Rapamycin rescues vascular, metabolic and learning deficits in apolipoprotein E4 transgenic mice with pre-symptomatic Alzheimer's disease

Ai-Ling Lin, Jordan B Jahrling, Wei Zhang, Nicholas DeRosa, Vikas Bakshi, Peter Romero, Veronica Galvan, Arlan Richardson, Ai-Ling Lin, Jordan B Jahrling, Wei Zhang, Nicholas DeRosa, Vikas Bakshi, Peter Romero, Veronica Galvan, Arlan Richardson

Abstract

Apolipoprotein E ɛ4 allele is a common susceptibility gene for late-onset Alzheimer's disease. Brain vascular and metabolic deficits can occur in cognitively normal apolipoprotein E ɛ4 carriers decades before the onset of Alzheimer's disease. The goal of this study was to determine whether early intervention using rapamycin could restore neurovascular and neurometabolic functions, and thus impede pathological progression of Alzheimer's disease-like symptoms in pre-symptomatic Apolipoprotein E ɛ4 transgenic mice. Using in vivo, multimodal neuroimaging, we found that apolipoprotein E ɛ4 mice treated with rapamycin had restored cerebral blood flow, blood-brain barrier integrity and glucose metabolism, compared to age- and gender-matched wild-type controls. The preserved vasculature and metabolism were associated with amelioration of incipient learning deficits. We also found that rapamycin restored the levels of the proinflammatory cyclophilin A in vasculature, which may contribute to the preservation of cerebrovascular function in the apolipoprotein E ɛ4 transgenics. Our results show that rapamycin improves functional outcomes in this mouse model and may have potential as an effective intervention to block progression of vascular, metabolic and early cognitive deficits in human Apolipoprotein E ɛ4 carriers. As rapamycin is FDA-approved and neuroimaging is readily used in humans, the results of the present study may provide the basis for future Alzheimer's disease intervention studies in human subjects.

Keywords: APOE4; Alzheimer’s disease; Brain imaging; blood–brain barrier; cerebral blood flow; cerebral glucose metabolism; cognition; inflammation; rapamycin.

© The Author(s) 2015.

Figures

Figure 1.
Figure 1.
Experimental design and timeline. APOE4 transgenic mice can have vascular defects as early as two weeks (0.5 months of age); metabolic/synaptic dysfunctions at four months of age; memory decline at 12 months of age., We obtained female mice at one month of age to test the hypothesis that restoring vascular functions can further impede the decline of metabolic and cognitive functions. After baseline cerebral blood flow (CBF) was measured, rapamycin diet was continuously supplied for six months. CBF was measured longitudinally using MRI after 1, 3, and 6 months of feeding. At the end-point of the study (i.e. mice at 7 months of age), blood–brain barrier (BBB) integrity was evaluated using MRI and cerebral metabolic rate of glucose (CMRGlc) was measured by PET (N = 6). A separate group of mice (N = 15) underwent behavioral assessment with Morris water maze (MWM). After MWM, mice were sacrificed and brain tissues were used for mechanistic pathway analyses using Western blot.
Figure 2.
Figure 2.
Rapamycin restoring brain vascular functions of APOE4 mice. (a) Representative baseline (pre-treated) CBF images of a WT-control and APOE4-control mice. The color code indicates the level of CBF in a linear scale; (b) comparison of CBF between pre- and one-month-post-treatment of a APOE4 mouse; (c) the time course of the global CBF changes among the three groups; (d) cortical CBF (in ml/g/min) of the mice at seven months of age; (e) hippocampal CBF (in ml/g/min) of the mice at seven months of age; (f) temporal lobe CBF (in ml/g/min) of the mice at seven months of age; (g) BBB leakage (indicated by the arrows) of the mice at seven months of age. Data are presented as mean ± standard error of the mean. *P < 0.05; **P < 0.01; ***P < 0.001;# no difference between WT-control and APOE4-Rapa. n.s.: non-significant; APOE4: apolipoprotein E4.
Figure 3.
Figure 3.
Rapamycin restoring brain metabolic functions of APOE4 mice. (a) CMRGlc maps of mice at seven months of age; the color code indicates the level of CMRGlc in a linear scale. Quantitative CMRGlc in the (b) cortex; (c) hippocampus; and, (d) temporal lobe of the three groups of mice. Data are presented as mean ± standard error of the mean. **P < 0.01; ***P < 0.001; n.s.: non-significant; APOE4: apolipoprotein E4.
Figure 4.
Figure 4.
Rapamycin ameliorates incipient learning phenotypes of APOE4 mice. (a) Time in seconds to reach a hidden platform. Time F (3, 117) = 37.51, P < 0.0001; Treatment F (2, 39) = 0.496, P = 0.613; Interaction F (6, 117) = 5.919, P < 0.0001; (b) total distance swam during trial. Distance F (3, 117) = 30.06, P < 0.0001; treatment F (2, 39) = 0.079, P = 0.024; interaction F (6, 117) = 5.704, P < 0.0001; (c) number of times mice crossed over the platform location in the probe trial. n.s., P = 0.091; (d) average swim speeds during training. Speed F (3, 117) = 0.416, P = 0.742; treatment F (2, 39) = 1.59, P = 0.217; interaction F (6, 117) = 4.986, P < 0.0001; (e) percent of trial spent floating. Time floating F (3, 117) = 0.821, P = 0.485; treatment F (2, 39) = 2.601, P = 0.087; interaction F (6, 117) = 1.19, P = 0.316; (f) percent of trial spent in thigmotaxis. Time in thigmotaxis F (3, 117) = 39.25, P < 0.0001; treatment F (2, 39) = 5.92, P = 0.006; interaction F (6, 117) = 2.212, P = 0.047. Data are presented mean ± standard error of the mean of 4 trials/animal/day. All asterisks (*) indicate a significant difference between WT-control vs. APOE4-control, all pound signs (#) indicate a significant difference between APOE4-control vs. APOE4-Rapa, and all money signs ($) indicate a significant difference between WT-control vs. APOE4-Rapa. Behavioral data were analyzed by two-way ANOVA followed by Tukey’s multiple comparisons test. APOE4: apolipoprotein E4.
Figure 5.
Figure 5.
Rapamycin restoring CypA levels in brain vasculature of APOE4 mice. (a,b) Immunoblots of cortical CypA lysates and the corresponding quantitative analyses; (c,d) immunoblots of microvascular CypA lysates and the corresponding quantitative analyses; (e,f) immunoblots of cortical NF-κB lysates and the corresponding quantitative analyses; (g,h) immunoblots of microvascular NF-κB lysates and the corresponding quantitative analyses. Data are presented as mean ± standard error of the mean. *P < 0.05. CypA: cyclophilin A; NF-κB: nuclear factor-κb.

References

    1. Liu CC, Kanekiyo T, Xu H, et al. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nature Rev Neurol 2013; 9: 106–118.
    1. Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 1993; 261: 921–923.
    1. Thambisetty M, Beason-Held L, An Y, et al. APOE epsilon4 genotype and longitudinal changes in cerebral blood flow in normal aging. Arch Neurol 2010; 67: 93–98.
    1. Reiman EM, Chen K, Alexander GE, et al. Correlations between apolipoprotein E epsilon4 gene dose and brain-imaging measurements of regional hypometabolism. Proc Natl Acad Sci USA 2005; 102: 8299–8302.
    1. Reiman EM, Caselli RJ, Chen K, et al. Declining brain activity in cognitively normal apolipoprotein E epsilon 4 heterozygotes: a foundation for using positron emission tomography to efficiently test treatments to prevent Alzheimer's disease. Proc Natl Acad Sci USA 2001; 98: 3334–3339.
    1. Fleisher AS, Chen K, Liu X, et al. Apolipoprotein E epsilon4 and age effects on florbetapir positron emission tomography in healthy aging and Alzheimer disease. Neurobiol Aging 2013; 34: 1–12.
    1. Reiman EM, Chen K, Alexander GE, et al. Functional brain abnormalities in young adults at genetic risk for late-onset Alzheimer's dementia. Proc Natl Acad Sci USA 2004; 101: 284–289.
    1. Bell RD, Winkler EA, Singh I, et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 2012; 485: 512–516.
    1. Austin BP, Nair VA, Meier TB, et al. Effects of hypoperfusion in Alzheimer's disease. J Alzheimer's Dis 2011; 26(Suppl 3): 123–133.
    1. Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer's disease and other disorders. Nature Rev Neurosci 2011; 12: 723–738.
    1. Alata W, Ye Y, St-Amour I, et al. Human apolipoprotein E varepsilon4 expression impairs cerebral vascularization and blood-brain barrier function in mice. J Cereb Blood Flow Metab 2015; 35: 86–94.
    1. Lin AL, Zheng W, Halloran JJ, et al. Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer's disease. J Cereb Blood Flow Metab 2013; 33: 1412–1421.
    1. Spilman P, Podlutskaya N, Hart MJ, et al. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease. PloS One 2010; 5: e9979.
    1. Mielke MM, Vemuri P, Rocca WA. Clinical epidemiology of Alzheimer's disease: assessing sex and gender differences. Clin Epidemiol 2014; 6: 37–48.
    1. Kilkenny C, Browne W, Cuthill IC, et al. Animal research: reporting in vivo experiments – the ARRIVE guidelines. J Cereb Blood Flow Metab 2011; 31: 991–993.
    1. Zlokovic BV. Cerebrovascular effects of apolipoprotein E: implications for Alzheimer disease. JAMA Neurol 2013; 70: 440–444.
    1. Salomon-Zimri S, Boehm-Cagan A, Liraz O, et al. Hippocampus-related cognitive impairments in Young apoE4 targeted replacement mice. Neurodegener Dis 2014; 13: 86–92.
    1. Yin JX, Turner GH, Lin HJ, et al. Deficits in spatial learning and memory is associated with hippocampal volume loss in aged apolipoprotein E4 mice. J Alzheimer's Dis 2011; 27: 89–98.
    1. Pulliam DA, Deepa SS, Liu Y, et al. Complex IV-deficient Surf1(-/-) mice initiate mitochondrial stress responses. Biochem J 2014; 462: 359–371.
    1. Liu Y, Diaz V, Fernandez E, et al. Rapamycin-induced metabolic defects are reversible in both lean and obese mice. Aging 2014; 6: 742–754.
    1. Galvan V, Gorostiza OF, Banwait S, et al. Reversal of Alzheimer's-like pathology and behavior in human APP transgenic mice by mutation of Asp664. Proc Natl Acad Sci USA 2006; 103: 7130–7135.
    1. Brommelhoff JA, Sultzer DL. Brain structure and function related to depression in Alzheimer's disease: contributions from neuroimaging research. J Alzheimer's Dis 2015; 45: 689–703.
    1. Kehoe EG, McNulty JP, Mullins PG, et al. Advances in MRI biomarkers for the diagnosis of Alzheimer's disease. Biomarker Med 2014; 8: 1151–1169.
    1. Lin AL, Zhang W, Gao X, et al. Caloric restriction increases ketone bodies metabolism and preserves blood flow in aging brain. Neurobiol Aging 2015; 36: 2296–2303.
    1. Eichenbaum H. Hippocampus: cognitive processes and neural representations that underlie declarative memory. Neuron 2004; 44: 109–120.
    1. Halliday MR, Rege SV, Ma Q, et al. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer's disease. J Cereb Blood Flow Metab Epub ahead of print 11 March 2015. DOI: 10.1038/jcbfm.2015.44.
    1. Steiner JP, Connolly MA, Valentine HL, et al. Neurotrophic actions of nonimmunosuppressive analogues of immunosuppressive drugs FK506, rapamycin and cyclosporin A. Nat Med 1997; 3: 421–428.
    1. Fox PT, Raichle ME, Mintun MA, et al. Nonoxidative glucose consumption during focal physiologic neural activity. Science 1988; 241: 462–464.
    1. Lin AL, Gao JH, Duong TQ, et al. Functional neuroimaging: a physiological perspective. Front Neuroenergetics 2010; 2: 1–5.
    1. Lin AL, Fox PT, Hardies J, et al. Nonlinear coupling between cerebral blood flow, oxygen consumption, and ATP production in human visual cortex. Proc Natl Acad Sci USA 2010; 107: 8446–8451.
    1. Lin AL, Coman D, Jiang L, et al. Caloric restriction impedes age-related decline of mitochondrial function and neuronal activity. J Cereb Blood Flow Metab 2014; 34: 1440–1443.
    1. Dash PK, Orsi SA, Moore AN. Spatial memory formation and memory-enhancing effect of glucose involves activation of the tuberous sclerosis complex-Mammalian target of rapamycin pathway. J Neurosci 2006; 26: 8048–8056.
    1. Poels MM, Ikram MA, Vernooij MW, et al. Total cerebral blood flow in relation to cognitive function: the Rotterdam Scan Study. J Cereb Blood Flow Metab 2008; 28: 1652–1655.
    1. Cunnane S, Nugent S, Roy M, et al. Brain fuel metabolism, aging, and Alzheimer's disease. Nutrition 2011; 27: 3–20.
    1. Nagata K, Buchan RJ, Yokoyama E, et al. Misery perfusion with preserved vascular reactivity in Alzheimer's disease. Ann N Y Acad Sci 1997; 826: 272–281.
    1. Nagata K, Kondoh Y, Atchison R, et al. Vascular and metabolic reserve in Alzheimer's disease. Neurobiol Aging 2000; 21: 301–307.
    1. Stranahan AM, Mattson MP. Metabolic reserve as a determinant of cognitive aging. J Alzheimer's Dis 2012; 30(Suppl 2): S5–S13.
    1. Lin AL, Pulliam DA, Deepa SS, et al. Decreased in vitro mitochondrial function is associated with enhanced brain metabolism, blood flow, and memory in Surf1-deficient mice. J Cereb Blood Flow Metab 2013; 33: 1605–1611.
    1. Youmans KL, Tai LM, Nwabuisi-Heath E, et al. APOE4-specific changes in Abeta accumulation in a new transgenic mouse model of Alzheimer disease. J Biol Chem 2012; 287: 41774–41786.
    1. Lin AL, Laird AR, Fox PT, et al. Multimodal MRI neuroimaging biomarkers for cognitive normal adults, amnestic mild cognitive impairment, and Alzheimer's disease. Neurol Res Int 2012; 2012: 907409.
    1. Lin AL, Rothman DL. What have novel imaging techniques revealed about metabolism in the aging brain?. Future Neurol 2014; 9: 341–354.
    1. Uh J, Lin AL, Lee K, et al. Validation of VASO cerebral blood volume measurement with positron emission tomography. Magn Reson Med 2011; 65: 744–749.
    1. Soefje SA, Karnad A, Brenner AJ. Common toxicities of mammalian target of rapamycin inhibitors. Target Oncol 2011; 6: 125–129.
    1. Hurez V, Dao V, Liu A, et al. Chronic mTOR inhibition in mice with rapamycin alters T, B, myeloid, and innate lymphoid cells and gut flora and prolongs life of immune-deficient mice. Aging Cell Epub ahead of print 28 August 2015. DOI: 10.1111/acel.12380.
    1. Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, et al. mTOR inhibition improves immune function in the elderly. Science translational medicine 2014; 6(268): 268ra179.
    1. Alvarado Y, Mita MM, Vemulapalli S, et al. Clinical activity of mammalian target of rapamycin inhibitors in solid tumors. Target Oncol 2011; 6: 69–94.
    1. Mita MM, Mita A, Rowinsky EK. The molecular target of rapamycin (mTOR) as a therapeutic target against cancer. Cancer Biol Therapy 2003; 2(4 Suppl 1): S169–S177.
    1. Richardson A, Galvan V, Lin AL, et al. How longevity research can lead to therapies for Alzheimer's disease: the rapamycin story. Exp Gerontol 2014; 68: 51–58.

Source: PubMed

3
Subscribe