9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Site-specific phosphorylation and caspase cleavage of GFAP are new markers of Alexander disease severity

      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

          Alexander disease (AxD) is a fatal neurodegenerative disorder caused by mutations in glial fibrillary acidic protein (GFAP), which supports the structural integrity of astrocytes. Over 70 GFAP missense mutations cause AxD, but the mechanism linking different mutations to disease-relevant phenotypes remains unknown. We used AxD patient brain tissue and induced pluripotent stem cell (iPSC)-derived astrocytes to investigate the hypothesis that AxD-causing mutations perturb key post-translational modifications (PTMs) on GFAP. Our findings reveal selective phosphorylation of GFAP-Ser13 in patients who died young, independently of the mutation they carried. AxD iPSC-astrocytes accumulated pSer13-GFAP in cytoplasmic aggregates within deep nuclear invaginations, resembling the hallmark Rosenthal fibers observed in vivo. Ser13 phosphorylation facilitated GFAP aggregation and was associated with increased GFAP proteolysis by caspase-6. Furthermore, caspase-6 was selectively expressed in young AxD patients, and correlated with the presence of cleaved GFAP. We reveal a novel PTM signature linking different GFAP mutations in infantile AxD.

          eLife digest

          Alexander disease is a rare and fatal neurodegenerative condition. It is caused by mutations in a gene that is crucial for the structure of astrocytes, a type of brain cell whose role is to support neurons. The gene codes for a protein called GFAP, which is made almost exclusively in astrocytes. In Alexander disease, mutated versions of the gene make GFAP collect in disordered clumps or aggregates, which interfere with the astrocytes’ normal activities.

          All Alexander disease patients develop GFAP aggregates, but the type of mutation they have in the gene for GFAP does not predict how their illness will progress. The age of onset of disease, for example, can vary between less than one year old to more than 70 years old. Battaglia et al. sought to understand how GFAP aggregates form in the cells of Alexander disease patients. One way that GFAP can be altered in the cell is by a process called phosphorylation. Enzymes called kinases add phosphate groups to GFAP, which can regulate the protein’s activity, stability and interactions with other proteins.

          Battaglia et al. found high levels of phosphorylation at one specific site in the GFAP protein in people who had very early onset of Alexander disease. This phosphorylation was not related to any particular mutation in the gene for GFAP. An added phosphate group at this location in the protein made GFAP more likely to be broken into two pieces by an enzyme called caspase-6. One of the breakdown products is already known to play a role in aggregation. Young patients with Alexander disease had high levels of GFAP breakdown products and caspase-6. The phosphorylated protein and this enzyme were found to accumulate in astrocyte aggregates.

          The findings provide a basis for investigating new strategies to treat Alexander disease that target phosphorylation – as removing or preventing the addition of phosphate groups can be done with drugs. But before exploring how to do this, it will be necessary to find out which enzyme is responsible for phosphorylating GFAP at this particular position. These studies may also give a broader understanding of other GFAP-like proteins (called intermediate filament proteins), which are involved in over 70 human diseases.

          Related collections

          Most cited references51

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

          Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system.

          Glial fibrillary acidic protein (GFAP) is the hallmark intermediate filament (IF; also known as nanofilament) protein in astrocytes, a main type of glial cells in the central nervous system (CNS). Astrocytes have a range of control and homeostatic functions in health and disease. Astrocytes assume a reactive phenotype in acute CNS trauma, ischemia, and in neurodegenerative diseases. This coincides with an upregulation and rearrangement of the IFs, which form a highly complex system composed of GFAP (10 isoforms), vimentin, synemin, and nestin. We begin to unravel the function of the IF system of astrocytes and in this review we discuss its role as an important crisis-command center coordinating cell responses in situations connected to cellular stress, which is a central component of many neurological diseases.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Cleavage at the caspase-6 site is required for neuronal dysfunction and degeneration due to mutant huntingtin.

            Cleavage of huntingtin (htt) has been characterized in vitro, and accumulation of caspase cleavage fragments represents an early pathological change in brains of Huntington's disease (HD) patients. However, the relationship between htt proteolysis and the pathogenesis of HD is unknown. To determine whether caspase cleavage of htt is a key event in the neuronal dysfunction and selective neurodegeneration in HD, we generated YAC mice expressing caspase-3- and caspase-6-resistant mutant htt. Mice expressing mutant htt, resistant to cleavage by caspase-6 but not caspase-3, maintain normal neuronal function and do not develop striatal neurodegeneration. Furthermore, caspase-6-resistant mutant htt mice are protected against neurotoxicity induced by multiple stressors including NMDA, quinolinic acid (QA), and staurosporine. These results are consistent with proteolysis of htt at the caspase-6 cleavage site being an important event in mediating neuronal dysfunction and neurodegeneration and highlight the significant role of htt proteolysis and excitotoxicity in HD.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Post-translational modifications of intermediate filament proteins: mechanisms and functions.

              Intermediate filaments (IFs) are cytoskeletal and nucleoskeletal structures that provide mechanical and stress-coping resilience to cells, contribute to subcellular and tissue-specific biological functions, and facilitate intracellular communication. IFs, including nuclear lamins and those in the cytoplasm (keratins, vimentin, desmin, neurofilaments and glial fibrillary acidic protein, among others), are functionally regulated by post-translational modifications (PTMs). Proteomic advances highlight the enormous complexity and regulatory potential of IF protein PTMs, which include phosphorylation, glycosylation, sumoylation, acetylation and prenylation, with novel modifications becoming increasingly appreciated. Future studies will need to characterize their on-off mechanisms, crosstalk and utility as biomarkers and targets for diseases involving the IF cytoskeleton.
                Bookmark

                Author and article information

                Contributors
                Role: Senior Editor
                Role: Reviewing Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                04 November 2019
                2019
                : 8
                : e47789
                Affiliations
                [1 ]deptDepartment of Cell Biology and Physiology University of North Carolina Chapel HillUnited States
                [2 ]deptDepartment of Pharmacology University of North Carolina Chapel HillUnited States
                [3 ]deptHuman Pluripotent Stem Cell Core University of North Carolina Chapel HillUnited States
                [4 ]deptDepartment of Biosciences University of Durham DurhamUnited Kingdom
                [5 ]deptDepartment of Pathology University of North Carolina Chapel HillUnited States
                [6 ]Thermo Fisher Scientific CarlsbadUnited States
                [7 ]deptDepartment of Pathology Columbia University New YorkUnited States
                [8 ]deptInstitute of Molecular Medicine National Tsing Hua University HsinchuTaiwan, Republic of China
                [9 ]deptDepartment of Physiology Mie University Graduate School of Medicine MieJapan
                Texas Children's Hospital United States
                Howard Hughes Medical Institute, Stanford University United States
                Howard Hughes Medical Institute, Stanford University United States
                Duke University Medical Center United States
                Author notes
                [†]

                Department of Neurology, Mayo clinic, Rochester, United States.

                Author information
                http://orcid.org/0000-0002-7909-7743
                https://orcid.org/0000-0003-0644-4123
                https://orcid.org/0000-0001-7663-4585
                Article
                47789
                10.7554/eLife.47789
                6927689
                31682229
                e5107338-553e-4362-b343-dee24659cf28
                © 2019, Battaglia et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 18 April 2019
                : 04 November 2019
                Funding
                Funded by: Elise's Corner Fund;
                Award ID: Research Grant
                Award Recipient :
                Funded by: United Leukodystrophy Foundation;
                Award ID: Research Grant
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000001, National Science Foundation;
                Award ID: Graduate Research Fellowship
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100007890, University of North Carolina at Chapel Hill;
                Award ID: Department of Cell Biology and Physiology
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: P30 CA016086
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Cell Biology
                Human Biology and Medicine
                Custom metadata
                GFAP protein carries a distinct post-translational modification signature that facilitates its pathogenic accumulation and aggregation in astrocytes of patients who succumb to Alexander Disease very early in life.

                Life sciences
                post-translational modification,protein aggregation,neurodegeneration,rare disease,induced pluripotent stem cells,astrocytes,human

                Comments

                Comment on this article