全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Silencing of Amyloid Precursor Protein Expression Using a New Engineered Delta Ribozyme

DOI: 10.1155/2012/947147

Full-Text   Cite this paper   Add to My Lib

Abstract:

Alzheimer's disease (AD) etiological studies suggest that an elevation in amyloid-β peptides (Aβ) level contributes to aggregations of the peptide and subsequent development of the disease. The major constituent of these amyloid peptides is the 1 to 40–42 residue peptide (Aβ40?42) derived from amyloid protein precursor (APP). Most likely, reducing Aβ levels in the brain may block both its aggregation and neurotoxicity and would be beneficial for patients with AD. Among the several possible ways to lower Aβ accumulation in the cells, we have selectively chosen to target the primary step in the Aβ cascade, namely, to reduce APP gene expression. Toward this end, we engineered specific SOFA-HDV ribozymes, a new generation of catalytic RNA tools, to decrease APP mRNA levels. Additionally, we demonstrated that APP-ribozymes are effective at decreasing APP mRNA and protein levels as well as Aβ levels in neuronal cells. Our results could lay the groundwork for a new protective treatment for AD. 1. Introduction Alzheimer’s disease (AD) is a degenerative disorder of the human central nervous system (CNS). Its clinical and neuropathological features are defined by a progressive loss of cognitive function and by the onset of a slowly progressive impairment of memory during mid- to late-adult life. The neuropathological hallmarks of AD include the accumulation and aggregation of amyloid-β peptide (Aβ), neurofibrillary tangles, astrocytic gliosis, and reductions in the numbers of both neurons and synapses in many areas of the brain, particularly in the cerebral cortex and hippocampus [1]. Strong evidence from multiple studies suggests that defects in Aβ regulation are one of the central biochemical events leading to the development of AD [2]. The neurotoxic Aβ fragment originates from the amyloid protein precursor (APP) following sequential cleavages by β (BACE) and γ-secretases (presenilin complex). Observations on the physiological processing of APP and on the effects of pathogenic mutations in the APP and/or the presenilin genes have led to the hypothesis that aberrant processing of APP into Aβ peptides is linked to AD [3]. We have previously reported strong evidence indicating that the amyloid cascade is an early and critical event in the neurodegeneration associated with AD. For example, cell lines and/or transgenic mice expressing mutant presenilin 1 (PS1), presenilin 2 (PS2), or APP exhibit an accelerated rate of neurotoxic Aβ formation [4]. Thus, the three known genetic causes of familial AD affect Aβ metabolism. Moreover, the ε4 allele of apolipoprotein E, a

References

[1]  G. Levesque, R. Sherrington, and P. St George-Hyslop, Molecular Genetic of Alzheimer Disease, Elsevier Science, Amsterdam, The Netherlands, 2002.
[2]  J. Hardy and D. J. Selkoe, “The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics,” Science, vol. 297, no. 5580, pp. 353–356, 2002.
[3]  M. Citron, C. B. Eckman, T. S. Diehl et al., “Additive effects of PS1 and APP mutations on secretion of the 42- residue amyloid β-protein,” Neurobiology of Disease, vol. 5, no. 2, pp. 107–116, 1998.
[4]  M. Citron, D. Westaway, W. Xia et al., “Mutant presenilins of Alzheimer's disease increase production of 42-residue amyloid β-protein in both transfected cells and transgenic mice,” Nature Medicine, vol. 3, no. 1, pp. 67–72, 1997.
[5]  D. M. Holtzman, A. M. Fagan, B. Mackey et al., “Apolipoprotein E facilitates neuritic and cerebrovascular plaque formation in an Alzheimer's disease model,” Annals of Neurology, vol. 47, no. 6, pp. 739–747, 2000.
[6]  R. Deane, A. Sagare, K. Hamm et al., “apoE isoform-specific disruption of amyloid β peptide clearance from mouse brain,” Journal of Clinical Investigation, vol. 118, no. 12, pp. 4002–4013, 2008.
[7]  M. Asif-Ullah, M. Lévesque, G. Robichaud, and J. P. Perreault, “Development of ribozyme-based gene-inactivations; the example of the hepatitis delta virus ribozyme,” Current Gene Therapy, vol. 7, no. 3, pp. 205–216, 2007.
[8]  D. Lévesque, S. Choufani, and J. P. Perreault, “Delta ribozyme benefits from a good stability in vitro that becomes outstanding in vivo,” RNA, vol. 8, no. 4, pp. 464–477, 2002.
[9]  L. J. Bergeron and J. P. Perreault, “Target-dependent on/off switch increases ribozyme fidelity,” Nucleic Acids Research, vol. 33, no. 4, pp. 1240–1248, 2005.
[10]  L. Bergeron, C. Reymond, and J. P. Perreault, “Functional characterization of the SOFA delta ribozyme,” RNA, vol. 11, no. 12, pp. 1858–1868, 2005.
[11]  G. A. Robichaud, J. P. Perreault, and R. J. Ouellette, “Development of an isoform-specific gene suppression system: the study of the human Pax-5B transcriptional element,” Nucleic Acids Research, vol. 36, no. 14, pp. 4609–4620, 2008.
[12]  K. Fiola, J. P. Perreault, and B. Cousineau, “Gene targeting in the gram-positive bacterium Lactococcus lactis, using various delta ribozymes,” Applied and Environmental Microbiology, vol. 72, no. 1, pp. 869–879, 2006.
[13]  M. V. Lévesque, D. Lévesque, F. P. Brière, and J. P. Perreault, “Investigating a new generation of ribozymes in order to target HCV,” PLoS ONE, vol. 5, no. 3, Article ID e9627, 2010.
[14]  C. Reymond, M. Bisaillon, and J. P. Perreault, “Monitoring of an RNA multistep folding pathway by isothermal titration calorimetry,” Biophysical Journal, vol. 96, no. 1, pp. 132–140, 2009.
[15]  L. J. Bergeron and J. P. Perreault, “Development and comparison of procedures for the selection of delta ribozyme cleavage sites within the hepatitis B virus,” Nucleic Acids Research, vol. 30, no. 21, pp. 4682–4691, 2002.
[16]  S. S. Hébert, V. Bourdages, C. Godin, M. Ferland, M. Carreau, and G. Lévesque, “Presenilin-1 interacts directly with the β-site amyloid protein precursor cleaving enzyme (BACE1),” Neurobiology of Disease, vol. 13, no. 3, pp. 238–245, 2003.
[17]  Q. Yu, D. B. Pecchia, S. L. Kingsley, J. E. Heckman, and J. M. Burke, “Cleavage of highly structured viral RNA molecules by combinatorial libraries of hairpin ribozymes. The most effective ribozymes are not predicted by substrate selection rules,” Journal of Biological Chemistry, vol. 273, no. 36, pp. 23524–23533, 1998.
[18]  J. F. Lucier, L. J. Bergeron, F. P. Brière, R. Ouellette, S. A. Elela, and J. P. Perreault, “RiboSubstrates: a web application addressing the cleavage specificities of ribozymes in designated genomes,” BMC Bioinformatics, vol. 7, article 480, 2006.
[19]  G. A. Higgins, D. A. Lewis, S. Bahmanyar et al., “Differential regulation of amyloid-β-protein mRNA expression within hippocampal neuronal subpopulations in Alzheimer disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 85, no. 4, pp. 1297–1301, 1988.
[20]  R. D. Moir and R. E. Tanzi, “LRP-mediated clearance of Aβ is inhibited by KPI-containing isoforms of APP,” Current Alzheimer Research, vol. 2, no. 2, pp. 269–273, 2005.
[21]  C. Reymond, J. D. Beaudoin, and J. P. Perreault, “Modulating RNA structure and catalysis: lessons from small cleaving ribozymes,” Cellular and Molecular Life Sciences, vol. 66, no. 24, pp. 3937–3950, 2009.
[22]  Y. Huang and R. J. Maraia, “Comparison of the RNA polymerase III transcription machinery in Schizosaccharomyces pombe, Saccharomyces cerevisiae and human,” Nucleic Acids Research, vol. 29, no. 13, pp. 2675–2690, 2001.
[23]  A. L. Jackson and P. S. Linsley, “Noise amidst the silence: off-target effects of siRNAs?” Trends in Genetics, vol. 20, no. 11, pp. 521–524, 2004.
[24]  X. Lin, X. Ruan, M. G. Anderson et al., “siRNA-mediated off-target gene silencing triggered by a 7?nt complementation,” Nucleic Acids Research, vol. 33, no. 14, pp. 4527–4535, 2005.
[25]  V. A. Alvarez, D. A. Ridenour, and B. L. Sabatini, “Retraction of synapses and dendritic spines induced by off-target effects of RNA interference,” Journal of Neuroscience, vol. 26, no. 30, pp. 7820–7825, 2006.
[26]  A. D. Judge, V. Sood, J. R. Shaw, D. Fang, K. McClintock, and I. MacLachlan, “Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA,” Nature Biotechnology, vol. 23, no. 4, pp. 457–462, 2005.
[27]  T. Laitala-Leinonen, “Update on the development of microRNA and siRNA molecules as regulators of cell physiology,” Recent Patents on DNA and Gene Sequences, vol. 4, no. 2, pp. 113–121, 2010.
[28]  I. H. Shih and M. D. Been, “Catalytic strategies of the hepatitis delta virus ribozymes,” Annual Review of Biochemistry, vol. 71, pp. 887–917, 2002.
[29]  L. Bergeron, J. Ouellet, and J. P. Perreault, “Ribozyme-based gene-inactivation systems require a fine comprehension of their substrate specificities; the case of delta ribozyme,” Current Medicinal Chemistry, vol. 10, no. 23, pp. 2589–2597, 2003.
[30]  L. J. Scherer and J. J. Rossi, “Approaches for the sequence-specific knockdown of mRNA,” Nature Biotechnology, vol. 21, no. 12, pp. 1457–1465, 2003.
[31]  K. Salehi-Ashtiani, A. Lupták, A. Litovchick, and J. W. Szostak, “A genomewide search for ribozymes reveals an HDV-like sequence in the human CPEB3 gene,” Science, vol. 313, no. 5794, pp. 1788–1792, 2006.
[32]  W. J. Netzer, C. Powell, Y. Nong et al., “Lowering beta-amyloid levels rescues learning and memory in a Down syndrome mouse model,” PloS one, vol. 5, no. 6, p. e10943, 2010.
[33]  D. M. Wilcock, N. Gharkholonarehe, W. E. Van Nostrand, J. Davis, M. P. Vitek, and C. A. Colton, “Amyloid reduction by amyloid-β vaccination also reduces mouse tau pathology and protects from neuron loss in two mouse models of Alzheimer's disease,” Journal of Neuroscience, vol. 29, no. 25, pp. 7957–7965, 2009.
[34]  P. S. Aisen, S. Gauthier, B. Vellas et al., “Alzhemed: a potential treatment for Alzheimer's disease,” Current Alzheimer Research, vol. 4, no. 4, pp. 473–478, 2007.
[35]  C. Janus, J. Pearson, J. McLaurin et al., “Aβ peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease,” Nature, vol. 408, no. 6815, pp. 979–982, 2000.
[36]  J. McLaurin, R. Cecal, M. E. Kierstead et al., “Therapeutically effective antibodies against amyloid-β peptide target amyloid-β residues 4-10 and inhibit cytotoxicity and fibrillogenesis,” Nature Medicine, vol. 8, no. 11, pp. 1263–1269, 2002.
[37]  D. Boche, N. Denham, C. Holmes, and J. A. R. Nicoll, “Neuropathology after active Aβ42 immunotherapy: implications for Alzheimer's disease pathogenesis,” Acta Neuropathologica, vol. 120, no. 3, pp. 369–384, 2010.
[38]  H. Wang, L. Song, F. Laird, P. C. Wong, and H. K. Lee, “BACE1 knock-outs display deficits in activity-dependent potentiation of synaptic transmission at mossy fiber to CA3 synapses in the hippocampus,” Journal of Neuroscience, vol. 28, no. 35, pp. 8677–8681, 2008.
[39]  A. V. Savonenko, T. Melnikova, F. M. Laird, K. A. Stewart, D. L. Price, and P. C. Wong, “Alteration of BACE1-dependent NRG1/ErbB4 signaling and schizophrenia-like phenotypes in BACE1-null mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 14, pp. 5585–5590, 2008.
[40]  F. M. Laird, H. Cai, A. V. Savonenko et al., “BACE1, a major determinant of selective vulnerability of the brain to amyloid-β amyloidogenesis, is essential for cognitive, emotional, and synaptic functions,” Journal of Neuroscience, vol. 25, no. 50, pp. 11693–11709, 2005.
[41]  M. Willem, A. N. Garratt, B. Novak et al., “Control of peripheral nerve myelination by the β-secretase BACE1,” Science, vol. 314, no. 5799, pp. 664–666, 2006.
[42]  X. Hu, C. W. Hicks, W. He et al., “Bace1 modulates myelination in the central and peripheral nervous system,” Nature Neuroscience, vol. 9, no. 12, pp. 1520–1525, 2006.
[43]  D. Y. Kim, B. W. Carey, H. Wang et al., “BACE1 regulates voltage-gated sodium channels and neuronal activity,” Nature Cell Biology, vol. 9, no. 7, pp. 755–764, 2007.
[44]  Z. Zhu, Z. Y. Sun, Y. Ye et al., “Discovery of cyclic acylguanidines as highly potent and selective β-site amyloid cleaving enzyme (BACE) inhibitors: part I—inhibitor design and validation,” Journal of Medicinal Chemistry, vol. 53, no. 3, pp. 951–965, 2010.
[45]  D. W. Klaver, M. C. J. Wilce, H. Cui et al., “Is BACE1 a suitable therapeutic target for the treatment of Alzheimer's disease? Current strategies and future directions,” Biological Chemistry, vol. 391, no. 8, pp. 849–859, 2010.
[46]  B. P. Imbimbo and I. Peretto, “Semagacestat, a γ-secretase inhibitor for the potential treatment of Alzheimer's disease,” Current Opinion in Investigational Drugs, vol. 10, no. 7, pp. 721–730, 2009.
[47]  D. B. Henley, P. C. May, R. A. Dean, and E. R. Siemers, “Development of semagacestat (LY450139), a functional γ-secretase inhibitor, for the treatment of Alzheimer's disease,” Expert Opinion on Pharmacotherapy, vol. 10, no. 10, pp. 1657–1664, 2009.
[48]  A. Extance, “Alzheimer's failure raises questions about disease-modifying strategies,” Nature Reviews Drug Discovery, vol. 9, no. 10, pp. 749–750, 2010.
[49]  L. McConlogue, M. Buttini, J. P. Anderson et al., “Partial reduction of BACE1 has dramatic effects on Alzheimer plaque and synaptic pathology in APP transgenic mice,” Journal of Biological Chemistry, vol. 282, no. 36, pp. 26326–26334, 2007.
[50]  J. M. Castellano, J. Kim, F. R. Stewart et al., “Human apoE isoforms differentially regulate brain amyloid-β peptide clearance,” Science Translational Medicine, vol. 3, no. 89, article ra57, 2011.
[51]  M. Takahashi, H. Miyoshi, I. M. Verma, and F. H. Gage, “Rescue from photoreceptor degeneration in the rd mouse by human immunodeficiency virus vector-mediated gene transfer,” Journal of Virology, vol. 73, no. 9, pp. 7812–7816, 1999.
[52]  A. F. Hottinger, M. Azzouz, N. Deglon, P. Aebischer, and A. D. Zurn, “Complete and long-term rescue of lesioned adult motoneurons by lentiviral-mediated expression of glial cell line-derived neurotrophic factor in the facial nucleus,” Journal of Neuroscience, vol. 20, no. 15, pp. 5587–5593, 2000.
[53]  Z. Lai and R. O. Brady, “Gene transfer into the central nervous system in vivo using a recombinanat lentivirus vector,” Journal of Neuroscience Research, vol. 67, no. 3, pp. 363–371, 2002.
[54]  J. H. Kordower, M. E. Emborg, J. Bloch et al., “Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease,” Science, vol. 290, no. 5492, pp. 767–773, 2000.
[55]  B. J. Spencer and I. M. Verma, “Targeted delivery of proteins across the blood-brain barrier,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 18, pp. 7594–7599, 2007.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133