Non-Coding RNAs as Sensors of Oxidative Stress in Neurodegenerative Diseases
Abstract
:1. Introduction
2. Oxidative Stress in Neurodegenerative Diseases
2.1. Reactive Oxygen Species Production and Activities
2.2. Oxidative Stress in Neurodegeneration
3. Regulatory Non-Coding RNAs in the Normal Function of the Nervous System
3.1. microRNAs
3.2. tRNA-Derived Fragments
3.3. Long Non-Coding RNAs
3.4. Circular RNAs (circRNAs)
4. NcRNAs and Oxidative Stress Management in Neurodegenerative Diseases
4.1. Alzheimer’s Disease
4.2. Parkinson’s Disease
4.3. Huntington’s Disease
4.4. Amyotrophic Lateral Sclerosis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Yaribeygi, H.; Panahi, Y.; Javadi, B.; Sahebkar, A. The underlying role of oxidative stress in neurodegeneration: A mechanistic review. CNS Neurol. Disord.-Drug Targets 2018, 17, 207–215. [Google Scholar] [CrossRef] [PubMed]
- Feingold, E.A.; Good, P.J.; Guyer, M.S.; Kamholz, S.; Liefer, L.; Wetterstrand, K.; Collins, F.S.; Gingeras, T.R.; Kampa, D.; Sekinger, E.A.; et al. The ENCODE (ENCyclopedia of DNA Elements) project. Science 2004, 306, 636–640. [Google Scholar]
- ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 2012, 489, 57–74. [Google Scholar] [CrossRef] [PubMed]
- Nie, L.; Wu, H.-J.; Hsu, J.-M.; Chang, S.-S.; Labaff, A.M.; Li, C.-W.; Wang, Y.; Hsu, J.L.; Hung, M.-C. Long non-coding RNAs: Versatile master regulators of gene expression and crucial players in cancer. Am. J. Transl. Res. 2012, 4, 127–150. [Google Scholar] [PubMed]
- Hombach, S.; Kretz, M. Non-coding RNAs: Classification, biology and functioning. Adv. Exp. Med. Biol. 2016, 937, 3–17. [Google Scholar] [CrossRef] [PubMed]
- Salta, E.; De Strooper, B. Non-coding RNAs with essential roles in neurodegenerative disorders. Lancet Neurol. 2012, 11, 189–200. [Google Scholar] [CrossRef]
- Qureshi, I.A.; Mehler, M.F. Emerging roles of non-coding RNAs in brain evolution, development, plasticity and disease. Nat. Rev. Neurosci. 2012, 13, 528–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, Y.-Y.; Kuo, H.-C. Functional roles and networks of non-coding RNAs in the pathogenesis of neurodegenerative diseases. J. Biomed. Sci. 2020, 27, 1–23. [Google Scholar] [CrossRef] [Green Version]
- Ma, F.; Zhang, X.; Yin, K.-J. MicroRNAs in central nervous system diseases: A prospective role in regulating blood-brain barrier integrity. Exp. Neurol. 2020, 323, 113094. [Google Scholar] [CrossRef]
- Duran, R.C.-D.; Wei, H.; Kim, D.H.; Wu, J.Q. Invited review: Long non-coding RNAs: Important regulators in the development, function and disorders of the central nervous system. Neuropathol. Appl. Neurobiol. 2019, 45, 538–556. [Google Scholar] [CrossRef] [Green Version]
- Esteller, M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011, 12, 861–874. [Google Scholar] [CrossRef] [PubMed]
- Dröge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef]
- Beckman, K.B.; Ames, B.N. The free radical theory of aging matures. Physiol. Rev. 1998, 78, 547–581. [Google Scholar] [CrossRef] [Green Version]
- Bush, A.I. Metals and neuroscience. Curr. Opin. Chem. Biol. 2000, 4, 184–191. [Google Scholar] [CrossRef]
- Jan, A.T.; Azam, M.; Siddiqui, K.; Ali, A.; Choi, I.; Haq, Q.M.R. Heavy metals and human health: Mechanistic insight into toxicity and counter defense system of antioxidants. Int. J. Mol. Sci. 2015, 16, 29592–29630. [Google Scholar] [CrossRef] [Green Version]
- Calderón-Garcidueñas, L.; Leray, E.; Heydarpour, P.; Torres-Jardón, R.; Reis, J. Air pollution, a rising environmental risk factor for cognition, neuroinflammation and neurodegeneration: The clinical impact on children and beyond. Rev. Neurol. 2016, 172. [Google Scholar] [CrossRef]
- Harwell, B. Biochemistry of oxidative stress. Biochem Soc Trans. 2007, 35 (Pt 5), 1147–1150. [Google Scholar]
- Echtay, K.S.; Roussel, D.; St-Pierre, J.; Jekabsons, M.B.; Cadenas, S.; Stuart, J.A.; Harper, J.A.; Roebuck, S.J.; Morrison, A.; Pickering, S.; et al. Superoxide activates mitochondrial uncoupling proteins. Nat. Cell Biol. 2002, 415, 96–99. [Google Scholar] [CrossRef] [PubMed]
- Halliwell, B. Free radicals, antioxidants, and human disease: Curiosity, cause, or consequence? Lancet 1994, 344, 721–724. [Google Scholar] [CrossRef]
- Fulda, S.; Gorman, A.M.; Hori, O.; Samali, A. Cellular stress responses: Cell survival and cell death. Int. J. Cell Biol. 2010, 2010, 1–23. [Google Scholar] [CrossRef] [Green Version]
- Adibhatla, R.M.; Hatcher, J.F. Lipid oxidation and peroxidation in cns health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2010, 12, 125–169. [Google Scholar] [CrossRef] [PubMed]
- Fu, H.; Hardy, J.; Duff, K.E. Selective vulnerability in neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1350–1358. [Google Scholar] [CrossRef]
- Smith, D.G.; Cappai, R.; Barnham, K.J. The redox chemistry of the Alzheimer’s disease amyloid β peptide. Biochim. Biophys. Acta (BBA)-Biomembr. 2007, 1768, 1976–1990. [Google Scholar] [CrossRef] [Green Version]
- Dinkova-Kostova, A.T.; Talalay, P.; Sharkey, J.; Zhang, Y.; Holtzclaw, W.D.; Wang, X.J.; David, E.; Schiavoni, K.H.; Finlayson, S.; Mierke, D.F.; et al. An exceptionally potent inducer of cytoprotective enzymes: Elucidation of the structural features that determine inducer potency and reactivity with Keap1. J. Biol. Chem. 2010, 285, 33747–33755. [Google Scholar] [CrossRef] [Green Version]
- Ghafouri-Fard, S.; Shoorei, H.; Taheri, M. Non-coding RNAs are involved in the response to oxidative stress. Biomed. Pharmacother. 2020, 127, 110228. [Google Scholar] [CrossRef] [PubMed]
- Finkel, T.; Holbrook, N.J. Oxidants, oxidative stress and the biology of ageing. Nat. Cell Biol. 2000, 408, 239–247. [Google Scholar] [CrossRef]
- Martinez, A.; Pamplona, R.; Ferrer, I.; Portero-Otin, M. Protein targets of oxidative damage in human neurodegenerative diseases with abnormal protein aggregates. Brain Pathol. 2010, 20, 281–297. [Google Scholar] [CrossRef]
- Sedelnikova, O.A.; Redon, C.E.; Dickey, J.S.; Nakamura, A.J.; Georgakilas, A.G.; Bonner, W.M. Role of oxidatively induced DNA lesions in human pathogenesis. Mutat. Res./Rev. Mutat. Res. 2010, 704, 152–159. [Google Scholar] [CrossRef] [Green Version]
- Korolainen, M.A.; Nyman, T.A.; Nyyssonen, P.; Hartikainen, E.S.; Pirttila, T. Multiplexed proteomic analysis of oxidation and concentrations of cerebrospinal fluid proteins in alzheimer disease. Clin. Chem. 2007, 53, 657–665. [Google Scholar] [CrossRef] [Green Version]
- Smith, M.A.; Sayre, L.M.; Anderson, V.E.; Harris, P.L.; Beal, M.F.; Kowall, N.; Perry, G. Cytochemical demonstration of oxidative damage in alzheimer disease by immunochemical enhancement of the carbonyl reaction with 2,4-dinitrophenylhydrazine. J. Histochem. Cytochem. 1998, 46, 731–735. [Google Scholar] [CrossRef] [Green Version]
- Davies, M.J. The oxidative environment and protein damage. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2005, 1703, 93–109. [Google Scholar] [CrossRef]
- Peña-Bautista, C.; Vento, M.; Baquero, M.; Cháfer-Pericás, C. Lipid peroxidation in neurodegeneration. Clin. Chim. Acta 2019, 497, 178–188. [Google Scholar] [CrossRef] [PubMed]
- Xiao, M.; Zhong, H.; Xia, L.; Tao, Y.; Yin, H. Pathophysiology of mitochondrial lipid oxidation: Role of 4-hydroxynonenal (4-HNE) and other bioactive lipids in mitochondria. Free Radic. Biol. Med. 2017, 111, 316–327. [Google Scholar] [CrossRef]
- Bruce-Keller, A.J.; Li, Y.J.; Lovell, M.A.; Kraemer, P.J.; Gary, D.S.; Brown, R.R.; Markesbery, W.R.; Mattson, M.P. 4-Hydroxynonenal, a product of lipid peroxidation, damages cholinergic neurons and impairs visuospatial memory in rats. J. Neuropathol. Exp. Neurol. 1998, 57, 257–267. [Google Scholar] [CrossRef] [Green Version]
- Keller, J.N.; Pang, Z.; Geddes, J.W.; Begley, J.G.; Germeyer, A.; Waeg, G.; Mattson, M.P. Impairment of glucose and glutamate transport and induction of mitochondrial oxidative stress and dysfunction in synaptosomes by amyloid β-peptide: Role of the lipid peroxidation product 4-hydroxynonenal. J. Neurochem. 2002, 69, 273–284. [Google Scholar] [CrossRef]
- Markesbery, W.R.; Lovell, M.A. DNA oxidation in Alzheimer’s disease. Antioxid. Redox Signal. 2006, 8, 2039–2045. [Google Scholar] [CrossRef]
- Lovell, M.A.; Markesbery, W.R. Oxidative DNA damage in mild cognitive impairment and late-stage Alzheimer’s disease. Nucleic Acids Res. 2007, 35, 7497–7504. [Google Scholar] [CrossRef] [Green Version]
- Lovell, M.A.; Gabbita, S.P.; Markesbery, W.R. Increased DNA oxidation and decreased levels of repair products in Alzheimer’s disease ventricular CSF. J. Neurochem. 1999, 72, 771–776. [Google Scholar] [CrossRef]
- Nunomura, A.; Perry, G.; Pappolla, M.A.; Wade, R.; Hirai, K.; Chiba, S.; Smith, M.A. RNA oxidation is a prominent feature of vulnerable neurons in Alzheimer’s disease. J. Neurosci. 1999, 19, 1959–1964. [Google Scholar] [CrossRef]
- Byun, K.; Yoo, Y.; Son, M.; Lee, J.; Jeong, G.-B.; Park, Y.M.; Salekdeh, G.H.; Lee, B. Advanced glycation end-products produced systemically and by macrophages: A common contributor to inflammation and degenerative diseases. Pharmacol. Ther. 2017, 177, 44–55. [Google Scholar] [CrossRef]
- Li, J.; Liu, D.; Sun, L.; Lu, Y.; Zhang, Z. Advanced glycation end products and neurodegenerative diseases: Mechanisms and perspective. J. Neurol. Sci. 2012, 317, 1–5. [Google Scholar] [CrossRef]
- Münch, G.; Cunningham, A.M.; Riederer, P.; Braak, H. Advanced glycation endproducts are associated with Hirano bodies in Alzheimer’s disease. Brain Res. 1998, 796, 307–310. [Google Scholar] [CrossRef]
- Gandhi, J.; Antonelli, A.C.; Afridi, A.; Vatsia, S.; Joshi, G.; Romanov, V.; Murray, I.V.; Khan, S.A. Protein misfolding and aggregation in neurodegenerative diseases: A review of pathogeneses, novel detection strategies, and potential therapeutics. Rev. Neurosci. 2019, 30, 339–358. [Google Scholar] [CrossRef]
- Kumar, V.; Sami, N.; Kashav, T.; Islam, A.; Ahmad, F.; Hassan, M.I. Protein aggregation and neurodegenerative diseases: From theory to therapy. Eur. J. Med. Chem. 2016, 124, 1105–1120. [Google Scholar] [CrossRef]
- Lévy, E.; El Banna, N.; Baïlle, D.; Heneman-Masurel, A.; Truchet, S.; Rezaei, H.; Huang, M.-E.; Béringue, V.; Martin, D.; Vernis, L. Causative links between protein aggregation and oxidative stress: A review. Int. J. Mol. Sci. 2019, 20, 3896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rousseau, A.; Bertolotti, A. Regulation of proteasome assembly and activity in health and disease. Nat. Rev. Mol. Cell Biol. 2018, 19, 697–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seifert, U.; Bialy, L.P.; Ebstein, F.; Bech-Otschir, D.; Voigt, A.; Schröter, F.; Prozorovski, T.; Lange, N.; Steffen, J.; Rieger, M.; et al. Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell 2010, 142, 613–624. [Google Scholar] [CrossRef] [Green Version]
- Ciechanover, A.; Brundin, P. The ubiquitin proteasome system in neurodegenerative diseases: Sometimes the chicken, sometimes the egg. Neuron 2003, 40, 427–446. [Google Scholar] [CrossRef] [Green Version]
- Green, E.D.; Watson, J.D.; Collins, F. Human genome project: Twenty-five years of big biology. Nat. Cell Biol. 2015, 526, 29–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- The ENCODE Project Consortium; Snyder, M.P.; Gingeras, T.R.; Moore, J.E.; Weng, Z.; Gerstein, M.B.; Ren, B.; Hardison, R.C.; Stamatoyannopoulos, J.A.; Graveley, B.R.; et al. Perspectives on ENCODE. Nature 2020, 583, 693–698. [Google Scholar]
- Mattick, J.S. Deconstructing the dogma: A new view of the evolution and genetic programming of complex organisms. Ann. N. Y. Acad. Sci. 2009. [Google Scholar] [CrossRef]
- Yang, J.X.; Rastetter, R.H.; Wilhelm, D. Non-coding RNAs: An introduction. Adv. Exp. Med. Biol. 2016, 886, 13–32. [Google Scholar]
- Quan, Z.; Zheng, D.; Qing, H. Regulatory roles of long non-coding rnas in the central nervous system and associated neurodegenerative diseases. Front. Cell. Neurosci. 2017, 11, 175. [Google Scholar] [CrossRef]
- Mercer, T.R.; Dinger, M.E.; Sunkin, S.M.; Mehler, M.F.; Mattick, J.S. Specific expression of long noncoding RNAs in the mouse brain. Proc. Natl. Acad. Sci. USA 2008, 105, 716–721. [Google Scholar] [CrossRef] [Green Version]
- Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
- Bhaskaran, M.; Mohan, M. MicroRNAs: History, biogenesis, and their evolving role in animal development and disease. Vet. Pathol. 2014, 51, 759–774. [Google Scholar] [CrossRef] [Green Version]
- Zlotorynski, E. Insights into the kinetics of microRNA biogenesis and turnover. Nat. Rev. Mol. Cell Biol. 2019, 20, 511. [Google Scholar] [CrossRef]
- Cuellar, T.L.; Davis, T.H.; Nelson, P.T.; Loeb, G.B.; Harfe, B.D.; Ullian, E.; McManus, M.T. Dicer loss in striatal neurons produces behavioral and neuroanatomical phenotypes in the absence of neurodegeneration. Proc. Natl. Acad. Sci. USA 2008, 105, 5614–5619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rajman, M.; Schratt, G. MicroRNAs in neural development: From master regulators to fine-tuners. Development 2017, 144, 2310–2322. [Google Scholar] [CrossRef] [Green Version]
- Fineberg, S.K.; Kosik, K.S.; Davidson, B.L. MicroRNAs potentiate neural development. Neuron 2009, 64, 303–309. [Google Scholar] [CrossRef] [Green Version]
- Bernstein, E.; Kim, S.Y.; Carmell, A.M.; Murchison, E.P.; Alcorn, H.L.; Li, M.Z.; Mills, A.A.; Elledge, S.J.; Anderson, K.V.; Hannon, G.J. Dicer is essential for mouse development. Nat. Genet. 2003, 35, 215–217. [Google Scholar] [CrossRef]
- Davis, T.H.; Cuellar, T.L.; Koch, S.M.; Barker, A.J.; Harfe, B.D.; McManus, M.T.; Ullian, E.M. Conditional loss of dicer disrupts cellular and tissue morphogenesis in the cortex and hippocampus. J. Neurosci. 2008, 28, 4322–4330. [Google Scholar] [CrossRef] [Green Version]
- Chmielarz, P.; Konovalova, J.; Najam, S.S.; Alter, H.; Piepponen, T.P.; Erfle, H.; Sonntag, K.C.; Schütz, G.; Vinnikov, I.A.; Domanskyi, A. Dicer and microRNAs protect adult dopamine neurons. Cell Death Dis. 2017, 8, e2813. [Google Scholar] [CrossRef] [Green Version]
- Hu, Z.; Li, Z. MiRNAs in synapse development and synaptic plasticity. Curr. Opin. Neurobiol. 2017, 45, 24–31. [Google Scholar] [CrossRef] [PubMed]
- Strazisar, M.; Cammaerts, S.; Van Der Ven, K.; Forero, A.D.; Lenaerts, A.-S.; Nordin, A.; Almeida-Souza, L.; Genovese, G.; Timmerman, V.; Liekens, A.; et al. MIR137 variants identified in psychiatric patients affect synaptogenesis and neuronal transmission gene sets. Mol. Psychiatry 2015, 20, 472–481. [Google Scholar] [CrossRef]
- The Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium. Genome-wide association study identifies five new schizophrenia loci. Nat. Genet. 2011, 43, 969–976. [Google Scholar] [CrossRef] [PubMed]
- Loohuis, N.F.O.; Ba, W.; Stoerchel, P.H.; Kos, A.; Jager, A.; Schratt, G.; Martens, G.J.; Van Bokhoven, H.; Kasri, N.N.; Aschrafi, A.; et al. MicroRNA-137 controls ampa-receptor-mediated transmission and mGluR-Dependent LTD. Cell Rep. 2015, 11, 1876–1884. [Google Scholar] [CrossRef] [Green Version]
- Harraz, M.M.; Eacker, S.M.; Wang, X.; Dawson, T.M.; Dawson, V.L. MicroRNA-223 is neuroprotective by targeting glutamate receptors. Proc. Natl. Acad. Sci. USA 2012, 109, 18962–18967. [Google Scholar] [CrossRef] [Green Version]
- Edbauer, D.; Neilson, J.R.; Foster, K.A.; Wang, C.-F.; Seeburg, D.P.; Batterton, M.N.; Tada, T.; Dolan, B.M.; Sharp, P.A.; Sheng, M. Regulation of Synaptic Structure and Function by FMRP-Associated MicroRNAs miR-125b and miR-132. Neuron 2010, 65, 373–384. [Google Scholar] [CrossRef] [Green Version]
- Hu, Z.; Yu, D.; Gu, Q.-H.; Yang, Y.; Tu, K.; Zhu, J.; Li, Z. miR-191 and miR-135 are required for long-lasting spine remodelling associated with synaptic long-term depression. Nat. Commun. 2014, 5, 1–17. [Google Scholar] [CrossRef] [Green Version]
- Schratt, G.M.; Tuebing, F.; Nigh, E.A.; Kane, C.G.; Sabatini, M.E.; Kiebler, M.; Greenberg, M.E. A brain-specific microRNA regulates dendritic spine development. Nat. Cell Biol. 2006, 439, 283–289. [Google Scholar] [CrossRef]
- Antoniou, A.; Khudayberdiev, S.; Idziak, A.; Bicker, S.; Jacob, R.; Schratt, G. The dynamic recruitment of TRBP to neuronal membranes mediates dendritogenesis during development. EMBO Rep. 2017, 19, e44853. [Google Scholar] [CrossRef]
- Fiore, R.; Khudayberdiev, S.; Christensen, M.; Siegel, G.; Flavell, S.W.; Kim, T.-K.; Greenberg, M.E.; Schratt, G.M. Mef2-mediated transcription of the miR379–410 cluster regulates activity-dependent dendritogenesis by fine-tuning Pumilio2 protein levels. EMBO J. 2009, 28, 697–710. [Google Scholar] [CrossRef] [PubMed]
- Marler, K.J.; Suetterlin, P.; Dopplapudi, A.; Rubikaite, A.; Adnan, J.; Maiorano, N.A.; Lowe, A.S.; Thompson, I.D.; Pathania, M.; Bordey, A.; et al. BDNF promotes axon branching of retinal ganglion cells via miRNA-132 and p250GAP. J. Neurosci. 2014, 34, 969–979. [Google Scholar] [CrossRef]
- Zhao, X.; He, X.; Han, X.; Yu, Y.; Ye, F.; Chen, Y.; Hoang, T.; Xu, X.; Mi, Q.-S.; Xin, M.; et al. MicroRNA-Mediated control of oligodendrocyte differentiation. Neuron 2010, 65, 612–626. [Google Scholar] [CrossRef] [Green Version]
- Dugas, J.C.; Cuellar, T.L.; Scholze, A.; Ason, B.; Ibrahim, A.; Emery, B.; Zamanian, J.L.; Foo, L.C.; McManus, M.T.; Barres, B.A. Dicer1 and miR-219 are required for normal oligodendrocyte differentiation and myelination. Neuron 2010, 65, 597–611. [Google Scholar] [CrossRef] [Green Version]
- Pogue, A.; Cui, J.; Li, Y.; Zhao, Y.; Culicchia, F.; Lukiw, W. Micro RNA-125b (miRNA-125b) function in astrogliosis and glial cell proliferation. Neurosci. Lett. 2010, 476, 18–22. [Google Scholar] [CrossRef]
- Taj, S.H.; Kho, W.; Riou, A.; Wiedermann, D.; Hoehn, M. MiRNA-124 induces neuroprotection and functional improvement after focal cerebral ischemia. Biomaterials 2016, 91, 151–165. [Google Scholar] [CrossRef]
- Guo, Y.; Hong, W.; Wang, X.; Zhang, P.; Körner, H.; Tu, J.; Wei, W. MicroRNAs in microglia: How do MicroRNAs affect activation, inflammation, polarization of microglia and mediate the interaction between microglia and glioma? Front. Mol. Neurosci. 2019, 12, 125. [Google Scholar] [CrossRef]
- Kumar, P.; Kuscu, C.; Dutta, A. biogenesis and function of transfer RNA-related fragments (tRFs). Trends Biochem. Sci. 2016, 41, 679–689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamasaki, S.; Ivanov, P.; Hu, G.-F.; Anderson, P. Angiogenin cleaves tRNA and promotes stress-induced translational repression. J. Cell Biol. 2009, 185, 35–42. [Google Scholar] [CrossRef] [Green Version]
- Kumar, P.; Mudunuri, S.B.; Anaya, J.; Dutta, A. TRFdb: A database for transfer RNA fragments. Nucleic Acids Res. 2015, 43, D141–D145. [Google Scholar] [CrossRef]
- Guzzi, N.; Bellodi, C. Novel insights into the emerging roles of tRNA-derived fragments in mammalian development. RNA Biol. 2020, 17, 1214–1222. [Google Scholar] [CrossRef] [Green Version]
- Karaiskos, S.; Grigoriev, A. Dynamics of tRNA fragments and their targets in aging mammalian brain. F1000Research 2016, 5, 2758. [Google Scholar] [CrossRef]
- Zhang, S.; Li, H.; Zheng, L.; Li, H.; Feng, C.; Zhang, W. Identification of functional tRNA-derived fragments in senescence-accelerated mouse prone 8 brain. Aging 2019, 11, 10485–10498. [Google Scholar] [CrossRef]
- Krishna, S.; Yim, D.G.; Lakshmanan, V.; Tirumalai, V.; Koh, J.L.; Park, J.E.; Cheong, J.K.; Low, J.L.; Lim, M.J.; Sze, S.K.; et al. Dynamic expression of tRNA-derived small RNAs define cellular states. EMBO Rep. 2019, 20. [Google Scholar] [CrossRef]
- Yu, M.; Lu, B.; Zhang, J.; Ding, J.; Liu, P.; Lu, Y. tRNA-derived RNA fragments in cancer: Current status and future perspectives. J. Hematol. Oncol. 2020, 13, 1–14. [Google Scholar] [CrossRef]
- Maute, R.L.; Schneider, C.; Sumazin, P.; Holmes, A.; Califano, A.; Basso, K.; Dalla-Favera, R. tRNA-derived microRNA modulates proliferation and the DNA damage response and is down-regulated in B cell lymphoma. Proc. Natl. Acad. Sci. USA 2013, 110, 1404–1409. [Google Scholar] [CrossRef] [Green Version]
- Guan, L.; Karaiskos, S.; Grigoriev, A. Inferring targeting modes of Argonaute-loaded tRNA fragments. RNA Biol. 2019, 17, 1070–1080. [Google Scholar] [CrossRef]
- Kumar, P.; Anaya, J.; Mudunuri, S.B.; Dutta, A. Meta-analysis of tRNA derived RNA fragments reveals that they are evolutionarily conserved and associate with AGO proteins to recognize specific RNA targets. BMC Biol. 2014, 12, 78. [Google Scholar] [CrossRef] [PubMed]
- Gonskikh, Y.; Gerstl, M.; Kos, M.; Borth, N.; Schosserer, M.; Grillari, J.; Polacek, N. Modulation of mammalian translation by a ribosome-associated tRNA half. RNA Biol. 2020, 17, 1125–1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wolozin, B.; Ivanov, P. Stress granules and neurodegeneration. Nat. Rev. Neurosci. 2019, 20, 649–666. [Google Scholar] [CrossRef]
- Ivanov, P.; O’Day, E.; Emara, M.M.; Wagner, G.; Lieberman, J.; Anderson, P.J. G-quadruplex structures contribute to the neuroprotective effects of angiogenin-induced tRNA fragments. Proc. Natl. Acad. Sci. USA 2014, 111, 18201–18206. [Google Scholar] [CrossRef] [Green Version]
- Ivanov, P.; Emara, M.M.; Villen, J.; Gygi, S.P.; Anderson, P. Angiogenin-induced tRNA fragments inhibit translation initiation. Mol. Cell 2011, 43, 613–623. [Google Scholar] [CrossRef] [Green Version]
- Lyons, S.M.; Achorn, C.; Kedersha, N.L.; Anderson, P.J.; Ivanov, P. YB-1 regulates tiRNA-induced Stress Granule formation but not translational repression. Nucleic Acids Res. 2016, 44, 6949–6960. [Google Scholar] [CrossRef]
- Salvatori, B.; Biscarini, S.; Morlando, M. Non-coding RNAs in nervous system development and disease. Front. Cell Dev. Biol. 2020, 8, 8. [Google Scholar] [CrossRef]
- Ulitsky, I.; Bartel, D.P. LincRNAs: Genomics, evolution, and mechanisms. Cell 2013, 154, 26–46. [Google Scholar] [CrossRef] [Green Version]
- Wang, K.C.; Chang, H.Y. Molecular mechanisms of long noncoding RNAs. Mol. Cell 2011, 43, 904–914. [Google Scholar] [CrossRef] [Green Version]
- An, H.; Williams, N.G.; Shelkovnikova, T.A. NEAT1 and paraspeckles in neurodegenerative diseases: A missing lnc found? Non-Coding RNA Res. 2018, 3, 243–252. [Google Scholar] [CrossRef]
- Zhao, Y.; Sun, L.; Wang, R.R.; Hu, J.-F.; Cui, J. The effects of mitochondria-associated long noncoding RNAs in cancer mitochondria: New players in an old arena. Crit. Rev. Oncol. 2018, 131, 76–82. [Google Scholar] [CrossRef]
- Chen, L.-L. Linking long noncoding RNA localization and function. Trends Biochem. Sci. 2016, 41, 761–772. [Google Scholar] [CrossRef]
- Briggs, J.A.; Wolvetang, E.J.; Mattick, J.S.; Rinn, J.L.; Barry, G. Mechanisms of long non-coding RNAs in mammalian nervous system development, plasticity, disease, and evolution. Neuron 2015, 88, 861–877. [Google Scholar] [CrossRef] [Green Version]
- Pollard, K.S.; Salama, S.R.; Lambert, N.; Lambot, M.-A.; Coppens, S.; Pedersen, J.S.; Katzman, S.; King, B.; Onodera, C.; Siepel, A.; et al. An RNA gene expressed during cortical development evolved rapidly in humans. Nat. Cell Biol. 2006, 443, 167–172. [Google Scholar] [CrossRef]
- Bernard, D.; Prasanth, K.V.; Tripathi, V.; Colasse, S.; Nakamura, T.; Xuan, Z.; Zhang, M.Q.; Sedel, F.; Jourdren, L.; Coulpier, F.; et al. A long nuclear-retained non-coding RNA regulates synaptogenesis by modulating gene expression. EMBO J. 2010, 29, 3082–3093. [Google Scholar] [CrossRef] [Green Version]
- Hart, R.P.; Goff, L.A. Long noncoding RNAs: Central to nervous system development. Int. J. Dev. Neurosci. 2016, 55, 109–116. [Google Scholar] [CrossRef] [Green Version]
- Keihani, S.; Kluever, V.; Mandad, S.; Bansal, V.; Rahman, R.; Fritsch, E.; Gomes, L.C.; Gärtner, A.; Kügler, S.; Urlaub, H.; et al. The long noncoding RNA neuroLNC regulates presynaptic activity by interacting with the neurodegeneration-associated protein TDP-43. Sci. Adv. 2019, 5, eaay2670. [Google Scholar] [CrossRef] [Green Version]
- Yang, C.; Tang, R.; Ma, X.; Wang, Y.; Luo, D.; Xu, Z.; Zhu, Y.; Yang, L. Tag SNPs in long non-coding RNA H19 contribute to susceptibility to gastric cancer in the Chinese Han population. Oncotarget 2015, 6, 15311–15320. [Google Scholar] [CrossRef] [Green Version]
- Han, C.-L.; Ge, M.; Liu, Y.-P.; Zhao, X.-M.; Wang, K.-L.; Chen, N.; Meng, W.-J.; Hu, W.; Zhang, J.-G.; Li, L.; et al. LncRNA H19 contributes to hippocampal glial cell activation via JAK/STAT signaling in a rat model of temporal lobe epilepsy. J. Neuroinflamm. 2018, 15, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Bond, A.M.; VanGompel, M.J.W.; Sametsky, E.A.; Clark, M.F.; Savage, J.C.; Disterhoft, J.F.; Kohtz, J.D. Balanced gene regulation by an embryonic brain ncRNA is critical for adult hippocampal GABA circuitry. Nat. Neurosci. 2009, 12, 1020–1027. [Google Scholar] [CrossRef] [Green Version]
- Ng, S.-Y.; Bogu, G.K.; Soh, B.S.; Stanton, L.W. The long noncoding RNA RMST interacts with SOX2 to regulate neurogenesis. Mol. Cell 2013, 51, 349–359. [Google Scholar] [CrossRef] [Green Version]
- Reddy, A.S.; O’Brien, D.; Pisat, N.; Weichselbaum, C.T.; Sakers, K.; Lisci, M.; Dalal, J.S.; Dougherty, J.D. A comprehensive analysis of cell type–specific nuclear RNA from neurons and glia of the brain. Biol. Psychiatry 2017, 81, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Ramos, A.D.; Andersen, R.E.; Liu, S.J.; Nowakowski, T.J.; Hong, S.J.; Gertz, C.C.; Salinas, R.D.; Zarabi, H.; Kriegstein, A.R.; Lim, D.A. The long noncoding RNA pnky regulates neuronal differentiation of embryonic and postnatal neural stem cells. Cell Stem Cell 2015, 16, 439–447. [Google Scholar] [CrossRef] [Green Version]
- Kleaveland, B.; Shi, C.Y.; Stefano, J.; Bartel, D.P. A network of noncoding regulatory RNAs Acts in the mammalian brain. Cell 2018, 174, 350–362.e17. [Google Scholar] [CrossRef] [Green Version]
- Jeck, W.R.; Sorrentino, J.A.; Wang, K.; Slevin, M.K.; Burd, C.E.; Liu, J.; Marzluff, W.F.; Sharpless, N.E. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 2013, 19, 141–157. [Google Scholar] [CrossRef] [Green Version]
- Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.W.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The biogenesis, biology and characterization of circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef]
- Yu, C.-Y.; Kuo, H.-C. The emerging roles and functions of circular RNAs and their generation. J. Biomed. Sci. 2019, 26, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Mehta, S.L.; Dempsey, R.J.; Vemuganti, R. Role of circular RNAs in brain development and CNS diseases. Prog. Neurobiol. 2020, 186, 101746. [Google Scholar] [CrossRef]
- You, X.; Vlatkovic, I.; Babic, A.; Will, T.J.; Epstein, I.; Tushev, G.; Akbalik, G.; Wang, M.; Glock, C.; Quedenau, C.; et al. Neural circular RNAs are derived from synaptic genes and regulated by development and plasticity. Nat. Neurosci. 2015, 18, 603–610. [Google Scholar] [CrossRef] [Green Version]
- Piwecka, M.; Glažar, P.; Hernandez-Miranda, L.R.; Memczak, S.; Wolf, S.A.; Rybak-Wolf, A.; Filipchyk, A.; Klironomos, F.; Jara, C.A.C.; Fenske, P.; et al. Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science 2017, 357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Du, L.; Bai, Y.; Han, B.; He, C.; Gong, L.; Huang, R.; Shen, L.; Chao, J.; Liu, P.; et al. CircDYM ameliorates depressive-like behavior by targeting miR-9 to regulate microglial activation via HSP90 ubiquitination. Mol. Psychiatry 2018, 25, 1175–1190. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Chen, X.; Li, Z.; Ye, W.; Ding, H.; Li, P.; Aung, L.H.H. Role of RNA oxidation in neurodegenerative diseases. Int. J. Mol. Sci. 2020, 21, 5022. [Google Scholar] [CrossRef]
- Shcherbik, N.; Pestov, D.G. The impact of oxidative stress on ribosomes: From injury to regulation. Cells 2019, 8, 1379. [Google Scholar] [CrossRef] [Green Version]
- Leisegang, M.S.; Schröder, K.; Brandes, R.P. Redox regulation and noncoding RNAs. Antioxidants Redox Signal. 2018, 29, 793–812. [Google Scholar] [CrossRef] [PubMed]
- Chu, L.; Su, M.Y.; Maggi, L.B., Jr.; Lu, L.; Mullins, C.; Crosby, S.; Huang, G.; Chng, W.J.; Vij, R.; Tomasson, M.H. Multiple myeloma–associated chromosomal translocation activates orphan snoRNA ACA11 to suppress oxidative stress. J. Clin. Investig. 2012, 122, 2793–2806. [Google Scholar] [CrossRef]
- Sahoo, T.; Del Gaudio, D.; German, J.R.; Shinawi, M.; Peters, S.U.; Person, R.E.; Garnica, A.D.; Cheung, S.W.; Beaudet, A.L. Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat. Genet. 2008, 40, 719–721. [Google Scholar] [CrossRef] [Green Version]
- Taft, R.J.; Glazov, E.A.; Lassmann, T.; Hayashizaki, Y.; Carninci, P.; Mattick, J.S. Small RNAs derived from snoRNAs. RNA 2009, 15, 1233–1240. [Google Scholar] [CrossRef] [Green Version]
- Barker, W.W.; Luis, C.A.; Kashuba, A.; Luis, M.; Harwood, D.G.; Loewenstein, D.; Waters, C.; Jimison, P.; Shepherd, E.; Sevush, S.; et al. Relative frequencies of alzheimer disease, lewy body, vascular and frontotemporal dementia, and hippocampal sclerosis in the state of florida brain bank. Alzheimer Dis. Assoc. Disord. 2002, 16, 203–212. [Google Scholar] [CrossRef] [PubMed]
- Tönnies, E.; Trushina, E. Oxidative stress, synaptic dysfunction, and Alzheimer’s disease. J. Alzheimer’s Dis. 2017, 57, 1105–1121. [Google Scholar] [CrossRef] [Green Version]
- Lanoiselée, H.-M.; Nicolas, G.; Wallon, D.; Rovelet-Lecrux, A.; Lacour, M.; Rousseau, S.; Richard, A.-C.; Pasquier, F.; Rollin-Sillaire, A.; Martinaud, O.; et al. APP, PSEN1, and PSEN2 mutations in early-onset Alzheimer disease: A genetic screening study of familial and sporadic cases. PLoS Med. 2017, 14, e1002270. [Google Scholar] [CrossRef] [Green Version]
- Van Cauwenberghe, C.; Van Broeckhoven, C.; Sleegers, K. The genetic landscape of Alzheimer disease: Clinical implications and perspectives. Genet. Med. 2016, 18, 421–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Millan, M.J. Linking deregulation of non-coding RNA to the core pathophysiology of Alzheimer’s disease: An integrative review. Prog. Neurobiol. 2017, 156, 1–68. [Google Scholar] [CrossRef]
- Zhao, J.; Yue, D.; Zhou, Y.; Jia, L.; Wang, H.; Guo, M.; Xu, H.; Chen, C.; Zhang, J.; Xu, L. The role of MicroRNAs in Aβ deposition and tau phosphorylation in Alzheimer’s disease. Front. Neurol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
- Smith, P.Y.; Hernandez-Rapp, J.; Jolivette, F.; Lecours, C.; Bisht, K.; Goupil, C.; Dorval, V.; Parsi, S.; Morin, F.; Planel, E.; et al. miR-132/212 deficiency impairs tau metabolism and promotes pathological aggregation in vivo. Hum. Mol. Genet. 2015, 24, 6721–6735. [Google Scholar] [CrossRef] [Green Version]
- Sierksma, A.; Lu, A.; Salta, E.; vanden Eynden, E.; Callaerts-Vegh, Z.; D’Hooge, R.; Blum, D.; Buée, L.; Fiers, M.; De Strooper, B. Deregulation of neuronal miRNAs induced by amyloid-β or TAU pathology. Mol. Neurodegener. 2018, 13, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Amakiri, N.; Kubosumi, A.; Tran, J.; Reddy, P.H. Amyloid beta and MicroRNAs in Alzheimer’s disease. Front. Neurosci. 2019, 13, 430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, J.; Yoon, H.; Chung, D.-E.; Brown, J.L.; Belmonte, K.C.; Kim, J. MiR-186 is decreased in aged brain and suppresses BACE1 expression. J. Neurochem. 2016, 137, 436–445. [Google Scholar] [CrossRef] [Green Version]
- Zhang, R.; Zhang, Q.; Niu, J.; Lu, K.; Xie, B.; Cui, D.; Xu, S. Screening of microRNAs associated with Alzheimer’s disease using oxidative stress cell model and different strains of senescence accelerated mice. J. Neurol. Sci. 2014, 338, 57–64. [Google Scholar] [CrossRef]
- Zhou, Y.; Wang, Z.-F.; Li, W.; Hong, H.; Chen, J.; Tian, Y.; Liu, Z.-Y. Protective effects of microRNA-330 on amyloid β-protein production, oxidative stress, and mitochondrial dysfunction in Alzheimer’s disease by targeting VAV1 via the MAPK signaling pathway. J. Cell. Biochem. 2018, 119, 5437–5448. [Google Scholar] [CrossRef]
- Zhang, L.; Dong, H.; Si, Y.; Wu, N.; Cao, H.; Mei, B.; Meng, B. MiR-125b promotes tau phosphorylation by targeting the neural cell adhesion molecule in neuropathological progression. Neurobiol. Aging 2019, 73, 41–49. [Google Scholar] [CrossRef]
- Jin, Y.; Tu, Q.; Liu, M. MicroRNA-125b regulates Alzheimer’s disease through SphK1 regulation. Mol. Med. Rep. 2018, 18, 2373–2380. [Google Scholar] [CrossRef]
- Li, J.J.; Dolios, G.; Wang, R.; Liao, F.-F. Soluble beta-amyloid peptides, but not insoluble fibrils, have specific effect on neuronal MicroRNA expression. PLoS ONE 2014, 9, e90770. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.-X.; Rajeev, B.W.; Stromberg, A.J.; Ren, N.; Tang, G.; Huang, Q.; Rigoutsos, I.; Nelson, P.T. The expression of MicroRNA miR-107 decreases early in Alzheimer’s disease and may accelerate disease progression through regulation of β-site amyloid precursor protein-cleaving enzyme 1. J. Neurosci. 2008, 28, 1213–1223. [Google Scholar] [CrossRef]
- De La Monte, S.M.; Wands, J.R. Molecular indices of oxidative stress and mitochondrial dysfunction occur early and often progress with severity of Alzheimer’s disease. J. Alzheimer’s Dis. 2006, 9, 167–181. [Google Scholar] [CrossRef]
- Prendecki, M.; Florczak-Wyspianska, J.; Kowalska, M.; Ilkowski, J.; Grzelak, T.; Bialas, K.; Kozubski, W.; Dorszewska, J. APOE genetic variants and apoE, miR-107 and miR-650 levels in Alzheimer’s disease. Folia Neuropathol. 2019, 57, 106–116. [Google Scholar] [CrossRef] [PubMed]
- Liang, C.; Zhu, H.; Xu, Y.; Huang, L.; Ma, C.; Deng, W.; Liu, Y.; Qin, C. MicroRNA-153 negatively regulates the expression of amyloid precursor protein and amyloid precursor-like protein 2. Brain Res. 2012, 1455, 103–113. [Google Scholar] [CrossRef]
- Wang, L.; Liu, J.; Wang, Q.; Jiang, H.; Zeng, L.; Li, Z.; Liu, R. MicroRNA-200a-3p mediates neuroprotection in Alzheimer-Related deficits and attenuates amyloid-beta overproduction and tau hyperphosphorylation via coregulating BACE1 and PRKACB. Front. Pharmacol. 2019, 10, 806. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Chen, W.; Yi, Y.; Tong, Q. miR-219-5p inhibits tau phosphorylation by targeting TTBK1 and GSK-3β in Alzheimer’s disease. J. Cell. Biochem. 2018, 120, 9936–9946. [Google Scholar] [CrossRef]
- El Fatimy, R.; Li, S.; Chen, Z.; Mushannen, T.; Gongala, S.; Wei, Z.; Balu, D.T.; Rabinovsky, R.; Cantlon, A.; Elkhal, A.; et al. MicroRNA-132 provides neuroprotection for tauopathies via multiple signaling pathways. Acta Neuropathol. 2018, 136, 537–555. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Wang, G.; Huang, Y.; Wang, L.-L.; Zhang, Y.-F.; Xu, J.; Zhou, Y.; Lourenco, G.F.; Zhang, B.; Wang, Y.; Ren, R.-J.; et al. MicroRNA-146a suppresses ROCK1 allowing hyperphosphorylation of tau in Alzheimer’s disease. Sci. Rep. 2016, 6, 26697. [Google Scholar] [CrossRef]
- Thompson, D.M.; Lu, C.; Green, P.J.; Parker, R. TRNA cleavage is a conserved response to oxidative stress in eukaryotes. RNA 2008, 14, 2095–2103. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Fang, Y.; Cheng, X.; Lian, Y.-J.; Xu, H.-L. Silencing of long noncoding RNA SOX21-AS1 relieves neuronal oxidative stress injury in mice with Alzheimer’s disease by upregulating FZD3/5 via the wnt signaling pathway. Mol. Neurobiol. 2018, 56, 3522–3537. [Google Scholar] [CrossRef]
- Guo, C.-C.; Jiao, C.-H.; Gao, Z.-M. Silencing of LncRNA BDNF-AS attenuates Aβ25-35-induced neurotoxicity in PC12 cells by suppressing cell apoptosis and oxidative stress. Neurol. Res. 2018, 40, 795–804. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Zhou, T.; Wang, T.; Wang, B. Suppression of lncRNA-ATB prevents amyloid-β-induced neurotoxicity in PC12 cells via regulating miR-200/ZNF217 axis. Biomed. Pharmacother. 2018, 108, 707–715. [Google Scholar] [CrossRef]
- Wang, X.; Wang, C.; Geng, C.; Zhao, K. LncRNA XIST knockdown attenuates Aβ25-35-induced toxicity, oxidative stress, and apoptosis in primary cultured rat hippocampal neurons by targeting miR-132. Int. J. Clin. Exp. Pathol. 2018, 11, 3915. [Google Scholar]
- Gao, Y.; Zhang, N.; Lv, C.; Li, N.; Li, X.; Li, W. lncRNA SNHG1 knockdown alleviates amyloid-β-induced neuronal injury by regulating ZNF217 via sponging miR-361-3p in Alzheimer’s disease. J. Alzheimer’s Dis. 2020. [Google Scholar] [CrossRef]
- Li, X.; Wang, S.-W.; Li, X.-L.; Yu, F.-Y.; Cong, H.-M. Knockdown of long non-coding RNA TUG1 depresses apoptosis of hippocampal neurons in Alzheimer’s disease by elevating microRNA-15a and repressing ROCK1 expression. Inflamm. Res. 2020, 69, 897–910. [Google Scholar] [CrossRef]
- Akhter, R. Circular RNA and Alzheimer’s disease. Adv. Exp. Med. Biol. 2018, 1087, 239–243. [Google Scholar]
- Li, Y.; Fan, H.; Sun, J.; Ni, M.; Zhang, L.; Chen, C.; Hong, X.; Fang, F.; Zhang, W.; Ma, P. Circular RNA expression profile of Alzheimer’s disease and its clinical significance as biomarkers for the disease risk and progression. Int. J. Biochem. Cell Biol. 2020, 123, 105747. [Google Scholar] [CrossRef]
- Dube, U.; Del-Aguila, J.L.; Li, Z.; Budde, J.P.; Jiang, S.; Hsu, S.; Ibanez, L.; Fernandez, M.V.; Farias, F.; Norton, J.; et al. An atlas of cortical circular RNA expression in Alzheimer disease brains demonstrates clinical and pathological associations. Nat. Neurosci. 2019, 22, 1903–1912. [Google Scholar] [CrossRef]
- Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388. [Google Scholar] [CrossRef] [PubMed]
- Lukiw, W.J. Circular RNA (circRNA) in Alzheimer’s disease (AD). Front. Genet. 2013, 4, 307. [Google Scholar] [CrossRef] [Green Version]
- Shi, Z.; Chen, T.; Yao, Q.; Zheng, L.; Zhang, Z.; Wang, J.; Hu, Z.; Cui, H.; Han, Y.; Han, X.; et al. The circular RNA ci RS-7 promotes APP and BACE 1 degradation in an NF-κB-dependent manner. FEBS J. 2017, 284, 1096–1109. [Google Scholar] [CrossRef]
- Lu, Y.; Tan, L.; Wang, X. Circular HDAC9/microRNA-138/Sirtuin-1 pathway mediates synaptic and amyloid precursor protein processing deficits in Alzheimer’s disease. Neurosci. Bull. 2019, 35, 877–888. [Google Scholar] [CrossRef] [PubMed]
- Jellinger, K.A. Neuropathobiology of non-motor symptoms in Parkinson disease. J. Neural Transm. 2015, 122, 1429–1440. [Google Scholar] [CrossRef]
- Przedborski, S. The two-century journey of Parkinson disease research. Nature Rev. Neurosci. 2017, 18, 251. [Google Scholar] [CrossRef]
- Tysnes, O.-B.; Storstein, A. epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef]
- Inzelberg, R.; Schecthman, E.; Paleacu, D.; Zach, L.; Bonwitt, R.; Carasso, R.L.; Nisipeanu, P. Onset and progression of disease in familial and sporadic Parkinson’s disease. Am. J. Med Genet. 2003, 255–258. [Google Scholar] [CrossRef]
- Eblesa, J.; Etrigo-Damas, I.; Equiroga-Varela, A.; Jackson-Lewis, V.R. Oxidative stress and Parkinson’s disease. Front. Neuroanat. 2015, 9, 91. [Google Scholar] [CrossRef] [Green Version]
- Wei, Z.; Li, X.; Li, X.; Liu, Q.; Cheng, Y. Oxidative stress in Parkinson’s disease: A systematic review and meta-analysis. Front. Mol. Neurosci. 2018, 11, 236. [Google Scholar] [CrossRef]
- Caudle, W.M.; Richardson, J.R.; Wang, M.Z.; Taylor, T.N.; Guillot, T.S.; McCormack, A.L.; Colebrooke, R.E.; Di Monte, D.A.; Emson, P.C.; Miller, G.W. Reduced vesicular storage of dopamine causes progressive nigrostriatal neurodegeneration. J. Neurosci. 2007, 27, 8138–8148. [Google Scholar] [CrossRef]
- Hwang, D.-Y.; Hong, S.; Jeong, J.-W.; Choi, S.; Kim, H.; Kim, J.; Kim, K.-S. Vesicular monoamine transporter 2 and dopamine transporter are molecular targets of Pitx3 in the ventral midbrain dopamine neurons. J. Neurochem. 2009, 111, 1202–1212. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Inoue, K.; Ishii, J.; Vanti, W.B.; Voronov, S.V.; Murchison, E.; Hannon, G.; Abeliovich, A. A microRNA feedback circuit in midbrain dopamine neurons. Science 2007, 317, 1220–1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.-L.; Chou, A.-H.; Wu, A.-S.; Chen, S.-Y.; Weng, Y.-H.; Kao, Y.-C.; Yeh, T.-H.; Chu, P.-J.; Lu, C.-S. PARK6 PINK1 mutants are defective in maintaining mitochondrial membrane potential and inhibiting ROS formation of substantia nigra dopaminergic neurons. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2011, 1812, 674–684. [Google Scholar] [CrossRef] [Green Version]
- Wood-Kaczmar, A.; Gandhi, S.; Yao, Z.; Abramov, A.S.Y.; Miljan, E.A.; Keen, G.; Stanyer, L.; Hargreaves, I.; Klupsch, K.; Deas, E.; et al. PINK1 Is necessary for long term survival and mitochondrial function in human dopaminergic neurons. PLoS ONE 2008, 3, e2455. [Google Scholar] [CrossRef]
- Kim, J.; Fiesel, F.C.; Belmonte, K.C.; Hudec, R.; Wang, W.-X.; Kim, C.; Nelson, P.T.; Springer, W.; Kim, J. MiR-27a and miR-27b regulate autophagic clearance of damaged mitochondria by targeting PTEN-induced putative kinase 1 (PINK1). Mol. Neurodegener. 2016, 11, 1–16. [Google Scholar] [CrossRef] [Green Version]
- Prajapati, P.; Sripada, L.; Singh, R.; Bhatelia, K.; Singh, R.; Singh, R. TNF-α regulates miRNA targeting mitochondrial complex-I and induces cell death in dopaminergic cells. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2015, 1852, 451–461. [Google Scholar] [CrossRef] [Green Version]
- Hayashi, T.; Ishimori, C.; Takahashi-Niki, K.; Taira, T.; Kim, Y.-C.; Maita, H.; Maita, C.; Ariga, H.; Iguchi-Ariga, S.M. DJ-1 binds to mitochondrial complex I and maintains its activity. Biochem. Biophys. Res. Commun. 2009, 390, 667–672. [Google Scholar] [CrossRef] [Green Version]
- Ariga, H.; Takahashi-Niki, K.; Kato, I.; Maita, H.; Niki, T.; Iguchi-Ariga, S.M.M. Neuroprotective function of DJ-1 in Parkinson’s disease. Oxidative Med. Cell. Longev. 2013, 2013, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, J.; Sullards, M.C.; Olzmann, J.A.; Rees, H.D.; Weintraub, S.T.; Bostwick, D.E.; Gearing, M.; Levey, A.I.; Chin, L.-S.; Li, L. Oxidative damage of DJ-1 is linked to sporadic parkinson and Alzheimer diseases. J. Biol. Chem. 2006, 281, 10816–10824. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Gao, C.; Sun, Q.; Pan, H.; Huang, P.; Ding, J.; Chen, S. MicroRNA-4639 is a regulator of DJ-1 expression and a potential early diagnostic marker for Parkinson’s disease. Front. Aging Neurosci. 2017, 9, 232. [Google Scholar] [CrossRef]
- Oh, S.E.; Park, H.-J.; He, L.; Skibiel, C.; Junn, E.; Mouradian, M.M. The Parkinson’s disease gene product DJ-1 modulates miR-221 to promote neuronal survival against oxidative stress. Redox Biol. 2018, 19, 62–73. [Google Scholar] [CrossRef]
- Xiong, R.; Wang, Z.; Zhao, Z.; Li, H.; Chen, W.; Zhang, B.; Wang, L.; Wu, L.; Li, W.; Ding, J.; et al. MicroRNA-494 reduces DJ-1 expression and exacerbates neurodegeneration. Neurobiol. Aging 2014, 35, 705–714. [Google Scholar] [CrossRef] [PubMed]
- Miñones-Moyano, E.; Porta, S.; Escaramís, G.; Rabionet, R.; Iraola, S.; Kagerbauer, B.; Espinosa-Parrilla, Y.; Ferrer, I.; Estivill, X.; Martí, E. MicroRNA profiling of Parkinson’s disease brains identifies early downregulation of miR-34b/c which modulate mitochondrial function. Hum. Mol. Genet. 2011, 20, 3067–3078. [Google Scholar] [CrossRef]
- Pantano, L.; Friedländer, M.R.; Escaramís, G.; Lizano, E.; Pallarès-Albanell, J.; Ferrer, I.; Estivill, X.; Martí, E. Specific small-RNA signatures in the amygdala at premotor and motor stages of Parkinson’s disease revealed by deep sequencing analysis. Bioinformatice 2015, 32, 673–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Junn, E.; Lee, K.W.; Byeong, S.J.; Chan, T.W.; Im, J.Y.; Mouradian, M.M. Repression of α-synuclein expression and toxicity by microRNA-7. Proc. Natl. Acad. Sci. USA 2009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McMillan, K.J.; Murray, T.K.; Bengoa-Vergniory, N.; Cordero-Llana, O.; Cooper, J.; Buckley, A.; Wade-Martins, R.; Uney, J.B.; O’Neill, M.J.; Wong, L.F.; et al. Loss of MicroRNA-7 regulation leads to α-synuclein accumulation and dopaminergic neuronal loss in vivo. Mol. Ther. 2017, 25, 2404–2414. [Google Scholar] [CrossRef] [Green Version]
- Kabaria, S.; Choi, D.C.; Chaudhuri, A.D.; Jain, M.R.; Li, H.; Junn, E. MicroRNA-7 activates Nrf2 pathway by targeting Keap1 expression. Free. Radic. Biol. Med. 2015, 89, 548–556. [Google Scholar] [CrossRef] [Green Version]
- Narasimhan, M.; Patel, D.; Vedpathak, D.; Rathinam, M.; Henderson, G.; Mahimainathan, L. Identification of novel microRNAs in post-transcriptional control of Nrf2 expression and redox homeostasis in neuronal, SH-SY5Y cells. PLoS ONE 2012, 7, e51111. [Google Scholar] [CrossRef] [Green Version]
- Van Es, M.A.; Schelhaas, H.J.; Van Vught, P.W.J.; Ticozzi, N.; Andersen, P.M.; Groen, E.J.N.; Schulte, C.; Blauw, H.M.; Koppers, M.; Diekstra, F.P.; et al. Angiogenin variants in Parkinson disease and amyotrophic lateral sclerosis. Ann. Neurol. 2011, 70, 964–973. [Google Scholar] [CrossRef]
- Steidinger, T.U.; Standaert, D.G.; Yacoubian, T.A. A neuroprotective role for angiogenin in models of Parkinson’s disease. J. Neurochem. 2010, 116, 334–341. [Google Scholar] [CrossRef] [Green Version]
- Steidinger, T.U.; Slone, S.R.; Ding, H.; Standaert, D.G.; Yacoubian, T.A. Angiogenin in Parkinson disease models: Role of akt phosphorylation and evaluation of AAV-mediated angiogenin expression in MPTP treated mice. PLoS ONE 2013, 8, e56092. [Google Scholar] [CrossRef]
- Blanco, S.; Dietmann, S.; Flores, J.V.; Hussain, S.; Kutter, C.; Humphreys, P.; Lukk, M.; Lombard, P.; Treps, L.; Popis, M.; et al. Aberrant methylation of t RNA s links cellular stress to neuro-developmental disorders. EMBO J. 2014, 33, 2020–2039. [Google Scholar] [CrossRef]
- Pallarès-Albanell, J.; Zomeño-Abellán, M.T.; Escaramís, G.; Pantano, L.; Soriano, A.; Segura, M.F.; Martí, E. A high-throughput screening identifies microRNA inhibitors that influence neuronal maintenance and/or response to oxidative stress. Mol. Ther.-Nucleic Acids 2019, 17, 374–387. [Google Scholar] [CrossRef] [Green Version]
- Hanada, T.; Weitzer, S.; Mair, B.; Bernreuther, C.; Wainger, B.J.; Ichida, J.K.; Hanada, R.; Orthofer, M.; Cronin, S.J.; Komnenovic, V.; et al. CLP1 links tRNA metabolism to progressive motor-neuron loss. Nature 2013, 495, 474–480. [Google Scholar] [CrossRef]
- Wang, L.; Yang, H.; Wang, Q.; Zhang, Q.; Wang, Z.; Zhang, Q.; Wu, S.; Li, H. Paraquat and MPTP induce alteration in the expression profile of long noncoding RNAs in the substantia nigra of mice: Role of the transcription factor Nrf2. Toxicol. Lett. 2018, 291, 11–28. [Google Scholar] [CrossRef]
- Cai, L.; Tu, L.; Li, T.; Yang, X.; Ren, Y.; Gu, R.; Zhang, Q.; Yao, H.; Qu, X.; Wang, Q.; et al. Downregulation of lncRNA UCA1 ameliorates the damage of dopaminergic neurons, reduces oxidative stress and inflammation in Parkinson’s disease through the inhibition of the PI3K/Akt signaling pathway. Int. Immunopharmacol. 2019, 75, 105734. [Google Scholar] [CrossRef]
- Liu, W.; Zhang, Q.; Zhang, J.; Pan, W.; Zhao, J.; Xu, Y. Long non-coding RNA MALAT1 contributes to cell apoptosis by sponging miR-124 in Parkinson disease. Cell Biosci. 2017, 7, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Zhai, K.; Liu, B.; Gao, L. Long Non-coding RNA TUG1 Promotes Parkinson’s disease via modulating MiR-152-3p/PTEN pathway. Hum. Gene Ther. 2020. [Google Scholar] [CrossRef]
- Kraus, T.F.J.; Haider, M.; Spanner, J.; Steinmaurer, M.; Dietinger, V.; Kretzschmar, H.A. Altered long noncoding RNA expression precedes the course of Parkinson’s disease—A preliminary report. Mol. Neurobiol. 2016, 54, 2869–2877. [Google Scholar] [CrossRef]
- Ding, X.-M.; Zhao, L.-J.; Qiao, H.-Y.; Wu, S.-L.; Wang, X.-H. Long non-coding RNA-p21 regulates MPP+-induced neuronal injury by targeting miR-625 and derepressing TRPM2 in SH-SY5Y cells. Chem. Interact. 2019, 307, 73–81. [Google Scholar] [CrossRef] [PubMed]
- Simchovitz, A.; Hanan, M.; Niederhoffer, N.; Madrer, N.; Yayon, N.; Bennett, E.R.; Greenberg, D.S.; Kadener, S.; Soreq, H. NEAT1 is overexpressed in Parkinson’s disease substantia nigra and confers drug-inducible neuroprotection from oxidative stress. FASEB J. 2019, 33, 11223–11234. [Google Scholar] [CrossRef] [Green Version]
- Simchovitz, A.; Hanan, M.; Yayon, N.; Lee, S.; Bennett, E.R.; Greenberg, D.S.; Kadener, S.; Soreq, H. A lncRNA survey finds increases in neuroprotective LINC-PINT in Parkinson’s disease substantia nigra. Aging Cell 2020, 19, e13115. [Google Scholar] [CrossRef] [Green Version]
- Hanan, M.; Simchovitz, A.; Yayon, N.; Vaknine, S.; Cohen-Fultheim, R.; Karmon, M.; Madrer, N.; Rohrlich, T.M.; Maman, M.; Bennett, E.R.; et al. A Parkinson’s disease Circ RNA s resource reveals a link between circ SLC 8A1 and oxidative stress. EMBO Mol. Med. 2020, 12. [Google Scholar] [CrossRef]
- Jia, E.; Zhou, Y.; Liu, Z.; Wang, L.; Ouyang, T.; Pan, M.; Liu, Z.; Ge, Q. transcriptomic profiling of circular RNA in different brain regions of Parkinson’s disease in a mouse model. Int. J. Mol. Sci. 2020, 21, 3006. [Google Scholar] [CrossRef] [Green Version]
- Feng, Z.; Zhang, L.; Wang, S.; Hong, Q. Circular RNA circDLGAP4 exerts neuroprotective effects via modulating miR-134-5p/CREB pathway in Parkinson’s disease. Biochem. Biophys. Res. Commun. 2020, 522, 388–394. [Google Scholar] [CrossRef]
- Sang, Q.; Liu, X.; Wang, L.; Qi, L.; Sun, W.; Wang, W.; Sun, Y.; Zhang, H. CircSNCA downregulation by pramipexole treatment mediates cell apoptosis and autophagy in Parkinson’s disease by targeting miR-7. Aging 2018, 10, 1281–1293. [Google Scholar] [CrossRef] [PubMed]
- McColgan, P.; Tabrizi, S.J. Huntington’s disease: A clinical review. Eur. J. Neurol. 2018, 25, 24–34. [Google Scholar] [CrossRef] [PubMed]
- Ross, C.A.; Tabrizi, S.J. Huntington’s disease: From molecular pathogenesis to clinical treatment. Lancet Neurol. 2011, 10, 83–98. [Google Scholar] [CrossRef]
- Bañez-Coronel, M.; Ayhan, F.; Tarabochia, A.D.; Zu, T.; Perez, B.A.; Tusi, S.K.; Pletnikova, O.; Borchelt, D.R.; Ross, C.A.; Margolis, R.L.; et al. RAN translation in Huntington disease. Neuron 2015, 88, 667–677. [Google Scholar] [CrossRef] [Green Version]
- Báñez-Coronel, M.; Porta, S.; Kagerbauer, B.; Mateu-Huertas, E.; Pantano, L.; Ferrer, I.; Guzmán, M.; Estivill, X.; Martí, E. A Pathogenic mechanism in Huntington’s disease involves small CAG-Repeated RNAs with neurotoxic activity. PLoS Genet. 2012, 8, e1002481. [Google Scholar] [CrossRef] [Green Version]
- Martí, E. RNA toxicity induced by expanded CAG repeats in Huntington’s disease. Brain Pathol. 2016, 26, 779–786. [Google Scholar] [CrossRef]
- Martí, E.; Pantano, L.; Bañez-Coronel, M.; Llorens, F.; Miñones-Moyano, E.; Porta, S.; Sumoy, L.; Ferrer, I.; Estivill, X. A myriad of miRNA variants in control and Huntington’s disease brain regions detected by massively parallel sequencing. Nucleic Acids Res. 2010, 38, 7219–7235. [Google Scholar] [CrossRef]
- Savas, J.N.; Makusky, A.; Ottosen, S.; Baillat, D.; Then, F.; Krainc, D.; Shiekhattar, R.; Markey, S.P.; Tanese, N. Huntington’s disease protein contributes to RNA-mediated gene silencing through association with Argonaute and P bodies. Proc. Nat.Acad. Sci. USA 2008. [Google Scholar] [CrossRef] [Green Version]
- Pircs, K.; Petri, R.; Madsen, S.; Brattås, P.L.; Vuono, R.; Ottosson, D.R.; St-Amour, I.; Hersbach, B.A.; Matusiak-Brückner, M.; Lundh, S.H.; et al. Huntingtin aggregation impairs autophagy, leading to argonaute-2 accumulation and global microRNA dysregulation. Cell Rep. 2018, 24, 1397–1406. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.-T.; Chu, K.; Im, W.-S.; Yoon, H.-J.; Im, J.-Y.; Park, J.-E.; Park, K.-H.; Jung, K.-H.; Lee, S.K.; Kim, M.; et al. Altered microRNA regulation in Huntington’s disease models. Exp. Neurol. 2011, 227, 172–179. [Google Scholar] [CrossRef]
- Packer, A.N.; Xing, Y.; Harper, S.Q.; Jones, L.; Davidson, B.L. The bifunctional microRNA miR-9/miR-9* regulates REST and coREST and is downregulated in Huntington’s disease. J. Neurosci. 2008, 28, 14341–14346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Langfelder, P.; Gao, F.; Wang, N.; Howland, D.; Kwak, S.; Vogt, T.F.; Aaronson, J.S.; Rosinski, J.; Coppola, G.; Horvath, S.; et al. MicroRNA signatures of endogenous Huntingtin CAG repeat expansion in mice. PLoS ONE 2018, 13, e0190550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wong, A.H.-K.; Veremeyko, T.; Patel, N.; Lemere, C.A.; Walsh, M.M.; Esau, C.; Vanderburg, C.R.; Krichevsky, A.M. De-repression of FOXO3a death axis by microRNA-132 and -212 causes neuronal apoptosis in Alzheimer’s disease. Hum. Mol. Genet. 2013, 22, 3077–3092. [Google Scholar] [CrossRef] [Green Version]
- Zhu, H.; Wang, X.; Chen, S. Downregulation of MiR-218-5p protects against oxygen-glucose deprivation/reperfusion-induced injuries of PC12 cells via upregulating n-myc downstream regulated gene 4 (NDRG4). Med Sci. Monit. Int. Med. J. Exp. Clin. Res. 2020, 26, e920101-1. [Google Scholar] [CrossRef]
- Johnson, R. Long non-coding RNAs in Huntington’s disease neurodegeneration. Neurobiol. Dis. 2012, 46, 245–254. [Google Scholar] [CrossRef]
- Chanda, K.; Das, S.; Chakraborty, J.; Bucha, S.; Maitra, A.; Chatterjee, R.; Mukhopadhyay, D.; Bhattacharyya, N.P. Altered levels of long NcRNAs Meg3 and Neat1 in cell and animal models of Huntington’s disease. RNA Biol. 2018, 15, 1348–1363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, R.H.; Al-Chalabi, A. Amyotrophic lateral sclerosis. N. Engl. J. Med. 2001, 344, 1688–1700. [Google Scholar]
- Kang, S.H.; Li, Y.; Fukaya, M.; Lorenzini, I.; Cleveland, D.W.; Ostrow, L.W.; Rothstein, J.D.; Bergles, D.E. Degeneration and impaired regeneration of gray matter oligodendrocytes in amyotrophic lateral sclerosis. Nat. Neurosci. 2013, 16, 571–579. [Google Scholar] [CrossRef] [Green Version]
- Philips, T.; Rothstein, J.D. Glial cells in amyotrophic lateral sclerosis. Exp. Neurol. 2014, 262, 111–120. [Google Scholar] [CrossRef] [Green Version]
- Rosen, D.R.; Siddique, T.; Patterson, D.; Figlewicz, D.A.; Sapp, P.C.; Hentati, A.; Donaldson, D.H.; Goto, J.; O’Regan, J.P.; Deng, H.-X.; et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993, 362, 59–62. [Google Scholar] [CrossRef]
- Douglas, A.G.L. Non-coding RNA in C9orf72-related amyotrophic lateral sclerosis and frontotemporal dementia: A perfect storm of dysfunction. Non-Coding RNA Res. 2018, 3, 178–187. [Google Scholar] [CrossRef]
- Neumann, M.; Sampathu, D.M.; Kwong, L.K.; Truax, A.C.; Micsenyi, M.C.; Chou, T.T.; Bruce, J.; Schuck, T.; Grossman, M.; Clark, C.M.; et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006, 314, 130–133. [Google Scholar] [CrossRef] [Green Version]
- Deng, H.-X.; Chen, W.; Hong, S.-T.; Boycott, K.M.; Gorrie, G.H.; Siddique, N.; Yang, Y.; Fecto, F.; Shi, Y.; Zhai, H.; et al. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature 2011, 477, 211–215. [Google Scholar] [CrossRef] [Green Version]
- Bosco, D.A.; Morfini, G.; Karabacak, N.M.; Song, Y.; Gros-Louis, F.; Pasinelli, P.; Goolsby, H.; Fontaine, B.A.; Lemay, N.; McKenna-Yasek, D.; et al. Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS. Nat. Neurosci. 2010, 13, 1396–1403. [Google Scholar] [CrossRef] [Green Version]
- Robberecht, W. Oxidative stress in amyotrophic lateral sclerosis. J. Neurol. 2000, 247. [Google Scholar] [CrossRef] [PubMed]
- Joilin, G.; Leigh, P.N.; Newbury, S.F.; Hafezparast, M. An overview of microRNAs as biomarkers of ALS. Front. Neurol. 2019, 10, 186. [Google Scholar] [CrossRef] [Green Version]
- Kovanda, A.; Leonardis, L.; Zidar, J.; Koritnik, B.; Dolenc-Groselj, L.; Kovacic, S.R.; Curk, T.; Rogelj, B. Differential expression of microRNAs and other small RNAs in muscle tissue of patients with ALS and healthy age-matched controls. Sci. Rep. 2018, 8, 5609. [Google Scholar] [CrossRef]
- Ricci, C.; Marzocchi, C.; Battistini, S. MicroRNAs as biomarkers in amyotrophic lateral sclerosis. Cells 2018, 7, 219. [Google Scholar] [CrossRef] [Green Version]
- Rizzuti, M.; Filosa, G.; Melzi, V.; Calandriello, L.; Dioni, L.; Bollati, V.; Bresolin, N.; Comi, G.P.; Barabino, S.; Nizzardo, M.; et al. MicroRNA expression analysis identifies a subset of downregulated miRNAs in ALS motor neuron progenitors. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Waller, R.; Wyles, M.; Heath, P.R.; Kazoka, M.; Wollff, H.; Shaw, P.J.; Kirby, J. Small RNA sequencing of sporadic amyotrophic lateral sclerosis cerebrospinal fluid reveals differentially expressed miRNAs Related to neural and glial activity. Front. Neurosci. 2018, 11, 731. [Google Scholar] [CrossRef]
- Ravnik-Glavač, M.; Glavač, D. Circulating RNAs as potential biomarkers in amyotrophic lateral sclerosis. Int. J. Mol. Sci. 2020, 21, 1714. [Google Scholar] [CrossRef] [Green Version]
- Koval, E.D.; Shaner, C.; Zhang, P.; Du Maine, X.; Fischer, K.; Tay, J.; Chau, B.N.; Wu, G.F.; Miller, T.M. Method for widespread microRNA-155 inhibition prolongs survival in ALS-model mice. Hum. Mol. Genet. 2013, 22, 4127–4135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Wei, Q.; Gu, X.; Chen, Y.; Chen, X.; Cao, B.; Ou, R.; Shang, H.-F. Decreased glycogenolysis by miR-338-3p promotes regional glycogen accumulation within the spinal cord of amyotrophic lateral sclerosis mice. Front. Mol. Neurosci. 2019, 12, 114. [Google Scholar] [CrossRef] [Green Version]
- Kim, D.; Nguyen, M.D.; Dobbin, M.M.; Fischer, A.; Sananbenesi, F.; Rodgers, J.T.; Delalle, I.; Baur, J.A.; Sui, G.; Armour, S.M.; et al. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. EMBO J. 2007, 26, 3169–3179. [Google Scholar] [CrossRef]
- Shioya, M.; Obayashi, S.; Tabunoki, H.; Arima, K.; Saito, Y.; Ishida, T.; Satoh, J. Aberrant microRNA expression in the brains of neurodegenerative diseases: MiR-29a decreased in Alzheimer disease brains targets neurone navigator 3. Neuropathol. Appl. Neurobiol. 2010, 36, 320–330. [Google Scholar] [CrossRef]
- Aschrafi, A.; Kar, A.N.; Natera-Naranjo, O.; MacGibeny, M.A.; Gioio, A.E.; Kaplan, B.B. MicroRNA-338 regulates the axonal expression of multiple nuclear-encoded mitochondrial mRNAs encoding subunits of the oxidative phosphorylation machinery. Cell. Mol. Life Sci. 2012, 69, 4017–4027. [Google Scholar] [CrossRef]
- Emde, A.; Eitan, C.; Liou, L.; Libby, R.T.; Rivkin, N.; Magen, I.; Reichenstein, I.; Oppenheim, H.; Eilam, R.; Silvestroni, A.; et al. Dysregulated mi RNA biogenesis downstream of cellular stress and ALS-causing mutations: A new mechanism for ALS. EMBO J. 2015, 34, 2633–2651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paladino, S.; Conte, A.; Caggiano, R.; Pierantoni, G.M.; Faraonio, R. Nrf2 pathway in age-related neurological disorders: Insights into microRNAs. Cell. Physiol. Biochem. 2018, 47, 1951–1976. [Google Scholar] [CrossRef]
- Wang, N.; Zhang, L.; Lu, Y.; Zhang, M.; Zhang, Z.; Wang, K.; Lv, J. Down-regulation of microRNA-142-5p attenuates oxygen-glucose deprivation and reoxygenation-induced neuron injury through up-regulating Nrf2/ARE signaling pathway. Biomed. Pharmacother. 2017, 89, 1187–1195. [Google Scholar] [CrossRef] [PubMed]
- Greenway, M.J.; Andersen, P.M.; Russ, C.; Ennis, S.; Cashman, S.; Donaghy, C.; Patterson, V.; Swingler, R.; Kieran, D.; Prehn, J.; et al. ANG mutations segregate with familial and ’sporadic’ amyotrophic lateral sclerosis. Nat. Genet. 2006, 38, 411–413. [Google Scholar] [CrossRef]
- Wolozin, B. Physiological protein aggregation run amuck: Stress granules and the genesis of neurodegenerative disease. Discov. Med. 2014, 17, 47–52. [Google Scholar]
- Wolozin, B. Regulated protein aggregation: Stress granules and neurodegeneration. Mol. Neurodegener. 2012, 7, 56. [Google Scholar] [CrossRef] [Green Version]
- Joilin, G.; Gray, E.; Thompson, A.G.; Bobeva, Y.; Talbot, K.; Weishaupt, J.; Ludolph, A.; Malaspina, A.; Leigh, P.N.; Newbury, S.F.; et al. Identification of a potential non-coding RNA biomarker signature for amyotrophic lateral sclerosis. Brain Commun. 2020, 2, fcaa053. [Google Scholar] [CrossRef]
- Birger, A.; Ben-Dor, I.; Ottolenghi, M.; Turetsky, T.; Gil, Y.; Sweetat, S.; Perez, L.; Belzer, V.; Casden, N.; Steiner, D.; et al. Human iPSC-derived astrocytes from ALS patients with mutated C9ORF72 show increased oxidative stress and neurotoxicity. EBioMedicine 2019, 50, 274–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nishimoto, Y.; Nakagawa, S.; Hirose, T.; Okano, H.J.; Takao, M.; Shibata, S.; Suyama, S.; Kuwako, K.-I.; Imai, T.; Murayama, S.; et al. The long non-coding RNA nuclear-enriched abundant transcript 1_2 induces paraspeckle formation in the motor neuron during the early phase of amyotrophic lateral sclerosis. Mol. Brain 2013, 6, 31. [Google Scholar] [CrossRef] [Green Version]
- Wang, C.; Duan, Y.; Duan, G.; Wang, Q.; Zhang, K.; Deng, X.; Qian, B.; Gu, J.; Ma, Z.; Zhang, S.; et al. Stress induces dynamic, cytotoxicity-antagonizing TDP-43 nuclear bodies via paraspeckle lncRNA NEAT1-mediated liquid-liquid phase separation. Mol. Cell 2020, 79, 443–458.e7. [Google Scholar] [CrossRef]
- Gupta, P.; Bhattacharjee, S.; Sharma, A.R.; Sharma, G.; Lee, S.-S.; Chakraborty, C. miRNAs in Alzheimer disease—A therapeutic perspective. Curr. Alzheimer Res. 2017, 14, 1198–1206. [Google Scholar] [CrossRef]
- Adams, B.D.; Parsons, C.; Walker, L.; Zhang, W.C.; Slack, F.J. Targeting noncoding RNAs in disease. J. Clin. Investig. 2017, 127, 761–771. [Google Scholar] [CrossRef]
- Jin, H.Y.; Gonzalez-Martin, A.; Miletic, A.V.; Lai, M.; Knight, S.; Sabouri-Ghomi, M.; Head, S.R.; Macauley, M.S.; Rickert, R.C.; Xiao, C. Transfection of microRNA mimics should be used with caution. Front. Genet. 2015, 6, 340. [Google Scholar] [CrossRef] [Green Version]
- He, M.; Liu, Y.; Wang, X.; Zhang, M.Q.; Hannon, G.J.; Huang, Z.J. Cell-type-based analysis of microRNA profiles in the mouse brain. Neuron 2012, 73, 35–48. [Google Scholar] [CrossRef] [Green Version]
- Espay, A.J.; Brundin, P.; Lang, A.E. Precision medicine for disease modification in Parkinson disease. Nat. Rev. Neurol. 2017, 13, 119–126. [Google Scholar] [CrossRef] [PubMed]
- Dawson, T.M.; Golde, T.E.; Lagier-Tourenne, C. Animal models of neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1370–1379. [Google Scholar] [CrossRef]
- Derrien, T.; Johnson, R.; Bussotti, G.; Tanzer, A.; Djebali, S.; Tilgner, H.; Guernec, G.; Martin, D.; Merkel, A.; Knowles, D.G.; et al. The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 2012, 22, 1775–1789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, X.; Yeo, G.; Muotri, A.R.; Kuwabara, T.; Gage, F.H. Noncoding rnas in the mammalian central nervous system. Annu. Rev. Neurosci. 2006, 29, 77–103. [Google Scholar] [CrossRef] [Green Version]
- Friedländer, M.R.; Lizano, E.; Houben, A.J.S.; Bezdan, D.; Báñez-Coronel, M.; Kudla, G.; Mateu-Huertas, E.; Kagerbauer, B.; González, J.; Chen, K.C.; et al. Evidence for the biogenesis of more than 1000 novel human microRNAs. Genome Biol. 2014, 15, R57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engle, S.J.; Blaha, L.; Kleiman, R.J. Best practices for translational disease modeling using human iPSC-derived neurons. Neuron 2018, 100, 783–797. [Google Scholar] [CrossRef] [Green Version]
- Tolosa, E.; Botta-Orfila, T.; Morató, X.; Calatayud, C.; Ferrer-Lorente, R.; Martí, M.-J.; Fernández, M.; Gaig, C.; Raya, Á.; Consiglio, A.; et al. MicroRNA alterations in iPSC-derived dopaminergic neurons from Parkinson disease patients. Neurobiol. Aging 2018, 69, 283–291. [Google Scholar] [CrossRef]
- Grenier, K.; Kao, J.; Diamandis, P. Three-dimensional modeling of human neurodegeneration: Brain organoids coming of age. Mol. Psychiatry 2019, 25, 254–274. [Google Scholar] [CrossRef]
- Katerji, M.; Filippova, M.; Duerksen-Hughes, P. Approaches and methods to measure oxidative stress in clinical samples: Research applications in the cancer field. Oxidative Med. Cell. Longev. 2019, 2019, 1–29. [Google Scholar] [CrossRef] [Green Version]
- O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [Google Scholar] [CrossRef]
- van den Berg, M.; Krauskopf, J.; Ramaekers, J.G.; Kleinjans, J.C.S.; Prickaerts, J.; Briedé, J.J. Circulating microRNAs as potential biomarkers for psychiatric and neurodegenerative disorders. Prog. Neurobiol. 2020, 185, 101732. [Google Scholar] [CrossRef]
- Brennan, S.; Keon, M.; Liu, B.; Su, Z.; Saksena, N.K. Panoramic visualization of circulating microRNAs across Neurodegenerative diseases in humans. Mol. Neurobiol. 2019, 56, 7380–7407. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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
Gámez-Valero, A.; Guisado-Corcoll, A.; Herrero-Lorenzo, M.; Solaguren-Beascoa, M.; Martí, E. Non-Coding RNAs as Sensors of Oxidative Stress in Neurodegenerative Diseases. Antioxidants 2020, 9, 1095. https://doi.org/10.3390/antiox9111095
Gámez-Valero A, Guisado-Corcoll A, Herrero-Lorenzo M, Solaguren-Beascoa M, Martí E. Non-Coding RNAs as Sensors of Oxidative Stress in Neurodegenerative Diseases. Antioxidants. 2020; 9(11):1095. https://doi.org/10.3390/antiox9111095
Chicago/Turabian StyleGámez-Valero, Ana, Anna Guisado-Corcoll, Marina Herrero-Lorenzo, Maria Solaguren-Beascoa, and Eulàlia Martí. 2020. "Non-Coding RNAs as Sensors of Oxidative Stress in Neurodegenerative Diseases" Antioxidants 9, no. 11: 1095. https://doi.org/10.3390/antiox9111095
APA StyleGámez-Valero, A., Guisado-Corcoll, A., Herrero-Lorenzo, M., Solaguren-Beascoa, M., & Martí, E. (2020). Non-Coding RNAs as Sensors of Oxidative Stress in Neurodegenerative Diseases. Antioxidants, 9(11), 1095. https://doi.org/10.3390/antiox9111095