contributions: J.H. and J.X. designed research; J.H., M.X., P.K.K.Y., J.L., Z.L., and S.K.C. performed research; M.X. measured the activity of MAO-B and did the in vitro binding, in vitro ubiquitination assay, and TH staining in striatum; M.X. and Z.L. did the cell death assay in ventral midbrain dopaminergic neurons; J.H., M.X., P.K.K.Y., and S.K.C. performed the MPTP assay; M.X., J.L., and K.K.C. measured dopamine and MPP+ levels; J.H. conducted the remaining experiments; K.K.C. contributed new reagents/analytic tools; J.H. and J.X. analyzed data; J.H. and J.X. wrote the paper; and J.H., M.X., P.K.K.Y., J.L., Z.L., K.K.C., S.K.C., and J.X. contributed to the discussion and commented on the manuscript.

                1J.H. and M.X. contributed equally to this work.

                2Present address: Department of Neurobiology, Systems Neuroscience Institute, University of Pittsburgh, Pittsburgh, PA 15261.

                Author information
                http://orcid.org/0000-0002-5366-5757
                Article
                201716506
                10.1073/pnas.1716506115
                6064985
                29987020
                3e65d0dc-90e5-4b92-8511-a55a9ec57cfe
                Copyright © 2018 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 9
                Funding
                Funded by: Research Grants Council, University Grants Committee (RGC, UGC) 501100002920
                Award ID: 16146516
                Award ID: 16102914
                Award ID: 663613
                Award ID: N_HKUST625/15
                Award ID: HKUST10/CRF/12R
                Award ID: C4011-14R
                Award ID: T13-607/12R and AoE/M-05/
                Award Recipient : Jun Xia
                Categories
                PNAS Plus
                Biological Sciences
                Neuroscience
                PNAS Plus

                parkinson’s disease,parkin,e3 ligase,pick1,bar domain
                parkinson’s disease, parkin, e3 ligase, pick1, bar domain

                Comments

                Comment on this article