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...

Cholesterol Potentiates β-Amyloid Genesis in Cultured Human Umbilical Vein Endothelial Cells

DOI: 10.4236/aad.2016.52002, PP. 23-34

Keywords: Aging, Alzheimer’s Disease, Cholesterol, Neurodegeneration, Vascular Dementia

Full-Text   Cite this paper   Add to My Lib

Abstract:

Cerebral Amyloid Angiopathy (CAA) occurs commonly among the elderly and almost invariably in patients with Alzheimer’s Disease (AD). The β-amyloid peptides (Aβ) are produced via the amy-loidogenic processing of β-Amyloid Precursor Protein (APP) by β-secretase-1 (BACE1) and γ- secretase. Vascular endothelial cells are lately shown to possess the molecular machinery of Aβ production, which might participate in the development of CAA. Hypercholesterolemia is considered a risk factor for AD, whereas less is known if cholesterol may modulate endothelial Aβ production. In the present study we verified the amyloidogenic capability of Human Umbilical Vein Endothelial Cells (HUVECs) in vitro and explored the effect of cholesterol exposure on their amy-loidogenic potential. Cholesterol treatments at 12.5 and 25 mg/dL significantly elevated APP, BACE1 and APP β-CTF protein levels and β-site APP cleavage activity in cell lysates, and Aβ40 levels in culture medium. However, coincubation with cholesterol at 50 and 100 mg/dL attenuated the viability of the cultured cells and diminished their amyloidogenic capability. These findings suggest that high cholesterol exposure is stressful to vascular endothelial cells, and at a certain dosage range can promote an amyloidogenic response in these cells.

References

[1]  Castellani, R.J., Smith, M.A., Perry, G. and Friedland, R.P. (2004) Cerebral Amyloid Angiopathy: Major Contributor or Decorative Response to Alzheimer’s Disease Pathogenesis. Neurobiology Aging, 25, 599-602. http://dx.doi.org/10.1016/j.neurobiolaging.2003.12.019
[2]  Thal, D.R., Griffin, W.S., de Vos, R.A. and Ghebremedhin, E. (2008) Cerebral Amyloid Angiopathy and Its Relationship to Alzheimer’s Disease. Acta Neuropathologica, 115, 599-609.
http://dx.doi.org/10.1007/s00401-008-0366-2
[3]  Weller, R.O., Boche, D. and Nicoll, J.A. (2009) Microvasculature Changes and Cerebral Amyloid Angiopathy in Alzheimer’s Disease and Their Potential Impact on Therapy. Acta Neuropathologica, 118, 87-102.
http://dx.doi.org/10.1007/s00401-009-0498-z
[4]  Honjo, K., Black, S.E. and Verhoeff, N.P. (2012) Alzheimer’s Disease, Cerebrovascular Disease, and the β-Amyloid Cascade. Canadian Journal of Neurological Sciences, 39, 712-728.
http://dx.doi.org/10.1017/S0317167100015547
[5]  Pfeifer, L.A., White, L.R., Ross, G.W., Petrovitch, H. and Launer, L.J. (2002) Cerebral Amyloid Angiopathy and Cognitive Function: The HAAS Autopsy Study. Neurology, 58, 1629-1634.
http://dx.doi.org/10.1212/WNL.58.11.1629
[6]  Jellinger, K.A. and Attems, J. (2003) Incidence of Cerebrovascular Lesions in Alzheimer’s Disease: A Postmortem Study. Acta Neuropathologica, 105, 14-17.
[7]  Keage, H.A., Carare, R.O., Friedland, R.P., Ince, P.G., Love, S., Nicoll, J.A., Wharton, S.B., Weller, R.O. and Brayne, C. (2009) Population Studies of Sporadic Cerebral Amyloid Angiopathy and Dementia: A Systematic Review. BMC Neurology, 9, 3.
http://dx.doi.org/10.1186/1471-2377-9-3
[8]  Arvanitakis, Z., Leurgans, S.E., Wang, Z., Wilson, R.S., Bennett, D.A. and Schneider, J.A. (2011) Cerebral Amyloid Angiopathy Pathology and Cognitive Domains in Older Persons. Annals of Neurology, 69, 320-327. http://dx.doi.org/10.1002/ana.22112
[9]  Obrien, R.J. (2011) Vascular Dementia: Atherosclerosis, Cognition and Alzheimer’s Disease. Current Alzheimer Research, 8, 341-344.
http://dx.doi.org/10.2174/156720511795745267
[10]  Viswanathan, A. and Greenberg, S.M. (2011) Cerebral Amyloid Angiopathy in the Elderly. Annals of Neurology, 70, 871-880.
http://dx.doi.org/10.1002/ana.22516
[11]  Purrucker, J.C., Hund, E., Ringleb, P.A., Hartmann, C., Rohde, S., Schönland, S. and Steiner, T. (2013) Cerebral Amy- loid Angiopathy—An Underdiagnosed Entity in Younger Adults with Lobar Intracerebral Hemorrhage? Amyloid, 20, 45-47.
http://dx.doi.org/10.3109/13506129.2012.746937
[12]  Charidimou, A., Gang, Q. and Werring, D.J. (2012) Sporadic Cerebral Amyloid Angiopathy Revisited: Recent Insights into Pathophysiology and Clinical Spectrum. Journal of Neurology, Neurosurgery, and Psychiatry, 83, 124-137.
http://dx.doi.org/10.1136/jnnp-2011-301308
[13]  Allen, N., Robinson, A.C., Snowden, J., Davidson, Y.S. and Mann, D.M. (2014) Patterns of Cerebral Amyloid Angiopathy Define Histopathological Phenotypes in Alzheimer’s Disease. Neuropathology and Applied Neurobiology, 40, 136-148.
http://dx.doi.org/10.1111/nan.12070
[14]  Geddes, J.W. and Cotman, C.W. (1991) Plasticity in Alzheimer’s Disease: Too Much or Not Enough? Neurobiology of Aging, 12, 330-333.
http://dx.doi.org/10.1016/0197-4580(91)90011-8
[15]  Arendt, T. (2001) Alzheimer’s Disease as a Disorder of Mechanisms Underlying Structural Brain Self-Organization. Neuroscience, 102, 723-765.
http://dx.doi.org/10.1016/S0306-4522(00)00516-9
[16]  Castellani, R.J., Lee, H.G., Zhu, X., Perry, G. and Smith, M.A. (2008) Alzheimer Disease Pathology as a Host Response. Journal of Neuropathology and Experimental Neurology, 67, 523-531.
http://dx.doi.org/10.1097/NEN.0b013e318177eaf4
[17]  Li, J.M., Liu, C., Hu, X., Cai, Y., Ma, C., Luo, X.G. and Yan, X.X. (2014) Inverse Correlation between Alzheimer’s Disease and Cancer: Implication for a Strong Impact of Regenerative Propensity on Neurodegeneration? BMC Neurology, 14, 211.
http://dx.doi.org/10.1186/s12883-014-0211-2
[18]  Yan, X.X., Ma, C., Gai, W.P., Cai, H. and Luo, X.G. (2014) Can BACE1 Inhibition Mitigate Early Axonal Pathology in Neurological Diseases? Journal of Alzheimer’s Disease, 38, 705-718
[19]  Li, J.M., Cai, Y., Liu, F., Yang, L., Hu, X., Patrylo, P.R., Cai, H., Luo, X.G., Xiao, D. and Yan, X.X. (2015) Experimental Microembolism Induces Localized Neuritic Pathology in Guinea Pig Cerebrum. Oncotarget, 6, 10772-10785. http://dx.doi.org/10.18632/oncotarget.3599
[20]  Laird, F.M., Cai, H., Savonenko, A.V., Farah, M.H., He, K., Melnikova, T., Wen, H., Chiang, H.C., Xu, G., Koliatsos, V.E., Borchelt, D.R., Price, D.L., Lee, H.K. and Wong, P.C. (2005) BACE1, a Major Determinant of Selective Vulnerability of the Brain to Amyloid-Beta Amyloidogenesis, Is Essential for Cognitive, Emotional, and Synaptic Functions. Journal of Neuroscience, 25, 11693-11709.
http://dx.doi.org/10.1523/JNEUROSCI.2766-05.2005
[21]  Vassar, R., Kovacs, D.M., Yan, R. and Wong, P.C. (2009) The Beta-Secretase Enzyme BACE in Health and Alzheimer’s Disease: Regulation, Cell Biology, Function, and Therapeutic Potential. Journal of Neuroscience, 29, 12787- 12794. http://dx.doi.org/10.1523/JNEUROSCI.3657-09.2009
[22]  Liu, F., Xue, Z.Q., Deng, S.H., Kun, X., Luo, X.G., Patrylo, P.R., Rose, G.M., Cai, H., Struble, R.G., Cai, Y. and Yan, X.X. (2013) γ-Secretase Binding Sites in Aged and Alzheimer’s Disease Human Cerebrum: the Choroid Plexus as a Putative Origin of CSF Aβ. European Journal of Neuroscience, 37, 1714-1725.
http://dx.doi.org/10.1111/ejn.12159
[23]  Holsinger, R.M., McLean, C.A., Beyreuther, K., Masters, C.L. and Evin, G. (2002) Increased Expression of the Amyloid Precursor Beta-Secretase in Alzheimer’s Disease. Annals of Neurology, 51, 783-786.
http://dx.doi.org/10.1002/ana.10208
[24]  Tyler, S.J., Dawbarn, D., Wilcock, G.K. and Allen, S.J. (2002) Alpha- and Beta-Secretase: Profound Changes in Alzheimer’s Disease. Biochemical and Biophysical Research Communications, 299, 373-376.
http://dx.doi.org/10.1016/S0006-291X(02)02635-9
[25]  Fukumoto, H., Cheung, B.S., Hyman, B.T. and Irizarry, M.C. (2002) Beta-Secretase Protein and Activity are Increased in the Neocortex in Alzheimer Disease. Archives of Neurology, 59, 1381-1389.
http://dx.doi.org/10.1001/archneur.59.9.1381
[26]  Yang, L.B., Lindholm, K., Yan, R., Citron, M., Xia, W., Yang, X.L., Beach, T., Sue, L., Wong, P., Price, D., Li, R. and Shen, Y. (2003) Elevated Beta-Secretase Expression and Enzymatic Activity Detected in Sporadic Alzheimer Disease. Nature Medicine, 9, 3-4.
http://dx.doi.org/10.1038/nm0103-3
[27]  Li, R., Lindholm, K., Yang, L.B., Yue, X., Citron, M., Yan, R., Beach, T., Sue, L., Sabbagh, M., Cai, H., Wong, P., Price, D. and Shen, Y. (2004) Amyloid Beta Peptide Load Is Correlated with Increased Beta-Secretase Activity in Sporadic Alzheimer’s Disease Patients. Proceedings of the National Academy of Sciences of the United States of America, 101, 3632-3637.
http://dx.doi.org/10.1073/pnas.0205689101
[28]  Coulson, D.T., Beyer, N., Quinn, J.G., Brockbank, S., Hellemans, J., Irvine, G.B., Ravid, R. and Johnston, J.A. (2010) BACE1 mRNA Expression in Alzheimer’s Disease Postmortem Brain Tissue. Journal of Alzheimer’s Disease, 22, 1111-1122.
[29]  Cai, Y., Xiong, K., Zhang, X.M., Cai, H., Luo, X.G., Feng, J.C., Clough, R.W., Struble, R.G., Patrylo, P.R., Chu, Y., Kordower, J.H. and Yan, X.X. (2010) β-Secretase-1 Elevation in Aged Monkey and Alzheimer’s Disease Human Cerebral Cortex Occurs around the Vasculature in Partnership with Multisystem Axon Terminal Pathogenesis and β-Amyloid Accumulation. European Journal of Neuroscience, 32, 1223-1238.
http://dx.doi.org/10.1111/j.1460-9568.2010.07376.x
[30]  Cheng, X., He, P., Lee, T., Yao, H., Li, R. and Shen, Y. (2014) High Activities of BACE1 in Brains with Mild Cognitive Impairment. American Journal of Pathology, 184, 141-147.
http://dx.doi.org/10.1016/j.ajpath.2013.10.002
[31]  Zhao, J., Fu, Y., Yasvoina, M., Shao, P., Hitt, B., O’Connor, T., Logan, S., Maus, E., Citron, M., Berry, R., Binder, L. and Vassar, R. (2007) Beta-Site Amyloid Precursor Protein Cleaving Enzyme 1 Levels Become Elevated in Neurons around Amyloid Plaques: Implications for Alzheimer’s Disease Pathogenesis. Journal of Neuroscience, 27, 3639-3649.
http://dx.doi.org/10.1523/JNEUROSCI.4396-06.2007
[32]  Zhang, X.M., Cai, Y., Cai, H., Xiong, K., Luo, X.G., Feng, J.C., Clough, R.W., Struble, R.G., Patrylo, P.R. and Yan, X.X. (2009) BACE1 Elevation in Transgenic Mouse Models of Alzheimer’s Disease Is Associated with Synaptic/Axonal Pathology and Amyloidogenesis: Implications for Neuritic Plaque Development. European Journal of Neuroscience, 30, 2271-2283.
http://dx.doi.org/10.1111/j.1460-9568.2009.07017.x
[33]  Cai, Y., Zhang, X.M., Macklin, L.N., Cai, H., Luo, X.G., Oddo, S., Laferla, F.M., Struble, R.G., Rose, G.M., Patrylo, P.R. and Yan, X.X. (2012) BACE1 Elevation Is Involved in Amyloid Plaque Development in the Triple Transgenic Model of Alzheimer’s Disease: Differential Aβ Antibody Labeling of Early-Onset Axon Terminal Pathology. Neurotoxicity Research, 21, 160-174.
http://dx.doi.org/10.1007/s12640-011-9256-9
[34]  Kandalepas, P.C., Sadleir, K.R., Eimer, W.A., Zhao, J., Nicholson, D.A. and Vassar, R. (2013) The Alzheimer’s β-Secretase BACE1 Localizes to Normal Presynaptic Terminals and to Dystrophic Presynaptic Terminals Surrounding Amyloid Plaques. Acta Neuropathologica, 126, 329-352.
http://dx.doi.org/10.1007/s00401-013-1152-3
[35]  Simons, E.R., Marshall, D.C., Long, H.J., Otto, K., Billingslea, A., Tibbles, H., Wells, J., Eisenhauer, P., Fine, R.E., Cribbs, D.H., Davies, T.A. and Abraham, C.R. (1998) Blood Brain Barrier Endothelial Cells Express Candidate Amyloid Precursor Protein-Cleaving Secretases. Amyloid, 5, 153-162.
http://dx.doi.org/10.3109/13506129809003841
[36]  Austin, S.A., Santhanam, A.V. and Katusic, Z.S. (2010) Endothelial Nitric Oxide Modulates Expression and Processing of Amyloid Precursor Protein. Circulation Research, 107, 1498-502.
http://dx.doi.org/10.1161/CIRCRESAHA.110.233080
[37]  Kitazume, S., Tachida, Y., Kato, M., Yamaguchi, Y., Honda, T., Hashimoto, Y., Wada, Y., Saito, T., Iwata, N., Saido, T. and Taniguchi, N. (2010) Brain Endothelial Cells Produce Amyloid β from Amyloid Precursor Protein 770 and Preferentially Secrete the O-Glycosylated Form. The Journal of Biological Chemistry, 285, 40097-40103.

http://dx.doi.org/10.1074/jbc.M110.144626
[38]  Ma, J.F., Wang, H.M., Li, Q.Y., Zhang, Y., Pan, J., Qiang, Q., Xin, X.Y., Tang, H.D., Ding, J.Q. and Chen, S.D. (2010) Starvation Triggers Abeta42 Generation from Human Umbilical Vascular Endothelial Cells. FEBS Letters, 584, 3101- 3106.
http://dx.doi.org/10.1016/j.febslet.2010.05.048
[39]  Bulbarelli, A., Lonati, E., Brambilla, A., Orlando, A., Cazzaniga, E., Piazza, F., Ferrarese, C., Masserini, M. and Sancini, G. (2012) Aβ42 Production in Brain Capillary Endothelial Cells after Oxygen and Glucose Deprivation. Molecular and Cellular Neuroscience, 49, 415-422.
http://dx.doi.org/10.1016/j.mcn.2012.01.007
[40]  Cheng, X., He, P., Yao, H., Dong, Q., Li, R. and Shen, Y. (2014) Occludin Deficiency with BACE1 Elevation in Cerebral Amyloid Angiopathy. Neurology, 82, 1707-1715.
http://dx.doi.org/10.1212/WNL.0000000000000403
[41]  Xue, Z.Q., He, Z.W., Yu, J.J., Cai, Y., Qiu, W.Y., Pan, A., Gai, W.P., Cai, H., Luo, X.G., Ma, C. and Yan, X.X. (2015) Non-Neuronal and Neuronal BACE1 Elevation in Association with Angiopathic and Leptomeningeal β-Amyloid Deposition in the Human Brain. BMC Neurology, 15, 71.
http://dx.doi.org/10.1186/s12883-015-0327-z
[42]  Devraj, K., Poznanovic, S., Spahn, C., Schwall, G., Harter, P.N., Mittelbronn, M., Antoniello, K., Paganetti. P., Muhs, A., Heilemann, M., Hawkins, R.A., Schrattenholz, A. and Liebner, S. (2015) BACE-1 Is Expressed in the Blood-Brain Barrier Endothelium and Is Upregulated in a Murine Model of Alzheimer’s Disease. Journal of Cerebral Blood Flow and Metabolism.
http://dx.doi.org/10.1177/0271678x15606463
[43]  Utermann, G., Kindermann, I., Kaffarnik, H. and Steinmetz, A. (1984) Apolipoprotein E Phenotypes and Hyperlipidemia. Human Genetics, 65, 232-236.
http://dx.doi.org/10.1007/BF00286508
[44]  Li, G., Shofer, J.B., Kukull, W.A., Peskind, E.R., Tsuang, D.W., Breitner, J.C., McCormick, W., Bowen, J.D., Teri, L., Schellenberg, G.D. and Larson, E.B. (2005) Serum Cholesterol and Risk of Alzheimer Disease: A Community-Based Cohort Study. Neurology, 65, 1045-1050.
http://dx.doi.org/10.1212/01.wnl.0000178989.87072.11
[45]  Anstey, K.J., Lipnicki, D.M. and Low, L.F. (2008) Cholesterol as a Risk Factor for Dementia and Cognitive Decline: A Systematic Review of Prospective Studies with Meta-Analysis. American Journal of Geriatric Psychiatry, 16, 343- 354.
http://dx.doi.org/10.1097/01.JGP.0000310778.20870.ae
[46]  Mahley, R.W., Huang, Y. and Weisgraber, K.H. (2006) Putting Cholesterol in Its Place: ApoE and Reverse Cholesterol Transport. Journal of Clinical Investigation, 116, 1226-1229.
http://dx.doi.org/10.1172/JCI28632
[47]  Solomon, A., Kivipelto, M., Wolozin, B., Zhou, J. and Whitmer, R.A. (2009) Midlife Serum Cholesterol and Increased Risk of Alzheimer’s and Vascular Dementia Three Decades Later. Dementia and Geriatric Cognitive Disorder, 28, 75- 80.
http://dx.doi.org/10.1159/000231980
[48]  Leduc, V., Jasmin-Bélanger, S. and Poirier, J. (2010) APOE and Cholesterol Homeostasis in Alzheimer’s Disease. Trends in Molecular Medicine, 16, 469-477.
http://dx.doi.org/10.1016/j.molmed.2010.07.008
[49]  Shepardson, N.E., Shankar, G.M. and Selkoe, D.J. (2011) Cholesterol Level and Statin Use in Alzheimer Disease: II. Review of Human Trials and Recommendations. Archives of Neurology, 68, 1385-1392.
http://dx.doi.org/10.1001/archneurol.2011.242
[50]  Tolppanen, A.M., Solomon, A., Soininen, H. and Kivipelto, M. (2012) Midlife Vascular Risk Factors and Alzheimer’s Disease: Evidence from Epidemiological Studies. Journal of Alzheimer’s Disease, 32, 531-540.
[51]  Sadigh-Eteghad, S., Talebi, M. and Farhoudi, M. (2012) Association of Apolipoprotein E Epsilon 4 Allele with Sporadic Late Onset Alzheimer’s Disease. A Meta-Analysis. Neurosciences (Riyadh), 17, 321-326.
[52]  Silva, T., Teixeira, J., Remião, F. and Borges, F. (2013) Alzheimer’s Disease, Cholesterol, and Statins: The Junctions of Important Metabolic Pathways. Angewandte Chemie International Edition in English, 52, 1110-1121.http://dx.doi.org/10.1002/anie.201204964
[53]  Reed, B., Villeneuve, S., Mack, W., Decarli, C., Chui, H.C. and Jagust, W. (2014) Associations between Serum Cholesterol Levels and Cerebral Amyloidosis. JAMA Neurology, 71, 195-200.
http://dx.doi.org/10.1001/jamaneurol.2013.5390
[54]  Maulik, M., Westaway, D., Jhamandas, J.H. and Kar, S. (2013) Role of Cholesterol in APP Metabolism and Its Significance in Alzheimer’s Disease Pathogenesis. Molecular Neurobiology, 47, 37-63.
http://dx.doi.org/10.1007/s12035-012-8337-y
[55]  Yan, X.X., Li, T., Rominger, C.M., Prakash, S.R., Wong, P.C., Olson, R.E., Zaczek, R. and Li, Y.W. (2004) Binding Sites of Gamma-Secretase Inhibitors in Rodent Brain: Distribution, Postnatal Development, and Effect of Deafferentation. The Journal of Neuroscience, 24, 2942-2952.
http://dx.doi.org/10.1523/JNEUROSCI.0092-04.2004
[56]  De Strooper, B. and Chávez Gutiérrez, L. (2015) Learning by Failing: Ideas and Concepts to Tackle γ-Secretases in Alzheimer’s Disease and beyond. Annual Review of Pharmacology and Toxicology, 55, 419-437. http://dx.doi.org/10.1146/annurev-pharmtox-010814-124309
[57]  Kim, H.S., Park, C.H., Cha, S.H., Lee, J.H., Lee, S., Kim, Y., Rah, J.C., Jeong, S.J. and Suh, Y.H. (2000) Carboxyl-Terminal Fragment of Alzheimer’s APP Destabilizes Calcium Homeostasis and Renders Neuronal Cells Vulnerable to Excitotoxicity. The FASEB Journal, 14, 1508-1517.
http://dx.doi.org/10.1096/fj.14.11.1508
[58]  Saganich, M.J., Schroeder, B.E., Galvan, V., Bredesen, D.E., Koo, E.H. and Heinemann, S.F. (2006) Deficits in Synaptic Transmission and Learning in Amyloid Precursor Protein (APP) Transgenic Mice Require C-Terminal Cleavage of APP. The Journal of Neuroscience, 26, 13428-13436.
http://dx.doi.org/10.1523/JNEUROSCI.4180-06.2006
[59]  Lauritzen, I., Pardossi-Piquard, R., Bauer, C., Brigham, E., Abraham, J.D., Ranaldi, S., Fraser, P., St-George-Hyslop, P., Le Thuc, O., Espin, V., Chami, L., Dunys, J. and Checler, F. (2012) The β-Secretase-Derived C-Terminal Fragment of βAPP, C99, but Not Aβ, Is a Key Contributor to Early Intraneuronal Lesions in Triple-Transgenic Mouse Hippocampus. The Journal of Neuroscience, 32, 16243-16255.
http://dx.doi.org/10.1523/JNEUROSCI.2775-12.2012
[60]  Jiang, Y., Rigoglioso, A., Peterhoff, C.M., Pawlik, M., Sato, Y., Bleiwas, C., Stavrides, P., Smiley, J.F., Ginsberg, S.D., Mathews, P.M., Levy, E. and Nixon, R.A. (2016) Partial BACE1 Reduction in a Down Syndrome Mouse Model Blocks Alzheimer-Related Endosomal Anomalies and Cholinergic Neurodegeneration: Role of APP-CTF. Neurobiology of Aging, 39, 90-98.
http://dx.doi.org/10.1016/j.neurobiolaging.2015.11.013
[61]  Barão, S., Moechars, D., Lichtenthaler, S.F. and De Strooper, B. (2016) BACE1 Physiological Functions May Limit Its Use as Therapeutic Target for Alzheimer’s Disease. Trends in Neurosciences, 39, 158-269.
http://dx.doi.org/10.1016/j.tins.2016.01.003
[62]  Nicoll, J.A. and McCarron, M.O. (2001) APOE Gene Polymorphism as a Risk Factor for Cerebral Amyloid Angiopathy-Related Hemorrhage. Amyloid, 8, 51-55.
[63]  Kurata, T., Kawai, H., Miyazaki, K., Kozuki, M., Morimoto, N., Ohta, Y., Ikeda, Y. and Abe, K. (2012) Statins Have Therapeutic Potential for the Treatment of Alzheimer’s Disease, Likely via Protection of the Neurovascular Unit in the AD Brain. Journal of the Neurological Sciences, 322, 59-63.
http://dx.doi.org/10.1016/j.jns.2012.06.011

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413