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

      Striatal Mutant Huntingtin Protein Levels Decline with Age in Homozygous Huntington’s Disease Knock-In Mouse Models

      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.

          Abstract

          Background:

          Huntington’s disease (HD) is a progressive neurodegenerative disorder associated with aging, caused by an expanded polyglutamine (polyQ) repeat within the Huntingtin (HTT) protein. In HD, degeneration of the striatum and atrophy of the cortex are observed while cerebellum is less affected.

          Objective:

          To test the hypothesis that HTT protein levels decline with age, which together with HTT mutation could influence disease progression.

          Methods:

          Using whole brain cell lysates, a unique method of SDS-PAGE and western analysis was used to quantitate HTT protein, which resolves as a monomer and as a high molecular weight species that is modulated by the presence of transglutaminase 2. HTT levels were measured in striatum, cortex and cerebellum in congenic homozygous Q140 and HdhQ150 knock-in mice and WT littermate controls.

          Results:

          Mutant HTT in both homozygous knock-in HD mouse models and WT HTT in control striatal and cortical tissues significantly declined in a progressive manner over time. Levels of mutant HTT in HD cerebellum remained high during aging.

          Conclusions:

          A general decline in mutant HTT levels in striatum and cortex is observed that may contribute to disease progression in homozygous knock-in HD mouse models through reduction of HTT function. In cerebellum, sustained levels of mutant HTT with aging may be protective to this tissue which is less overtly affected in HD.

          Related collections

          Most cited references37

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

          Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila.

          Proteins with expanded polyglutamine repeats cause Huntington's disease and other neurodegenerative diseases. Transcriptional dysregulation and loss of function of transcriptional co-activator proteins have been implicated in the pathogenesis of these diseases. Huntington's disease is caused by expansion of a repeated sequence of the amino acid glutamine in the abnormal protein huntingtin (Htt). Here we show that the polyglutamine-containing domain of Htt, Htt exon 1 protein (Httex1p), directly binds the acetyltransferase domains of two distinct proteins: CREB-binding protein (CBP) and p300/CBP-associated factor (P/CAF). In cell-free assays, Httex1p also inhibits the acetyltransferase activity of at least three enzymes: p300, P/CAF and CBP. Expression of Httex1p in cultured cells reduces the level of the acetylated histones H3 and H4, and this reduction can be reversed by administering inhibitors of histone deacetylase (HDAC). In vivo, HDAC inhibitors arrest ongoing progressive neuronal degeneration induced by polyglutamine repeat expansion, and they reduce lethality in two Drosophila models of polyglutamine disease. These findings raise the possibility that therapy with HDAC inhibitors may slow or prevent the progressive neurodegeneration seen in Huntington's disease and other polyglutamine-repeat diseases, even after the onset of symptoms.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Full-length human mutant huntingtin with a stable polyglutamine repeat can elicit progressive and selective neuropathogenesis in BACHD mice.

            To elucidate the pathogenic mechanisms in Huntington's disease (HD) elicited by expression of full-length human mutant huntingtin (fl-mhtt), a bacterial artificial chromosome (BAC)-mediated transgenic mouse model (BACHD) was developed expressing fl-mhtt with 97 glutamine repeats under the control of endogenous htt regulatory machinery on the BAC. BACHD mice exhibit progressive motor deficits, neuronal synaptic dysfunction, and late-onset selective neuropathology, which includes significant cortical and striatal atrophy and striatal dark neuron degeneration. Power analyses reveal the robustness of the behavioral and neuropathological phenotypes, suggesting BACHD as a suitable fl-mhtt mouse model for preclinical studies. Additional analyses of BACHD mice provide novel insights into how mhtt may elicit neuropathogenesis. First, unlike previous fl-mhtt mouse models, BACHD mice reveal that the slowly progressive and selective pathogenic process in HD mouse brains can occur without early and diffuse nuclear accumulation of aggregated mhtt (i.e., as detected by immunostaining with the EM48 antibody). Instead, a relatively steady-state level of predominantly full-length mhtt and a small amount of mhtt N-terminal fragments are sufficient to elicit the disease process. Second, the polyglutamine repeat within fl-mhtt in BACHD mice is encoded by a mixed CAA-CAG repeat, which is stable in both the germline and somatic tissues including the cortex and striatum at the onset of neuropathology. Therefore, our results suggest that somatic repeat instability does not play a necessary role in selective neuropathogenesis in BACHD mice. In summary, the BACHD model constitutes a novel and robust in vivo paradigm for the investigation of HD pathogenesis and treatment.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              HDAC6 and microtubules are required for autophagic degradation of aggregated huntingtin.

              CNS neurons are endowed with the ability to recover from cytotoxic insults associated with the accumulation of proteinaceous polyglutamine aggregates via a process that appears to involve capture and degradation of aggregates by autophagy. The ubiquitin-proteasome system protects cells against proteotoxicity by degrading soluble monomeric misfolded aggregation-prone proteins but is ineffective against, and impaired by, non-native protein oligomers. Here we show that autophagy is induced in response to impaired ubiquitin proteasome system activity. We show that ATG proteins, molecular determinants of autophagic vacuole formation, and lysosomes are recruited to pericentriolar cytoplasmic inclusion bodies by a process requiring an intact microtubule cytoskeleton and the cytoplasmic deacetylase HDAC6. These data suggest that HDAC6-dependent retrograde transport on microtubules is used by cells to increase the efficiency and selectivity of autophagic degradation.
                Bookmark

                Author and article information

                Journal
                J Huntingtons Dis
                J Huntingtons Dis
                JHD
                Journal of Huntington's Disease
                IOS Press (Nieuwe Hemweg 6B, 1013 BG Amsterdam, The Netherlands )
                1879-6397
                1879-6400
                19 May 2018
                1 June 2018
                2018
                : 7
                : 2
                : 137-150
                Affiliations
                [a ] Department of Neurology, David Geffen School of Medicine, University of California Los Angeles , Los Angeles, CA, USA
                [b ] Centre for Integrative Biology, University of Trento , Trento, Italy
                [c ] Department of Neuroscience, University of Virginia School of Medicine , Charlottesville, VA, USA
                [d ] Department of Neurobiology and Behavior, University of California , Irvine, Irvine, CA, USA
                [e ] Burke Medical Research Institute , White Plains, NY, USA
                [f ]Brain and Mind Research Institute, Weill Medical College of Cornell University , New York, NY, USA
                [g ] Department of Neurology, Weill Medical College of Cornell University , New York, NY, USA
                [h ] Department of Psychiatry and Human Behavior, University of California , Irvine, Irvine, CA, USA
                [i ] Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee , Knoxville, TN, USA
                [j ] Institute of Memory Impairments and Neurological Disorders, University of California , Irvine, Irvine, CA, USA
                Author notes
                [* ]Corresponding author: Joan S. Steffan, PhD, Department of Psychiatry and Human Behavior, 3218 Biological Sciences III, University of California, Irvine, Irvine, CA 92697-4545, USA. Tel.: +1 949 824 6365; Fax: +1 949 824 2577; E-mail: jssteffa@ 123456uci.edu .
                Article
                JHD170274
                10.3233/JHD-170274
                6002862
                29843246
                552c9fe2-bd2b-4f7f-b3d5-03410fe7bd77
                © 2018 – IOS Press and the authors. All rights reserved

                This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial (CC BY-NC 4.0) License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                Categories
                Research Report

                autophagy,huntingtin,huntington’s disease,neurodegeneration

                Comments

                Comment on this article