2,014
Views
6
CrossRef citations to date
0
Altmetric
Review

Critical role of mitosis in spontaneous late-onset Alzheimer’s disease; from a Shugoshin 1 cohesinopathy mouse model

, , & ORCID Icon
Pages 2321-2334 | Received 22 Jul 2018, Accepted 20 Aug 2018, Published online: 20 Sep 2018

References

  • Jack CR Jr., Bennett DA, Blennow K, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:535–562.
  • Frisoni GB, Boccardi M, Barkhof F, et al. Strategic roadmap for an early diagnosis of Alzheimer’s disease based on biomarkers. Lancet Neurol. 2017;16:661–676.
  • Jansen WJ, Ossenkoppele R, Tijms BM, et al. Association of cerebral amyloid-β aggregation with cognitive functioning in persons without dementia. JAMA Psychiatry. 2018;75:84–95.
  • Jan AT, Azam M, Rahman S, et al. Perspective insights into disease progression, diagnostics, and therapeutic approaches in Alzheimer’s disease: a judicious update. Front Aging Neurosci. 2017;9:356.
  • Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356.
  • Hardy J. The discovery of Alzheimer-causing mutations in the APP gene and the formulation of the “amyloid cascade hypothesis”. FEBS J. 2017;284:1040–1044.
  • Kametani F, Hasegawa M. Reconsideration of amyloid hypothesis and Tau hypothesis in Alzheimer’s disease. Front Neurosci. 2018;12:25.
  • Alzheimer’s organization [cited 2018 July 20]. Available from: https://www.alz.org/research/science/alzheimers_research.asp
  • Scheltens P, Blennow K, Breteler MM, et al. Alzheimer’s disease. Lancet. 2016;388:505–517.
  • Cummings J, Aisen PS, DuBois B, et al. Drug development in Alzheimer’s disease: the path to 2025. Alzheimers Res Ther. 2016;8:39.
  • Qian X, Hamad B, Dias-Lalcaca G. The Alzheimer disease market. Nat Rev Drug Discov. 2015;14:675–676.
  • Cummings JL, Morstorf T, Zhong K. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther. 2014;6:37.
  • Carroll J Another Alzheimer’s drug flops in pivotal clinical trial Endpoints News. cited 2017 Feb 15. Available from: http://www.sciencemag.org/news/2017/02/another-alzheimers-drug-flops-pivotal-clinical-trial
  • Szeto JY, Lewis SJ. Current treatment options for Alzheimer’s disease and Parkinson’s disease dementia. Curr Neuropharmacol. 2016;14(4):326–338.
  • Logovinsky V, Satlin A, Lai R, et al. Safety and tolerability of BAN2401–a clinical study in Alzheimer’s disease with a protofibril selective Aβ antibody. Alzheimers Res Ther. 2016;8:14.
  • Söllvander S, Nikitidou E, Gallasch L, et al. The Aβ protofibril selective antibody mAb158 prevents accumulation of Aβ in astrocytes and rescues neurons from Aβ-induced cell death. J Neuroinflammation. 2018;15:98.
  • Hall AM, Roberson ED. Mouse models of Alzheimer’s disease. Brain Res Bull. 2012;88:3–12.
  • Sasaguri H, Nilsson P, Hashimoto S, et al. APP mouse models for Alzheimer’s disease preclinical studies. EMBO J. 2017;36:2473–2487.
  • Kitazawa M, Medeiros R, Laferla FM. Transgenic mouse models of Alzheimer disease: developing a better model as a tool for therapeutic interventions. Curr Pharm Des. 2012;18:1131–1147.
  • Ameen-Ali KE, Wharton SB, Simpson JE, et al. Review: neuropathology and behavioural features of transgenic murine models of Alzheimer’s disease. Neuropathol Appl Neurobiol. 2017;43:553–570.
  • Egan KJ, Vesterinen HM, Beglopoulos V, et al. From a mouse: systematic analysis reveals limitations of experiments testing interventions in Alzheimer’s disease mouse models. Evid Based Preclin Med. 2016;3:e00015.
  • Wirths O. Altered neurogenesis in mouse models of Alzheimer disease. Neurogenesis (Austin). 2017;4:e1327002.
  • Huang CC, Chung CM, Leu HB, et al. Diabetes mellitus and the risk of Alzheimer’s disease: a nationwide population-based study. PLoS One. 2014;9:e87095.
  • Hou Y, Song H, Croteau DL, et al. Genome instability in Alzheimer disease. Mech Ageing Dev. 2017;161:83–94.
  • Lee SL, Thomas P, Fenech M. Genome instability biomarkers and blood micronutrient risk profiles associated with mild cognitive impairment and Alzheimer’s disease. Mutat Res. 2015;776:54–83.
  • Hartley D, Blumenthal T, Carrillo M, et al. Down syndrome and Alzheimer’s disease: common pathways, common goals. Alzheimers Dement. 2015;11(6):700–709.
  • Chan GK, Jablonski SA, Sudakin V, et al. Human BUBR1 is a mitotic checkpoint kinase that monitors CENP-E functions at kinetochores and binds the cyclosome/APC. J Cell Biol. 1999;146:941–954.
  • Tang Z, Bharadwaj R, Li B, et al. Mad2-independent inhibition of APCCdc20 by the mitotic checkpoint protein BubR1. Dev Cell. 2001;1:227–237.
  • Kapanidou M, Lee S, Bolanos-Garcia VM. BubR1 kinase: protection against aneuploidy and premature aging. Trends Mol Med. 2015;21:364–372.
  • Dai W, Wang Q, Liu T, et al. Slippage of mitotic arrest and enhanced tumor development in mice with BubR1 haploinsufficiency. Cancer Res. 2004;64:440–445.
  • Rao CV, Yang YM, Swamy MV, et al. Colonic tumorigenesis in BubR1±ApcMin/+ compound mutant mice is linked to premature separation of sister chromatids and enhanced genomic instability. Proc Natl Acad Sci U S A. 2005;102(12):4365–4370.
  • Baker DJ, Jeganathan KB, Cameron JD, et al. BubR1 insufficiency causes early onset of aging-associated phenotypes and infertility in mice. Nat Genet. 2004;36(7):744–749.
  • Baker DJ, Wijshake T, Tchkonia T, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–236.
  • Baker DJ, Dawlaty MM, Wijshake T, et al. Increased expression of BubR1 protects against aneuploidy and cancer and extends healthy lifespan. Nat Cell Biol. 2013;15:96–102.
  • Choi CI, Yoo KH, Hussaini SM, et al. The progeroid gene BubR1 regulates axon myelination and motor function. Aging (Albany NY). 2016;8:2667–2688.
  • Yang Z, Jun H, Choi CI, et al. Age-related decline in BubR1 impairs adult hippocampal neurogenesis. Aging Cell. 2017;16:598–601.
  • Zivković L, Spremo-Potparević B, Djelić N, et al. Analysis of premature centromere division (PCD) of the chromosome 18 in peripheral blood lymphocytes in Alzheimer disease patients. Mech Ageing Dev. 2006;127:892–896.
  • Zivković L, Spremo-Potparević B, Plecas-Solarović B, et al. Premature centromere division of metaphase chromosomes in peripheral blood lymphocytes of Alzheimer’s disease patients: relation to gender and age. J Gerontol A Biol Sci Med Sci. 2010;65:1269–1274.
  • Bajic V, Spremo-Potparevic B, Zivkovic L, et al. Cohesion and the aneuploid phenotype in Alzheimer’s disease: a tale of genome instability. Neurosci Biobehav Rev. 2015;55:365–374.
  • Boveri T. Concerning the origin of malignant tumours by Theodor Boveri. Translated and annotated by Henry Harris. J Cell Sci. 2008;121(Suppl 1):1–84.
  • Schvartzman JM, Sotillo R, Benezra R. Mitotic chromosomal instability and cancer: mouse modelling of the human disease. Nat Rev Cancer. 2010;10(2):102–115.
  • Rao CV, Yamada HY, Yao Y, et al. Enhanced genomic instabilities caused by deregulated microtubule dynamics and chromosome segregation: a perspective from genetic studies in mice. Carcinogenesis. 2009;30:1469–1474.
  • Foijer F, Draviam VM, Sorger PK, et al. Studying chromosome instability in the mouse. Biochim Biophys Acta. 2008;1786:73–82.
  • Ricke RM, van Ree JH, van Deursen JM. Whole chromosome instability and cancer: a complex relationship. Trends Genet. 2008;24:457–466.
  • Rao CV, Yamada HY. Genomic instability and colon carcinogenesis: from the perspective of genes. Front Oncol. 2013;3:130.
  • Weaver BA, Silk AD, Montagna C, et al. Aneuploidy acts both oncogenically and as a tumor suppressor. Cancer Cell. 2007;11:25–36.
  • Weaver BA, Cleveland DW. The role of aneuploidy in promoting and suppressing tumors. J Cell Biol. 2009;185:935–937.
  • Zasadil LM, Britigan EM, Ryan SD, et al. High rates of chromosome missegregation suppress tumor progression but do not inhibit tumor initiation. Mol Biol Cell. 2016;27:1981–1989.
  • Silk AD, Zasadil LM, Holland AJ, et al. Chromosome missegregation rate predicts whether aneuploidy will promote or suppress tumors. Proc Natl Acad Sci USA. 2013;110:E4134–41.
  • Funk LC, Zasadil LM, Weaver BA. Living in CIN: mitotic infidelity and its consequences for tumor promotion and suppression. Dev Cell. 2016;39:638–652.
  • Salic A, Waters JC, Mitchison TJ. Vertebrate shugoshin links sister centromere cohesion and kinetochore microtubule stability in mitosis. Cell. 2004;118:567–578.
  • McGuinness BE, Hirota T, Kudo NR, et al. Shugoshin prevents dissociation of cohesin from centromeres during mitosis in vertebrate cells. PLoS Biol. 2005;3:e86.
  • Schöckel L, Möckel M, Mayer B, et al. Cleavage of cohesin rings coordinates the separation of centrioles and chromatids. Nat Cell Biol. 2011;13:966–972.
  • Wang X, Yang Y, Duan Q, et al. sSgo1, a major splice variant of Sgo1, functions in centriole cohesion where it is regulated by Plk1. Dev Cell. 2008;14:331–341.
  • Tsang WY, Dynlacht BD. sSgo1, a guardian of centriole cohesion. Dev Cell. 2008;14:320–322.
  • Yamada HY, Yao Y, Wang X, et al. Haploinsufficiency of SGO1 results in deregulated centrosome dynamics, enhanced chromosomal instability and colon tumorigenesis. Cell Cycle. 2012;11:479–488.
  • Yamada HY, Zhang Y, Reddy A, et al. Tumor-promoting/progressing role of additional chromosome instability in hepatic carcinogenesis in Sgo1 (Shugoshin 1) haploinsufficient mice. Carcinogenesis. 2015;36:429–440.
  • Rao CV, Sanghera S, Zhang Y, et al. Antagonizing pathways leading to differential dynamics in colon carcinogenesis in Shugoshin1 (Sgo1)-haploinsufficient chromosome instability model. Mol Carcinog. 2016;55:600–610.
  • Iwaizumi M, Shinmura K, Mori H, et al. Human Sgo1 downregulation leads to chromosomal instability in colorectal cancer. Gut. 2009;58:249–260.
  • Kim MS, An CH, Yoo NJ, et al. Frameshift mutations of chromosome cohesion-related genes SGOL1 and PDS5B in gastric and colorectal cancers with high microsatellite instability. Hum Pathol. 2013;44:2234–2240.
  • Kahyo T, Iwaizumi M, Shinmura K, et al. A novel tumor-derived SGOL1 variant causes abnormal mitosis and unstable chromatid cohesion. Oncogene. 2011;30:4453–4463.
  • Matsuura S, Kahyo T, Shinmura K, et al. SGOL1 variant B induces abnormal mitosis and resistance to taxane in non-small cell lung cancers. Sci Rep. 2013;3:3012.
  • Wang LH, Yen CJ, Li TN, et al. Sgo1 is a potential therapeutic target for hepatocellular carcinoma. Oncotarget. 2015;6:2023–2033.
  • Yamada HY, Kumar G, Zhang Y, et al. Systemic chromosome instability in Shugoshin-1 mice resulted in compromised glutathione pathway, activation of Wnt signaling and defects in immune system in the lung. Oncogenesis. 2016;5:e256.
  • Rao CV, Sanghera S, Zhang Y, et al. Systemic chromosome instability resulted in colonic transcriptomic changes in metabolic, proliferation, and stem cell regulators in Sgo1-/+ mice. Cancer Research. 2016;76:630–642.
  • Sheltzer JM, Torres EM, Dunham MJ, et al. Transcriptional consequences of aneuploidy. Proc Natl Acad Sci U S A. 2012;109:12644–12649.
  • Torres EM, Sokolsky T, Tucker CM, et al. Effects of aneuploidy on cellular physiology and cell division in haploid yeast. Science. 2007;317:916–924.
  • Williams BR, Prabhu VR, Hunter KE, et al. Aneuploidy affects proliferation and spontaneous immortalization in mammalian cells. Science. 2008;322:703–709.
  • Santaguida S, Vasile E, White E, et al. Aneuploidy-induced cellular stresses limit autophagic degradation. Genes Dev. 2015;29:2010–2021.
  • Chetaille P, Preuss C, Burkhard S, et al. Mutations in SGOL1 cause a novel cohesinopathy affecting heart and gut rhythm. Nat Genet. 2014;46(11):1245–1249.
  • Song AT, Galli A, Leclerc S, et al. Characterization of Sgo1 expression in developing and adult mouse. Gene Expr Patterns. 2017;25-26:36–45.
  • Song AT, Galli A, Leclerc S, et al. Dataset of Sgo1 expression in cardiac, gastrointestinal, hepatic and neuronal tissue in mouse. Data Brief. 2017;13:731–737.
  • Rao CV, Asch AS, Yamada HY. Emerging links among Chromosome Instability (CIN), cancer, and aging. Molecular Carcinogenesis. 2017;56:791–803.
  • Rao CV, Farooqui M, Zhang Y, et al. Spontaneous development of Alzheimer’s disease-associated brain pathology in a Shugoshin 1 mouse cohesinopathy model. Aging Cell. 2018;17(4):e12797. PMID: 29943428.
  • Valero J, Bernardino L, Cardoso FL, et al. Impact of neuroinflammation on hippocampal neurogenesis: relevance to aging and Alzheimer’s disease. J Alzheimers Dis. 2017;60:S161–S168.
  • Heneka MT, Carson MJ, El Khoury J, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405.
  • Sawikr Y, Yarla NS, Peluso I, et al. Neuroinflammation in Alzheimer’s disease: the preventive and therapeutic potential of polyphenolic nutraceuticals. Adv Protein Chem Struct Biol. 2017;108:33–57.
  • Praticò D. Oxidative stress hypothesis in Alzheimer’s disease: a reappraisal. Trends Pharmacol Sci. 2008;29:609–615.
  • Zhao Y, Zhao B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxid Med Cell Longev. 2013;2013:316523.
  • García-Blanco A, Baquero M, Vento M, et al. Potential oxidative stress biomarkers of mild cognitive impairment due to Alzheimer disease. J Neurol Sci. 2017;373:295–302.
  • Connolly NMC, Theurey P, Adam-Vizi V, et al. Guidelines on experimental methods to assess mitochondrial dysfunction in cellular models of neurodegenerative diseases. Cell Death Differ. 2018;25:542–572.
  • Wang JM, Sun C. Calcium and neurogenesis in Alzheimer’s disease. Front Neurosci. 2010;4:194.
  • Leija-Salazar M, Piette C, Proukakis C. Review: somatic mutations in neurodegeneration. Neuropathol Appl Neurobiol. 2018;44(3):267–285.
  • Duncan T, Valenzuela M. Alzheimer’s disease, dementia, and stem cell therapy. Stem Cell Res Ther. 2017;8:111.
  • Schwartz M, Deczkowska A. Neurological disease as a failure of brain-immune crosstalk: the multiple faces of neuroinflammation. Trends Immunol. 2016;37:668–679.
  • Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14:133–150.
  • Nalivaeva NN, Turner AJ. Role of ageing and oxidative stress in regulation of amyloid-degrading enzymes and development of neurodegeneration. Curr Aging Sci. 2017;10:32–40.
  • Adlard PA, Tran BA, Finkelstein DI, et al. A review of β-amyloid neuroimaging in Alzheimer’s disease. Front Neurosci. 2014;8:327.
  • Avila J, Llorens-Martín M, Pallas-Bazarra N, et al. Cognitive decline in neuronal aging and Alzheimer’s disease: role of NMDA receptors and associated proteins. Front Neurosci. 2017;11:626.
  • Foster TC, Kyritsopoulos C, Kumar A. Central role for NMDA receptors in redox mediated impairment of synaptic function during aging and Alzheimer’s disease. Behav Brain Res. 2017;322:223–232.
  • Bredesen D. The end of Alzheimer’s: the first program to prevent and reverse cognitive decline. New York (NY): Avery; 2017. ISBN-10: 0735216207.
  • Bredesen DE. Metabolic profiling distinguishes three subtypes of Alzheimer’s disease. Aging (Albany NY). 2015;7:595–600.
  • Kondratick CM, Vandré DD. Alzheimer’s disease neurofibrillary tangles contain mitosis-specific phosphoepitopes. J Neurochem. 1996;67:2405–2416.
  • Delobel P, Flament S, Hamdane M, et al. Abnormal Tau phosphorylation of the Alzheimer-type also occurs during mitosis. J Neurochem. 2002;83:412–420.
  • Judge M, Hornbeck L, Potter H, et al. Mitosis-specific phosphorylation of amyloid precursor protein at threonine 668 leads to its altered processing and association with centrosomes. Mol Neurodegener. 2011;6:80.
  • Ogawa O, Zhu X, Lee HG, et al. Ectopic localization of phosphorylated histone H3 in Alzheimer’s disease: a mitotic catastrophe? Acta Neuropathol. 2003;105:524–528.
  • Folch J, Junyent F, Verdaguer E, et al. Role of cell cycle re-entry in neurons: a common apoptotic mechanism of neuronal cell death. Neurotox Res. 2012;22:195–207.
  • Yang Y, Herrup K. Cell division in the CNS: protective response or lethal event in post-mitotic neurons? Biochim Biophys Acta. 2007;1772:457–466.
  • Zhu X, Lee HG, Perry G, et al. Alzheimer disease, the two-hit hypothesis: an update. Biochim Biophys Acta. 2007;1772:494–502.
  • Zhu X, Raina AK, Perry G, et al. Alzheimer’s disease: the two-hit hypothesis. Lancet Neurol. 2004;3:219–226.
  • Webber KM, Raina AK, Marlatt MW, et al. The cell cycle in Alzheimer disease: a unique target for neuropharmacology. Mech Ageing Dev. 2005;126:1019–1025.
  • Vincent I, Bu B, Hudson K, et al. Constitutive Cdc25B tyrosine phosphatase activity in adult brain neurons with M phase-type alterations in Alzheimer disease. Neuroscience. 2001;105:639–650.
  • Ding XL, Husseman J, Tomashevski A, et al. The cell cycle Cdc25A tyrosine phosphatase is activated in degenerating postmitotic neurons in Alzheimer disease. Am J Pathol. 2000;157:1983–1990.
  • Tomashevski A, Husseman J, Jin LW, et al. Constitutive Wee1 activity in adult brain neurons with M phase-type alterations in Alzheimer neurodegeneration. J Alzheimers Dis. 2001;3:195–207.
  • Seward ME, Swanson E, Norambuena A, et al. Amyloid-β signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer’s disease. J Cell Sci. 2013;126:1278–1286.
  • Csiszar A, Tarantini S, Fülöp GA, et al. Hypertension impairs neurovascular coupling and promotes microvascular injury: role in exacerbation of Alzheimer’s disease. Geroscience. 2017. PMID: 28853030.
  • Tucsek Z, Noa Valcarcel-Ares M, Tarantini S, et al. Hypertension-induced synapse loss and impairment in synaptic plasticity in the mouse hippocampus mimics the aging phenotype: implications for the pathogenesis of vascular cognitive impairment. Geroscience. 2017. PMID: 28664509.
  • Orehek AJ. The micron stroke hypothesis of Alzheimer’s disease and dementia. Med Hypotheses. 2012;78:562–570.
  • Shi S, Liang D, Chen Y, et al. Gx-50 reduces β-amyloid-induced TNF-α, IL-1β, NO, and PGE2 expression and inhibits NF-κB signaling in a mouse model of Alzheimer’s disease. Eur J Immunol. 2016;46(3):665–676.
  • Li X, Long J, He T, et al. Integrated genomic approaches identify major pathways and upstream regulators in late onset Alzheimer’s disease. Sci Rep. 2015;5:12393.
  • Arendt T, Holzer M, Stöbe A, et al. Activated mitogenic signaling induces a process of dedifferentiation in Alzheimer’s disease that eventually results in cell death. Ann N Y Acad Sci. 2000;920:249–255.
  • Yang Y, Wang X, Dai W. Human Sgo1 is an excellent target for induction of apoptosis of transformed cells. Cell Cycle. 2006;5:896–901.
  • Bordiuk OL, Smith K, Morin PJ, et al. Cell proliferation and neurogenesis in adult mouse brain. PLoS One. 2014;9:e111453.
  • Wang W, Bu B, Xie M, et al. Neural cell cycle dysregulation and central nervous system diseases. Prog Neurobiol. 2009;89:1–17.
  • Rosenkrantz JL, Carbone L. Investigating somatic aneuploidy in the brain: why we need a new model. Chromosoma. 2017;126:337–350.
  • Kline AD, Krantz ID, Deardorff MA, et al. Cornelia de Lange syndrome and molecular implications of the cohesin complex: abstracts from the 7th biennial scientific and educational symposium 2016. Am J Med Genet A. 2017;173:1172–1185.
  • Kumar R, Corbett MA, Van Bon BW, et al. Increased STAG2 dosage defines a novel cohesinopathy with intellectual disability and behavioral problems. Hum Mol Genet. 2015;24:7171–7181.
  • Nigg EA, Čajánek L, Arquint C. The centrosome duplication cycle in health and disease. FEBS Lett. 2014;588:2366–2372.
  • Bajić VP, Spremo-Potparević B, Zivković L, et al. Is the time dimension of the cell cycle re-entry in AD regulated by centromere cohesion dynamics? Biosci Hypotheses. 2008;1:156–161.
  • Van Dam D, De Deyn PP. Non human primate models for Alzheimer’s disease-related research and drug discovery. Expert Opin Drug Discov. 2017;12(2):187–200.
  • Chikara S, Nagaprashantha LD, Singhal J, et al. Oxidative stress and dietary phytochemicals: role in cancer chemoprevention and treatment. Cancer Lett. 2018;413:122–134.
  • Wilson DW, Nash P, Buttar HS, et al. The role of food antioxidants, benefits of functional foods, and influence of feeding habits on the health of the older person: an overview. Antioxidants (Basel). 2017;6:E81.
  • Rao CV, Pal S, Mohammed A, et al. Biological effects and epidemiological consequences of arsenic exposure, and reagents that can ameliorate arsenic damage in vivo. Oncotarget. 2017;8:57605–57621.
  • O’Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol. 2016;13:417–430.
  • Salmela AL, Kallio MJ. Mitosis as an anti-cancer drug target. Chromosoma. 2013;122:431–449.
  • Rao CV, Kurkjian CD, Yamada HY. Mitosis-targeting natural products for cancer prevention and therapy. Curr Drug Targets. 2012;13:1820–1830.
  • Motwani M, Li X, Schwartz GK. Flavopiridol, a cyclin-dependent kinase inhibitor, prevents spindle inhibitor-induced endoreduplication in human cancer cells. Clin Cancer Res. 2000;6:924–932.
  • Srikumar T, Padmanabhan J. Potential use of flavopiridol in treatment of chronic diseases. Adv Exp Med Biol. 2016;929:209–228.
  • Zeidner JF, Karp JE. Clinical activity of alvocidib (flavopiridol) in acute myeloid leukemia. Leuk Res. 2015;39(12):1312–1318.
  • Jorda EG, Verdaguer E, Canudas AM, et al. Neuroprotective action of flavopiridol, a cyclin-dependent kinase inhibitor, in colchicine-induced apoptosis. Neuropharmacology. 2003;45:672–683.
  • Leggio GM, Catania MV, Puzzo D, et al. The antineoplastic drug flavopiridol reverses memory impairment induced by Amyloid-ß1-42 oligomers in mice. Pharmacol Res. 2016;106:10–20.
  • Kodaira H, Kusuhara H, Ushiki J, et al. Kinetic analysis of the cooperation of P-glycoprotein (P-gp/Abcb1) and breast cancer resistance protein (Bcrp/Abcg2) in limiting the brain and testis penetration of erlotinib, flavopiridol, and mitoxantrone. J Pharmacol Exp Ther. 2010;333:788–796.
  • Wadman M. NIH gambles on recycled drugs. Nature. 2013;499:263–264.
  • National NIH Center for Advancing Translational Science [cited 2018 July 20]. Available from: https://ncats.nih.gov/preclinical/repurpose
  • Kaufman AC, Salazar SV, Haas LT, et al. Fyn inhibition rescues established memory and synapse loss in Alzheimer mice. Ann Neurol. 2015;77:953–971.
  • Folch J, Petrov D, Ettcheto M, et al. Current research therapeutic strategies for Alzheimer’s disease treatment. Neural Plast. 2016;2016:8501693.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.