Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2017

Open Access 01-12-2017 | Research article

Tetraspanin 6: a pivotal protein of the multiple vesicular body determining exosome release and lysosomal degradation of amyloid precursor protein fragments

Authors: Francesc X. Guix, Ragna Sannerud, Fedor Berditchevski, Amaia M. Arranz, Katrien Horré, An Snellinx, Amantha Thathiah, Takaomi Saido, Takashi Saito, Sundaresan Rajesh, Michael Overduin, Samir Kumar-Singh, Enrico Radaelli, Nikky Corthout, Julien Colombelli, Sébastien Tosi, Sebastian Munck, Isabel H. Salas, Wim Annaert, Bart De Strooper

Published in: Molecular Neurodegeneration | Issue 1/2017

Login to get access

Abstract

Background

The mechanisms behind Aβ-peptide accumulation in non-familial Alzheimer’s disease (AD) remain elusive. Proteins of the tetraspanin family modulate Aβ production by interacting to γ-secretase.

Methods

We searched for tetraspanins with altered expression in AD brains. The function of the selected tetraspanin was studied in vitro and the physiological relevance of our findings was confirmed in vivo.

Results

Tetraspanin-6 (TSPAN6) is increased in AD brains and overexpression in cells exerts paradoxical effects on Amyloid Precursor Protein (APP) metabolism, increasing APP-C-terminal fragments (APP-CTF) and Aβ levels at the same time. TSPAN6 affects autophagosome-lysosomal fusion slowing down the degradation of APP-CTF. TSPAN6 recruits also the cytosolic, exosome-forming adaptor syntenin which increases secretion of exosomes that contain APP-CTF.

Conclusions

TSPAN6 is a key player in the bifurcation between lysosomal-dependent degradation and exosome mediated secretion of APP-CTF. This corroborates the central role of the autophagosomal/lysosomal pathway in APP metabolism and shows that TSPAN6 is a crucial player in APP-CTF turnover.
Appendix
Available only for authorised users
Literature
2.
3.
go back to reference De Strooper B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol Rev. 2010;90:465–94.CrossRefPubMed De Strooper B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol Rev. 2010;90:465–94.CrossRefPubMed
4.
go back to reference Chávez-Gutiérrez L, Bammens L, Benilova I, Vandersteen A, Benurwar M, Borgers M, et al. The mechanism of γ-secretase dysfunction in familial Alzheimer disease. EMBO J. 2012;31:2261–74.CrossRefPubMedPubMedCentral Chávez-Gutiérrez L, Bammens L, Benilova I, Vandersteen A, Benurwar M, Borgers M, et al. The mechanism of γ-secretase dysfunction in familial Alzheimer disease. EMBO J. 2012;31:2261–74.CrossRefPubMedPubMedCentral
5.
go back to reference Guix FX, Wahle T, Vennekens K, Snellinx A, Chávez-Gutiérrez L, Ill-Raga G, et al. Modification of γ-secretase by nitrosative stress links neuronal ageing to sporadic Alzheimer's disease. EMBO Mol Med. 2012;4:660–73.CrossRefPubMedPubMedCentral Guix FX, Wahle T, Vennekens K, Snellinx A, Chávez-Gutiérrez L, Ill-Raga G, et al. Modification of γ-secretase by nitrosative stress links neuronal ageing to sporadic Alzheimer's disease. EMBO Mol Med. 2012;4:660–73.CrossRefPubMedPubMedCentral
6.
go back to reference Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, et al. ApoE influences amyloid-β (Aβ) clearance despite minimalapoE/Aβ association in physiological conditions. Proc Natl Acad Sci U S A. 2013;110:1807–16.CrossRef Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, et al. ApoE influences amyloid-β (Aβ) clearance despite minimalapoE/Aβ association in physiological conditions. Proc Natl Acad Sci U S A. 2013;110:1807–16.CrossRef
7.
go back to reference Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E, et al. Clearance systems in the brain implications for Alzheimer disease. Nat Rev Neurol. 2015;11:457–70.CrossRefPubMedPubMedCentral Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E, et al. Clearance systems in the brain implications for Alzheimer disease. Nat Rev Neurol. 2015;11:457–70.CrossRefPubMedPubMedCentral
8.
go back to reference Zhao Z, Sagare AP, Ma Q, Halliday MR, Kong P, Kisler K, et al. Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance. Nat Neurosci. 2015;18:978–87.CrossRefPubMedPubMedCentral Zhao Z, Sagare AP, Ma Q, Halliday MR, Kong P, Kisler K, et al. Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance. Nat Neurosci. 2015;18:978–87.CrossRefPubMedPubMedCentral
9.
go back to reference Roh JH, Jiang H, Finn MB, Stewart FR, Mahan TE, Cirrito JR, et al. Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer’s disease. J Exp Med. 2014;211:2487–96.CrossRefPubMedPubMedCentral Roh JH, Jiang H, Finn MB, Stewart FR, Mahan TE, Cirrito JR, et al. Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer’s disease. J Exp Med. 2014;211:2487–96.CrossRefPubMedPubMedCentral
10.
go back to reference Sagare AP, Bell RD, Zlokovic BV. Neurovascular defects and faulty amyloid-beta vascular clearance in Alzheimer’s disease. Adv Alzheimer’s Dis. 2012;3:87–100. Sagare AP, Bell RD, Zlokovic BV. Neurovascular defects and faulty amyloid-beta vascular clearance in Alzheimer’s disease. Adv Alzheimer’s Dis. 2012;3:87–100.
11.
go back to reference Szaruga M, Veugelen S, Benurwar M, Lismont S, Sepulveda-Falla D, Lleo A, et al. Qualitative changes in human γ-secretase underlie familial Alzheimer’s disease. J Exp Med. 2015;212:2003–13.CrossRefPubMedPubMedCentral Szaruga M, Veugelen S, Benurwar M, Lismont S, Sepulveda-Falla D, Lleo A, et al. Qualitative changes in human γ-secretase underlie familial Alzheimer’s disease. J Exp Med. 2015;212:2003–13.CrossRefPubMedPubMedCentral
12.
go back to reference Marquer C, Laine J, Dauphinot L, Hanbouch L, Lemercier-Neuillet C, Pierrot N, et al. Increasing membrane cholesterol of neurons in culture recapitulates Alzheimer’s disease early phenotypes. Mol Neurodegener. 2014;9:60.CrossRefPubMedPubMedCentral Marquer C, Laine J, Dauphinot L, Hanbouch L, Lemercier-Neuillet C, Pierrot N, et al. Increasing membrane cholesterol of neurons in culture recapitulates Alzheimer’s disease early phenotypes. Mol Neurodegener. 2014;9:60.CrossRefPubMedPubMedCentral
13.
go back to reference Vetrivel KS, Cheng H, Kim SH, Chen Y, Barnes NY, Parent AT, et al. Spatial segregation of gamma-secretase and substrates in distinct membrane domains. J Biol Chem. 2005;280:25892–900.CrossRefPubMedPubMedCentral Vetrivel KS, Cheng H, Kim SH, Chen Y, Barnes NY, Parent AT, et al. Spatial segregation of gamma-secretase and substrates in distinct membrane domains. J Biol Chem. 2005;280:25892–900.CrossRefPubMedPubMedCentral
14.
go back to reference Rajendran L, Schneider A, Schlechtingen G, Weidlich S, Ries J, Braxmeier T, et al. Efficient inhibition of the Alzheimer’s disease beta-secretase by membrane targeting. Science. 2008;320:520–3.CrossRefPubMed Rajendran L, Schneider A, Schlechtingen G, Weidlich S, Ries J, Braxmeier T, et al. Efficient inhibition of the Alzheimer’s disease beta-secretase by membrane targeting. Science. 2008;320:520–3.CrossRefPubMed
15.
go back to reference Holmes O, Paturi S, Ye W, Wolfe MS, Selkoe DJ. Effects of membrane lipids on the activity and processivity of purified γ-secretase. Biochemistry. 2012;51:3565–75.CrossRefPubMedPubMedCentral Holmes O, Paturi S, Ye W, Wolfe MS, Selkoe DJ. Effects of membrane lipids on the activity and processivity of purified γ-secretase. Biochemistry. 2012;51:3565–75.CrossRefPubMedPubMedCentral
16.
go back to reference Thathiah A, Horré K, Snellinx A, Vandewyer E, Huang Y, Ciesielska M, et al. β-arrestin 2 regulates Aβ generation and γ-secretase activity in Alzheimer’s disease. Nat Med. 2013;19:43–9.CrossRefPubMed Thathiah A, Horré K, Snellinx A, Vandewyer E, Huang Y, Ciesielska M, et al. β-arrestin 2 regulates Aβ generation and γ-secretase activity in Alzheimer’s disease. Nat Med. 2013;19:43–9.CrossRefPubMed
17.
go back to reference Chen AC, Kim S, Shepardson N, Patel S, Hong S, Selkoe DJ. Physical and functional interaction between the α- and γ-secretases : a new model of regulated intramembrane proteolysis. J Cell Biol. 2015;211:1157–76.CrossRefPubMedPubMedCentral Chen AC, Kim S, Shepardson N, Patel S, Hong S, Selkoe DJ. Physical and functional interaction between the α- and γ-secretases : a new model of regulated intramembrane proteolysis. J Cell Biol. 2015;211:1157–76.CrossRefPubMedPubMedCentral
18.
go back to reference Odintsova E, Berditchevski F. Tetraspanins as regulators of protein trafficking. Traffic. 2007;8:89–96.CrossRefPubMed Odintsova E, Berditchevski F. Tetraspanins as regulators of protein trafficking. Traffic. 2007;8:89–96.CrossRefPubMed
19.
go back to reference Hemler ME. Tetraspanin functions and associated microdomains. Nat Rev Mol Cell Biol. 2005;6:801–11.CrossRefPubMed Hemler ME. Tetraspanin functions and associated microdomains. Nat Rev Mol Cell Biol. 2005;6:801–11.CrossRefPubMed
20.
go back to reference Wakabayashi T, Craessaerts K, Bammens L, Bentahir M, Borgions F, Herdewijn P, et al. Analysis of the gamma-secretase interactome and validation of its association with tetraspanin-enriched microdomains. Nat Cell Biol. 2009;11:1340–6.CrossRefPubMed Wakabayashi T, Craessaerts K, Bammens L, Bentahir M, Borgions F, Herdewijn P, et al. Analysis of the gamma-secretase interactome and validation of its association with tetraspanin-enriched microdomains. Nat Cell Biol. 2009;11:1340–6.CrossRefPubMed
21.
go back to reference Dunn CD, Sulis ML, Ferrando AA, Greenwald I. A conserved tetraspanin subfamily promotes notch signaling in Caenorhabditis elegans and in human cells. Proc Natl Acad Sci U S A. 2010;107:5907–12.CrossRefPubMedPubMedCentral Dunn CD, Sulis ML, Ferrando AA, Greenwald I. A conserved tetraspanin subfamily promotes notch signaling in Caenorhabditis elegans and in human cells. Proc Natl Acad Sci U S A. 2010;107:5907–12.CrossRefPubMedPubMedCentral
22.
go back to reference Dornier E, Coumailleau F, Ottavi JF, Moretti J, Boucheix C, Mauduit P, et al. TspanC8 tetraspanins regulate ADAM10/kuzbanian trafficking and promote notch activation in flies and mammals. J Cell Biol. 2012;199:481–96.CrossRefPubMedPubMedCentral Dornier E, Coumailleau F, Ottavi JF, Moretti J, Boucheix C, Mauduit P, et al. TspanC8 tetraspanins regulate ADAM10/kuzbanian trafficking and promote notch activation in flies and mammals. J Cell Biol. 2012;199:481–96.CrossRefPubMedPubMedCentral
23.
go back to reference Haining EJ, Yang J, Bailey RL, Khan K, Collier R, Tsai S, et al. The TspanC8 subgroup of tetraspanins interacts with a disintegrin and metalloprotease 10 (ADAM10) and regulates its maturation and cell surface expression. J Biol Chem. 2012;287:39753–65.CrossRefPubMedPubMedCentral Haining EJ, Yang J, Bailey RL, Khan K, Collier R, Tsai S, et al. The TspanC8 subgroup of tetraspanins interacts with a disintegrin and metalloprotease 10 (ADAM10) and regulates its maturation and cell surface expression. J Biol Chem. 2012;287:39753–65.CrossRefPubMedPubMedCentral
24.
go back to reference Jouannet S, Saint-Pol J, Fernandez L, Nguyen V, Charrin S, Boucheix C, et al. TspanC8 tetraspanins differentially regulate the cleavage of ADAM10 substrates, notch activation and ADAM10 membrane compartmentalization. Cell Mol Life Sci. 2015;73:1895–915.CrossRefPubMedPubMedCentral Jouannet S, Saint-Pol J, Fernandez L, Nguyen V, Charrin S, Boucheix C, et al. TspanC8 tetraspanins differentially regulate the cleavage of ADAM10 substrates, notch activation and ADAM10 membrane compartmentalization. Cell Mol Life Sci. 2015;73:1895–915.CrossRefPubMedPubMedCentral
25.
go back to reference Bassani S, Cingolani LA, Valnegri P, Folci A, Zapata J, Gianfelice A, et al. The X-linked intellectual disability protein TSPAN7 regulates excitatory synapse development and AMPAR trafficking. Neuron. 2012;73:1143–58.CrossRefPubMedPubMedCentral Bassani S, Cingolani LA, Valnegri P, Folci A, Zapata J, Gianfelice A, et al. The X-linked intellectual disability protein TSPAN7 regulates excitatory synapse development and AMPAR trafficking. Neuron. 2012;73:1143–58.CrossRefPubMedPubMedCentral
26.
go back to reference Charrin S, Le Naour F, Silvie O, Milhiet PE, Boucheix C, Rubinstein E. Lateral organization of membrane proteins: tetraspanins spin their web. Biochem J. 2009;420:133–54.CrossRefPubMed Charrin S, Le Naour F, Silvie O, Milhiet PE, Boucheix C, Rubinstein E. Lateral organization of membrane proteins: tetraspanins spin their web. Biochem J. 2009;420:133–54.CrossRefPubMed
28.
go back to reference Strohecker AM, Joshi S, Possemato R, Abraham RT, Sabatini DM, White E. Identification of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase as a novel autophagy regulator by high content shRNA screening. Oncogene. 2015;34:5662–76.CrossRefPubMedPubMedCentral Strohecker AM, Joshi S, Possemato R, Abraham RT, Sabatini DM, White E. Identification of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase as a novel autophagy regulator by high content shRNA screening. Oncogene. 2015;34:5662–76.CrossRefPubMedPubMedCentral
29.
go back to reference Bossers K, Wirz KT, Meerhoff GF, Essing AH, Van Dongen JW, Houba P, et al. Concerted changes in transcripts in the prefrontal cortex precede neuropathology in Alzheimer’s disease. Brain. 2010;133:3699–723.CrossRefPubMed Bossers K, Wirz KT, Meerhoff GF, Essing AH, Van Dongen JW, Houba P, et al. Concerted changes in transcripts in the prefrontal cortex precede neuropathology in Alzheimer’s disease. Brain. 2010;133:3699–723.CrossRefPubMed
30.
go back to reference Blalock EM, Geddes JW, Chen KC, Porter NM, Markesbery WR, Landfield PW. Incipient Alzheimer’s disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses. Proc Natl Acad Sci U S A. 2004;101:2173–8.CrossRefPubMedPubMedCentral Blalock EM, Geddes JW, Chen KC, Porter NM, Markesbery WR, Landfield PW. Incipient Alzheimer’s disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses. Proc Natl Acad Sci U S A. 2004;101:2173–8.CrossRefPubMedPubMedCentral
31.
go back to reference Miller JA, Woltjer RL, Goodenbour JM, Horvath S, Geschwind DH. Genes and pathways underlying regional and cell type changes in Alzheimer’s disease. Genome Med. 2013;5(5):48.CrossRefPubMedPubMedCentral Miller JA, Woltjer RL, Goodenbour JM, Horvath S, Geschwind DH. Genes and pathways underlying regional and cell type changes in Alzheimer’s disease. Genome Med. 2013;5(5):48.CrossRefPubMedPubMedCentral
32.
go back to reference Kumar-Singh S, Pirici D, McGowan E, Serneels S, Ceuterick C, Hardy J, Duff K, Dickson D, Van Broeckhoven C. Dense-core plaques in Tg2576 and PSAPP mouse models of Alzheimer’s disease are centered on vessel walls. Am J Pathol. 2005;167:527–43.CrossRefPubMedPubMedCentral Kumar-Singh S, Pirici D, McGowan E, Serneels S, Ceuterick C, Hardy J, Duff K, Dickson D, Van Broeckhoven C. Dense-core plaques in Tg2576 and PSAPP mouse models of Alzheimer’s disease are centered on vessel walls. Am J Pathol. 2005;167:527–43.CrossRefPubMedPubMedCentral
33.
go back to reference Zoncu R, Bar-Peled L, Efeyan A, Wang S, Sancak Y, Sabatini DM. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H-ATPase. Science. 2011;334:678–83.CrossRefPubMedPubMedCentral Zoncu R, Bar-Peled L, Efeyan A, Wang S, Sancak Y, Sabatini DM. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H-ATPase. Science. 2011;334:678–83.CrossRefPubMedPubMedCentral
34.
go back to reference Kakuda N, Funamoto S, Yagishita S, Takami M, Osawa S, Dohmae N, Ihara Y. Equimolar production of amyloid beta-protein and amyloid precursor protein intracellular domain from beta-carboxyl-terminal fragment by gamma-secretase. J Biol Chem. 2006;281:14776–86.CrossRefPubMed Kakuda N, Funamoto S, Yagishita S, Takami M, Osawa S, Dohmae N, Ihara Y. Equimolar production of amyloid beta-protein and amyloid precursor protein intracellular domain from beta-carboxyl-terminal fragment by gamma-secretase. J Biol Chem. 2006;281:14776–86.CrossRefPubMed
35.
go back to reference Sannerud R, Declerck I, Peric A, Raemaekers T, Menendez G, Zhou L, et al. ADP ribosylation factor 6 (ARF6) controls amyloid precursor protein (APP) processing by mediating the endosomal sorting of BACE1. Proc Natl Acad Sci U S A. 2011;108:E559–68.CrossRefPubMedPubMedCentral Sannerud R, Declerck I, Peric A, Raemaekers T, Menendez G, Zhou L, et al. ADP ribosylation factor 6 (ARF6) controls amyloid precursor protein (APP) processing by mediating the endosomal sorting of BACE1. Proc Natl Acad Sci U S A. 2011;108:E559–68.CrossRefPubMedPubMedCentral
36.
go back to reference Bolte S, Cordelières FP. A guided tour into subcellular colocalization analysis in light microscopy. J Microsc. 2006;224:213–32.CrossRefPubMed Bolte S, Cordelières FP. A guided tour into subcellular colocalization analysis in light microscopy. J Microsc. 2006;224:213–32.CrossRefPubMed
37.
go back to reference Kang BS, Cooper DR, Jelen F, Devedjiev Y, Derewenda U, Dauter Z, et al. PDZ tandem of human syntenin: crystal structure and functional properties. Structure. 2003;11:459–68.CrossRefPubMed Kang BS, Cooper DR, Jelen F, Devedjiev Y, Derewenda U, Dauter Z, et al. PDZ tandem of human syntenin: crystal structure and functional properties. Structure. 2003;11:459–68.CrossRefPubMed
38.
go back to reference Kraulis PJ, Domaille PJ, Campbell-Burk SL, Van Aken T, Laue ED. Solution structure and dynamics of ras p21. GDP determined by heteronuclear three- and four-dimensional NMR spectroscopy. Biochemistry. 1994;33:3515–31.CrossRefPubMed Kraulis PJ, Domaille PJ, Campbell-Burk SL, Van Aken T, Laue ED. Solution structure and dynamics of ras p21. GDP determined by heteronuclear three- and four-dimensional NMR spectroscopy. Biochemistry. 1994;33:3515–31.CrossRefPubMed
39.
go back to reference Latysheva N, Muratov G, Rajesh S, Padgett M, Hotchin NA, Overduin M, et al. Syntenin-1 is a new component of tetraspanin-enriched microdomains: mechanisms and consequences of the interaction of syntenin-1 with CD63. Mol Cell Biol. 2006;26:7707–18.CrossRefPubMedPubMedCentral Latysheva N, Muratov G, Rajesh S, Padgett M, Hotchin NA, Overduin M, et al. Syntenin-1 is a new component of tetraspanin-enriched microdomains: mechanisms and consequences of the interaction of syntenin-1 with CD63. Mol Cell Biol. 2006;26:7707–18.CrossRefPubMedPubMedCentral
40.
go back to reference Cierpicki T, Bushweller JH, Derewenda ZS. Probing the supramodular architecture of a multidomain protein: the structure of syntenin in solution. Structure. 2005;13:319–27.CrossRefPubMed Cierpicki T, Bushweller JH, Derewenda ZS. Probing the supramodular architecture of a multidomain protein: the structure of syntenin in solution. Structure. 2005;13:319–27.CrossRefPubMed
41.
go back to reference Saito T, Matsuba Y, Mihira N, Takano J, Nilsson P, Itohara S, et al. Single App knock-in mouse models of Alzheimer’s disease. Nat Neurosci. 2014;17:661–3.CrossRefPubMed Saito T, Matsuba Y, Mihira N, Takano J, Nilsson P, Itohara S, et al. Single App knock-in mouse models of Alzheimer’s disease. Nat Neurosci. 2014;17:661–3.CrossRefPubMed
42.
go back to reference Stenmark H, Parton RG, Steele-Mortimer O, Lütcke A, Gruenberg J, Zerial M. Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis. EMBO J. 1994;13:1287–96.PubMedPubMedCentral Stenmark H, Parton RG, Steele-Mortimer O, Lütcke A, Gruenberg J, Zerial M. Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis. EMBO J. 1994;13:1287–96.PubMedPubMedCentral
44.
go back to reference Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10:925–37.CrossRefPubMed Stuffers S, Sem Wegner C, Stenmark H, Brech A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 2009;10:925–37.CrossRefPubMed
45.
go back to reference Baietti MF, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14:677–85.CrossRefPubMed Baietti MF, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14:677–85.CrossRefPubMed
46.
go back to reference Doyle DA, Lee A, Lewis J, Kim E, Sheng M, MacKinnon R. Crystal structures of a complexed and peptide-free membrane protein-binding domain: molecular basis of peptide recognition by PDZ. Cell. 1996;85:1067–76.CrossRefPubMed Doyle DA, Lee A, Lewis J, Kim E, Sheng M, MacKinnon R. Crystal structures of a complexed and peptide-free membrane protein-binding domain: molecular basis of peptide recognition by PDZ. Cell. 1996;85:1067–76.CrossRefPubMed
47.
go back to reference Chen X, Li M, Chen D, Gao W, Guan JL, Komatsu M, et al. Autophagy induced by calcium phosphate precipitates involves endoplasmic reticulum membranes in autophagosome biogenesis. PLoS One. 2012;7:e52347.CrossRefPubMedPubMedCentral Chen X, Li M, Chen D, Gao W, Guan JL, Komatsu M, et al. Autophagy induced by calcium phosphate precipitates involves endoplasmic reticulum membranes in autophagosome biogenesis. PLoS One. 2012;7:e52347.CrossRefPubMedPubMedCentral
48.
go back to reference Orsi A, Razi M, Dooley HC, Robinson D, Weston AE, Collinson LM, et al. Dynamic and transient interactions of Atg9 with autophagosomes, but not membrane integration, are required for autophagy. Mol Biol Cell. 2012;23:1860–73.CrossRefPubMedPubMedCentral Orsi A, Razi M, Dooley HC, Robinson D, Weston AE, Collinson LM, et al. Dynamic and transient interactions of Atg9 with autophagosomes, but not membrane integration, are required for autophagy. Mol Biol Cell. 2012;23:1860–73.CrossRefPubMedPubMedCentral
49.
go back to reference Bruno P, Calastretti A, Priulla M, Asnaghi L, Scarlatti F, Nicolin A, et al. Cell survival under nutrient stress is dependent on metabolic conditions regulated by Akt and not by autophagic vacuoles. Cell Signal. 2007;19:2118–26.CrossRefPubMed Bruno P, Calastretti A, Priulla M, Asnaghi L, Scarlatti F, Nicolin A, et al. Cell survival under nutrient stress is dependent on metabolic conditions regulated by Akt and not by autophagic vacuoles. Cell Signal. 2007;19:2118–26.CrossRefPubMed
51.
go back to reference Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 1995;375:754–60.CrossRefPubMed Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 1995;375:754–60.CrossRefPubMed
52.
go back to reference Dobrowolski R, Vick P, Ploper D, Gumper I, Snitkin H, Sabatini DD, et al. Presenilin deficiency or lysosomal inhibition enhances Wnt signaling through relocalization of GSK3 to the late-endosomal compartment. Cell Rep. 2012;2:1316–28.CrossRefPubMedPubMedCentral Dobrowolski R, Vick P, Ploper D, Gumper I, Snitkin H, Sabatini DD, et al. Presenilin deficiency or lysosomal inhibition enhances Wnt signaling through relocalization of GSK3 to the late-endosomal compartment. Cell Rep. 2012;2:1316–28.CrossRefPubMedPubMedCentral
53.
go back to reference Esselens C, Oorschot V, Baert V, Raemaekers T, Spittaels K, Serneels L, et al. Presenilin 1 mediates the turnover of telencephalin in hippocampal neurons via an autophagic degradative pathway. J Cell Biol. 2004;166:1041–54.CrossRefPubMedPubMedCentral Esselens C, Oorschot V, Baert V, Raemaekers T, Spittaels K, Serneels L, et al. Presenilin 1 mediates the turnover of telencephalin in hippocampal neurons via an autophagic degradative pathway. J Cell Biol. 2004;166:1041–54.CrossRefPubMedPubMedCentral
54.
go back to reference Lee JH, Yu WH, Kumar A, Lee S, Mohan PS, Peterhoff CM, et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell. 2010;141:1146–58.CrossRefPubMedPubMedCentral Lee JH, Yu WH, Kumar A, Lee S, Mohan PS, Peterhoff CM, et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell. 2010;141:1146–58.CrossRefPubMedPubMedCentral
55.
go back to reference Jiang Y, Mullaney KA, Peterhoff CM, Che S, Schmidt SD, Boyer-Boiteau A, et al. Alzheimer’s-related endosome dysfunction in down syndrome is Aβ-independent but requires APP and is reversed by BACE-1 inhibition. Proc Natl Acad Sci U S A. 2010;107:1630–5.CrossRefPubMed Jiang Y, Mullaney KA, Peterhoff CM, Che S, Schmidt SD, Boyer-Boiteau A, et al. Alzheimer’s-related endosome dysfunction in down syndrome is Aβ-independent but requires APP and is reversed by BACE-1 inhibition. Proc Natl Acad Sci U S A. 2010;107:1630–5.CrossRefPubMed
56.
go back to reference Israel MA, Yuan Bardy SHC, Reyna SM, Mu Y, Herrera C, Hefferan MP, et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature. 2012;482:216–20.PubMedPubMedCentral Israel MA, Yuan Bardy SHC, Reyna SM, Mu Y, Herrera C, Hefferan MP, et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature. 2012;482:216–20.PubMedPubMedCentral
57.
go back to reference Kim S, Sato Y, Mohan PS, Peterhoff C, Pensalfini A, Rigoglioso A, et al. Evidence that the rab5 effector APPL1 mediates APP-βCTF-induced dysfunction of endosomes in down syndrome and Alzheimer’s disease. Mol Psychiatry. 2015;21:707–16.CrossRefPubMedPubMedCentral Kim S, Sato Y, Mohan PS, Peterhoff C, Pensalfini A, Rigoglioso A, et al. Evidence that the rab5 effector APPL1 mediates APP-βCTF-induced dysfunction of endosomes in down syndrome and Alzheimer’s disease. Mol Psychiatry. 2015;21:707–16.CrossRefPubMedPubMedCentral
58.
go back to reference Sannerud R, Esselens C, Ejsmont P, Mattera R, Rochin L, Tharkeshwar AK, et al. Restricted location of PSEN2 γ-secretase determines substrate specificity and generates an intracellular Aβ pool. Cell. 2016;8674:30568–2. Sannerud R, Esselens C, Ejsmont P, Mattera R, Rochin L, Tharkeshwar AK, et al. Restricted location of PSEN2 γ-secretase determines substrate specificity and generates an intracellular Aβ pool. Cell. 2016;8674:30568–2.
Metadata
Title
Tetraspanin 6: a pivotal protein of the multiple vesicular body determining exosome release and lysosomal degradation of amyloid precursor protein fragments
Authors
Francesc X. Guix
Ragna Sannerud
Fedor Berditchevski
Amaia M. Arranz
Katrien Horré
An Snellinx
Amantha Thathiah
Takaomi Saido
Takashi Saito
Sundaresan Rajesh
Michael Overduin
Samir Kumar-Singh
Enrico Radaelli
Nikky Corthout
Julien Colombelli
Sébastien Tosi
Sebastian Munck
Isabel H. Salas
Wim Annaert
Bart De Strooper
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2017
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-017-0165-0

Other articles of this Issue 1/2017

Molecular Neurodegeneration 1/2017 Go to the issue