Extracellular Vesicle as a Source of Alzheimer’s Biomarkers: Opportunities and Challenges
Abstract
:1. Extracellular Vesicles: An Overview
1.1. Biogenesis and Secretion
1.2. Constituents of EVs: Proteins, miRNAs, Lipids and other Nucleic Acids
2. Physiological Role of EVs in Brain
3. Alzheimer’s Disease (AD), a Proteinopathy
4. Pathological Roles of Exosomes in AD
5. Biochemical Biomarkers for Early Diagnosis of Alzheimer’s Disease
5.1. Non-Amyloid Protein Biomarkers in CSF
5.2. Protein Biomarkers in Plasma and Circulating EVs
5.3. microRNA in EVs of Plasma and CSF
6. Consideration to Develop AD Biomarkers in EVs
6.1. Standardization of Pre-Analytical Sources of Variability for EV AD Biomarkers
6.2. Yield and Purity of Isolated EVs from Biofluid
7. Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Sharma, P.; Schiapparelli, L.; Cline, H.T. Exosomes function in cell-cell communication during brain circuit development. Curr. Opin. Neurobiol. 2013, 23, 997–1004. [Google Scholar] [CrossRef] [Green Version]
- Faure’, J.; Lachenal, G.; Court, M.; Hirrlinger, J.; Chatellard-Causse, C.; Blot, B.; Grange, J.; Schoehn, G.; Goldberg, Y.; Boyer, V.; et al. Exosomes are released by cultured cortical neurones. Mol. Cell. Neurosci. 2006, 31, 642–648. [Google Scholar] [CrossRef]
- Janas, A.M.; Sapoń, K.; Janas, T.; Stowell, M.H.; Janas, T. Exosomes and other extracellular vesicles in neural cells and neurodegenerative diseases. Biochim. Biophys. Acta-Biomembr. 2016, 1858, 1139–1151. [Google Scholar] [CrossRef]
- Von Bartheld, C.S.; Altick, A.L. Multivesicular bodies in neurons: Distribution, protein content and trafficking functions. Prog. Neurobiol. 2011, 93, 313–340. [Google Scholar] [CrossRef] [PubMed]
- Bronisz, A.; Wang, Y.; Nowicki, M.O.; Peruzzi, P.; Ansari, K.I.; Ogawa, D.; Balaj, L.; De Rienzo, G.; Mineo, M.; Nakano, I.; et al. Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res. 2014, 74, 738–750. [Google Scholar] [CrossRef]
- Surgucheva, I.; Sharov, V.S.; Surguchov, A. γ-Synuclein: Seeding of α-synuclein aggregation and transmission between cells. Biochemistry 2012, 51, 4743–4754. [Google Scholar] [CrossRef]
- van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
- Budnik, V.; Ruiz-Cañada, C.; Wendler, F. Extracellular vesicles round off communication in the nervous system. Nat. Rev. Neurosci. 2016, 17, 160–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Babst, M. MVB vesicle formation: ESCRT-dependent, ESCRT-independent and everything in between. Curr. Opin. Cell Biol. 2011, 23, 452–457. [Google Scholar] [CrossRef] [PubMed]
- Hurley, J.H. ESCRTs are everywhere. EMBO J. 2015, 34, 2398–2407. [Google Scholar] [CrossRef] [Green Version]
- Daleke, D.L. Regulation of transbilayer plasma membrane phospholipid asymmetry. J. Lipid Res. 2003, 44, 233–242. [Google Scholar] [CrossRef]
- Stachowiak, J.C.; Brodsky, F.M.; Miller, E.A. A cost—Benefit analysis of the physical mechanisms of membrane curvature. Nat. Cell Biol. 2013, 15, 1019–1027. [Google Scholar] [CrossRef]
- Mcmahon, H.T.; Boucrot, E. Membrane curvature at a glance. J. Cell Sci. 2015, 128, 1065–1070. [Google Scholar] [CrossRef] [Green Version]
- Muralidharan-Chari, V.; Clancy, J.; Plou, C.; Romao, M.; Chavrier, P.; Raposo, G.; D’Souza-Schorey, C. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr. Biol. 2009, 19, 1875–1885. [Google Scholar] [CrossRef]
- Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef]
- Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
- Maas, S.L.; Breakefield, X.O.; Weaver, A.M. Extracellular vesicles: Unique intercellular delivery vehicles. Trends Cell Biol. 2017, 27, 172–188. [Google Scholar] [CrossRef]
- Colombo, M.; Moita, C.; van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Théry, C.; Raposo, G. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 2013, 126, 5553–5565. [Google Scholar] [CrossRef] [Green Version]
- van Niel, G.; Charrin, S.; Simoes, S.; Romao, M.; Rochin, L.; Saftig, P.; Marks, M.S.; Rubinstein, E.; Raposo, G. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev. Cell 2011, 21, 708–721. [Google Scholar] [CrossRef]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
- Stuffers, S.; Sem Wegner, C.; Stenmark, H.; Brech, A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic 2009, 10, 925–937. [Google Scholar] [CrossRef]
- Tricarico, C.; Clancy, J.; D’Souza-Schorey, C. Biology and biogenesis of shed microvesicles. Small GTPases 2017, 8, 220–232. [Google Scholar] [CrossRef]
- Buschow, S.I.; Nolte-‘t Hoen, E.N.; van Niel, G.; Pols, M.S.; ten Broeke, T.; Lauwen, M.; Ossendorp, F.; Melief, C.J.; Raposo, G.; Wubbolts, R.; et al. MHC II in dendritic cells is targeted to lysosomes or T cell-induced exosomes via distinct multivesicular body pathways. Traffic 2009, 10, 1528–1542. [Google Scholar] [CrossRef]
- Piper, R.C.; Luzio, J.P.; Luzio, J.P. Late endosomes: Sorting and partitioning in multivesicular bodies. Traffic 2001, 2, 612–621. [Google Scholar] [CrossRef]
- Stenmark, H. Rab GTPases as coordinators of vesicle traffic. Nat. Rev. Mol. Cell Biol. 2009, 10, 513–525. [Google Scholar] [CrossRef]
- Hsu, C.; Morohashi, Y.; Yoshimura, S.I.; Manrique-Hoyos, N.; Jung, S.Y.; Lauterbach, M.A.; Bakhti, M.; Grønborg, M.; Möbius, W.; Rhee, J.S.; et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 2010, 189, 223–232. [Google Scholar] [CrossRef]
- Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef]
- Jahn, R.; Scheller, R.H. SNAREs—Engines for membrane fusion. Nat. Rev. Mol. Cell Biol. 2006, 7, 631–643. [Google Scholar] [CrossRef]
- Mulcahy, L.A.; Pink, R.C.; Carter, D.R.F. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef]
- Moon, P.-G.; Lee, J.-E.; Cho, Y.-E.; Lee, S.J.; Chae, Y.S.; Jung, J.H.; Kim, I.-S.; Park, H.Y.; Baek, M.-C. Fibronectin on circulating extracellular vesicles as a liquid biopsy to detect breast cancer. Oncotarget 2016, 7, 40189–40199. [Google Scholar] [CrossRef] [Green Version]
- Soung, Y.H.; Ford, S.; Zhang, V.; Chung, J. Exosomes in cancer diagnostics. Cancers 2017, 9, 8. [Google Scholar] [CrossRef]
- D’Alessandra, Y.; Devanna, P.; Limana, F.; Straino, S.; Di Carlo, A.; Brambilla, P.G.; Rubino, M.; Carena, M.C.; Spazzafumo, L.; De Simone, M.; et al. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur. Heart J. 2010, 31, 2765–2773. [Google Scholar] [CrossRef] [Green Version]
- Saman, S.; Kim, W.; Raya, M.; Visnick, Y.; Miro, S.; Saman, S.; Jackson, B.; McKee, A.C.; Alvarez, V.E.; Lee, N.C.Y.; et al. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J. Biol. Chem. 2012, 287, 3842–3849. [Google Scholar] [CrossRef]
- Bunggulawa, E.J.; Wang, W.; Yin, T.; Wang, N.; Durkan, C.; Wang, Y.; Wang, G. Recent advancements in the use of exosomes as drug delivery systems. J. Nanobiotechnol. 2018, 16, 81. [Google Scholar] [CrossRef]
- Willms, E.; Johansson, H.J.; Mäger, I.; Lee, Y.; Blomberg, K.E.M.; Sadik, M.; Alaarg, A.; Smith, C.I.E.; Lehtiö, J.; El Andaloussi, S.; et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci. Rep. 2016, 6, 22519. [Google Scholar] [CrossRef] [Green Version]
- Pisitkun, T.; Shen, R.-F.; Knepper, M.A. Identification and proteomic profiling of exosomes in human urine. Proc. Natl. Acad. Sci. USA 2004, 101, 13368–13373. [Google Scholar] [CrossRef] [Green Version]
- Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef]
- Subra, C.; Laulagnier, K.; Perret, B.; Record, M. Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies. Biochimie 2007, 89, 205–212. [Google Scholar] [CrossRef]
- Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Théry, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977. [Google Scholar] [CrossRef] [Green Version]
- Simpson, R.J.; Jensen, S.S.; Lim, J.W.E. Proteomic profiling of exosomes: Current perspectives. Proteomics 2008, 8, 4083–4099. [Google Scholar] [CrossRef]
- Chiasserini, D.; van Weering, J.R.T.; Piersma, S.R.; Pham, T.V.; Malekzadeh, A.; Teunissen, C.E.; de Wit, H.; Jiménez, C.R. Proteomic analysis of cerebrospinal fluid extracellular vesicles: A comprehensive dataset. J. Proteomics 2014, 106, 191–204. [Google Scholar] [CrossRef]
- Zhang, J.; Li, S.; Li, L.; Li, M.; Guo, C.; Yao, J.; Mi, S. Exosome and exosomal microRNA: Trafficking, sorting and function. Genom. Proteomics Bioinform. 2015, 13, 17–24. [Google Scholar] [CrossRef]
- Vlassov, A.V.; Magdaleno, S.; Setterquist, R.; Conrad, R. Exosomes: Current knowledge of their composition, biological functions and diagnostic and therapeutic potentials. Biochim. Biophys. Acta-Gen. Subj. 2012, 1820, 940–948. [Google Scholar] [CrossRef]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
- Thind, A.; Wilson, C. Exosomal miRNAs as cancer biomarkers and therapeutic targets. J. Extracell. Vesicles 2016, 5, 31292. [Google Scholar] [CrossRef]
- Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef]
- Rabinowits, G.; Gerçel-Taylor, C.; Day, J.M.; Taylor, D.D.; Kloecker, G.H. Exosomal microRNA: A diagnostic marker for lung cancer. Clin. Lung Cancer 2009, 10, 42–46. [Google Scholar] [CrossRef]
- Mathivanan, S.; Simpson, R.J. ExoCarta: A compendium of exosomal proteins and RNA. Proteomics 2009, 9, 4997–5000. [Google Scholar] [CrossRef]
- Kim, D.K.; Kang, B.; Kim, O.Y.; Choi, D.S.; Lee, J.; Kim, S.R.; Go, G.; Yoon, Y.J.; Kim, J.H.; Jang, S.C.; et al. EVpedia: An integrated database of high-throughput data for systemic analyses of extracellular vesicles. J. Extracell. Vesicles 2013, 2, 20384. [Google Scholar] [CrossRef]
- Iraci, N.; Leonardi, T.; Gessler, F.; Vega, B.; Pluchino, S. Focus on extracellular vesicles: Physiological role and signalling properties of extracellular membrane vesicles. Int. J. Mol. Sci. 2016, 17, 171. [Google Scholar] [CrossRef]
- Frühbeis, C.; Fröhlich, D.; Kuo, W.P.; Krämer-Albers, E.-M. Extracellular vesicles as mediators of neuron-glia communication. Front. Cell. Neurosci. 2013, 7, 182. [Google Scholar] [CrossRef]
- Rajendran, L.; Bali, J.; Barr, M.M.; Court, F.A.; Krämer-Albers, E.-M.; Picou, F.; Raposo, G.; van der Vos, K.E.; van Niel, G.; Wang, J.; et al. Emerging Roles of Extracellular Vesicles in the Nervous System. J. Neurosci. 2014, 34, 15482–15489. [Google Scholar] [CrossRef] [Green Version]
- Morris, G.P.; Clark, I.A.; Zinn, R.; Vissel, B. Microglia: A new frontier for synaptic plasticity, learning and memory and neurodegenerative disease research. Neurobiol. Learn. Mem. 2013, 105, 40–53. [Google Scholar] [CrossRef]
- Bahrini, I.; Song, J.H.; Diez, D.; Hanayama, R. Neuronal exosomes facilitate synaptic pruning by up-regulating complement factors in microglia. Sci. Rep. 2015, 5, 7989. [Google Scholar] [CrossRef] [Green Version]
- Frühbeis, C.; Fröhlich, D.; Kuo, W.P.; Amphornrat, J.; Thilemann, S.; Saab, A.S.; Kirchhoff, F.; Möbius, W.; Goebbels, S.; Nave, K.A.; et al. Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 2013, 11, e1001604. [Google Scholar] [CrossRef]
- Morel, L.; Regan, M.; Higashimori, H.; Ng, S.K.; Esau, C.; Vidensky, S.; Rothstein, J.; Yang, Y. Neuronal exosomal miRNA-dependent translational regulation of astroglial glutamate transporter GLT1. J. Biol. Chem. 2013, 288, 7105–7116. [Google Scholar] [CrossRef]
- Chivet, M.; Javalet, C.; Laulagnier, K.; Blot, B.; Hemming, F.J.; Sadoul, R. Exosomes secreted by cortical neurons upon glutamatergic synapse activation specifically interact with neurons. J. Extracell. Vesicles 2014, 3, 24722. [Google Scholar] [CrossRef]
- Koniusz, S.; Andrzejewska, A.; Muraca, M.; Srivastava, A.K.; Janowski, M.; Lukomska, B. Extracellular vesicles in physiology, pathology and therapy of the immune and central nervous system, with focus on extracellular vesicles derived from mesenchymal stem cells as therapeutic tools. Front. Cell. Neurosci. 2016, 10, 109. [Google Scholar] [CrossRef]
- Lachenal, G.; Pernet-Gallay, K.; Chivet, M.; Hemming, F.J.; Belly, A.; Bodon, G.; Blot, B.; Haase, G.; Goldberg, Y.; Sadoul, R. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol. Cell. Neurosci. 2010, 46, 409–418. [Google Scholar] [CrossRef]
- Drago, F.; Lombardi, M.; Prada, I.; Gabrielli, M.; Joshi, P.; Cojoc, D.; Franck, J.; Fournier, I.; Vizioli, J.; Verderio, C. ATP modifies the proteome of extracellular vesicles released by microglia and influences their action on astrocytes. Front. Pharmacol. 2017, 8, 910. [Google Scholar] [CrossRef]
- Krämer-Albers, E.-M.; Bretz, N.; Tenzer, S.; Winterstein, C.; Möbius, W.; Berger, H.; Nave, K.-A.; Schild, H.; Trotter, J. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? Proteomics Clin. Appl. 2007, 1, 1446–1461. [Google Scholar] [CrossRef]
- Danzer, K.M.; Kranich, L.R.; Ruf, W.P.; Cagsal-Getkin, O.; Winslow, A.R.; Zhu, L.; Vanderburg, C.R.; McLean, P.J. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 2012, 7, 42. [Google Scholar] [CrossRef]
- Vingtdeux, V.; Hamdane, M.; Loyens, A.; Gelé, P.; Drobeck, H.; Bégard, S.; Galas, M.C.; Delacourte, A.; Beauvillain, J.C.; Buée, L.; et al. Alkalizing drugs induce accumulation of amyloid precursor protein by-products in luminal vesicles of multivesicular bodies. J. Biol. Chem. 2007, 282, 18197–18205. [Google Scholar] [CrossRef]
- Dinkins, M.B.; Enasko, X.J.; Hernandez, C.; Wang, G.; Kong, J.; Helwa, I.; Liu, Y.; Terry, A.V.; Bieberich, E. Neutral sphingomyelinase-2 deficiency ameliorates Alzheimer’s disease pathology and improves cognition in the 5XFAD mouse. J. Neurosci. 2016, 36, 8653–8667. [Google Scholar] [CrossRef]
- Sinha, M.S.; Ansell-Schultz, A.; Civitelli, L.; Hildesjö, C.; Larsson, M.; Lannfelt, L.; Ingelsson, M.; Hallbeck, M. Alzheimer’s disease pathology propagation by exosomes containing toxic amyloid-beta oligomers. Acta Neuropathol. 2018, 136, 41–56. [Google Scholar] [CrossRef] [Green Version]
- Asai, H.; Ikezu, S.; Tsunoda, S.; Medalla, M.; Luebke, J.; Haydar, T.; Wolozin, B.; Butovsky, O.; Kügler, S.; Ikezu, T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015, 18, 1584–1593. [Google Scholar] [CrossRef] [Green Version]
- Spires-Jones, T.L.; Hyman, B.T. The intersection of amyloid beta and tau at synapses in Alzheimer’s disease. Neuron 2014, 82, 756–771. [Google Scholar] [CrossRef] [Green Version]
- De Strooper, B.; Karran, E. The cellular phase of Alzheimer’s disease. Cell 2016, 164, 603–615. [Google Scholar] [CrossRef]
- Hanger, D.P.; Anderton, B.H.; Noble, W. Tau phosphorylation: The therapeutic challenge for neurodegenerative disease. Trends Mol. Med. 2009, 15, 112–119. [Google Scholar] [CrossRef]
- Uddin, M.S.; Stachowiak, A.; Al Mamun, A.; Tzvetkov, N.T.; Takeda, S.; Atanasov, A.G.; Bergantin, L.B.; Abdel-Daim, M.M.; Stankiewicz, A.M. Autophagy and Alzheimer’s disease: From molecular mechanisms to therapeutic implications. Front. Aging Neurosci. 2018, 10, 4. [Google Scholar] [CrossRef]
- Nixon, R.A. Autophagy, amyloidogenesis and Alzheimer disease. J. Cell Sci. 2007, 120, 4081–4091. [Google Scholar] [CrossRef] [Green Version]
- Saido, T.; Leissring, M.A. Proteolytic degradation of amyloid β-protein. Cold Spring Harb. Perspect. Biol. 2013, 2, a006379. [Google Scholar] [CrossRef]
- Nilsson, P.; Loganathan, K.; Sekiguchi, M.; Matsuba, Y.; Hui, K.; Tsubuki, S.; Tanaka, M.; Iwata, N.; Saito, T.; Saido, T.C. Aβ secretion and plaque formation depend on autophagy. Cell Rep. 2013, 5, 61–69. [Google Scholar] [CrossRef]
- Dolan, P.J.; Johnson, G.V.W. A caspase cleaved form of tau is preferentially degraded through the autophagy pathway. J. Biol. Chem. 2010, 285, 21978–21987. [Google Scholar] [CrossRef]
- Menon, M.B.; Dhamija, S. Beclin 1 phosphorylation – at the center of autophagy regulation. Front. Cell Dev. Biol. 2018, 6, 137. [Google Scholar] [CrossRef]
- Pickford, F.; Masliah, E.; Britschgi, M.; Lucin, K.; Narasimhan, R.; Jaeger, P.A.; Small, S.; Spencer, B.; Rockenstein, E.; Levine, B.; et al. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid β accumulation in mice. J. Clin. Investig. 2008, 118, 2190–2199. [Google Scholar] [CrossRef] [Green Version]
- Lucin, K.M.; O’Brien, C.E.; Bieri, G.; Czirr, E.; Mosher, K.I.; Abbey, R.J.; Mastroeni, D.F.; Rogers, J.; Spencer, B.; Masliah, E.; et al. Microglial beclin 1 regulates retromer trafficking and phagocytosis and is impaired in Alzheimer’s disease. Neuron 2013, 79, 873–886. [Google Scholar] [CrossRef] [Green Version]
- Guerreiro, R.J.; Gustafson, D.R.; Hardy, J. The genetic architecture of Alzheimer’s disease: Beyond APP, PSENS and APOE. Neurobiol. Aging 2012, 33, 437–456. [Google Scholar] [CrossRef]
- Somavarapu, A.K.; Kepp, K.P. Loss of stability and hydrophobicity of presenilin 1 mutations causing Alzheimer’s disease. J. Neurochem. 2016, 137, 101–111. [Google Scholar] [CrossRef]
- Ikeda, M.; Yonemura, K.; Kakuda, S.; Tashiro, Y.; Fujita, Y.; Takai, E.; Hashimoto, Y.; Makioka, K.; Furuta, N.; Ishiguro, K.; et al. Cerebrospinal fluid levels of phosphorylated tau and Aβ1-38/Aβ1-40/Aβ1-42 in Alzheimer’s disease with PS1 mutations. Amyloid 2013, 20, 107–112. [Google Scholar] [CrossRef]
- Lee, J.-H.; Yu, W.H.; Kumar, A.; Lee, S.; Mohan, P.S.; Peterhoff, C.M.; Wolfe, D.M.; Martinez-Vicente, M.; Massey, A.C.; Sovak, G.; et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell 2010, 141, 1146–1158. [Google Scholar] [CrossRef]
- Cataldo, A.M.; Peterhoff, C.M.; Schmidt, S.D.; Terio, N.B.; Duff, K.; Beard, M.; Mathews, P.M.; Nixon, R.A. Presenilin mutations in familial Alzheimer disease and transgenic mouse models accelerate neuronal lysosomal pathology. J. Neuropathol. Exp. Neurol. 2004, 63, 821–830. [Google Scholar] [CrossRef]
- Tooze, S.A.; Abada, A.; Elazar, Z. Endocytosis and autophagy: Exploitation or cooperation? Cold Spring Harb. Perspect. Biol. 2014, 6, 1–15. [Google Scholar] [CrossRef]
- Baixauli, F.; López-Otín, C.; Mittelbrunn, M. Exosomes and autophagy: Coordinated mechanisms for the maintenance of cellular fitness. Front. Immunol. 2014, 5, 1–6. [Google Scholar] [CrossRef]
- Xu, J.; Camfield, R.; Gorski, S.M. The interplay between exosomes and autophagy—Partners in crime. J. Cell Sci. 2018, 131, jcs215210. [Google Scholar] [CrossRef]
- Guo, H.; Chitiprolu, M.; Roncevic, L.; Javalet, C.; Hemming, F.J.; Trung, M.T.; Meng, L.; Latreille, E.; de Souza, C.T.; McCulloch, D.; et al. Atg5 disassociates the V1V0-ATPase to promote exosome production and tumor metastasis independent of canonical macroautophagy. Dev. Cell 2017, 43, 716–730. [Google Scholar] [CrossRef]
- Murrow, L.; Malhotra, R.; Debnath, J. ATG12-ATG3 interacts with Alix to promote basal autophagic flux and late endosome function. Nat. Cell Biol. 2015, 17, 300–310. [Google Scholar] [CrossRef]
- Lashley, T.; Schott, J.M.; Weston, P.; Murray, C.E.; Wellington, H.; Keshavan, A.; Foti, S.C.; Foiani, M.; Toombs, J.; Rohrer, J.D.; et al. Molecular biomarkers of Alzheimer’s disease: Progress and prospects. Dis. Model. Mech. 2018, 11, dmm031781. [Google Scholar] [CrossRef]
- Toledo, J.B.; Brettschneider, J.; Grossman, M.; Arnold, S.E.; Hu, W.T.; Xie, S.X.; Lee, V.M.-Y.; Shaw, L.M.; Trojanowski, J.Q. CSF biomarkers cutoffs: The importance of coincident neuropathological diseases. Acta Neuropathol. 2012, 124, 23–35. [Google Scholar] [CrossRef]
- Guzman, V.A.; Carmichael, O.T.; Schwarz, C.; Tosto, G.; Zimmerman, M.E.; Brickman, A.M. White matter hyperintensities and amyloid are independently associated with entorhinal cortex volume among individuals with mild cognitive impairment. Alzheimer’s Dement. 2013, 9, S124–S131. [Google Scholar] [CrossRef] [Green Version]
- Kester, M.I.; Goos, J.D.C.; Teunissen, C.E.; Benedictus, M.R.; Bouwman, F.H.; Wattjes, M.P.; Barkhof, F.; Scheltens, P.; van der Flier, W.M. Associations between cerebral small-vessel disease and Alzheimer disease pathology as measured by cerebrospinal fluid biomarkers. JAMA Neurol. 2014, 71, 855–862. [Google Scholar] [CrossRef]
- Gao, J.; Wang, L.; Huntley, M.L.; Perry, G.; Wang, X. Pathomechanisms of TDP-43 in neurodegeneration. J. Neurochem. 2018, 146, 7–20. [Google Scholar] [CrossRef] [Green Version]
- James, B.D.; Wilson, R.S.; Boyle, P.A.; Trojanowski, J.Q.; Bennett, D.A.; Schneider, J.A. TDP-43 stage, mixed pathologies and clinical Alzheimer’s-type dementia. Brain 2016, 139, 2983–2993. [Google Scholar] [CrossRef]
- Salminen, A.; Kauppinen, A.; Kaarniranta, K. Hypoxia/ischemia activate processing of amyloid precursor protein: Impact of vascular dysfunction in the pathogenesis of Alzheimer’s disease. J. Neurochem. 2017, 140, 536–549. [Google Scholar] [CrossRef]
- Zhang, X.; Zhou, K.; Wang, R.; Cui, J.; Lipton, S.A.; Liao, F.-F.; Xu, H.; Zhang, Y.-W. Hypoxia-inducible factor 1α (HIF-1α)-mediated hypoxia increases BACE1 expression and β-amyloid generation. J. Biol. Chem. 2007, 282, 10873–10880. [Google Scholar] [CrossRef]
- Villa, J.C.; Chiu, D.; Brandes, A.H.; Escorcia, F.E.; Villa, C.H.; Maguire, W.F.; Hu, C.-J.; de Stanchina, E.; Simon, M.C.; Sisodia, S.S.; et al. Nontranscriptional role of Hif-1α in activation of γ-secretase and notch signaling in breast cancer. Cell Rep. 2014, 8, 1077–1092. [Google Scholar] [CrossRef]
- Sperling, R.A.; Aisen, P.S.; Beckett, L.A.; Bennett, D.A.; Craft, S.; Fagan, A.M.; Iwatsubo, T.; Jack, C.R.; Kaye, J.; Montine, T.J.; et al. Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 280–292. [Google Scholar] [CrossRef]
- McKhann, G.M.; Knopman, D.S.; Chertkow, H.; Hyman, B.T.; Jack, C.R.; Kawas, C.H.; Klunk, W.E.; Koroshetz, W.J.; Manly, J.J.; Mayeux, R.; et al. The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 263–269. [Google Scholar] [CrossRef] [Green Version]
- Blennow, K.; Mattsson, N.; Schöll, M.; Hansson, O.; Zetterberg, H. Amyloid biomarkers in Alzheimer’s disease. Trends Pharmacol. Sci. 2015, 36, 297–309. [Google Scholar] [CrossRef]
- Reitz, C.; Mayeux, R. Alzheimer disease: Epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem. Pharmacol. 2014, 88, 640–651. [Google Scholar] [CrossRef] [Green Version]
- Dubois, B.; Feldman, H.H.; Jacova, C.; Hampel, H.; Molinuevo, J.L.; Blennow, K.; Dekosky, S.T.; Gauthier, S.; Selkoe, D.; Bateman, R.; et al. Advancing research diagnostic criteria for Alzheimer’s disease: The IWG-2 criteria. Lancet Neurol. 2014, 13, 614–629. [Google Scholar] [CrossRef]
- Shi, L.; Baird, A.L.; Westwood, S.; Hye, A.; Dobson, R.; Thambisetty, M.; Lovestone, S. A decade of blood biomarkers for Alzheimer’s disease research: An evolving field, improving study designs and the challenge of replication. J. Alzheimer’s Dis. 2018, 62, 1181–1198. [Google Scholar] [CrossRef]
- Mattsson, N.; Andreasson, U.; Persson, S.; Carrillo, M.C.; Collins, S.; Chalbot, S.; Cutler, N.; Dufour-Rainfray, D.; Fagan, A.M.; Heegaard, N.H.H.; et al. CSF biomarker variability in the Alzheimer’s Association quality control program. Alzheimer’s Dement. 2013, 9, 251–261. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.J.; Desplats, P.; Sigurdson, C.; Tsigelny, I.; Masliah, E. Cell-to-cell transmission of non-prion protein aggregates. Nat. Rev. Neurol. 2010, 6, 702–706. [Google Scholar] [CrossRef]
- Aguzzi, A.; Baumann, F.; Bremer, J. The prion’s elusive reason for being. Annu. Rev. Neurosci. 2008, 31, 439–477. [Google Scholar] [CrossRef]
- Meyer-luehmann, M.; Coomaraswamy, J.; Bolmont, T.; Kaeser, S.; Schaefer, C.; Kilger, E.; Neuenschwander, A.; Abramowski, D.; Jaton, A.L.; Vigouret, J.; et al. Supporting online material for exogenous induction of cerebral β-amyloidogenesis is governed by agent and host. Science 2006, 313, 1781–1785. [Google Scholar] [CrossRef]
- Eisele, Y.S.; Bolmont, T.; Heikenwalder, M.; Langer, F.; Jacobson, L.H.; Yan, Z.-X.; Roth, K.; Aguzzi, A.; Staufenbiel, M.; Walker, L.C.; et al. Induction of cerebral β-amyloidosis: Intracerebral versus systemic Aβ inoculation. Proc. Natl. Acad. Sci. USA 2009, 106, 12926–12931. [Google Scholar] [CrossRef] [Green Version]
- Dinkins, M.B.; Dasgupta, S.; Wang, G.; Zhu, G.; Bieberich, E. Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol. Aging 2014, 35, 1792–1800. [Google Scholar] [CrossRef]
- Rajendran, L.; Honsho, M.; Zahn, T.; Keller, P.; Geiger, K.; Verkade, P.; Simons, K. Alzheimer’s disease β-amyloid peptides are released in association with exosomes. Proc. Natl. Acad. Sci. USA 2006, 103, 11172–11177. [Google Scholar] [CrossRef]
- Hayashi, H.; Kimura, N.; Yamaguchi, H.; Hasegawa, K.; Yokoseki, T.; Shibata, M.; Yamamoto, N.; Michikawa, M.; Yoshikawa, Y.; Terao, K.; et al. A seed for Alzheimer amyloid in the brain. J. Neurosci. 2004, 24, 4894–4902. [Google Scholar] [CrossRef]
- Yuyama, K.; Sun, H.; Mitsutake, S.; Igarashi, Y. Sphingolipid-modulated exosome secretion promotes clearance of amyloid-β by microglia. J. Biol. Chem. 2012, 287, 10977–10989. [Google Scholar] [CrossRef]
- Yuyama, K.; Sun, H.; Sakai, S.; Mitsutake, S.; Okada, M.; Tahara, H.; Furukawa, J.I.; Fujitani, N.; Shinohara, Y.; Igarashi, Y. Decreased amyloid-β pathologies by intracerebral loading of glycosphingolipid-enriched exosomes in Alzheimer model mice. J. Biol. Chem. 2014, 289, 24488–24498. [Google Scholar] [CrossRef]
- Iranifar, E.; Seresht, B.M.; Momeni, F.; Fadaei, E.; Mehr, M.H.; Ebrahimi, Z.; Rahmati, M.; Kharazinejad, E.; Mirzaei, H. Exosomes and microRNAs: New potential therapeutic candidates in Alzheimer disease therapy. J. Cell. Physiol. 2018, 234, 2296–2305. [Google Scholar] [CrossRef]
- McKeever, P.M.; Schneider, R.; Taghdiri, F.; Weichert, A.; Multani, N.; Brown, R.A.; Boxer, A.L.; Karydas, A.; Miller, B.; Robertson, J.; et al. MicroRNA expression levels are altered in the cerebrospinal fluid of patients with young-onset Alzheimer’s disease. Mol. Neurobiol. 2018, 55, 8826–8841. [Google Scholar] [CrossRef]
- Hébert, S.S.; Horré, K.; Nicolaï, L.; Papadopoulou, A.S.; Mandemakers, W.; Silahtaroglu, A.N.; Kauppinen, S.; Delacourte, A.; De Strooper, B. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer’s disease correlates with increased BACE1/β-secretase expression. Proc. Natl. Acad. Sci. USA 2008, 105, 6415–6420. [Google Scholar] [CrossRef]
- Nelson, P.T.; Wang, W.X. MiR-107 is reduced in Alzheimer’s disease brain neocortex: Validation study. J. Alzheimer’s Dis. 2010, 21, 75–79. [Google Scholar] [CrossRef]
- Zhu, H.C.; Wang, L.M.; Wang, M.; Song, B.; Tan, S.; Teng, J.F.; Duan, D.X. MicroRNA-195 downregulates Alzheimer’s disease amyloid-β production by targeting BACE1. Brain Res. Bull. 2012, 88, 596–601. [Google Scholar] [CrossRef]
- Patel, N.; Hoang, D.; Miller, N.; Ansaloni, S.; Huang, Q.; Rogers, J.T.; Lee, J.C.; Saunders, A.J. MicroRNAs can regulate human APP levels. Mol. Neurodegener. 2008, 3, 10. [Google Scholar] [CrossRef]
- Liu, C.G.; Song, J.; Zhang, Y.Q.; Wang, P.C. MicroRNA-193b is a regulator of amyloid precursor protein in the blood and cerebrospinal fluid derived exosomal microRNA-193b is a biomarker of Alzheimer’s disease. Mol. Med. Rep. 2015, 10, 2395–2400. [Google Scholar] [CrossRef]
- Manzine, P.R.; Pelucchi, S.; Horst, M.A.; Vale, F.A.C.; Pavarini, S.C.I.; Audano, M.; Mitro, N.; Di Luca, M.; Marcello, E.; Cominetti, M.R. microRNA 221 targets ADAM10 mRNA and is downregulated in Alzheimer’s disease. J. Alzheimer’s Dis. 2018, 61, 113–123. [Google Scholar] [CrossRef]
- Lau, P.; Bossers, K.; Janky, R.; Salta, E.; Frigerio, C.S.; Barbash, S.; Rothman, R.; Sierksma, A.S.R.; Thathiah, A.; Greenberg, D.; et al. Alteration of the microRNA network during the progression of Alzheimer’s disease. EMBO Mol. Med. 2013, 5, 1613–1634. [Google Scholar] [CrossRef]
- Ma, X.; Liu, L.; Meng, J. MicroRNA-125b promotes neurons cell apoptosis and Tau phosphorylation in Alzheimer’s disease. Neurosci. Lett. 2017, 661, 57–62. [Google Scholar] [CrossRef]
- Absalon, S.; Kochanek, D.M.; Raghavan, V.; Krichevsky, A.M. MiR-26b, upregulated in Alzheimer’s disease, activates cell cycle entry, tau-phosphorylation and apoptosis in postmitotic neurons. J. Neurosci. 2013, 33, 14645–14659. [Google Scholar] [CrossRef]
- Atkinson, A.J.; Colburn, W.A.; DeGruttola, V.G.; DeMets, D.L.; Downing, G.J.; Hoth, D.F.; Oates, J.A.; Peck, C.C.; Schooley, R.T.; Biomarkers Definitions Working Group; et al. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 2001, 69, 89–95. [Google Scholar] [CrossRef]
- Blennow, K.; Hampel, H. CSF markers for incipient Alzheimer’s disease. Lancet Neurol. 2003, 2, 605–613. [Google Scholar] [CrossRef]
- Shaw, L.M.; Vanderstichele, H.; Knapik-Czajka, M.; Clark, C.M.; Aisen, P.S.; Petersen, R.C.; Blennow, K.; Soares, H.; Simon, A.; Lewczuk, P.; et al. Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann. Neurol. 2009, 65, 403–413. [Google Scholar] [CrossRef]
- Mattsson, N.; Zetterberg, H.; Hansson, O.; Andreasen, N.; Parnetti, L.; Jonsson, M.; Herukka, S.K.; van der Flier, W.M.; Blankenstein, M.A.; Ewers, M.; et al. CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. JAMA 2009, 302, 385–393. [Google Scholar] [CrossRef]
- Kester, M.I.; Teunissen, C.E.; Crimmins, D.L.; Herries, E.M.; Ladenson, J.H.; Scheltens, P.; van der Flier, W.M.; Morris, J.C.; Holtzman, D.M.; Fagan, A.M. Neurogranin as a cerebrospinal fluid biomarker for synaptic loss in symptomatic Alzheimer disease. JAMA Neurol. 2015, 72, 1275–1280. [Google Scholar] [CrossRef]
- Kvartsberg, H.; Duits, F.H.; Ingelsson, M.; Andreasen, N.; Öhrfelt, A.; Andersson, K.; Brinkmalm, G.; Lannfelt, L.; Minthon, L.; Hansson, O.; et al. Cerebrospinal fluid levels of the synaptic protein neurogranin correlates with cognitive decline in prodromal Alzheimer’s disease. Alzheimer’s Dement. 2015, 11, 1180–1190. [Google Scholar] [CrossRef]
- Lista, S.; Hampel, H. Synaptic degeneration and neurogranin in the pathophysiology of Alzheimer’s disease. Expert Rev. Neurother. 2017, 17, 47–57. [Google Scholar] [CrossRef]
- Portelius, E.; Zetterberg, H.; Skillbäck, T.; Törnqvist, U.; Andreasson, U.; Trojanowski, J.Q.; Weiner, M.W.; Shaw, L.M.; Mattsson, N.; Blennow, K. Cerebrospinal fluid neurogranin: Relation to cognition and neurodegeneration in Alzheimer’s disease. Brain 2015, 138, 3373–3385. [Google Scholar] [CrossRef]
- Wellington, H.; Paterson, R.W.; Portelius, E.; Törnqvist, U.; Magdalinou, N.; Fox, N.C.; Blennow, K.; Schott, J.M.; Zetterberg, H. Increased CSF neurogranin concentration is specific to Alzheimer disease. Neurology 2016, 86, 829–835. [Google Scholar] [CrossRef] [Green Version]
- Portelius, E.; Olsson, B.; Höglund, K.; Cullen, N.C.; Kvartsberg, H.; Andreasson, U.; Zetterberg, H.; Sandelius, Å.; Shaw, L.M.; Lee, V.M.; et al. Cerebrospinal fluid neurogranin concentration in neurodegeneration: Relation to clinical phenotypes and neuropathology. Acta Neuropathol. 2018, 136, 363–376. [Google Scholar] [CrossRef]
- Alcolea, D.; Vilaplana, E.; Suárez-Calvet, M.; Illán-Gala, I.; Blesa, R.; Clarimón, J.; Lladó, A.; Sánchez-Valle, R.; Molinuevo, J.L.; García-Ribas, G.; et al. CSF sAPPβ, YKL-40 and neurofilament light in frontotemporal lobar degeneration. Neurology 2017, 89, 178–188. [Google Scholar] [CrossRef]
- Mattsson, N.; Insel, P.S.; Palmqvist, S.; Portelius, E.; Zetterberg, H.; Weiner, M.; Blennow, K.; Hansson, O. Cerebrospinal fluid tau, neurogranin and neurofilament light in Alzheimer’s disease. EMBO Mol. Med. 2016, 8, 1184–1196. [Google Scholar] [CrossRef]
- Koyama, A.; Okereke, O.I.; Yang, T.; Blacker, D.; Selkoe, D.J.; Grodstein, F. Plasma amyloid-β as a predictor of dementia and cognitive decline. Arch. Neurol. 2012, 69, 824–831. [Google Scholar] [CrossRef]
- Rissman, R.A.; Trojanowski, J.Q.; Shaw, L.M.; Aisen, P.S. Longitudinal plasma amyloid beta as a biomarker of Alzheimer’s disease. J. Neural Transm. 2012, 119, 843–850. [Google Scholar] [CrossRef] [Green Version]
- Yang, C.C.; Yang, S.Y.; Chieh, J.J.; Horng, H.E.; Hong, C.Y.; Yang, H.C.; Chen, K.H.; Shih, B.Y.; Chen, T.F.; Chiu, M.J. Biofunctionalized magnetic nanoparticles for specifically detecting biomarkers of Alzheimer’s disease in vitro. ACS Chem. Neurosci. 2011, 2, 500–505. [Google Scholar] [CrossRef]
- Janelidze, S.; Stomrud, E.; Palmqvist, S.; Zetterberg, H.; van Westen, D.; Jeromin, A.; Song, L.; Hanlon, D.; Tan Hehir, C.A.; Baker, D.; et al. Plasma β-amyloid in Alzheimer’s disease and vascular disease. Sci. Rep. 2016, 6, 26801. [Google Scholar] [CrossRef] [Green Version]
- Mattsson, N.; Zetterberg, H.; Janelidze, S.; Insel, P.S.; Andreasson, U.; Stomrud, E.; Palmqvist, S.; Baker, D.; Tan Hehir, C.A.; Jeromin, A.; et al. Plasma tau in Alzheimer disease. Neurology 2016, 87, 1827–1835. [Google Scholar] [CrossRef] [Green Version]
- Nakamura, A.; Kaneko, N.; Villemagne, V.L.; Kato, T.; Doecke, J.; Doré, V.; Fowler, C.; Li, Q.-X.; Martins, R.; Rowe, C.; et al. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature 2018, 554, 249–254. [Google Scholar] [CrossRef]
- Fiandaca, M.S.; Kapogiannis, D.; Mapstone, M.; Boxer, A.; Eitan, E.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Federoff, H.J.; Miller, B.L.; et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimer’s Dement. 2015, 11, 600–607. [Google Scholar] [CrossRef]
- Kapogiannis, D.; Boxer, A.; Schwartz, J.B.; Abner, E.L.; Biragyn, A.; Masharani, U.; Frassetto, L.; Petersen, R.C.; Miller, B.L.; Goetzl, E.J. Dysfunctionally phosphorylated type 1 insulin receptor substrate in neural-derived blood exosomes of preclinical Alzheimer’s disease. FASEB J. 2014, 29, 589–596. [Google Scholar] [CrossRef] [Green Version]
- Mullins, R.J.; Mustapic, M.; Goetzl, E.J.; Kapogiannis, D. Exosomal biomarkers of brain insulin resistance associated with regional atrophy in Alzheimer’s disease. Hum. Brain Mapp. 2017, 38, 1933–1940. [Google Scholar] [CrossRef]
- Goetzl, E.J.; Boxer, A.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Miller, B.L.; Kapogiannis, D. Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease. Neurology 2015, 85, 40–47. [Google Scholar] [CrossRef] [Green Version]
- Goetzl, E.J.; Kapogiannis, D.; Schwartz, J.B.; Lobach, I.V.; Goetzl, L.; Abner, E.L.; Jicha, G.A.; Karydas, A.M.; Boxer, A.; Miller, B.L. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 2016, 30, 4141–4148. [Google Scholar] [CrossRef]
- Goetzl, E.J.; Mustapic, M.; Kapogiannis, D.; Eitan, E.; Lobach, I.V.; Goetzl, L.; Schwartz, J.B.; Miller, B.L. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB J. 2016, 30, 3853–3859. [Google Scholar] [CrossRef]
- Hébert, S.S.; Horré, K.; Nicolaï, L.; Bergmans, B.; Papadopoulou, A.S.; Delacourte, A.; De Strooper, B. MicroRNA regulation of Alzheimer’s Amyloid precursor protein expression. Neurobiol. Dis. 2009, 33, 422–428. [Google Scholar] [CrossRef]
- Kim, J.; Yoon, H.; Ramírez, C.M.; Lee, S.M.; Hoe, H.S.; Fernández-Hernando, C.; Kim, J. MiR-106b impairs cholesterol efflux and increases Aβ levels by repressing ABCA1 expression. Exp. Neurol. 2012, 235, 476–483. [Google Scholar] [CrossRef]
- Ben Halima, S.; Siegel, G.; Rajendran, L. miR-186 in Alzheimer’s disease: A big hope for a small RNA? J. Neurochem. 2016, 137, 308–311. [Google Scholar] [CrossRef]
- Frigerio, C.S.; Lau, P.; Salta, E.; Tournoy, J.; Bossers, K.; Vandenberghe, R.; Wallin, A.; Bjerke, M.; Zetterberg, H.; Blennow, K.; et al. Reduced expression of hsa-miR-27a-3p in CSF of patients with Alzheimer disease. Neurology 2013, 81, 2103–2106. [Google Scholar] [CrossRef]
- Müller, M.; Kuiperij, H.B.; Versleijen, A.A.M.; Chiasserini, D.; Farotti, L.; Baschieri, F.; Parnetti, L.; Struyfs, H.; De Roeck, N.; Luyckx, J.; et al. Validation of microRNAs in cerebrospinal fluid as biomarkers for different forms of dementia in a multicenter study. J. Alzheimer’s Dis. 2016, 52, 1321–1333. [Google Scholar] [CrossRef]
- Alexandrov, P.N.; Dua, P.; Hill, J.M.; Bhattacharjee, S.; Zhao, Y.; Lukiw, W.J. microRNA (miRNA) speciation in Alzheimer’s disease (AD) cerebrospinal fluid (CSF) and extracellular fluid (ECF). Int. J. Biochem. Mol. Biol. 2012, 3, 365–373. [Google Scholar]
- Denk, J.; Boelmans, K.; Siegismund, C.; Lassner, D.; Arlt, S.; Jahn, H. MicroRNA profiling of CSF reveals potential biomarkers to detect Alzheimer`s disease. PLoS ONE 2015, 10, e0126423. [Google Scholar] [CrossRef]
- Li, W.; Li, X.; Xin, X.; Kan, P.-C.; Yan, Y. MicroRNA-613 regulates the expression of brain-derived neurotrophic factor in Alzheimer’s disease. Biosci. Trends 2016, 10, 372–377. [Google Scholar] [CrossRef]
- Lusardi, T.A.; Phillips, J.I.; Wiedrick, J.T.; Harrington, C.A.; Lind, B.; Lapidus, J.A.; Quinn, J.F.; Saugstad, J.A. MicroRNAs in human cerebrospinal fluid as biomarkers for Alzheimer’s disease. J. Alzheimer’s Dis. 2017, 55, 1223–1233. [Google Scholar] [CrossRef]
- Wiedrick, J.T.; Phillips, J.I.; Lusardi, T.A.; McFarland, T.J.; Lind, B.; Sandau, U.S.; Harrington, C.A.; Lapidus, J.A.; Galasko, D.R.; Quinn, J.F.; et al. Validation of microRNA biomarkers for Alzheimer’s disease in human cerebrospinal fluid. J. Alzheimer’s Dis. 2019, 67, 875–891. [Google Scholar] [CrossRef]
- Leidinger, P.; Backes, C.; Deutscher, S.; Schmitt, K.; Mueller, S.C.; Frese, K.; Haas, J.; Ruprecht, K.; Paul, F.; Stähler, C.; et al. A blood based 12-miRNA signature of Alzheimer disease patients. Genome Biol. 2013, 14, R78. [Google Scholar] [CrossRef]
- Williams, Z.; Ben-Dov, I.Z.; Elias, R.; Mihailovic, A.; Brown, M.; Rosenwaks, Z.; Tuschlb, T. Comprehensive profiling of circulating microRNA via small RNA sequencing of cDNA libraries reveals biomarker potential and limitations. Proc. Natl. Acad. Sci. USA 2013, 110, 4255–4260. [Google Scholar] [CrossRef] [Green Version]
- Laurent, L.C.; Abdel-Mageed, A.B.; Adelson, P.D.; Arango, J.; Balaj, L.; Breakefield, X.; Carlson, E.; Carter, B.S.; Majem, B.; Chen, C.C.; et al. Meeting report: Discussions and preliminary findings on extracellular RNA measurement methods from laboratories in the NIH Extracellular RNA Communication Consortium. J. Extracell. Vesicles 2015, 4, 26533. [Google Scholar] [CrossRef]
- Crossland, R.E.; Norden, J.; Bibby, L.A.; Davis, J.; Dickinson, A.M. Evaluation of optimal extracellular vesicle small RNA isolation and qRT-PCR normalisation for serum and urine. J. Immunol. Methods 2016, 429, 39–49. [Google Scholar] [CrossRef]
- Keller, A.; Backes, C.; Haas, J.; Leidinger, P.; Maetzler, W.; Deuschle, C.; Berg, D.; Ruschil, C.; Galata, V.; Ruprecht, K.; et al. Validating Alzheimer’s disease micro RNAs using next-generation sequencing. Alzheimer’s Dement. 2016, 12, 565–576. [Google Scholar] [CrossRef]
- Tan, L.; Yu, J.-T.; Liu, Q.-Y.; Tan, M.-S.; Zhang, W.; Hu, N.; Wang, Y.-L.; Sun, L.; Jiang, T.; Tan, L. Circulating miR-125b as a biomarker of Alzheimer’s disease. J. Neurol. Sci. 2014, 336, 52–56. [Google Scholar] [CrossRef]
- Tan, L.; Yu, J.-T.; Tan, M.-S.; Liu, Q.-Y.; Wang, H.-F.; Zhang, W.; Jiang, T.; Tan, L. Genome-wide serum microRNA expression profiling identifies serum biomarkers for Alzheimer’s disease. J. Alzheimer’s Dis. 2014, 40, 1017–1027. [Google Scholar] [CrossRef]
- Dong, H.; Li, J.; Huang, L.; Chen, X.; Li, D.; Wang, T.; Hu, C.; Xu, J.; Zhang, C.; Zen, K.; et al. Serum microRNA profiles serve as novel biomarkers for the diagnosis of Alzheimer’s disease. Dis. Markers 2015, 2015, 625659. [Google Scholar] [CrossRef]
- Xie, B.; Liu, Z.; Jiang, L.; Liu, W.; Song, M.; Zhang, Q.; Zhang, R.; Cui, D.; Wang, X.; Xu, S. Increased serum miR-206 level predicts conversion from amnestic mild cognitive impairment to Alzheimer’s disease: A 5-year follow-up study. J. Alzheimer’s Dis. 2017, 55, 509–520. [Google Scholar] [CrossRef]
- Xie, B.; Zhou, H.; Zhang, R.; Song, M.; Yu, L.; Wang, L.; Liu, Z.; Zhang, Q.; Cui, D.; Wang, X.; et al. Serum miR-206 and miR-132 as potential circulating biomarkers for Mild Cognitive Impairment. J. Alzheimer’s Dis. 2015, 45, 721–731. [Google Scholar] [CrossRef]
- Jia, L.H.; Liu, Y.N. Downregulated serum miR-223 servers as biomarker in Alzheimer’s disease. Cell Biochem. Funct. 2016, 34, 233–237. [Google Scholar] [CrossRef]
- Wang, T.; Chen, K.; Li, H.; Dong, S.; Su, N.; Liu, Y.; Cheng, Y.; Dai, J.; Yang, C.; Xiao, S. The feasibility of utilizing plasma MiRNA107 and BACE1 messenger RNA gene expression for clinical diagnosis of amnestic mild cognitive impairment. J. Clin. Psychiatry 2015, 76, 135–141. [Google Scholar] [CrossRef]
- Sheinerman, K.S.; Tsivinsky, V.G.; Abdullah, L.; Crawford, F.; Umansky, S.R. Plasma microRNA biomarkers for detection of mild cognitive impairment: Biomarker validation study. Aging 2013, 5, 925–938. [Google Scholar] [CrossRef]
- Bhatnagar, S.; Chertkow, H.; Schipper, H.M.; Yuan, Z.; Shetty, V.; Jenkins, S.; Jones, T.; Wang, E. Increased microRNA-34c abundance in Alzheimer’s disease circulating blood plasma. Front. Mol. Neurosci. 2014, 7, 1–11. [Google Scholar] [CrossRef]
- Yang, T.T.; Liu, C.G.; Gao, S.C.; Zhang, Y.; Wang, P.C. The serum exosome derived microRNA-135a, -193b and -384 were potential Alzheimer’s disease biomarkers. Biomed. Environ. Sci. 2018, 31, 87–96. [Google Scholar]
- Cheng, L.; Doecke, J.D.; Sharples, R.A.; Villemagne, V.L.; Fowler, C.J.; Rembach, A.; Martins, R.N.; Rowe, C.C.; Macaulay, S.L.; Masters, C.L.; et al. Prognostic serum miRNA biomarkers associated with Alzheimer’s disease shows concordance with neuropsychological and neuroimaging assessment. Mol. Psychiatry 2015, 20, 1188–1196. [Google Scholar] [CrossRef]
- Lugli, G.; Cohen, A.M.; Bennett, D.A.; Shah, R.C.; Fields, C.J.; Hernandez, A.G.; Smalheiser, N.R. Plasma exosomal miRNAs in persons with and without Alzheimer disease: Altered expression and prospects for biomarkers. PLoS ONE 2015, 10, e0139233. [Google Scholar] [CrossRef]
- Villa, C.; Fenoglio, C.; De Riz, M.; Clerici, F.; Marcone, A.; Benussi, L.; Ghidoni, R.; Gallone, S.; Cortini, F.; Serpente, M.; et al. Role of hnRNP-A1 and miR-590-3p in neuronal death: Genetics and expression analysis in patients with Alzheimer disease and frontotemporal lobar degeneration. Rejuvenation Res. 2011, 14, 275–281. [Google Scholar] [CrossRef]
- Momen-Heravi, F.; Balaj, L.; Alian, S.; Trachtenberg, A.J.; Hochberg, F.H.; Skog, J.; Kuo, W.P. Impact of biofluid viscosity on size and sedimentation efficiency of the isolated microvesicles. Front. Physiol. 2012, 3, 162. [Google Scholar] [CrossRef]
- Witwer, K.W.; Buzás, E.I.; Bemis, L.T.; Bora, A.; Lässer, C.; Lötvall, J.; Nolte-’t Hoen, E.N.; Piper, M.G.; Sivaraman, S.; Skog, J.; et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles 2013, 2, 20360. [Google Scholar] [CrossRef]
- Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.F.; Wiegmans, A.P.; Leimgruber, A.; Möller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef] [Green Version]
- Van Deun, J.; Mestdagh, P.; Sormunen, R.; Cocquyt, V.; Vermaelen, K.; Vandesompele, J.; Bracke, M.; De Wever, O.; Hendrix, A. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J. Extracell. Vesicles 2014, 3, 24858. [Google Scholar] [CrossRef]
- Gemmell, C.H.; Sefton, M.V.; Yeo, E.L. Platelet-derived microparticle formation involves glycoprotein IIb-IIIa Inhibition by RGDS and a Glanzmann’s thrombasthenia defect. J. Biol. Chem. 1993, 268, 14586–14589. [Google Scholar]
- George, J.; Thoi, L.L.; Mcmanus, M.; Reimann, T.A. Isolation of human platelet membrane microparticles from plasma and serum. Blood 1982, 60, 834–840. [Google Scholar]
- Lacroix, R.; Judicone, C.; Mooberry, M.; Boucekine, M.; Key, N.S.; Dignat-George, F.; Ambrozic, A.; Bailly, N.; Buffat, C.; Buzas, E.; et al. Standardization of pre-analytical variables in plasma microparticle determination: Results of the International Society on Thrombosis and Haemostasis SSC Collaborative workshop. J. Thromb. Haemost. 2013, 11, 1190–1193. [Google Scholar] [CrossRef]
- György, B.; Pálóczi, K.; Kovács, A.; Barabás, E.; Beko, G.; Várnai, K.; Pállinger, É.; Szabó-Taylor, K.; Szabó, T.G.; Kiss, A.A.; et al. Improved circulating microparticle analysis in acid-citrate dextrose (ACD) anticoagulant tube. Thromb. Res. 2014, 133, 285–292. [Google Scholar] [CrossRef] [Green Version]
- Lötvall, J.; Hil, A.F.; Hochberg, F.; Buzάs, E.I.; Di Vizio, D.; Gardiner, C.; Gho, Y.S.; Kurochkin, I.V.; Mathivanan, S.; Quesenberry, P.; et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2014, 3, 26913. [Google Scholar] [CrossRef]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
- Cheng, L.; Sun, X.; Scicluna, B.J.; Coleman, B.M.; Hill, A.F. Characterization and deep sequencing analysis of exosomal and non-exosomal miRNA in human urine. Kidney Int. 2014, 86, 433–444. [Google Scholar] [CrossRef]
- Quek, C.; Bellingham, S.A.; Jung, C.H.; Scicluna, B.J.; Shambrook, M.C.; Sharples, R.A.; Cheng, L.; Hill, A.F. Defining the purity of exosomes required for diagnostic profiling of small RNA suitable for biomarker discovery. RNA Biol. 2017, 14, 245–258. [Google Scholar] [CrossRef]
- Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [Green Version]
- Sódar, B.W.; Kovács, Á.; Visnovitz, T.; Pállinger, É.; Vékey, K.; Pocsfalvi, G.; Turiák, L.; Buzás, E.I. Best practice of identification and proteomic analysis of extracellular vesicles in human health and disease. Expert Rev. Proteomics 2017, 14, 1073–1090. [Google Scholar] [CrossRef]
- Gallien, S.; Duriez, E.; Demeure, K.; Domon, B. Selectivity of LC-MS/MS analysis: Implication for proteomics experiments. J. Proteomics 2013, 81, 148–158. [Google Scholar] [CrossRef]
Body Fluid | Study Groups | miRNA Analysis Method | miRNA | Sensitivity | Specificity | Reference |
---|---|---|---|---|---|---|
Whole blood | NC# (21) MCI (18) AD (94) | qRT-PCR of miRNA discovered by NGS from 22 HC and 48 AD | 12 miRNAs 1 | 95.1 | 91.5 | [162] |
Serum | NC (150) AD (105) | qRT-PCR | miR-125b | 81 | 68 | [163] |
Serum | NC (50, 155) AD (50, 158) | qRT-PCR following Illumina sequencing | miR-342-3p 6 miRNAs 2 | 85 81 | 71 68 | [164] |
Serum | NC (48, 75) AD (48, 79) | qRT-PCR following Solexa sequencing | 4 miRNAs (miR-31, miR-93, miR-143, miR-146a) | n.a.$ (AUC = 71 – 75) | [165] | |
Serum | MCI-MCI (330) MCI-AD (128) | qRT-PCR | miR-206 | 95.3 | 77.8 | [166] |
Serum | NC (76) MCI (66) | qRT-PCR | miR-206 miR-132 Combined | 86.4 69.7 85.5 | 76.3 100 98.5 | [167] |
Serum | NC (62) AD (84) | qRT-PCR | miR-29 + miR-223 miR-125b + miR-223 | n.a. (AUC = 0.826) n.a. (AUC = 0.879) | [168] | |
Plasma | NC (81) MCI (116) AD (97) | qRT-PCR | miR-107 | 98.3 | 82.7 | [169] |
Plasma | NC (50) MCI (50) | qRT-PCR | miR-132 family (3 pairs) miR-134 family (3 pairs) | 84–94 (96) 76–88(80) | 96–98 (96) 80–90(94) | [170] |
Plasma | NC (85) AD (78) | qRT-PCR | miR-34c miR-34a | 92 84 | 96 74 | [171] |
Exosome (serum) | NC (228) MCI (101) AD (107) | qRT-PCR | miR-135a miR-193b miR-384 Combined | 90 78 85 99 | 95 77 90 95 | [172] |
Exosome (serum) | NC (36) MCI (8) AD (16) | qRT-PCR following discovery by sequencing from 23 HC, 3 MCI and 23 AD | 16 miRNAs 3 | 87 | 77 | [173] |
Exosome (plasma) | NC (35) AD (35) | Illumina deep sequencing | Panel of 7 miRNAs 4 | n.a. (AD prediction rate = 89%) | [174] | |
Exosome (plasma, CSF) | NC (7) MCI (43) AD (51) | qRT-PCR | miR-193b | n.a. | [119] | |
CSF | NC (40) MCI (37) AD (57) | qRT-PCR | No tested miRNA deregulated in AD (miR-29a, miR-125b-5p, miR-146a-5p, miR-16-5p, miR-24-3p) | n.a. | n.a. | [152] |
PBMC | NC (344) AD (287) | qRT-PCR | miR-590-3p | n.a. | n.a. | [175] |
Developmental Process | Current or Future Challenges | Required Strategies |
---|---|---|
1. EV preparation |
|
|
2. Biomarker discovery |
|
|
3. Clinical validation |
|
|
4. Clinical application |
|
|
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lee, S.; Mankhong, S.; Kang, J.-H. Extracellular Vesicle as a Source of Alzheimer’s Biomarkers: Opportunities and Challenges. Int. J. Mol. Sci. 2019, 20, 1728. https://doi.org/10.3390/ijms20071728
Lee S, Mankhong S, Kang J-H. Extracellular Vesicle as a Source of Alzheimer’s Biomarkers: Opportunities and Challenges. International Journal of Molecular Sciences. 2019; 20(7):1728. https://doi.org/10.3390/ijms20071728
Chicago/Turabian StyleLee, Seongju, Sakulrat Mankhong, and Ju-Hee Kang. 2019. "Extracellular Vesicle as a Source of Alzheimer’s Biomarkers: Opportunities and Challenges" International Journal of Molecular Sciences 20, no. 7: 1728. https://doi.org/10.3390/ijms20071728
APA StyleLee, S., Mankhong, S., & Kang, J.-H. (2019). Extracellular Vesicle as a Source of Alzheimer’s Biomarkers: Opportunities and Challenges. International Journal of Molecular Sciences, 20(7), 1728. https://doi.org/10.3390/ijms20071728