9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Phosphorylated Presenilin 1 decreases β-amyloid by facilitating autophagosome–lysosome fusion

      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.

          Significance

          Autophagy is a catabolic process involving the formation of double-membrane–bound organelles called autophagosomes, which participate in the degradation of intracellular material through fusion with lysosomes. We have found a level of regulation of autophagosomal–lysosomal fusion where Presenilin 1 (PS1) phosphorylated at Ser367 specifically binds Annexin A2, which, through successive binding steps, facilitates this fusion. Lack of phosphorylation on PS1 1 Ser367 causes accumulation of partially fused autophagosomes and lysosomes in mouse brain and reduced autophagic flux. This inhibition of autophagy leads to decreased βCTF degradation and accumulation of toxic Aβ-peptide in the brain. This signaling pathway offers new potential drug targets for Alzheimer’s disease.

          Abstract

          Presenilin 1 (PS1), the catalytic subunit of the γ-secretase complex, cleaves βCTF to produce Aβ. We have shown that PS1 regulates Aβ levels by a unique bifunctional mechanism. In addition to its known role as the catalytic subunit of the γ-secretase complex, selective phosphorylation of PS1 on Ser367 decreases Aβ levels by increasing βCTF degradation through autophagy. Here, we report the molecular mechanism by which PS1 modulates βCTF degradation. We show that PS1 phosphorylated at Ser367, but not nonphosphorylated PS1, interacts with Annexin A2, which, in turn, interacts with the lysosomal N-ethylmaleimide–sensitive factor attachment protein receptor (SNARE) Vamp8. Annexin A2 facilitates the binding of Vamp8 to the autophagosomal SNARE Syntaxin 17 to modulate the fusion of autophagosomes with lysosomes. Thus, PS1 phosphorylated at Ser367 has an antiamyloidogenic function, promoting autophagosome–lysosome fusion and increasing βCTF degradation. Drugs designed to increase the level of PS1 phosphorylated at Ser367 should be useful in the treatment of Alzheimer’s disease.

          Related collections

          Most cited references19

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

          Calcium dyshomeostasis and intracellular signalling in Alzheimer's disease.

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

            ATG14 promotes membrane tethering and fusion of autophagosomes to endolysosomes.

            Autophagy, an important catabolic pathway implicated in a broad spectrum of human diseases, begins by forming double membrane autophagosomes that engulf cytosolic cargo and ends by fusing autophagosomes with lysosomes for degradation. Membrane fusion activity is required for early biogenesis of autophagosomes and late degradation in lysosomes. However, the key regulatory mechanisms of autophagic membrane tethering and fusion remain largely unknown. Here we report that ATG14 (also known as beclin-1-associated autophagy-related key regulator (Barkor) or ATG14L), an essential autophagy-specific regulator of the class III phosphatidylinositol 3-kinase complex, promotes membrane tethering of protein-free liposomes, and enhances hemifusion and full fusion of proteoliposomes reconstituted with the target (t)-SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) syntaxin 17 (STX17) and SNAP29, and the vesicle (v)-SNARE VAMP8 (vesicle-associated membrane protein 8). ATG14 binds to the SNARE core domain of STX17 through its coiled-coil domain, and stabilizes the STX17-SNAP29 binary t-SNARE complex on autophagosomes. The STX17 binding, membrane tethering and fusion-enhancing activities of ATG14 require its homo-oligomerization by cysteine repeats. In ATG14 homo-oligomerization-defective cells, autophagosomes still efficiently form but their fusion with endolysosomes is blocked. Recombinant ATG14 homo-oligomerization mutants also completely lose their ability to promote membrane tethering and to enhance SNARE-mediated fusion in vitro. Taken together, our data suggest an autophagy-specific membrane fusion mechanism in which oligomeric ATG14 directly binds to STX17-SNAP29 binary t-SNARE complex on autophagosomes and primes it for VAMP8 interaction to promote autophagosome-endolysosome fusion.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              An atomic structure of human γ-secretase.

              Dysfunction of the intramembrane protease γ-secretase is thought to cause Alzheimer's disease, with most mutations derived from Alzheimer's disease mapping to the catalytic subunit presenilin 1 (PS1). Here we report an atomic structure of human γ-secretase at 3.4 Å resolution, determined by single-particle cryo-electron microscopy. Mutations derived from Alzheimer's disease affect residues at two hotspots in PS1, each located at the centre of a distinct four transmembrane segment (TM) bundle. TM2 and, to a lesser extent, TM6 exhibit considerable flexibility, yielding a plastic active site and adaptable surrounding elements. The active site of PS1 is accessible from the convex side of the TM horseshoe, suggesting considerable conformational changes in nicastrin extracellular domain after substrate recruitment. Component protein APH-1 serves as a scaffold, anchoring the lone transmembrane helix from nicastrin and supporting the flexible conformation of PS1. Ordered phospholipids stabilize the complex inside the membrane. Our structure serves as a molecular basis for mechanistic understanding of γ-secretase function.
                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
                3 July 2017
                22 May 2017
                22 May 2017
                : 114
                : 27
                : 7148-7153
                Affiliations
                [1] aLaboratory of Molecular and Cellular Neuroscience, The Rockefeller University , New York, NY 10065;
                [2] bDepartment of Internal Medicine, Yale University School of Medicine , New Haven, CT 06520;
                [3] cDepartment of Cell Biology, Yale University School of Medicine , New Haven, CT 06520
                Author notes
                1To whom correspondence may be addressed. Email: vbustos@ 123456rockefeller.edu or greengard@ 123456rockefeller.edu .

                Contributed by Paul Greengard, April 27, 2017 (sent for review March 30, 2017; reviewed by Yue-Ming Li and Sangram S. Sisodia)

                Author contributions: V.B., M.V.P., M.F., F.S.G., and P.G. designed research; V.B., M.V.P., A.B., and A.L. performed research; V.B. and M.V.P. contributed new reagents/analytic tools; V.B., M.V.P., A.B., A.L., M.F., F.S.G., and P.G. analyzed data; and V.B., M.V.P., M.F., F.S.G., and P.G. wrote the paper.

                Reviewers: Y.-M.L., Memorial Sloan–Kettering Cancer Center; and S.S.S., The University of Chicago.

                Author information
                http://orcid.org/0000-0002-4437-0893
                Article
                PMC5502640 PMC5502640 5502640 201705240
                10.1073/pnas.1705240114
                5502640
                28533369
                8c0e90dd-c87f-4844-b4da-49639c582e27

                Freely available online through the PNAS open access option.

                History
                Page count
                Pages: 6
                Funding
                Funded by: Fisher Center for Alzheimer's Research Foundation 100001621
                Award ID: 001
                Funded by: HHS | National Institutes of Health (NIH) 100000002
                Award ID: AG047781
                Funded by: U.S. Department of Defense (DOD) 100000005
                Award ID: W81XWH-09-1-0402
                Funded by: U.S. Department of Defense (DOD) 100000005
                Award ID: W81XWH-14-1-0045
                Funded by: HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) 100000062
                Award ID: DK098108
                Funded by: JPB Foundation 100007457
                Award ID: 475
                Categories
                Biological Sciences
                Neuroscience
                From the Cover

                autophagosome–lysosome fusion,Presenilin 1,phosphorylation,autophagy,Annexin A2

                Comments

                Comment on this article