You are on page 1of 7

Biochimica et Biophysica Acta 1822 (2012) 650–656

Contents lists available at SciVerse ScienceDirect

Biochimica et Biophysica Acta


journal homepage: www.elsevier.com/locate/bbadis

The senescence accelerated mouse (SAMP8) as a model for oxidative stress and
Alzheimer's disease ☆
John E. Morley a,⁎, Harvey James Armbrecht a, c, Susan A. Farr a, b, Vijaya B. Kumar a, c
a
Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
b
John Cochrane VA Medical Center, St. Louis, MO, USA
c
GRECC (Geriatric Research, Education and Clinical Center), VA Medical Center, St. Louis, MO, USA

a r t i c l e i n f o a b s t r a c t

Article history: The senescence accelerated mouse (SAMP8) is a spontaneous animal model of overproduction of amyloid
Received 2 September 2011 precursor protein (APP) and oxidative damage. It develops early memory disturbances and changes in the
Received in revised form 11 November 2011 blood–brain barrier resulting in decreased efflux of amyloid-β protein from the brain. It has a marked in-
Accepted 12 November 2011
crease in oxidative stress in the brain. Pharmacological treatments that reduce oxidative stress improve
Available online 26 November 2011
memory. Treatments that reduce amyloid-β (antisense to APP and antibodies to amyloid-β) not only im-
Keywords:
prove memory but reduce oxidative stress. Early changes in lipid peroxidative damage favor mitochondrial
Senescence accelerated mouse (SAMP8) dysfunction as being a trigger for amyloid-β overproduction in this genetically susceptible mouse strain.
Oxidative damage This sets in motion a cycle where the increased amyloid-beta further damages mitochondria. We suggest
Alzheimer's disease that this should be termed the Inflammatory-Amyloid Cycle and may well be similar to the mechanisms re-
Blood–brain barrier sponsible for the pathophysiology of Alzheimer's disease. This article is part of a Special Issue entitled: Anti-
Amyloid-beta oxidants and Antioxidant Treatment in Disease.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction An alternative hypothesis, called the mitochondrial cascade hy-


pothesis, was proposed by Swerdlow and Kahn in 2004 [2]. This
There are approximately 35.6 million people world-wide with de- hypothesis suggested that damage to mitochondrial DNA results in
mentia and this number is expected to increase to over 115 million by mitochondrial dysfunction in Alzheimer's disease. The increase in
2050. Alzheimer's disease is the most common neurodegenerative oxidative stress is in turn responsible for activation of the Aβ. This
disorder that produces dementia. Alzheimer's disease results in irre- has now been termed the modified Aβ cascade hypothesis [3,4].
versible loss of cortical neurons, especially in the associative cortex Alzheimer's patients have an increase in mitochondrial DNA muta-
and the hippocampus. The molecular findings in Alzheimer's disease tions [5]. In addition, their hippocampal neurons have a deficiency
consist of amyloid-beta protein (Aβ) deposits in senile plaques, intra- in cytochrome oxidase, the enzymatically active component of com-
cellular neurofibrillary tangles, cholinergic deficits and loss of synap- plex IV [6]. This leads to a decrease in ATP production and an increase
tic processes and dendritic spines. The classical belief is that the in oxidative stress resulting in synaptic dysfunction, apoptosis, and
pathology of Alzheimer's disease is due to overproduction of Aβ. tau hyperphosphorylation. These two hypotheses are contrasted in
This “amyloid hypothesis” suggests that Aβ activates phosphorylation Fig. 1.
of tau through glycogen synthase kinase (GSK) leading to neurofibril- Two basic types of rodent models for Alzheimer's disease exist.
lary tangles, directly causes memory deficits through effects of solu- These consist of the transgenic (over-producing human Amyloid
ble amyloid causing synaptic damage and memory impairment, Precursor Protein and associated secretases, tau and ApoE) mice
results in the amyloid plaques, damages the blood brain barrier trap- and the spontaneous senescence accelerated mouse P8 (SAMP8).
ping Aβ within the brain and produces cell death through apoptosis, There is evidence for increased oxidative damage in the transgenic
excitotoxicity, inflammation and oxidative stress [1]. mice which can be reversed to some extent by antioxidants [7–10].
In these models antioxidants also reduced Aβ and plaque burden
but not APP [11]. However, these models overexpress genes with
mutations seen in early onset Alzheimer's disease, which account
☆ This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treat- for less than 5% of all Alzheimer's disease. A spontaneous mouse
ment in Disease. model where the processes are not dominated by overproduction
⁎ Corresponding author at. Division of Geriatric Medicine, Saint Louis University
School of Medicine, 1402 S. Grand Blvd., M238, St. Louis, MO 63104, USA. Tel.: + 1
of Aβ would appear to be a better model to investigate the two hy-
314 977 8462; fax: + 1 314 771 8575. potheses of causation of Alzheimer's disease. This review will de-
E-mail address: morley@slu.edu (J.E. Morley). scribe such a model, the SAMP8, and also explore in depth the role

0925-4439/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbadis.2011.11.015
J.E. Morley et al. / Biochimica et Biophysica Acta 1822 (2012) 650–656 651

Fig. 1. A. The amyloid-beta peptide cascade hypothesis. In this version all changes in Alzheimer's disease are driven by overproduction of amyloid precursor protein. B. The modified
amyloid hypothesis where alternations in mitochondrial function drive the pathogenesis of Alzheimer's disease.

of oxidative damage in memory function. The SAMP8 mouse is a spe- animal are inappropriate for this purpose [18]. For this reason, we
cific model of memory dysfunction, other SAM mice strains are developed a backcross of SAMP8 to CD-1 and found that memory
models of senescence. deficits were no longer present in the 88% backcross mice. These
mice appear to be more appropriate controls for comparison with
2. The SAMP8 mouse the SAMP8.
Studies using a variety of learning techniques (passive avoidance,
The SAMP8 mouse was one of the accelerated senescence strains one way active avoidance, aversive T-maze, object recognition, lever
that spontaneously developed from breeding pairs of the AKR/J press appetitive task, Greek cross and Morris Water maze) have
series [12]. The fact that the breeding was not pathogen free and demonstrated both learning and retention deficits in the SAMP8
that significant outbreeding occurred, together appear to have mice [19–26]. These memory deficits can be reversed by multiple
been responsible for the development of this unique lineage. The pharmacological agents that modulate the neurotransmitters that
SAMP8 mouse develops early learning and memory deficits (be- appear to play a role in Alzheimer's disease, albeit with a shift in
tween 8 and 10 months) together with other characteristics sim- the dose response curve [27–29].
ilar to those seen in Alzheimer's disease. Other characteristics of Pathologically, the SAMP8 mice have a reduction in neuronal
these mice include an altered circadian rhythm [13], reduced anxi- spine density [30], an increase in astrocyte gliosis, an accumulation
ety behavior [14], immune dysfunction late in their lifespan [15] of periodic acid Schiff-positive granules and vacuoles in the magno-
and reduced lifespan [16]. Female mice have less robust memory cellular reticular formation [31–34]. There is also an early increase
changes than male SAMP8, and for this reason most studies have in lipofusion [35].
been carried out in male SAMP8 mice [17]. Microsatellite DNA SAMP8 mice overproduce a compound similar to amyloid-beta
markers have shown that the AKR strain classically used as a control protein [36–38]. Amyloid plaques occur late in life (~20 months) in
652 J.E. Morley et al. / Biochimica et Biophysica Acta 1822 (2012) 650–656

Table 1 cytosolic reductase that is thought to detoxify oxidative stress. RhoB


Comparison of Alzheimer's disease, SAMP8 mouse and transgenic mice models. was also decreased and is a GTPase involved in the response to stress
Alzheimer's SAMP8 Transgenic and apoptosis. JNK stress-activated protein kinase is a serine-
disease models threonine kinase which regulates c-JUN. It is involved in responses
Overproduction of amyloid-β Yes Yes Yes to stressors such as DNA damage and reactive oxygen species. Its
Amyloid plaques Yes Latea Yes levels were decreased with aging.
Phosphorylated tau Increased Increased In some models In the SAMP8 mice there is an increase with aging in neuronal
Cerebral amyloid angiopathy Yes Yes Yes
nitric oxide synthase [47,70]. This is believed to be responsible for in-
Neuron loss Yes Yes ?
Synaptic dysfunction Yes Yes Yes creased astrogliosis [70]. These changes in the astrocytes are associat-
Dendritic spine loss Yes Marked ? ed with an increase in glial fibrillary acidic protein (FGAP) [71]. In
Gliosis Yes Yes Yes Alzheimer's disease brains there are also high levels of GFAP and as-
Cholinergic deficit Yes Yes Yes trocyte hypertrophy [72].
Learning and memory impaired Yes Yes Yes
When SAMP8 mice age from 6 to 12 months, there is a marked in-
Circadian rhythm disturbances Yes Yes ?
Oxidative damage Yes 4 months 8 months crease in oxidative stress [73]. Both protein carbonyls (an index of
protein oxidation) and thiobarbituric acid reactive substance (a
? = uncertain.
a
Occur at 16 to 18 months. marker of lipid peroxidation) increase. This is accompanied by a de-
cline in the weakly immobilized to strongly immobilized ratio of the
protein-specific spin label Mal-6. Administration of either alpha-
these mice, suggesting that the plaques per se are not involved in the lipoic acid or n-acetyl cysteine for 4 weeks not only improved mem-
pathogenesis of the memory defect [38]. There is an overproduction ory but also reversed the measures of oxidative stress. These findings
of amyloid precursor protein and Aβ as shown with a number of an- support the concept that oxidative stress plays a role in the memory
tibodies [38]. The increase may not be in either Aβ1–40 or 1–42 dysfunction of SAMP8 mice.
though Manich et al. [39] found both these species in hippocampal
granules. Other characteristics of Alzheimer's disease shared by 4. Reversal of oxidative stress in SAMP8 mice
SAMP8 mice are hyperphosphorylation of tau [40], increased alpha
synuclein [41], an increase in presinilin [42], increased oxidative Proteomics has demonstrated increased oxidization and/or ni-
damage [43], decreased choline acetyl transferase activity [44], in- tration of certain proteins in Alzheimer's disease brains [74]. In
creased glutamate [45], altered NMDA function [46] and increased the SAMP8 mouse, proteomics demonstrated increased protein
neuronal nitric oxide synthase [47]. Low testosterone is associated carborylation of α-enolase, LDH-2, α-spectrin and DRP-2 [75].
with Alzheimer's disease and rapid progression of mild cognition im- The oxidative changes in α-enolase and DRP-2 have previously
pairment in males [48,49]. SAMP8 mice have low testosterone and been observed in Alzheimer's brains [76,77]. It is postulated that
testosterone replacement reduces APP [50]. these oxidative changes lead to abnormal brain metabolism and
Further evidence for an increase in APP in the SAMP8 comes from neurochemical changes. When SAMP8 mice were treated with
studies with antibodies and antisense developed to Aβ. These have alpha-lipoic acid we found a decrease in the specific carbonyl con-
been shown to cross the blood brain barrier and improve memory tent of lactate dehydrogenase B, dihydropyimidase-like protein 2
in the SAMP8 [51–53]. Antisense to APP has been shown to enter and α-enolase [78]. In addition, brain levels of neurofilament trip-
into cells, reduce Aβ and enhance memory [54,55]. It also increases let L protein, α-enolase and ubiquitous mitochondrial creatine ki-
egress of Aβ from the brain [56]. nase increased. Restoration of these proteins to their normal
Overall, the available studies in the SAMP8 suggest it might be an condition by alpha-lipoic acid may play a role in the improvement
ideal model to examine the interaction of oxidative damage and APP of memory observed in the SAMP8.
production. Table 1 compares the changes in the SAMP8, transgenic Lipid peroxidation is increased in SAMP8 mice as early as 2 months
mice and Alzheimer's. of age [79]. Lipid composition of the brain membranes appears to play
an important role in memory in the SAMP8 mice [80–82]. The levels
3. Oxidative damage and the SAMP8 of the antioxidant, alpha-tocopheral increase with aging in the
SAMP8 mouse [83]. Butterfield et al. [84] found that oxidation of
Mitochondria are the major generator of cellular energy and also lipids in the SAMP8 could be reversed by the antioxidants, N-Test-
the major source of potentially harmful reactive oxygen species. butyl-alpha-phenylnitrone and vitamin E.
Thus, as mitochondrial dysfunction occurs with aging, it plays a A number of other approaches to reducing oxidative stress in
major role in both the physiological aging process and neurode- SAMP8 mice have had success in improving function in these
generation [57,58]. This concept has been termed the free radical mice. Dietary restriction by 40% decreased superoxide radicals
theory of aging. Multiple studies have shown that oxidative damage and metabolic rate while extending the lifespan [85]. Both an
is a hallmark of Alzheimer's disease [59–63]. Thus, a valid model of onion extract and di-n-propyl-triscelfide decreased lipid hydroper-
Alzheimer's disease needs to demonstrate the presence of oxidative oxide in the hippocampus of SAMP8 mice and improved memory
stress. [86]. Similarly, the antioxidant oolong and green teas reversed
Oxidative damage has been universally observed in the SAMP8 memory deficits, decreased brain lipofusion and increased serum
[16,64]. Nomura et al. [65] were the first to find higher amounts of Trolox equivalent antioxidant activity [87]. Lotus seedpod
malondealdehyde and lower levels of superoxidase dismutase in proanthocyanidins (LSPC) improved memory in the Y-maze test
SAMP8 compared to SAMR1 mice. Other early findings included an [88]. LSPC decreased the elevated levels of malondialdehyde, nitric
increase in thiobarbituric acid reactivity and a decrease in glutathione oxide and nitric oxide synthase and increased the levels
catalyze, and glutamine synthase [66–68]. There was also an increase glutathione, glutathione peroxidase and superoxidase dismutase in
in the H2O2-producing enzyme, acylCoA oxidase as the SAMP8 ages. the brain and serum of SAMP8 mice. Mulberry extracts are rich in
In a microarray study Kumar et al. [69] found 4 genes involved in phenolics and anthocyanins. Feeding mulberry extracts to SAMP8s
the stress response and antioxidant metabolism are altered with for 12 weeks improved memory in avoidance testing, decreased
aging in the SAMP8 mouse. There was a decrease in heme oxygenase1 amyloid-beta protein, increased antioxidant enzyme activity and de-
which forms biliverdin from the heme ring and is associated with creased lipid oxidation [89]. Similarly, the phenolic rich extra virgin
Alzheimer's disease. Quinone oxireductase was increased. It is a olive oil improved memory and decreased oxidative damage in the
J.E. Morley et al. / Biochimica et Biophysica Acta 1822 (2012) 650–656 653

brains of SAMP8 mice [90]. Icariin, another antioxidant, improved in the SAMP8 mice [54]. This antisense also decreased lipid peroxida-
memory in the water maze and the step-down passive avoidance tion and protein oxidation in the brain of the SAMP8 mouse [100].
tests [91]. The antisense also decreased carbonyl levels of aldolase 3, coronin
1a and peroxiredoxin 2 [101]. These findings suggest that reducing
5. Effects of melatonin in SAMP8 mice Aβ can lead to reversal of the oxidative damage in SAMP8 mice.
It is believed that impairment of efflux of Aβ from the brain across
Melatonin is a well recognized powerful antioxidant and anti- the blood–brain barrier is a mechanism by which Aβ accumulates in
inflammatory agent [92]. It has beneficial effects by ameliorating the brains of persons with Alzheimer's disease [102]. Lipoprotein re-
both oxidative and nitrosative stress state. Melatonin has both pro- ceptor protein-1 (LRP-1) is the pump that produces efflux of the Aβ
and anti-inflammatory effects, promoting early inflammatory re- from the brain. In Alzheimer's brains there are elevated levels of the
sponses and attenuating later responses to protect against the chronic lipid peroxidation product, 4-hydroxy-2-nonenal which are aggregat-
effects of inflammation. The effects of melatonin in the SAMP8 mouse ed with LRP-1 [103]. The fact that SAMP8 mice also has a deficit in the
have been studied in detail. There is a marked aging-associated in- efflux of Aβ from the brain, suggests that this may be due to oxidative
crease in oxidative stress and inflammation in the livers of the damage of the blood brain barrier.
SAMP8 mice [93]. Melatonin decreased mRNA expression of TNFα,
IL-1β, iNOS and NKkB 1 and 2 in the liver. The protein levels of TNFα
and IL-1 β were also reduced. Thus, melatonin reduces inflammation 7. Hormesis and amyloid-beta
in SAMP8 mice.
With aging in the brain of the SAMP8 there is a reduction in ATP The law of hormesis suggests that low doses of a substance (toxin
production associated with diminished activity of the respiratory or physiological modulator) may have different effects on a biochem-
chain complexes [94]. Melatonin administration prevented mito- ical process or, more specifically, that low doses stimulate and high
chondrial impairment and maintained ATP production in the SAMP8 doses inhibit the same process. Recently, our group [104] and Puzzo
mice [95]. There is a decrease in the melatonin-1 receptor with et al. [105,106] have found that low doses (picomolar) of Aβ actually
aging in the SAMP8 mouse [96]. Melatonin reduced thiobarbituric enhance long term potentiation, increase hippocampal acetylcholine
acid reactive substances and protein carbonyl levels in the brains of production and enhance memory, while higher doses inhibit these
SAMP8 mice, while increasing glutathione peroxidase levels [97]. processes. Blocking the production of Aβ in young mice or cells has
Melatonin decreased cell loss in cerebral cortex in parallel with the the opposite effect. This suggests that the physiological role of Aβ
reduction in lipid and protein oxidative damage [98]. The tau hyper- may be memory enhancement with overproduction leading to cogni-
phosphorylation seen in SAMP8 mice was also reduced by melatonin. tive dysfunction and Alzheimer's disease.
Melatonin elevated hippocampal pyramidal volume and increased Aβ is a potent antioxidant in free oxidative systems [107]. This is
the numerical density and surface volume of mitochondria [99]. Mel- related to the ability of Aβ to bind to zinc, copper and iron [108].
atonin improved cognitive performance in the Y-maze and in the This led Atwood et al. [109] to suggest that the formation of amyloid
eight arm radial maze [99]. plaques could act to remove reactive oxygen species. They believed
that excessive Aβ accumulation may prevent metal ion chelation
6. Oxidative stress and amyloid-beta protein and result in increasing oxidative stress. Alternatively increased Aβ
may decrease oxidative stress by quenching hydroxyl radicals [110].
A 42mer phosphorothiolated antisense oligonucleotide directed No studies examining the effects of picomolar concentrations of Aβ
against the Aβ region of amyloid precursor protein improves memory on free radical generation have been undertaken.

Fig. 2. The neurotransmitter sequence of the effects of increased amyloid-beta is based on previous work by our group [1,16].
654 J.E. Morley et al. / Biochimica et Biophysica Acta 1822 (2012) 650–656

8. Conclusion [16] J.E. Morley, The SAMP8 mouse: a model of Alzheimer disease? Biogerontology 3
(2002) 57–60.
[17] J.F. Flood, S.A. Farr, F.E. Kaiser, J.E. Morley, Age-related impairment in learning but
Oxidative damage to neuronal cells is believed to play a central not memory in SAMP8 female mice, Pharmacol. Biochem. Behav. 50 (1995)
role in the pathogenesis of Alzheimer's disease. There is no ideal ani- 661–664.
[18] C. Xia, K. Higuchi, M. Shimizu, T. Matsushita, K. Kogishi, J. Wang, T. Chiba, M.F.
mal model to study Alzheimer's disease. However, the SAMP8 mouse Festing, M. Hosokawa, Genetic typing of the senescence-accelerated mouse
appears to be an excellent model to study the interactions between (SAM) strains with microsatellite markers, Mamm. Genome 10 (1999) 235–238.
Aβ overproduction and oxidative damage to brain tissue. [19] H. Yagi, S. Katoh, I. Akiguchi, T. Takeda, Age-related deterioration of ability of
acquisition in memory and learning in senescence-accelerated mouse; SAM-P/8
Fig. 2 provides a schematic of the alterations in the SAMP8 brain as an animal model of disturbances in recent memory, Brain Res. 474 (1988) 86–93.
that lead to memory failure and tissue damage. At present, there is in- [20] J.F. Flood, J.E. Morley, Learning and memory in the SAMP8 mouse, Neurosci.
adequate information to answer the chicken or egg question of Biobehav. Rev. 22 (1998) 1–20.
[21] M. Miyamoto, Y. Kiyota, M. Nishiyama, A. Nagaoka, Senescence-accelerated
whether Aβ drives increased mitochondrial dysfunction and oxida-
mouse (SAM): age-related reduced anxiety-like behavior in the SAM-P/8 strain,
tive damage or whether mitochondrial dysfunction leads to inflam- Physiol. Behav. 51 (1994) 979–985.
mation resulting in excess Aβ production. The fact that blocking Aβ [22] J.F. Flood, J.E. Morley, Age-related changes in footshock avoidance acquisition
production with an antisense improves memory and decreases oxida- and retention in senescence accelerated mouse (SAM), Neurobiol. Aging 14
(1993) 153–157.
tive damage would suggest that Aβ is the primary culprit. However, [23] J.F. Flood, J.E. Morley, Early onset of age-related impairment of aversive and ap-
decreasing oxidative damage with a variety of agents improves mem- petitive learning in the SAM-P/8 mouse, J. Gerontol. 47 (1994) 852–859.
ory and decreases brain damage suggesting the opposite conclusion. [24] M. Miyamoto, Characteristics of memory and behavioral disorders in SAMP8
mice, in: T. Takeda (Ed.), The SAM Model of Senescence, Elsevier Science B.V,
Thus, we suggest that either overproduction of Aβ or alternatively Amsterdam, 1994, pp. 61–66.
mitochondrial dysfunction or brain injury resulting in inflammation [25] G.H. Chen, C. Wang, H.Y. Yangchen, R.Y. Liu, J.N. Zhou, Age-related changes in
can trigger what then becomes a vicious cycle. The early lipid perox- anxiety are task-specific in the senescence-accelerated prone mouse 8, Physiol.
Behav. 91 (2007) 644–651.
idative damage seen in SAMP8 mice would favor mitochondrial [26] A. Markowska, E.L. Spangler, D.K. Ingram, Behavioral assessment senescence-
dysfunction as being the trigger for the amyloid-beta overproduction accelerated mouse (SAMP8 and R1), Physiol. Behav. 64 (1998) 15–26.
in a genetically susceptible mouse strain. We suggest this should be [27] J.F. Flood, J.E. Morley, M. La Reginna, Age-related changes in the pharmacological
improvement of retention in senescence accelerated mouse (SAM), Neurobiol.
termed the Inflammatory-Amyloid Cycle. Finally, we believe that low Aging 14 (1993) 159–166.
dose Aβ is physiologically important in maintaining memory and may, [28] J.F. Flood, S.A. Farr, K. Uezu, J.E. Morley, Age-related changes in septal serotonergic,
in fact, actually protect against oxidative damage to neurons. GABAergic and glutamatergic facilitation of retention in SAMP8 mice, Mech. Ageing
Dev. 105 (1998) 173–188.
[29] J.F. Flood, F.J. Harris, J.E. Morley, Age-related changes in hippocampal drug facilita-
References tion of memory processing in SAMP8 mice, Neurobiol. Aging 17 (1996) 15–24.
[30] H. Sugiyama, H. Akiyama, I. Akiguchi, M. Kameyama, T. Takeda, Loss of dendritic
[1] J.E. Morley, Alzheimer's disease: future treatments, J. Am. Med. Dir. Assoc. 12 spines in hippocampal CA1 pyramidal cells in senescence accelerated mouse
(2011) 1–7. (SAM)—a quantitative Golgi study, Clin. Neurol. 27 (1987) 841–845.
[2] R.H. Swerdlow, S.M. Khan, A “mitochondrial cascade hypothesis” for sporadic [31] H. Akiyama, M. Kameyama, I. Akiguchi, H. Sugiyama, T. Kawamata, H. Fukuyama,
Alzheimer's disease, Med. Hypotheses 63 (2004) 8–20. H. Kimura, M. Matsushita, T. Takeda, Periodic acid-Schiff (PAS)-positive granular
[3] T.E. Golde, D. Dickson, M. Hutton, Filling the gaps in the abeta cascade hypothesis structures increase in the brain of senescence accelerated mouse (SAM), Acta
of Alzheimer's disease, Curr. Alzheimer Res. 3 (2006) 421–430. Neuropathol. 72 (1986) 124–129.
[4] P.I. Moreira, M.S. Santos, C.R. Oliveira, J.C. Shenk, A. Nunomura, M.A. Smith, X. [32] S. Garcia-Matas, J. Gutierrez-Cuesta, A. Coto-Montes, R. Rubio-Acero, C. Diez-
Zhu, G. Perry, Alzheimer disease and the role of free radicals in the pathogenesis Vives, A. Camius, M. Pallas, C. Sanfeliu, R. Critofol, Dysfunction of astrocytes in
of the disease, CNS Neurol. Disord. Drug Targets 7 (2008) 3–10. senescence-accelerated mice SAMP8 reduces their neuroprotective capacity,
[5] P.E. Coskun, M.F. Beal, D.C. Wallace, Alzheimer's brains harbor somatic mtDNA Aging Cell 7 (2008) 630–640.
control-region mutations that suppress mitochondrial transcription and replica- [33] Y. Wu, A.Q. Zhang, D.T. Yew, Age related changes of various markers of astro-
tion, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) 10726–10731. cytes in senescence-accelerated mice hippocampus, Neurochem. Int. 46 (2005)
[6] D.A. Cottrell, E.L. Blakely, M.A. Johnson, P.G. Ince, D.M. Turnbull, Mitochondrial 565–574.
enzyme-deficient hippocampal neurons and choroidal cells in AD, Neurology [34] M. Jucker, L.C. Walker, P. Schwarb, J. Hengemihle, H. Kuo, A.D. Snow, F. Bamert,
57 (2001) 260–264. D.K. Ingram, Age-related deposition of glia-associated fibrillar material in brains
[7] J. Yao, H. Du, S. Yan, F. Fang, C. Wang, L.F. Lue, L. Guo, D. Chen, D.M. Stern, F.J. Gunn of C57BL/6 mice, Neuroscience 60 (1994) 875–889.
Moore, J. Xi Chen, O. Arancio, S.S. Yan, Inhibition of amyloid-beta (Abeta) peptide- [35] J.F. Flood, P.M. Morley, J.E. Morley, Age-related changes in learning, memory,
binding alcohol dehydrogenase-Abeta interaction reduces Abeta accumulatin and and lipofuscin as a function of the percentage of SAMP8 genes, Physiol. Behav.
improves mitochondrial function in a mouse model of Alzheimer's disease, 58 (1995) 819–822.
J. Neurosci. 31 (2011) 2313–2320. [36] M. Takemura, S. Nakamura, I. Akiguchi, M. Ueno, N. Oka, S. Ishikawa, A. Shimada, J.
[8] H. Du, L. Guo, S. Yan, A.A. Sosunov, A.A. Sosunov, G.M. McKhann, S.S. Yan, Early Kimura, T. Takeda, Beta/A4 protein-like immunoreactive granular structures in the
deficits in synaptic mitochondria in an Alzheimer's disease mouse model, Proc. brain of senescence-accelerated mouse, Am. J. Pathol. 142 (1993) 1887–1897.
Natl. Acad. Sci. U. S. A. 107 (2010) 18670–18675. [37] A. Fukunari, A. Kato, Y. Sasaki, T. Yoshimoto, S. Ishiura, K. Suzuki, T. Nakajima,
[9] D.H. Hyun, M.R. Mughal, H. Yang, J.H. Lee, E.J. Ko, N.D. Hunt, R. de Cabo, M.P. Colocalization of prolyl endopeptidase and amyloid β-peptide in brains of
Mattson, The plasma membrane redox system is impaired by amyloid β- senescence-accelerated mouse, Neurosci. Lett. 176 (1994) 201–204.
peptide and in the hippocampus and cerebral cortex of 3xTgAD, Exp. Neurol. [38] J.E. Morley, V.B. Kumar, A.E. Bernardo, S.A. Farr, K. Uezu, N. Tamosa, J.F. Flood,
225 (2010) 423–429. Beta-amyloid precursor polypeptide in SAMP8 mice affects learning and memory,
[10] R.A. Robinson, M.B. Lange, R. Sultana, V. Galvan, J. Fombonne, O. Gorostiza, J. Peptides 21 (2000) 1761–1767.
Zhang, G. Sarrier, J. Cai, W.M. Pierce, D.E. Bredesen, D.A. Butterfield, Differential [39] G. Manich, C. Mercader, J. Del Valle, J. Duran-Vilaregut, A. Camins, M. Pallas, J.
expression and redox proteomics analyses of an Alzheimer disease transgenic Vilaplana, C. Pelegri, Characterization of amyloid-β granules in the hippocampus
mouse model: effects of the amyloid-β peptide of amyloid precursor protein of SAMP8 mice, J. Alzheimers Dis. 25 (2011) 535–546.
(E), Neuroscience 177 (2011) 207–222. [40] A.M. Canudas, J. Gutierrez-Cuesta, M.I. Rodriguez, D. Acuna-Castroviejo, F.X. Sureda,
[11] E. Hamel, N. Nicolakakis, T. Aboulkassim, B. Ongali, X.K. Tong, Oxidative stress A. Camins, M. Pallas, Hyperphosphorylation of microtubule-associated protein
and cerebrovascular dysfunction in mouse models of Alzheimer's disease, Exp. tau in senescence-accelerated mouse (SAM), Mech. Ageing Dev. 126 (2005)
Physiol. 93 (2008) 116–120. 1300–1304.
[12] T. Takeda, M. Josokawa, S. Takeshita, M. Irino, M. Higuchi, T. Matsushita, Y. [41] B. Caballero, I. Vega-Naredo, V. Sierra, C. Huidobro-Fernandez, C. Soria-Valles,
Tomita, K. Yasushira, H. Hamamoto, K. Shimizu, M. Ishii, T. Yamamuro, A new D. De Gonzalo-Calvo, D. Tolivia, J. Gutierrez-Cuesta, M. Pallas, A. Camins, M.J.
murine model of accelerated senescence, Mech. Ageing Dev. 17 (1981) 183–194. Rodriguez-Colunga, A. Coto-Montes, Favorable effects of a prolonged treatment
[13] M. Nishiyama, H. Takahashi, M. Miyamato, Senescence-accelerated mouse with melatonin on the level of oxidative damage and neurodegeneration in
(SAMP8): an animal model for age-related circadian rhythm disorder, in: T. senescence-accelerated mice, J. Pineal. Res. 45 (2008) 302–311.
Takeda (Ed.), The SAM Model of Senescence, Elsevier Science BV, Amsterdam, [42] V.B. Kumar, M. Franko, W.A. Banks, P. Kasinadhuni, S.A. Farr, K. Vyas, V. Choudhuri,
1994, pp. 393–396. J.E. Morley, Increase in presenilin 1 (PS1) levels in senescence-accelerated mice
[14] M. Miyamoto, Characteristics of age-related behavioral changes in senescence- (SAMP8) may indirectly impair memory by affecting amyloid precursor protein
accelerated mouse SAMP8 and SAMP10, Exp. Gerontol. 32 (1997) 139–148. (APP) processing, J. Exp. Biol. 212 (Pt 4) (2009) 494–498.
[15] D.C. Powers, J.E. Morley, J.F. Flood, Age-related changes in LFA-1 expression, cell [43] D.A. Butterfield, H.F. Poon, The senescence-accelerated prone mouse (SAMP8): a
adhesion, and PHA-induced proliferation by lymphocytes from senescence- model of age-related cognitive decline with relevance to alterations of the gene
accelerated mouse (SAM)-P/8 and AMP-R/1 substrains, Cell. Immunol. 141 expression and protein abnormalities in Alzheimer's disease, Exp. Gerontol. 40
(1992) 444–456. (2005) 774–783.
J.E. Morley et al. / Biochimica et Biophysica Acta 1822 (2012) 650–656 655

[44] R. Strong, V. Reddy, J.E. Morley, Cholinergic deficits in the septal–hippocampal [69] V.B. Kumar, M.W. Franko, S.A. Farr, J.H. Armbrecht, J.E. Morley, Identification of age-
pathway of the SAM-P/8 senescence accelerated mouse, Brain Res. 966 (2003) dependent changes in expression of senescence-accelerated mouse (SAMP8) hip-
150–156. pocampal proteins by expression array analysis, Biochem. Biophy. Res. Commun.
[45] Y. Kitamura, X.H. Zhao, T. Ohnuki, M. Takei, Y. Nomura, Age-related changes in 272 (2000) 657–661.
transmitter glutamate and NMDA receptor/channels in the brain of [70] S. Han, J.A. Rudd, Z.Y. Hu, L. Zhang, D.T. Yew, M. Fang, Analysis of neuronal nitric
senescence-accelerated mouse, Neurosci. Lett. 137 (1992) 169–172. oxide synthase expression and increasing astrogliosis in the brain of senescence-
[46] Y. Nomura, Y. Kitamura, T. Ohnuki, T. Arima, Y. Yamanaka, K. Sasaki, Y. Oomura, Al- accelerated-prone 8 mice, Int. J. Neurosci. 120 (2010) 602–608.
terations in aceylcholine, NMDA, benzodiazepine receptors and protein kinase C in [71] Y. Wu, A.Q. Zhang, D.T. Yew, Age related changes of various markers of astrocytes I
the brain of the senescence-accelerated mouse: an animal model useful for studies senescence-accelerated mice hippocampus, Neurochem. Int. 46 (2005) 565–574.
on cognitive enhancers, Behav. Brain Res. 83 (1997) 51–55. [72] R.G. Nagele, J. Wegiel, V. Venkataraman, H. Imaki, K.C. Wang, J. Wegiel, Contribution
[47] A.K. Ali, W.A. Banks, V.B. Kumar, Gn Shah, J.L. Lynch, S.A. Farr, M.A. Fleegal- of glial cells to the development of amyloid plaques in Alzheimer's disease,
DeMotta, J.E. Morley, Nitric oxide activity and isoenzyme expression in the Neurobiol. Aging 25 (2004) 663–674.
senescence-accelerated mouse p8 model of Alzheimer's disease: effects of [73] S.A. Farr, H.F. Poon, D. Dogrukol-Ak, J. Drake, W.A. Banks, E. Eyerman, D.A. Butterfield,
anti-amyloid antibody and antisense treatments, J. Gerontol. A Biol. Sci. Med. J.E. Morley, The antioxidants alpha-lipoic acid and N-acetylcysteine reverse memory
Sci. 64 (2009) 1025–1030. impairment and brain oxidative stress in aged SAMP8 mice, J. Neurochem. 84 (2003)
[48] L.W. Chu, S. Tam, P.W. Lee, R.L. Wong, P.Y. Yik, W. Tsui, Y. Song, B.M. Cheung, J.E. 1173–1183.
Morley, K.S. Lam, Bioavailable testosterone is associated with a reduced risk of [74] D.A. Butterfield, T. Reed, R. Sultana, Roles of 3-nitrotyrosine- and 4-hydroxynonenal-
amnestic mild cognitive impairment in older men, Clin. Endocrinol. 68 (2008) modified brain proteins in the progression and pathogenesis of Alzheimer's disease,
589–598. Free. Radic. Res. 45 (2011) 59–72.
[49] L.W. Chu, S. Tam, R.L. Wong, P.Y. Yik, Y. Song, B.M. Cheung, J.E. Morley, K.S. Lam, [75] H.F. Poon, A. Castegna, S.A. Farr, V. Thongboonkerd, B.C. Lynnn, W.A. Banks, J.E.
Bioavailable testosterone predicts a lower risk of Alzheimer's disease in older Morley, J.B. Klein, D.A. Butterfield, Quantitative proteomics analysis of specific
men, J. Alzheimers Dis. (Aug 6 2010) [Epub ahead of print]. protein expression and oxidative modification in aged senescence-accelerated-
[50] J.F. Flood, S.A. Farr, F.E. Kaiser, M. La Regina, J.E. Morley, Age-related decrease of prone 8 mice brain, Neuroscience 126 (2004) 915–926.
plasma testosterone in SAMP8 mice: replacement improves age-related impairment [76] A. Castegna, M. Aksenov, V. Thongboonkerd, J.B. Klein, W.M. Pierce, R.
of learning and memory, Physiol. Behavior 57 (1995) 669–673. Booze, W.R. Markesbery, D.A. Butterfield, Proteomic identification of oxidatively
[51] J.E. Morley, S.A. Farr, J.F. Flood, Antibody to amyloid beta protein alleviates modified proteins in Alzheimer's disease brain. Part II: dihydropyrimidinase-
impaired acquisition, retention, and memory processing in SAMP8 mice, Neurobiol. related protein 2, alpha-enolase and heat shock cognate 71, J. Neurochem. 82
Learn Med. 78 (2002) 125–138. (2002) 1524–1532.
[52] W.A. Banks, S.A. Farr, J.E. Morley, K.M. Wolf, V. Geylis, M. Steinitz, Anti-amyloid [77] D.A. Butterfield, M.L. Lange, Multifunctional roles of enolase in Alzheimer's disease:
beta protein antibody passage across the blood–brain barrier in the SAMP8 beyond altered glucose metabolism, J. Neurochem. 111 (2009) 915–933.
mouse model of Alzheimer's disease: an age-related selective uptake with re- [78] H.F. Poon, S.A. Farr, V. Thongboonkerd, B.C. Lynn, W.A. Banks, J.E. Morley, J.B. Klein,
versal of learning impairment, Exp. Neurol. 206 (2007) 248–256. D.A. Butterfield, Proteomic analysis of specific brain proteins in aged SAMP8 mice
[53] Y. Zhang, J.S. He, J. Wang, F.X. Bao, S.Y. Pang, F. Yin, H.G. Hu, X.L. Pong, W.M. Sun, treated with alpha-lipoic acid: implications for aging and age-related neurodegen-
Y.P. Zheng, L.L. Hou, T. Hong, Administration of amyloid-β42 oligomer-specific erative disorders, Neurchem. Int. 46 (2005) 159–168.
monoclonal antibody improved memory performance in SAMP8 mice, J. Alzheimers [79] N. Nishiyama, Y. Zhou, H. Saito, Ameliorative effects of chronic treatment using
Dis. 23 (2011) 551–561. DX-9386, a traditional Chinese prescription, on learning performance and lipid
[54] V.B. Kumar, S.A. Farr, J.F. Flood, V. Kamlesh, M. Franko, W.A. Banks, J.E. Morley, peroxide content in senescence accelerated mouse, Biol. Pharm. Bull. 17 (1994)
Site-directed antisense oligonucleotide decreases the expression of amyloid 1481–1484.
precursor protein and reverses deficits in learning and memory in aged [80] J.E. Morley, S.A. Farr, V.B. Kumar, W.A. Banks, Alzheimer's disease through the eye
SAMP8 mice, Peptides 21 (2000) 1769–1775. of a mouse. Acceptance lecture for the 2001 Gayle A. Olson and Richard D. Olson
[55] W.A. Banks, S.A. Farr, W. Butt, V.B. Kumar, M.W. Franko, J.E. Morley, Delivery prize, Peptides 23 (2002) 589–599.
across the blood–brain barrier of antisense directed against amyloid beta: rever- [81] M.C. Hung, K. Shibasaki, R. Yoshida, M. Sato, K. Imaizumi, Learning behaviour and
sal of learning and memory deficits in mice overexpressing amyloid precursor cerebral protein kinase C, antioxidant status, lipid composition in senescence-
protein, J. Pharmacol. Exp. Ther. 297 (2001) 1113–1121. accelerated mouse: influence of a phosphatidylcholine-vitamin B12 diet, Br. J.
[56] W.A. Banks, V.B. Kimar, S.A. Farr, R. Nakaoke, J.M. Robinson, J.E. Morley, Impair- Nutr. 86 (2001) 163–171.
ments in brain-to-blood transport of amyloid-β and reabsorption of cerebrospinal [82] A.L. Petursdottir, S.A. Farr, J.E. Morley, W.A. Banks, G.V. Skuladottir, Effect of
fluid in an animal model of Alzheimer's disease are reversed by antisense directed dietary n−3 polyunsaturated fatty acids on brain lipid fatty acid composition,
against amyloid-β protein precursor, J. Alzheimers Dis. 23 (2011) 599–605. learning ability, and memory of senescence-accelerated mouse, J. Gerontol. A
[57] A.M. Pickrell, C.T. Moraes, What role does mitochondrial stress play in neurode- Biol. Sci. Med. Sci. 63 (2008) 1153–1160.
generative diseases? Methods Mol. Biol. 648 (2010) 63–78. [83] A.L. Petursdottir, S.A. Farr, J.E. Morley, W.A. Banks, G.V. Skuladottir, Lipid perox-
[58] C. Rodolfo, F. Ciccosanti, G.D. Giacomo, M. Piacentini, G.M. Fimia, Proteomic idation in brain during aging in the senescence-accelerated mouse (SAM),
analysis of mitochondrial dysfunction in neurodegenerative diseases, Expert Neurobiol. Aging 28 (2007) 1170–1178.
Rev. Proteomics 7 (2010) 519–542. [84] D.A. Butterfield, T. Koppal, B. Howard, R. Subramaniam, N. Hall, K. Hensley, S.
[59] R. Sultana, D.A. Butterfield, Oxidatively modified, mitochondria-relevant brain Yatin, K. Allen, M. Aksenov, M. Aksenova, J. Carney, Structural and functional
proteins in subjects with Alzheimer disease and mild cognitive impairment, changes in proteins induced by free radical-mediated oxidative stress and
J. Bioenerg. Biomembr. 41 (2009) 441–446. protective action of the antioxidants N-tert-butyl-alpha-phenylnitrone and
[60] H.H. Palacios, B.B. Yendluri, K. Parvathaneni, V.B. Shadlinski, M.E. Obrenovich, J. vitamin E, Ann. N. Y. Acad. Sci. 854 (1998) 448–462.
Leszek, D. Gokhman, K. Gasiorowski, V. Bragin, G. Aliev, Mitochondrion-specific [85] J.H. Choi, D. Kim, Effects of age and dietary restriction on lifespan and oxidative
antioxidants as drug treatments for Alzheimer disease, CNS Neurol. Disord. Drug stress of SAMP8 mice with learning and memory impairments, J. Nutr. Health
Targets 10 (2011) 149–162. Aging 4 (2000) 182–186.
[61] R. Sultana, M. Perluigi, D.A. Butterfield, Redox proteomics identification of oxi- [86] H. Nishimura, O. Higuchi, K. Tateshita, K. Tomobe, Y. Okuma Nomura, Antioxidative
datively modified proteins in Alzheimer's disease brain and invivo and in vitro activity and ameliorative effects of memory impairment of sulfur-containing com-
models of AD centered around Abeta (1–42), J. Chromatogr. B Analyt. Technol. pounds in Allium species, Biofactors 26 (2006) 135–146.
Biomed. Life Sci. 833 (2006) 3–11. [87] Y.C. Chan, K. Hosoda, C.J. Tsai, S. Yamamoto, M.F. Wang, Favorable effects of tea on
[62] D.A. Butterfield, Amyloid beta-peptide (1–42)-induced oxidative stress and reducing the cognitive deficits and brain morphological changes in senescence-
neurotoxicity: implications for neurodegeneration in Alzheimer's disease accelerated mice, J. Nutr. Sci. Vitaminol. (Tokyo) 52 (2006) 266–273.
brain. A review, Free. Radic. Res. 36 (2002) 1307–1313. [88] Y. Gong, L. Liu, B. Xie, Y. Liao, E. Yang, Z. Sun, Ameliorative effects of lotus seed-
[63] W.E. Muller, A. Eckert, C. Kurz, G.P. Eckert, K. Leuner, Mitochondrial dysfunction: pod proanthocyanidins on cognitive deficits and oxidative damage in
common final pathway in brain aging and Alzheimer's disease—therapeutic aspects, senescence-accelerated mice, Behav. Brain Res. 194 (2008) 100–107.
Mol. Neurobiol. 41 (2010) 159–171. [89] P.H. Shih, Y.C. Chan, J.W. Liao, M.F. Wang, G.C. Yen, Antioxidant and cognitive
[64] M. Pallas, A. Camins, M.A. Smith, G. Perry, H.G. Lee, G. Casadesis, From aging to promotion effects of anthocyanin-rich mulberry (Morus atropurpurea L.) on
Alzheimer's disease: unveiling “the switch” with the senescence-accelerated senescence-accelerated mice and prevention of Alzheimer's disease, J. Nutr.
mouse model (SAMP8), J. Alzheimers Dis. 15 (2008) 615–624. Biochem. 21 (2010) 598–605.
[65] Y. Nomura, B.X. Wang, S.B. Qi, T. Namba, S. Kaneko, Biochemical changes re- [90] S.A. Farr, T.O. Price, L.J. Dominguez, A. Motisi, F. Saiano, M.L. Niehoff, J.E. Morley,
lated to aging in the senescence-accelerated mouse, Exp. Gerontol. 24 W.A. Banks, N. Ercal, M. Barbagolo, Extra virgin olive oil improves learning and
(1989) 49–55. memory in SAMP8 mice, J Alzheimers Dis. 28 (2012) 81–92.
[66] J. Liu, A. Mori, Age-associated changes in superoxide dismutase activity, thrio- [91] X.L. He, W.Q. Zhou, M.G. Bi, G.H. Du, Neuroprotective effects of icariin on memory
barbituric acid reactivity and reduced glutathione level in the brain and liver impairment and neurochemical deficits in senescence-accelerated mouse pron
in senescence accelerated mice (SAM): a comparison with DDY mice, Mech. 8 (SAMP8) mice, Brain Res. 1334 (2010) 73–83.
Ageing Dev. 71 (1993) 23–30. [92] D. Acuna Castroviejo, L.C. Lopez, G. Escames, A. Lopez, J.A. Garcia, R.J. Reiter,
[67] E. Sato, N. Oda, N. Ozaki, S. Hashimoto, T. Kurokawa, S. Ishibashi, Early and transient Melatonin–mitochondria interplay in health and disease, Curr. Top. Med.
increase in oxidative stress in the cerebral cortex of senescence-accelerated mouse, Chem. 11 (2011) 221–240.
Mech. Ageing Dev. 86 (1996) 105–114. [93] S. Cuesta, R. Kireev, K. Forman, C. Garcia, G. Escames, C. Ariznavarreta, E. Vara,
[68] E. Sato, T. Kurokawea, N. Oda, S. Ishibashi, Early appearance of abnormality of J.A. Tresguerres, Melatonin improves inflammation processes in liver of
microperoxisomal enzymes in the cerebral cortex of senescence-accelerated senescence-accelerated prone male mice (SAMP8), Exp. Gerontol. 45 (2010)
mouse, Mech. Ageing Dev. 92 (1996) 175–184. 950–956.
656 J.E. Morley et al. / Biochimica et Biophysica Acta 1822 (2012) 650–656

[94] M. Carretero, G. Escames, L.C. Lopez, C. Venegas, J.C. Dayoub, L. Garcia, D. Acuna- [102] R. Deane, B.V. Ziokovic, Role of the blood–brain barrier in the pathogenesis of
Castroviego, Long-term melatonin administration protects brain mitochondria Alzheimer's disease, Curr. Alzheimer Res. 4 (2007) 191–197.
from aging, J. Pineal. Res. 47 (2009) 192–200. [103] J.B. Owen, R. Sultana, C.D. Aluise, M.A. Erickson, T.O. Price, G. Bu, W.A. Banks, D.A.
[95] M.I. Rodriguez, G. Escames, L.C. Lopez, A. Lopez, J.A. Garcia, F. Ortiz, V. Sanchez, M. Butterfield, Oxidative modification to LDL receptor-related protein 1 in hippo-
Romeu, O. Acuna-Castroviejo, Improved mitochondrial function and increased campus from subjects with Alzheimer disease: implications for Aβ accumulation
life span after chronic melatonin treatment in senescent prone mice, Exp. Gerontol. in AD brain, Free Radic. Biol. Med. 49 (2010) 1798–1803.
43 (2006) 749–756. [104] J.E. Morley, S.A. Farr, W.A. Banks, S.N. Johnson, K.A. Yamada, L. Xu, A physiological
[96] B. Caballero, I. Vega-Naredo, V. Sierra, C. Huidobro-Fernandez, C. Soria-Valles, D. role for amyloid-beta protein enhancement of learning and memory, J. Alzheimers
DeGonzalo-Calvo, D. Tolivia, J. Gutierrez-Cuesta, M. Pallas, A. Camins, M.J. Dis. 19 (2010) 441–449.
Rodriguez-Colunga, A. Coto-Montes, Favorable effects of a prolonged treatment [105] D. Puzzo, L. Privitera, M. Fa', A. Staniszewski, G. Hashimoto, F. Aziz, M. Sakurai,
with melatonin on the level of oxidative damage and neurodegeneration in E.M. Ribe, C.M. Troy, M. Mercken, S.S. Jung, A. Palmeri, O. Arancio, Endoge-
senescence-accelerated mice, J. Pineal. Res. 45 (2008) 302–311. nous amyloid-β is necessary for hippocampal synaptic plasticity and memory,
[97] Y. Okatani, A. Wakatsuki, R.J. Reiter, Y. Miyahara, Melatonin reduces oxidative Ann. Neurol. 69 (2011) 819–830.
damage of neural lipids and proteins in senescence-accelerated mouse, Neurobiol. [106] D. Puzzo, L. Privitera, E. Leznik, M. Fa, A. Staniszewski, A. Palmeri, O. Arancio,
Aging 23 (2002) 639–644. Picomolar amyloid-beta positively modulates synaptic plasticity and memory
[98] J. Gutierrez-Cuesta, F.X. Sureda, M. Romeu, A.M. Canudas, B. Caballero, A. coto- in hippocampus, J. Neurosci. 28 (2008) 14537–14545.
Montes, A. Camius, M. Pallas, Chronic administration of melatonin reduces cerebral [107] R. Baruch-Suchodolsky, B. Fischer, Abeta40, either soluble or aggregated, is a re-
injury biomarkers in SAMP8, J. Pineal. Res. 42 (2007) 394–402. markably potent antioxidant in cell-free oxidative systems, Biochemistry 48
[99] S. Cheng, C. Ma, H. Qu, W. Fan, J. Pang, H. He, Differential effects of melatonin on (2009) 4354–4370.
hippocampal neurodegeneration in different aged accelerated senescence prone [108] N.G. Nair, G. Perry, M.A. Smith, V.P. Reddy, NMR studies of zinc, copper, and iron
mouse-8, Neuro. Endocrinol. Lett. 29 (2008) 91–99. binding to histidine, the principal metal ion complexing site of amyloid-beta
[100] H.F. Poon, G. Joshi, R. Sultana, S.A. Farr, W.A. Banks, I.E. Morley, V. Calabrese, D.A. peptide, J. Alzheimers Dis. 20 (2010) 57–66.
Butterfield, Antisense directed at the Abeta region of APP decreases brain oxidative [109] C.S. Atwood, M.E. Obrenovich, T. Liu, H. Chan, G. Perry, M.A. Smith, R.N.
markers in aged senescence accelerated mice, Brain Res. 1018 (2004) 86–96. Martins, Amyloid-beta: a chameleon walking in two worlds: a review of the
[101] H.F. Poon, S.A. Farr, W.A. Banks, W.M. Pierce, J.B. Klein, J.E. Morley, D.A. Butterfield, Pro- trophic and toxic properties of amyloid-beta, Brain Res. Brain Res. Rev. 43
teomic identification of less oxidized brain proteins in aged senescence-accelerated (2003) 1–16.
mice following administration of antisense oligonuleotide directed at the Abeta region [110] A.L. Friedlich, L.L. Butcher, Involvement of free oxygen radicals in beta-
of amyloid precursor protein, Brain Res. Mol. Brain Res. 138 (2005) 8–16. amyloidosis: an hypothesis, Neurobiol. Aging 15 (1994) 443–455.

You might also like