Potential role of orexin and sleep modulation in the pathogenesis of Alzheimer's disease

Jee Hoon Roh, Hong Jiang, Mary Beth Finn, Floy R Stewart, Thomas E Mahan, John R Cirrito, Ashish Heda, B Joy Snider, Mingjie Li, Masashi Yanagisawa, Luis de Lecea, David M Holtzman, Jee Hoon Roh, Hong Jiang, Mary Beth Finn, Floy R Stewart, Thomas E Mahan, John R Cirrito, Ashish Heda, B Joy Snider, Mingjie Li, Masashi Yanagisawa, Luis de Lecea, David M Holtzman

Abstract

Age-related aggregation of amyloid-β (Aβ) is an upstream pathological event in Alzheimer's disease (AD) pathogenesis, and it disrupts the sleep-wake cycle. The amount of sleep declines with aging and to a greater extent in AD. Poor sleep quality and insufficient amounts of sleep have been noted in humans with preclinical evidence of AD. However, how the amount and quality of sleep affects Aβ aggregation is not yet well understood. Orexins (hypocretins) initiate and maintain wakefulness, and loss of orexin-producing neurons causes narcolepsy. We tried to determine whether orexin release or secondary changes in sleep via orexin modulation affect Aβ pathology. Amyloid precursor protein (APP)/Presenilin 1 (PS1) transgenic mice, in which the orexin gene is knocked out, showed a marked decrease in the amount of Aβ pathology in the brain with an increase in sleep time. Focal overexpression of orexin in the hippocampus in APP/PS1 mice did not alter the total amount of sleep/wakefulness and the amount of Aβ pathology. In contrast, sleep deprivation or increasing wakefulness by rescue of orexinergic neurons in APP/PS1 mice lacking orexin increased the amount of Aβ pathology in the brain. Collectively, modulation of orexin and its effects on sleep appear to modulate Aβ pathology in the brain.

© 2014 Roh et al.

Figures

Figure 1.
Figure 1.
Marked reduction of Aβ pathology in the APP/PS1/OR−/− mice compared with APP/PS1 mice. (A–I) The amount of Aβ pathology was noted at 3.5 (A–C) and 8.5 mo (D–F) in APP/PS1-21 mouse line and at 6 mo in APP/PS1δE9 mouse line (G–I). (J and K) Amount of wakefulness at 3 mo of age before the onset of Aβ pathology in the brain was compared between APP/PS1δE9 and APP/PS1δE9/OR−/− mice. Each mouse was investigated independently one time. Data are presented as mean ± SEM (n = 4–12 in each group, two-tailed Student’s t test in C, F, and I and Mann-Whitney test in J). *, P < 0.05; **, P < 0.01; and ***, P < 0.001. Bar, 500 µm.
Figure 2.
Figure 2.
Marked reduction of amyloid pathology in the APP/PS1/OR−/− mice compared with APP/PS1 mice. (A–L) Amyloid pathology measured by Aβ immunoreactivity and number of amyloid plaques after X-34 staining is noted at 3.5 (A, B, G, and H) and 8.5 mo (C, D, I, and J) in the APP/PS1-21 mouse line and at 6 mo in the APP/PS1δE9 mouse line (E, F, K, and L). Each mouse was investigated independently one time. Data are presented as mean ± SEM (n = 5–9 in each group, two-tailed Student’s t test). *, P < 0.05; **, P < 0.01; and ***, P < 0.001. Bar, 100 µm.
Figure 3.
Figure 3.
Strong reduction in Aβ species in APP/PS1 mice lacking orexin. (A–D) The amount of PBS-soluble and guanidine-soluble forms of Aβ40 and Aβ42 were compared in APP/PS1δE9/OR−/− and APP/PS1δE9 mice (left two columns) and in APP/PS1-21/OR−/− and APP/PS1-21 mice (right two columns). Results were obtained from the hippocampus (A and B) and from the cortex (C and D) of each group of mice. Each mouse was investigated independently one time. All samples were measured in triplicate. Data are presented as mean ± SEM (n = 5–9 in each group, two-tailed Student’s t test). *, P < 0.05; **, P < 0.01; and ***, P < 0.001.
Figure 4.
Figure 4.
No changes in Aβ deposition and amount of wakefulness by focal overexpression of orexin. (A–F) Amount of Aβ pathology was compared after focal injection of ubiquitin-driven orexin lentiviral vector versus ubiquitin-driven GFP lentiviral vector in the hippocampus from 3 to 6 mo (A–C) or 5 to 9 mo (D–F) in APP/PS1δE9 mice. (G) Levels of orexin in the hippocampus and CSF were compared after focal injection of orexin or GFP lentiviral vector driven by ubiquitin promoter. (H) The amount of wakefulness in APP/PS1δE9 mice was compared after focal injection of ubiquitin-driven orexin lentiviral vector versus ubiquitin-driven GFP lentiviral vector. Each mouse was investigated independently one time. Data are presented as mean ± SEM (n = 4–5 in each group, two-tailed Student’s t test in C, F, and H; one-way ANOVA, followed by Tukey’s post hoc test in G). *, P < 0.05. Bar, 500 µm.
Figure 5.
Figure 5.
Restoration of diurnal fluctuation of ISF Aβ40 via rescue of orexin expression in APP/PS1/OR−/− mice. (A and B) Diurnal fluctuation of ISF Aβ40 in young APP/PS1δE9 mice was compared with the diurnal fluctuation in the same mice when orexin was knocked out (APP/PS1δE9/OR−/− mice). (C–E) Diurnal fluctuation of ISF Aβ40 in the hippocampus and minutes awake per hour of APP/PS1-21/OR−/− mice treated with hypocretin/orexin promoter–driven lentiviral vector overexpressing orexin in the hypothalamus bilaterally were compared with mice treated with the same lentiviral vector expressing GFP. (F) Expression of orexin by immunostaining in orexinergic neurons in the hypothalamus of APP/PS1-21/OR−/− mice treated with hypocretin/orexin promoter–driven lentiviral vector expressing GFP or orexin (middle and right) compared with wild-type mice (left). Each mouse was investigated independently one time. All ISF Aβ measurements were performed in duplicate. Data are presented as mean ± SEM (n = 6 in each group, two-tailed Student’s t test in A–D and Mann–Whitney test in E). *, P < 0.05; and **, P < 0.01. Bar, 100 µm.
Figure 6.
Figure 6.
Increase in Aβ deposition and wakefulness by rescue of orexin expression in APP/PS1/OR−/− mice via expression of orexin with lentiviral vector driven by hypocretin/orexin promoter in the bilateral hypothalamus. (A–C) Amount of Aβ pathology in the hippocampus and cortex of the APP/PS1-21/OR−/− mice after bilateral injection of hypocretin promoter–driven orexin lentiviral vector in the hypothalamus of the APP/PS1/OR−/− mice was compared with results from injection of the same lentiviral vector overexpressing GFP. (D) The amount of wakefulness as measured by polysomnography was compared between the same groups of mice. Each mouse was investigated independently one time. Data are presented as mean ± SEM (n = 6–7 in each group, two-tailed Student’s t test in C; n = 3 in each group, Mann–Whitney test in D). *, P < 0.05. Bar, 500 µm.
Figure 7.
Figure 7.
Increased Aβ deposition with chronic sleep deprivation in APP/PS1-21/OR−/− mice. (A–F) The amount of Aβ plaques stained with HJ3.4B (A–C) and fibrillar Aβ stained with X-34 (D–F) were compared between APP/PS1-21/OR−/− mice exposed to a large platform and a small platform. Sleep deprivation experiments were performed using a small and large platform in a cage with water on the bottom, where a mouse cannot sleep on the small platform because of its size, whereas they can maintain a normal sleep–wake cycle on the larger platform. Mice exposed to small platforms (n = 3) and to large platforms (n = 3) were analyzed together within a set of experiments. The results are the sum of five repeats in different mice. Each mouse was investigated independently. Data are presented as mean ± SEM (n = 13–14 in each group, two-tailed Student’s t test). *, P < 0.05; and **, P < 0.01. Bars: (A) 500 µm; (D) 100 µm.

References

    1. Bero A.W., Yan P., Roh J.H., Cirrito J.R., Stewart F.R., Raichle M.E., Lee J.M., and Holtzman D.M.. 2011. Neuronal activity regulates the regional vulnerability to amyloid-β deposition. Nat. Neurosci. 14:750–756 10.1038/nn.2801
    1. Carter M.E., Yizhar O., Chikahisa S., Nguyen H., Adamantidis A., Nishino S., Deisseroth K., and de Lecea L.. 2010. Tuning arousal with optogenetic modulation of locus coeruleus neurons. Nat. Neurosci. 13:1526–1533 10.1038/nn.2682
    1. Chemelli R.M., Willie J.T., Sinton C.M., Elmquist J.K., Scammell T., Lee C., Richardson J.A., Williams S.C., Xiong Y., Kisanuki Y., et al. . 1999. Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation. Cell. 98:437–451 10.1016/S0092-8674(00)81973-X
    1. Cirrito J.R., May P.C., O’Dell M.A., Taylor J.W., Parsadanian M., Cramer J.W., Audia J.E., Nissen J.S., Bales K.R., Paul S.M., et al. . 2003. In vivo assessment of brain interstitial fluid with microdialysis reveals plaque-associated changes in amyloid-β metabolism and half-life. J. Neurosci. 23:8844–8853.
    1. Cirrito J.R., Kang J.E., Lee J., Stewart F.R., Verges D.K., Silverio L.M., Bu G., Mennerick S., and Holtzman D.M.. 2008. Endocytosis is required for synaptic activity-dependent release of amyloid-β in vivo. Neuron. 58:42–51 10.1016/j.neuron.2008.02.003
    1. de Lecea L., Kilduff T.S., Peyron C., Gao X., Foye P.E., Danielson P.E., Fukuhara C., Battenberg E.L., Gautvik V.T., Bartlett F.S. II, et al. . 1998. The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc. Natl. Acad. Sci. USA. 95:322–327 10.1073/pnas.95.1.322
    1. Holtzman D.M., Goate A., Kelly J., and Sperling R.. 2011. Mapping the road forward in Alzheimer’s disease. Sci. Transl. Med. 3:114ps48 10.1126/scitranslmed.3003529
    1. Jankowsky J.L., Fadale D.J., Anderson J., Xu G.M., Gonzales V., Jenkins N.A., Copeland N.G., Lee M.K., Younkin L.H., Wagner S.L., et al. . 2004. Mutant presenilins specifically elevate the levels of the 42 residue β-amyloid peptide in vivo: evidence for augmentation of a 42-specific γ secretase. Hum. Mol. Genet. 13:159–170 10.1093/hmg/ddh019
    1. Ju Y.E., and Holtzman D.M.. 2013. Sleep evaluation by actigraphy for patients with Alzheimer disease—reply. JAMA Neurol. 70:1074–1075 10.1001/jamaneurol.2013.3490
    1. Ju Y.E., Lucey B.P., and Holtzman D.M.. 2014. Sleep and Alzheimer disease pathology—a bidirectional relationship. Nat Rev Neurol. 10:115–119 10.1038/nrneurol.2013.269
    1. Kamenetz F., Tomita T., Hsieh H., Seabrook G., Borchelt D., Iwatsubo T., Sisodia S., and Malinow R.. 2003. APP processing and synaptic function. Neuron. 37:925–937 10.1016/S0896-6273(03)00124-7
    1. Kang J.E., Lim M.M., Bateman R.J., Lee J.J., Smyth L.P., Cirrito J.R., Fujiki N., Nishino S., and Holtzman D.M.. 2009. Amyloid-β dynamics are regulated by orexin and the sleep-wake cycle. Science. 326:1005–1007 10.1126/science.1180962
    1. Kim J., Castellano J.M., Jiang H., Basak J.M., Parsadanian M., Pham V., Mason S.M., Paul S.M., and Holtzman D.M.. 2009. Overexpression of low-density lipoprotein receptor in the brain markedly inhibits amyloid deposition and increases extracellular A β clearance. Neuron. 64:632–644 10.1016/j.neuron.2009.11.013
    1. Koenigsknecht-Talboo J., Meyer-Luehmann M., Parsadanian M., Garcia-Alloza M., Finn M.B., Hyman B.T., Bacskai B.J., and Holtzman D.M.. 2008. Rapid microglial response around amyloid pathology after systemic anti-Aβ antibody administration in PDAPP mice. J. Neurosci. 28:14156–14164 10.1523/JNEUROSCI.4147-08.2008
    1. Matsuki T., Nomiyama M., Takahira H., Hirashima N., Kunita S., Takahashi S., Yagami K., Kilduff T.S., Bettler B., Yanagisawa M., and Sakurai T.. 2009. Selective loss of GABAB receptors in orexin-producing neurons results in disrupted sleep/wakefulness architecture. Proc. Natl. Acad. Sci. USA. 106:4459–4464 10.1073/pnas.0811126106
    1. Nir Y., Staba R.J., Andrillon T., Vyazovskiy V.V., Cirelli C., Fried I., and Tononi G.. 2011. Regional slow waves and spindles in human sleep. Neuron. 70:153–169 10.1016/j.neuron.2011.02.043
    1. Ooms S., Overeem S., Besse K., Rikkert M.O., Verbeek M., and Claassen J.A.. 2014. Effect of 1 night of total sleep deprivation on cerebrospinal fluid β-amyloid 42 in healthy middle-aged men: a randomized clinical trial. JAMA Neurol. 71:971–977 10.1001/jamaneurol.2014.1173
    1. Radde R., Bolmont T., Kaeser S.A., Coomaraswamy J., Lindau D., Stoltze L., Calhoun M.E., Jäggi F., Wolburg H., Gengler S., et al. . 2006. Aβ42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 7:940–946 10.1038/sj.embor.7400784
    1. Raichle M.E., MacLeod A.M., Snyder A.Z., Powers W.J., Gusnard D.A., and Shulman G.L.. 2001. A default mode of brain function. Proc. Natl. Acad. Sci. USA. 98:676–682 10.1073/pnas.98.2.676
    1. Roh J.H., Huang Y., Bero A.W., Kasten T., Stewart F.R., Bateman R.J., and Holtzman D.M.. 2012. Disruption of the sleep-wake cycle and diurnal fluctuation of β-amyloid in mice with Alzheimer’s disease pathology. Sci. Transl. Med. 4:150ra122 10.1126/scitranslmed.3004291
    1. Sperling R.A., Aisen P.S., Beckett L.A., Bennett D.A., Craft S., Fagan A.M., Iwatsubo T., Jack C.R. Jr, Kaye J., Montine T.J., et al. . 2011. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 7:280–292 10.1016/j.jalz.2011.03.003
    1. Spira A.P., Gamaldo A.A., An Y., Wu M.N., Simonsick E.M., Bilgel M., Zhou Y., Wong D.F., Ferrucci L., and Resnick S.M.. 2013. Self-reported sleep and β-amyloid deposition in community-dwelling older adults. JAMA Neurol. 70:1537–1543.
    1. Vlassenko A.G., Vaishnavi S.N., Couture L., Sacco D., Shannon B.J., Mach R.H., Morris J.C., Raichle M.E., and Mintun M.A.. 2010. Spatial correlation between brain aerobic glycolysis and amyloid-β (Aβ) deposition. Proc. Natl. Acad. Sci. USA. 107:17763–17767 10.1073/pnas.1010461107
    1. Vyazovskiy V.V., Olcese U., Lazimy Y.M., Faraguna U., Esser S.K., Williams J.C., Cirelli C., and Tononi G.. 2009. Cortical firing and sleep homeostasis. Neuron. 63:865–878 10.1016/j.neuron.2009.08.024
    1. Vyazovskiy V.V., Olcese U., Hanlon E.C., Nir Y., Cirelli C., and Tononi G.. 2011. Local sleep in awake rats. Nature. 472:443–447 10.1038/nature10009
    1. Xie L., Kang H., Xu Q., Chen M.J., Liao Y., Thiyagarajan M., O’Donnell J., Christensen D.J., Nicholson C., Iliff J.J., et al. . 2013. Sleep drives metabolite clearance from the adult brain. Science. 342:373–377 10.1126/science.1241224
    1. Yamada K., Holth J.K., Liao F., Stewart F.R., Mahan T.E., Jiang H., Cirrito J.R., Patel T.K., Hochgräfe K., Mandelkow E.M., and Holtzman D.M.. 2014. Neuronal activity regulates extracellular tau in vivo. J. Exp. Med. 211:387–393 10.1084/jem.20131685
    1. Yan P., Bero A.W., Cirrito J.R., Xiao Q., Hu X., Wang Y., Gonzales E., Holtzman D.M., and Lee J.M.. 2009. Characterizing the appearance and growth of amyloid plaques in APP/PS1 mice. J. Neurosci. 29:10706–10714 10.1523/JNEUROSCI.2637-09.2009

Source: PubMed

3
订阅