Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2013

Open Access 01.12.2013 | Research article

Age-dependent roles of peroxisomes in the hippocampus of a transgenic mouse model of Alzheimer’s disease

verfasst von: Francesca Fanelli, Sara Sepe, Marcello D’Amelio, Cinzia Bernardi, Loredana Cristiano, AnnaMaria Cimini, Francesco Cecconi, Maria Paola Ceru', Sandra Moreno

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2013

Abstract

Background

Alzheimer’s Disease (AD) is a progressive neurodegenerative disease, especially affecting the hippocampus. Impairment of cognitive and memory functions is associated with amyloid β-peptide-induced oxidative stress and alterations in lipid metabolism. In this scenario, the dual role of peroxisomes in producing and removing ROS, and their function in fatty acids β-oxidation, may be critical. This work aims to investigating the possible involvement of peroxisomes in AD onset and progression, as studied in a transgenic mouse model, harboring the human Swedish familial AD mutation. We therefore characterized the peroxisomal population in the hippocampus, focusing on early, advanced, and late stages of the disease (3, 6, 9, 12, 18 months of age). Several peroxisome-related markers in transgenic and wild-type hippocampal formation were comparatively studied, by a combined molecular/immunohistochemical/ultrastructural approach.

Results

Our results demonstrate early and significant peroxisomal modifications in AD mice, compared to wild-type. Indeed, the peroxisomal membrane protein of 70 kDa and acyl-CoA oxidase 1 are induced at 3 months, possibly reflecting the need for efficient fatty acid β-oxidation, as a compensatory response to mitochondrial dysfunction. The concomitant presence of oxidative damage markers and the altered expression of antioxidant enzymes argue for early oxidative stress in AD. During physiological and pathological brain aging, important changes in the expression of peroxisome-related proteins, also correlating with ongoing gliosis, occur in the hippocampus. These age- and genotype-based alterations, strongly dependent on the specific marker considered, indicate metabolic and/or numerical remodeling of peroxisomal population.

Conclusions

Overall, our data support functional and biogenetic relationships linking peroxisomes to mitochondria and suggest peroxisomal proteins as biomarkers/therapeutic targets in pre-symptomatic AD.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1750-1326-8-8) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

FF made substantial contribution to conception and design of the study, carried out the morphological and molecular experiments and has been involved in drafting the manuscript. SS participated in performing immunoblotting experiments and statistical analyses. MD participated in interpretation of data and revised the manuscript critically. CB has been involved in morphological analysis and interpretation of data. LC participated in performing and interpreting immunofluorescence experiments. AC participated in the design of the study. FC revised the manuscript critically. MPC participated in design and coordination of the study and has been involved in revising the manuscript critically. SM conceived and designed the study, helped to draft the manuscript and gave final approval of the version to be published. All authors read and approved the final manuscript.
Abkürzungen
8-OH(d)G
8-hydroxy(deoxy)guanosine
Amyloid β
AD
Alzheimer’s disease
AOX
Acyl-CoA oxidase 1
CAT
Catalase
GFAP
Glial fibrillary acidic protein
GPX1
Selenium-dependent glutathione peroxidase
NeuN
Neuronal nuclei
Pex14p
Peroxin 14p
PGC-1α
Peroxisome proliferator-activated receptor γ coativator 1α
PMP70
Peroxisomal membrane protein of 70 kDa
PPAR
Peroxisome proliferator-activated receptor
PVDF
Polyvinylidene fluoride
RT
Room temperature
ROS
Reactive oxygen species
SDS
Sodium dodecyl phosphate
SOD1
CU,Zn-superoxide dismutase
SOD2
Mn-superoxide dismutase
Tg
Tg2576 transgenic mouse model of Alzheimer’s disease
THL
3-ketoacyl-CoA thiolase
VLCFA
Very long chain fatty acid
WB
Western blotting
WT
Wild-type

Background

Alzheimer’s disease (AD) is the most common form of dementia, characterized by progressive neurodegeneration, particularly affecting the hippocampal formation. Impairment of cognitive and memory functions is associated with amyloid β-peptide (Aβ) accumulation, increased oxidative stress, lipid metabolism alteration, and inflammation [15].
The central role of peroxisomes in reactive oxygen species (ROS) metabolism has emerged since their discovery [6]. In fact, peroxisomes participate in both ROS generation and removal, under physiological or pathological conditions. Moreover, peroxisomes are involved in a wide range of catabolic and anabolic functions, including β-oxidation of very long chain fatty acids (VLCFAs) [7, 8], biosynthesis of polyunsaturated fatty acids and plasmalogens [9, 10], and calcium homeostasis [11, 12]. Emergent studies have revealed that peroxisomes can also function as intracellular signaling compartments and organizing platforms that orchestrate important developmental decisions from inside the cell [13]. These dynamic and versatile organelles respond to physiological and pathological changes in cellular environment by adapting their morphology, number and enzyme content accordingly [14]. Peroxisomal dysfunction has been shown to be associated with cellular aging as well as with age-related degenerative diseases [13]. The potential role of peroxisomes in human AD, especially in relationship with lipid metabolism, has recently been suggested [15]. Indeed, a decrease in plasmalogens and, conversely, an increase in VLCFAs, were described in cases with advanced Braak stages [16]. Moreover, in an in vitro model of advanced AD a decrease of peroxisomes in hippocampal neurons was reported, while induction of peroxisomal proliferation attenuated Aβ-dependent toxicity [17]. We previously demonstrated that peroxisomes are involved in early stages of AD, as studied either in vivo, in a transgenic mouse model [18], or in vitro, on Aβ treated cortical neurons [19].
The aim of the present work was to investigate the role of peroxisomes during the progression of AD and in normal aging. To this purpose, we utilized the Tg2576 (Tg) mouse model, compared to its wild-type (WT) counterpart. Differently from other mouse models, this strain displays a slowly progressive AD pathology, offering the opportunity to study even subtle age-dependent alterations [2023]. By combined molecular and morphological approaches, we examined the expression of peroxisome-related proteins in the hippocampal formation at early and advanced AD stages (3, 6, 9, 12, and 18 months). We focused on the CA1 hippocampal region, as pyramidal cells in this field are the most profoundly affected cell type in human AD [24]. Specifically, the expression and immunolocalization patterns of peroxisomal membrane protein of 70 kDa (PMP70), peroxin 14p (Pex14p), catalase (CAT), acyl-CoA oxidase 1 (AOX), and 3-ketoacyl-CoA thiolase (THL) were studied, to get an insight into the biogenesis and functioning of peroxisomes in the diseased brain. Given the involvement of peroxisomes in the maintenance of the redox status, antioxidant enzymes other than CAT, namely selenium-dependent glutathione peroxidase (GPX1), Cu,Zn-superoxide dismutase (SOD1), and Mn-superoxide dismutase (SOD2) were also investigated in Tg animals and in their WT littermates. It is worth recalling that these proteins, though mainly localized to different cell compartments, were also found in peroxisomes [25]. At selected stages, markers for oxidative damage to lipids and nucleic acids - acrolein and 8-hydroxy(deoxy)guanosine (8-OH(d)G), respectively - were also studied.
Notably, the size and functions of peroxisomal population are regulated by a class of ligand-activated transcription factors (peroxisome proliferator-activated receptors, PPARs) [26]. Among these, we studied PPARα which is directly involved in peroxisomal induction [27], plays a neuroprotective role in age-related inflammation [28] and enhances memory consolidation [29]. It is worth-mentioning in this context that PPARα specific agonists have been demonstrated to exert a neuroprotective action against Aβ-mediated toxicity in vitro [17]. Finally, we examined the expression of PPARγ coactivator-1γ (PGC-1α), in view of its synergism with PPARα and its implication in peroxisomal remodeling and biogenesis [30, 31].

Results

The distribution of peroxisomes in Tg and WT hippocampal formation from 3-, 6-, 9-, 12-, and 18-month-old mice was investigated by analyzing the expression of peroxisomal membrane (PMP70, Pex14p) and matrix (CAT, AOX, THL) proteins. Markers of oxidative damage to nucleic acids (8-OH(d)G) and to lipids (acrolein), as well as antioxidant enzymes (SOD1, SOD2, GPX1), were also studied. Further, to get an insight into the transcriptional regulation of these genes, PPARα and PGC-1α expression was examined. At selected stages, the presence of PMP70 and PPARα in neuronal and/or astroglial cells was studied by immunofluorescence and their ultrastructural localization was investigated by immunoelectron microscopy.

Peroxisomal membrane proteins: Pex14p and PMP70

Pex14p is a membrane-anchored peroxin, involved in the peroxisomal import of matrix proteins [3234], and thus considered a dependable marker for peroxisomal number [35]. Pex14p hippocampal levels, as assessed by densitometric analysis of WB (Figure 1a), show no genotype-dependent differences throughout the examined period, except for 18 months of age, when remarkably higher protein concentration in Tg, compared to WT, is detected. Rather, age-related decreases are observed in both normal and diseased animals, namely between 3 and 6 months in the former, and between 6 and 9 months in the latter. This pattern is consistent with Pex14p immunohistochemical staining of pyramidal neurons in CA1 hippocampal field (Figure 1b).
PMP70, a major component of mammalian peroxisomal membranes, is also considered as a good marker for the overall size of peroxisomal population [36]. This ATP-binding cassette transporter, also known as ABCD3, is suggested to be responsible for the metabolic transport of long and branched-chain fatty acyl-CoAs [37]. Our WB data (Figure 2a) show a significant PMP70 induction in 3-month-old Tg hippocampus compared to control. A decrease at 6 months of age ensues in both genotypes, being especially dramatic in the diseased mice. Neither age- nor genotype-related variations are detected at 9 and 12 months, while a peak of PMP70 protein levels is observed at 18 months, particularly in the pathological genotype.
PMP70 immunohistochemical results on the CA1 hippocampal field are in agreement with molecular data (Figure 2b). Indeed, 3-month-old Tg pyramidal cell layer displays stronger immunoreactivity than its WT counterpart, while, at 6 months, decreased immunostaining is observed in both genotypes. At 9–12 months of age, immunostaining levels remain stable, and they remarkably increase in 18-month-old hippocampus, especially in the somata of Tg pyramidal neurons. PMP70 pre-embedding immunoelectron microscopy allowed us to identify positive peroxisomes in 3-month-old CA1 pyramidal neurons, in both WT and Tg animals (Figure 2c). The immunoreaction product appears confined to the membrane, as expected. Consistent with molecular and immunohistochemical data, ultrastructural analysis shows numerous PMP70 immunoreactive peroxisomes in hippocampal cells of young Tg mice.
The strong PMP70 immunoreactivity observed at 18 months suggests possible contribution by astrogliosis to peroxisome numerical increase. To address this issue, we performed GFAP immunohistochemistry and immunoblotting (Figures 3a and b), as well as double immunofluorescence of PMP70 in combination with GFAP (Figure 3c). As expected, astrogliosis is present in the aging hippocampus, particularly in the Tg animals, and also associates with senile plaques. Confocal images of 18-month-old WT and Tg CA1 fields show bright PMP70 immunofluorescence in GFAP and GFAP+ cells, in both genotypes. PMP70+/GFAP+ cells are especially numerous in Tg, demonstrating that peroxisomal increase in the AD senescent hippocampus is importantly contributed by astroglial cells.

Peroxisomal fatty acid β-oxidation enzymes

Even though the important role of peroxisomal β-oxidation in brain development and functioning is well recognized [8], no information on this metabolic pathway in AD is presently available. We therefore analyzed the expression of two major peroxisomal β-oxidation enzymes, namely AOX and THL at the onset and during the progression of AD. The former is the rate-limiting enzyme catalyzing the first step of the cycle, while THL is the last enzyme of the pathway. In 3-month-old Tg hippocampus, AOX expression is significantly higher than in its WT counterpart (Figure 4a). Starting from 6 months, protein levels significantly decrease and remain relatively low throughout the progression of disease. This pattern is supported by AOX immunohistochemical analysis, showing intense immunoreactivity in CA1 pyramidal cell somata of 3-month-old Tg hippocampus, not observed at subsequent stages (Figure 4b). The expression pattern of THL displays no genotype- or age-linked variations, except for 18 months, when hippocampal levels are strongly decreased in both genotypes, suggesting age-dependent impairment of peroxisomal β-oxidation (Figure 4c).

ROS-scavenging enzymes: CAT, GPX1, SOD1 and SOD2

Peroxisomes play a pivotal role in the protection against ROS, mainly through their CAT activity, but also thanks to their content in SOD1, SOD2, and GPX1 [25, 38, 39]. High CAT levels are found in 3-month-old hippocampus irrespective of the genotype, while at 6 months significantly higher values are observed in Tg, as compared to WT (Figure 5a). At 9 months, CAT expression in the Tg decreases to WT levels, which are maintained at later ages in both genotypes. These data are confirmed by immunohistochemical results, CA1 pyramidal neurons being especially CAT positive in 6-month-old Tg hippocampus (Figure 5b).
The other major H2O2–scavenging enzyme, GPX1, shows no significant variations during normal aging, while differences in its expression levels are seen in the pathological genotype, during AD progression. Indeed, at 3 months, GPX1 expression in Tg is lower than in WT, increasing thereafter, to reach significantly higher levels than WT at 9 months. In the aging Tg hippocampus a progressive decrease in GPX1 protein, below WT levels, is observed (Figure 5c). Immunohistochemical data are consistent with WB data (Figure 5d).
The molecular and morphological expression patterns of the two enzymes responsible for the dismutation of the superoxide anion to hydrogen peroxide – SOD1 and SOD2 – are shown in Figures 6 and 7, respectively. As to SOD1 (Figure 6a, b), in 3-month-old Tg hippocampus its levels are significantly lower than in WT, but progressively increase thereafter, reaching at 9 months significantly higher levels than its WT counterpart. SOD1 expression remains stable in the Tg at 12 months of age, when WT levels are instead maximally increased, and sharply decreases at 18 months in both genotypes.
SOD2 expression pattern during normal aging (Figure 7a, b) closely resembles that of SOD1 in WT hippocampus, while in Tg mice no age-dependent variations are observed. Interestingly, SOD2 levels are significantly higher in 3- and in 18-month-old Tg hippocampus, compared to their WT counterparts. Remarkably, at 18 months, SOD2 mainly localizes to glial cells surrounding senile plaques in Tg hippocampus (Figure 7c).

Oxidative stress markers: acrolein and 8-OH(d)G

To evaluate possible oxidative stress occurring in the hippocampus during normal and pathological aging, markers of oxidative damage to biomolecules were investigated by immunohistochemistry. Antibodies to acrolein, marker of lipid peroxidation (Figure 8a), and to 8-OH(d)G, marker of DNA/RNA oxidative modifications (Figure 8b), were tested at 3 and 18 months, as representative of early and advanced AD stages.
We found a similar immunoreactivity pattern for both markers, strongly related to the genotype, in that intense labeling in Tg CA1 pyramidal layer was observed, as early as 3 months, when WT neurons are faintly immunopositive in their cytoplasm. In 18-month-old hippocampus, oxidative damage markers are detected in both WT and Tg CA1 pyramidal cells, even though the former are consistently less immunoreactive than the latter. Intriguingly, the intensity of 8-OH(d)G immunoreaction at 18 months in Tg CA1 neurons appears lower than at 3 months, suggesting a lower degree of oxidative damage to nucleic acids in late AD.

PPARα and PGC-1α

Since the expression and activity of PPARα, major regulator of peroxisome biogenesis, is influenced by oxidative stress [28], we also addressed the putative involvement of this transcription factor at the onset and at late stages of AD. PPARα immunostaining of CA1 pyramidal cell layer shows significantly higher levels in 3-month-old Tg hippocampus, compared to its WT counterpart (Figure 9a). Triple immunofluorescence experiments of PPARα in combination with the neuronal marker NeuN and the astroglial marker GFAP, confirm high expression of the receptor in 3-month-old Tg neuronal cell nuclei and demonstrate its presence also in glial cell nuclei (Figure 9b). Pre-embedding immunoelectron microscopy further confirms the massive presence of PPARα within the nucleus of Tg hippocampal pyramidal neurons (Figure 9c).
Given the growing evidence for a PGC-1α-mediated transcriptional regulation in response to oxidative stress and in view of its roles in peroxisome biogenesis [31] and in promoting the nonamyloidogenic pathway of APP [40], we analyzed the immunohistochemical localization of this coactivator in the hippocampal CA1 region (Figure 10). At 3 months of age, PGC-1α appears localized in both the nucleus and the cytoplasm, including dendrites extending in the stratum radiatum, of WT and Tg hippocampus. Interestingly, at 6 months, immunoreactivity is highly concentrated in Tg neuronal nuclei, while in WT hippocampus nuclear localization is observed at 9 months. At the latest age considered, a general decrease in immunoreactivity is detected.

Discussion

The aim of the present study was to investigate the role of peroxisomes in AD onset and progression. In a previous work [18], peroxisomal involvement in early AD was demonstrated in the Tg2576 mouse model, also used in this work. Differently from that study, in which male animals were analyzed, we here utilized females, since epidemiological and histopathological data indicate this as the most AD-affected gender in mice and humans [41, 42]. We focused on the hippocampal formation, i.e., the primarily affected area of the diseased brain [43]. The expression of peroxisome-related proteins was studied by combined molecular and morphological approaches, at the onset (3 months) and during AD progression (6, 9, 12, 18 months) in Tg animals, compared to their WT counterparts.
Our results demonstrate that significant peroxisomal alterations occur in Tg mice, as early as 3 months. These early changes are consistent with behavioral, electrophysiological, ultrastructural and molecular modifications reported in this model at the same age [22]. Indeed, PMP70, long been considered marker of the numerical size of peroxisomal population [36] is upregulated, as assessed by WB. Consistently, PMP70 immunohistochemistry reveals especially intense immunostaining in the pyramidal layer of Tg hippocampal CA1 subdivision, and immunoelectron microscopy shows numerous positive peroxisomes in the somata of these neurons.
Differently from PMP70, the biogenesis marker Pex14p, also used to evaluate peroxisome number [35], is unchanged in 3-month-old Tg animals. The absence of correlation between PMP70 and Pex14p levels in early AD could either indicate specific concentration of PMP70 in a numerically unchanged peroxisomal population, or impoverishment of Pex14p in proliferated peroxisomes. In either case, the altered content in these proteins likely results in profound modifications of peroxisomal functions. Interestingly, Pex14p has recently been involved in microtubule-based peroxisome motility [44], suggesting that at this early AD stage, peroxisomes, endowed with a relatively low Pex14p/PMP70 ratio are altered in their transport along neurites. To this respect, it is worth mentioning that a recent work from Berger’s group demonstrates an increased peroxisomal volume density in neuronal cell bodies of human AD brain, associated with impaired peroxisome trafficking [16].
The specific increase in early AD of PMP70, presumably involved in fatty acyl-CoA import across peroxisomal membranes [37], could reflect the need for a more efficient acyl-CoA β-oxidation. In agreement with this hypothesis, AOX, rate-limiting enzyme of peroxisomal β-oxidation pathway, is upregulated in 3-month-old Tg hippocampus. This increase could thus represent a compensatory response to early Aβ-mediated mitochondrial insult, to cope with the impaired energy metabolism, occurring in AD [45, 46]. Indeed, abnormalities in glucose metabolism/mitochondrial function are invariant features of early AD [4649]. Consistently, positron emission tomography studies in AD patients report abnormal cerebral glucose utilization, particularly in the hippocampus, decades prior to the onset of histopathological and clinical features [50]. A consequent shift in brain metabolism, from a primarily aerobic glycolysis pathway to a ketogenic/fatty acid β-oxidation pathway is thought to occur [51]. In this context, we suggest that peroxisomal β-oxidation contributes to such metabolic change, providing mitochondria with acetyl-CoA and fatty acyl substrates. Induction of peroxisomal β-oxidation pathway may also be neuroprotective, by enhancing the clearance of potentially toxic VLCFAs and by promoting the biosynthesis of the neuroprotective docosahexaenoic acid (DHA) [15]. Relevantly, elevated levels of VLCFA have been found in human AD brain lesions [16] and the ability of these fatty acids to trigger oxidative stress and mitochondrial dysfunction on neural cells has recently been demonstrated [52, 53].
A biogenetic/metabolic relationship between peroxisomes and mitochondria has long been established and recently emphasized [13, 30, 31]. In yeast, experimentally induced or age-associated mitochondrial dysfunction activates the so-called retrograde (RTG) signaling pathway, leading to the transcription of RTG-target genes, including those required for peroxisome biogenesis and function [13]. Our results showing increased PPARα nuclear immunoreactivity allow us to speculate that this gene be included among the above mentioned RTG-target genes. The increased PPARα expression could in turn be responsible for the induction of its target genes PMP70 and AOX [54, 55]. Even though the molecular triggers of PPARα activation remain to be determined in our model, endogenous production of PPARα ligands, e.g., oxidized lipid molecules, is likely to occur due to Aβ-mediated ROS generation.
While, in principle, AOX induction in 3-month-old Tg hippocampus may represent a positive response to enhanced need for lipid substrates to be processed in mitochondria, this induction may also result in increased H2O2 production by the oxidase. Moreover, mitochondrial respiration supported by fatty acids generates substantial rates of ROS production [56], thus contributing to cellular redox status alteration. In this context, even the observed early increase of SOD2 expression, in agreement with reports demonstrating induction of mitochondrial genes in young Tg mice [57], may contribute itself to redox imbalance, through enhanced conversion of superoxide anion to H2O2.
Notably, GPX1, the major cytosolic H2O2–scavenging enzyme, is significantly down-regulated in our early AD samples, further exacerbating imbalance between generation and removal of hydrogen peroxide. GPX1 seems especially critical in AD, since its deletion increases susceptibility of neurons to Aβ-mediated damage, while its overexpression protects against neurodegeneration [58, 59]. The postulated pro-oxidant environment likely results in post-translational modifications of GPX1 itself, leading to its irreversible inactivation [60]. Even the low levels of SOD1 detected in the young Tg hippocampus may be explained taking into account that this cytosolic protein is selectively damaged by H2O2 [61]. Importantly, SOD1 deficiency in Tg2576 mice has been associated with oxidative damage, accelerated Aβ oligomerization and memory impairment [62].
Therefore, our data strongly support the idea that oxidative stress is the primary culprit in AD pathogenesis [63]. Consistently, 8-OH(d)G and acrolein, markers of oxidative modification to biomolecules [64, 65], show DNA/RNA oxidation and lipoperoxidation in the hippocampus of young Tg mice. Interestingly, 8-OH(d)G immunostaining is predominantly localized in the cytoplasm, suggesting that mitochondrial nucleic acids are primarily affected. To our knowledge, this is the first report of such a modification occurring at 3 months in this mouse strain. In this context, it is worth mentioning that acrolein has been suggested not only as a marker of lipoperoxidation, but also as initiator of oxidative stress [65].
Between 3 and 6 months of age, most peroxisomal markers decrease in WT hippocampal formation, indicating a numerical reduction and/or a metabolic change of the organelles in the physiological course of maturity. By contrast, in Tg mice of the same age, when several hallmarks of AD pathology make their appearance [21], marker-specific patterns are observed. In particular, while high levels of CAT and Pex14p are maintained, PMP70 and AOX are significantly decreased, suggesting metabolic, rather than numerical, modifications of peroxisomes. These changes, indicating decreased efficiency of peroxisomal β-oxidation, presumably lead to VLCFA accumulation, consistent with what reported in human AD [16]. Interestingly, pharmacological inhibition of peroxisomal β-oxidation, resulting in VLCFA accumulation, is positively correlated with Aβ40 levels in rat cerebral cortex [66].
The relatively high CAT levels observed in 6-month-old Tg hippocampus are conceivably sustained by transcription factors activated in response to oxidative stress. To this respect, the cellular redox state sensor PGC-1α may play a role in regulating CAT expression [67]. Our immunohistochemical data support this relationship, in that at 6 months intense PGC-1α nuclear staining, suggestive of its induction and activity, is observed in CA1 pyramidal neurons, exclusively in Tg. The decrease in PGC-1α nuclear immunostaining seen at later AD stages, also parallels the behavior of CAT.
The same transcriptional co-activator, which has been involved in aging and age-associated diseases [68], may be responsible for the pattern of other antioxidant enzymes. Indeed, the increased PGC-1α nuclear immunoreactivity observed around 9–12 months in Wt neurons is paralleled by the raise in SOD1 and SOD2 levels, occurring in 12-month-old normal hippocampus. Conversely, the dramatically low expression of PGC-1α at 12 months in AD neurons, when we first observe hippocampal amyloid deposits (not shown), is concomitant with the lower levels of SOD1, SOD2 and, particularly, GPX1, compared to WT. Accordingly, altered glutathione redox status has been associated to the onset of amyloid plaques in a mouse model of AD [69] and decreased activities of SOD1 and GPX1 have been reported in symptomatic human AD [62, 70]. By contrast, SOD2 overexpression in Tg mice results in diminished plaque formation [71].
In 18-month-old mouse hippocampus, genotype-dependent variations are observed for most peroxisomal and antioxidant proteins. At this stage, gliosis, known to occur during brain aging and exacerbated in AD [7274], should be taken into account, when interpreting WB quantitative data. In fact, astrogliosis may be at least in part responsible for the peaks of PMP70 and Pex14p observed in old Tg mice. An important contribution to the overall brain peroxisomal population by astrocytes, highlighted by our GFAP/PMP70 double immunofluorescence images, is consistent with the notion that peroxisomes are abundant in this cell type [7577]. Nevertheless, our immunohistochemical data on 18-month-old hippocampus clearly show enhanced positivity to PMP70 and Pex14p also in CA1 pyramidal neurons, markedly in the Tg, suggesting a global increase of peroxisome number in the hippocampal tissue, contributed by both neuronal and glial cells. Differently from the above membrane proteins, peroxisomal matrix enzymes, namely CAT, AOX and THL are not increased or even decreased, in 18-month-old hippocampus, possibly owing to age-related low efficiency of peroxisomal protein import [13] and in keeping with the impairment of brain peroxisomal acyl-CoA β-oxidation in physiological and pathological human aging [15, 16].
Oxidative stress is known to occur in age-related neurodegeneration [78, 79]. Our results on 18-month-old WT hippocampus show significantly lower levels of SOD1 and SOD2 proteins, while GPX1 is not decreased, with respect to previous ages. Thus, it seems that oxidative damage in the aging hippocampus may result from defective scavenging of superoxide anion, rather than hydrogen peroxide. In 18-month-old Tg hippocampus, GPX1 levels are significantly lower than in the corresponding WT, indicating impaired removal of H2O2, thus exacerbating the physiological age-related alteration of the cellular redox status. Concerning SODs, while SOD1 follows the decreasing trend of the age-matched WT hippocampus, SOD2 is not decreased in 18-month-old Tg, being significantly higher than in its WT counterpart. This finding can be explained by immunohistochemical data, showing particularly high concentration of SOD2 in glial cells surrounding amyloid plaques. Consistently, SOD2 - but not SOD1, CAT, or GPX1 - is induced in astroglia by activated microglia in an in vitro model of neuroinflammation [80].
A pro-oxidant cellular environment in the aged hippocampus is further demonstrated by the presence of oxidative damage markers (acrolein and 8-OH(d)G), particularly concentrated in Tg neurons. However, while in WT hippocampus both markers clearly show age-dependent increase, the same is not true for the Tg, where 8-OH(d)G immunoreactivity appears reduced in 18-month-old hippocampus, compared to the young. This observation is in agreement with data from other Authors indicating a negative correlation between 8-OH(d)G levels and histological amyloid burden [81, 82].

Conclusions

Overall, our results confirm and extend our previous observations on the early involvement of peroxisomes in AD pathogenesis [18]. In particular, we strongly suggest that these organelles constitute a first line of defense in support of mitochondria, primary targets of intracellular Aβ/ROS-mediated damage. This protective role specifically involves increased peroxisomal fatty acyl β-oxidation, likely providing mitochondria with acetyl-CoA and shortened acyl-CoAs. This conclusion may shed new light into the physiological significance of peroxisome metabolism in mature neurons, so far mainly related to the synthesis of plasma membrane components and to neurotransmission [710, 83, 84].
In brain aging, when oxidative stress sensu strictu begins, peroxisomes become target of ROS, their functions irreversibly decline, thus contributing themselves to the “pro-oxidant loop” [13]. Deepening knowledge of these processes, which are associated to aging and accelerated/exacerbated in AD, will add significantly to the dissection of pathogenic mechanisms of the disease. At the same time, the early response shown by peroxisomes supports the potential use of some of their proteins as biomarkers, to recognize the first signs of presymptomatic AD and to follow the effectiveness of novel therapeutic approaches, possibly involving peroxisomal induction. Indeed, preventing or treating the disease earlier than in its mild to moderate stages presently appears a major task, as stressed by the latest commentaries on the topic [85, 86].

Methods

Animals

Heterozygous female Tg mice [20] and WT littermates were used for all experiments. Male mice (C57B6 × SJL), hemizygous for human AβPP695 carrying the double mutation K670N and M671L (FADSwedish mutation), were purchased from Taconic Farms, Inc. (Germantown, NY, USA). The Tg colony is maintained and the genotyping is performed as previously described [18].
For immunoblotting analyses, Tg and WT female mice aging 3, 6, 9, 12, and 18 months were used. Six animals from each group were killed by cervical dislocation, brains were rapidly excised on an ice-cold plate, and the hippocampus was dissected out before pooling samples (three pools of two hippocampi each). For morphological studies, three Tg mice and three WT littermates for each age considered were deeply anesthetized with urethane (1 g/kg body weight, injected i.p.), before rapid killing by transcardial perfusion with the fixative solution.
Experiments were performed in accordance with the European Community’s Council Directive 86/609/EEC. Formal approval of these experiments was obtained from the Italian Ministry of Health (D.L.vo 116/92; Prot. No. 155-VI-1.1.). All efforts were made to minimize the number of animals used and their suffering.

Molecular analyses

Total homogenate preparation from hippocampal tissue

Protein extraction was performed by homogenizing tissue in lysis buffer (320 mM sucrose, 50 mM NaCl, 50 mM Tris–HCl, pH 7.5, 1% Triton X-100, 0.5 mM sodium orthovanadate, 5 mM β-glycerophosphate, 1% protease inhibitor), incubating on ice for 30 min. Homogenates were then centrifuged at 13,000 g for 10 min. The total protein content of the resulting supernatant was determined using a spectrophotometric assay, according to Bradford [87]. Samples were then diluted 3:4 in 200 mM Tris–HCl, pH 6.8, containing 40% glycerol, 20% β-mercaptoethanol, 4% sodium dodecyl phosphate (SDS), bromophenol blue.

Immunoblotting analyses

Western blotting (WB) experiments were performed as previously described [18]. Membranes were probed at 4°C overnight with the following primary antibodies: 1:10000 rabbit polyclonal anti-Pex14p (generous gift of Prof. R. Erdmann, Ruhr-Universität Bochum, Bochum, Germany); 1:1000 rabbit polyclonal anti-PMP70 (Sigma-Aldrich, St. Louis, MO, USA); 1:200 rabbit polyclonal anti-glial fibrillary acidic protein (anti-GFAP, Sigma-Aldrich); 1:1000 rabbit polyclonal anti-AOX (generous gift of Prof. A. Völkl, University of Heidelberg, Heidelberg, Germany); 1:1000 rabbit polyclonal anti-THL (generous gift of Prof. P. Van Veldhoven, Katholieke Universiteit Leuven, Leuven, Belgium); 1:2000 rabbit polyclonal anti-CAT (Rockland, Gilbertsville, PA, USA); 1:3000 rabbit polyclonal anti-GPX1 (Abnova, Taipei City, Taiwan); 1:1000 rabbit polyclonal anti-SOD1 (Abcam, Cambridge Science Park, Cambridge, UK); 1:10000 anti-SOD2 (Abcam); 1:3000 mouse monoclonal anti-β-actin (Sigma-Aldrich). This was followed by incubation with 1:2000 HRP-conjugated goat anti-rabbit or anti-mouse IgG secondary antibodies (Santa Cruz Biotechnology, Santa Cruz, CA, USA) in blocking solution, for 1 h at 4°C. Immunoreactive bands were visualized by a chemiluminescence detection kit (ECL Plus™ Western Blotting Detection Reagents, Amersham GE Healthcare, Little Chalfont, UK). The relative densities of immunoreactivity were determined by densitometry using the software ImageJ (NIH, Bethesda, MD, USA) and normalized with respect to β-actin. Data are mean of five different experiments.

Morphological analyses

Brain tissue preparation

Mice were perfusion-fixed and brains were paraffin-embedded, as previously described [18]. The left halves from 3-month-old Tg and WT brains were used for immunoelectron microscopy.

Immunohistochemistry

Serial, 5-μm-thick sagittal brain sections from Tg and WT animals were deparaffinized and processed for immunohistochemistry, as previously described [18]. The following primary antibodies were used: 1:500 rabbit polyclonal anti-Pex14p (generous gift of Prof. R. Erdmann); 1:200 rabbit polyclonal anti-PMP70 (Sigma-Aldrich); 1:300 rabbit polyclonal anti-GFAP (Dako, Glostrup, Denmark); 1:100 rabbit polyclonal anti-AOX (generous gift of Prof. A. Volkl, Heidelberg); 1:200 rabbit polyclonal anti-CAT (Rockland); 1:100 rabbit polyclonal anti-SOD1 and 1:1000 anti-SOD2 (Abcam); 1:500 rabbit polyclonal anti-acrolein (generous gift of Prof. K. Uchida, Nagoya University, Nagoya, Japan); 1:200 rabbit polyclonal anti-PPARα (ABR Affinity BioReagents, Golden, CO, USA); 1:50 mouse monoclonal anti-PGC-1α (Calbiochem), and 1:500 mouse monoclonal anti-8OH(d)G (QED Bioscience Inc., San Diego, CA, USA),. In control sections, the primary antibody was omitted. Biotinylated goat anti-rabbit IgG (Vector Laboratories, Burlingame, CA, USA), or biotinylated goat anti-mouse IgG (Vector) were used as secondary antibodies. Immuno-complexes were visualized, and slides were dehydrated and mounted, as previously reported [18]. Sections were observed under an Olympus BX 51 microscope, equipped with a Leica DFC 420 camera; electronic images were captured by a Leica Application Suite system, and composed in an Adobe Photoshop CS5 format.

Immunofluorescence

For double immunofluorescence experiments, sagittal paraffin sections from 18-month-old Tg and WT brains were deparaffinized and rehydrated, as above. Sections were incubated overnight at 4°C with a mixture of rabbit polyclonal anti-PMP70 (1:100, Sigma-Aldrich) and either mouse monoclonal anti-GFAP (1:200, Chemicon, Temecula, CA, USA), or mouse monoclonal anti-Neuronal Nuclei (anti-NeuN 1:100, Chemicon). Sections were thoroughly rinsed with PBS, then incubated for 1 h at RT with a mixture of 1:100 Alexa488 conjugated goat anti-rabbit IgG (Invitrogen) and 1:500 Alexa555 conjugated goat anti-mouse IgG (Invitrogen). All antibodies were diluted with PBS containing 4% BSA. Triple immunofluorescence experiments were performed on 3-month-old Tg and WT brains, using a mixture of primary antibodies (1:100 rabbit polyclonal anti-PPARα, 1:200 sheep polyclonal anti-GFAP, 1:100 mouse monoclonal anti-NeuN) and of secondary antibodies (1:100 Alexa633 conjugated goat anti-rabbit IgG, 1:500 Alexa488 conjugated goat anti-mouse IgG, and 1:100 Alexa546 conjugated goat anti-sheep IgG). Controls were performed by omitting the primary antibody. Slides were observed in a Leica TCS SP5 confocal microscope and electronic images, captured by a Leica Application Suite system, were composed in an Adobe Photoshop CS5 format.

Immunoelectron microscopy

Samples from 3-month-old Tg and WT brains were sagittally cut by a vibratome, and 100-μm-thick sections, were processed for pre-embedding immunolocalization, according to the previously described procedure [18]. Sections were incubated with either 1:500 anti-PMP70 or with 1:150 anti-PPARα, as primary antibodies, diluted in PBS. After immunocomplexes visualization, slices were osmium-postfixed, dehydrated, and flat-embedded in Epon as previously described [18]. CA1 hippocampal fields of the resin-embedded sections, identified in a light microscope, were remounted on Epon blanks, and further sectioned on a Reichert Ultracut S ultramicrotome. Ultrathin sections were briefly contrasted with uranyl acetate and lead citrate, and observed in a Philips CM120 electron microscope equipped with a Philips Megaview III camera. Electronic images were captured by AnalySys 2.0 software and composed in Adobe Photoshop CS5 format.

Statistical analysis

Statistical evaluation for WB experiments was performed using GraphPad Prism 4 software. For comparison between genotypes and among different ages, statistical analysis of WB densitometric values was accomplished using two-way analysis of variance (ANOVA) followed by the Bonferroni test to detect significant differences between groups. Means from independent experiments were then expressed as means ± SD. For all statistical analyses, P <0.05 was considered as statistically significant.

Acknowledgements

We wish to thank Prof. Ralph Erdmann (Ruhr-Universität Bochum, Bochum, Germany), for generously providing us with the anti-Pex14p, Prof. Alfred Volkl (University of Heidelberg, Germany) for anti-acyl CoA oxidase, Prof. Paul Van Veldhoven (Catholic University of Leuven, Belgium) for anti-thiolase, and Prof. Koji Uchida (Nagoya University, Aichi, Japan), for anti-acrolein.
We are indebted to Sara Porcellotti, Pamela Rosso, Federica Giardi and Anna Fracassi for technical and imaging assistance. The technical staff of UOC Pathological Anatomy of San Filippo Neri Hospital is also gratefully acknowledged for skilful assistance.
This work was partially supported by grants to SM from the University “Roma Tre” (“Internationalization and Research Training Project ” and “CAL”). MPC and AC are financially supported by RIA from University of L’Aquila and by FP-6 Integrated European Project LSHG-CT 768-512018. FC is financially supported by the Telethon Foundation (GGP10225), the Italian Ministry of University and Research (FIRB Accordi di Programma 2011) and the Italian Ministry of Health (“Ricerca Finalizzata” and “Ricerca Corrente”). MDA is financially supported by a grant from Alzheimer's Association (NIRG-11-204588).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

FF made substantial contribution to conception and design of the study, carried out the morphological and molecular experiments and has been involved in drafting the manuscript. SS participated in performing immunoblotting experiments and statistical analyses. MD participated in interpretation of data and revised the manuscript critically. CB has been involved in morphological analysis and interpretation of data. LC participated in performing and interpreting immunofluorescence experiments. AC participated in the design of the study. FC revised the manuscript critically. MPC participated in design and coordination of the study and has been involved in revising the manuscript critically. SM conceived and designed the study, helped to draft the manuscript and gave final approval of the version to be published. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Bonda DJ, Wang X, Perry G, Nunomura A, Tabaton M, Zhu X, Smith MA: Oxidative stress in Alzheimer disease: a possibility for prevention. Neuropharmacology. 2010, 59: 290-294. 10.1016/j.neuropharm.2010.04.005.CrossRefPubMed Bonda DJ, Wang X, Perry G, Nunomura A, Tabaton M, Zhu X, Smith MA: Oxidative stress in Alzheimer disease: a possibility for prevention. Neuropharmacology. 2010, 59: 290-294. 10.1016/j.neuropharm.2010.04.005.CrossRefPubMed
2.
Zurück zum Zitat Querfurth HW, LaFerla FM: Alzheimer’s disease. N Engl J Med. 2010, 362: 329-344. 10.1056/NEJMra0909142.CrossRefPubMed Querfurth HW, LaFerla FM: Alzheimer’s disease. N Engl J Med. 2010, 362: 329-344. 10.1056/NEJMra0909142.CrossRefPubMed
3.
5.
Zurück zum Zitat Walsh DM, Teplow DB: Alzheimer’s disease and the amyloid β-protein. Prog Mol Biol Transl Sci. 2012, 107: 101-124.CrossRefPubMed Walsh DM, Teplow DB: Alzheimer’s disease and the amyloid β-protein. Prog Mol Biol Transl Sci. 2012, 107: 101-124.CrossRefPubMed
6.
Zurück zum Zitat De Duve C, Baudhuin P: Peroxisomes (microbodies and related particles). Physiol Rev. 1966, 46: 323-357.PubMed De Duve C, Baudhuin P: Peroxisomes (microbodies and related particles). Physiol Rev. 1966, 46: 323-357.PubMed
7.
Zurück zum Zitat Poirier Y, Antonenkov VD, Glumoff T, Hiltunen JK: Peroxisomal beta-oxidation, a metabolic pathway with multiple functions. Biochim Biophys Acta. 2006, 1763: 1413-1426. 10.1016/j.bbamcr.2006.08.034.CrossRefPubMed Poirier Y, Antonenkov VD, Glumoff T, Hiltunen JK: Peroxisomal beta-oxidation, a metabolic pathway with multiple functions. Biochim Biophys Acta. 2006, 1763: 1413-1426. 10.1016/j.bbamcr.2006.08.034.CrossRefPubMed
8.
Zurück zum Zitat Wanders RJ, Waterham HR: Biochemistry of mammalian peroxisomes revisited. Annu Rev Biochem. 2006, 75: 295-332. 10.1146/annurev.biochem.74.082803.133329.CrossRefPubMed Wanders RJ, Waterham HR: Biochemistry of mammalian peroxisomes revisited. Annu Rev Biochem. 2006, 75: 295-332. 10.1146/annurev.biochem.74.082803.133329.CrossRefPubMed
9.
Zurück zum Zitat Sprecher H, Luthria DL, Mohammed BS, Baykousheva SP: Reevaluation of the pathways for the biosynthesis of polyunsaturated fatty acids. J Lipid Res. 1995, 36: 2471-2477.PubMed Sprecher H, Luthria DL, Mohammed BS, Baykousheva SP: Reevaluation of the pathways for the biosynthesis of polyunsaturated fatty acids. J Lipid Res. 1995, 36: 2471-2477.PubMed
10.
Zurück zum Zitat Wanders RJ, Ferdinandusse S, Brites P, Kemp S: Peroxisomes, lipid metabolism and lipotoxicity. Biochim Biophys Acta. 2010, 1801: 272-280. 10.1016/j.bbalip.2010.01.001.CrossRefPubMed Wanders RJ, Ferdinandusse S, Brites P, Kemp S: Peroxisomes, lipid metabolism and lipotoxicity. Biochim Biophys Acta. 2010, 1801: 272-280. 10.1016/j.bbalip.2010.01.001.CrossRefPubMed
11.
Zurück zum Zitat Drago I, Giacomello M, Pizzo P, Pozzan T: Calcium dynamics in the peroxisomal lumen of living cells. J Biol Chem. 2008, 283: 14384-14390. 10.1074/jbc.M800600200.CrossRefPubMed Drago I, Giacomello M, Pizzo P, Pozzan T: Calcium dynamics in the peroxisomal lumen of living cells. J Biol Chem. 2008, 283: 14384-14390. 10.1074/jbc.M800600200.CrossRefPubMed
12.
Zurück zum Zitat Lasorsa FM, Pinton P, Palmieri L, Scarcia P, Rottensteiner H, Rizzuto R, Palmieri F: Peroxisomes as novel players in cell calcium homeostasis. J Biol Chem. 2008, 283: 15300-15308. 10.1074/jbc.M800648200.PubMedCentralCrossRefPubMed Lasorsa FM, Pinton P, Palmieri L, Scarcia P, Rottensteiner H, Rizzuto R, Palmieri F: Peroxisomes as novel players in cell calcium homeostasis. J Biol Chem. 2008, 283: 15300-15308. 10.1074/jbc.M800648200.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Ribeiro D, Castro I, Fahimi HD, Schrader M: Peroxisome morphology in pathology. Histol Histopathol. 2012, 27: 661-676.PubMed Ribeiro D, Castro I, Fahimi HD, Schrader M: Peroxisome morphology in pathology. Histol Histopathol. 2012, 27: 661-676.PubMed
15.
Zurück zum Zitat Lizard G, Rouaud O, Demarquoy J, Cherkaoui-Malki M, Iuliano L: Potential roles of peroxisomes in Alzheimer’s disease and in dementia of the Alzheimer’s type. J Alzheimers Dis. 2012, 29: 241-254.PubMed Lizard G, Rouaud O, Demarquoy J, Cherkaoui-Malki M, Iuliano L: Potential roles of peroxisomes in Alzheimer’s disease and in dementia of the Alzheimer’s type. J Alzheimers Dis. 2012, 29: 241-254.PubMed
16.
Zurück zum Zitat Kou J, Kovacs GG, Höftberger R, Kulik W, Brodde A, Forss-Petter S, Hönigschnabl S, Gleiss A, Brugger B, Wanders R, Just W, Budka H, Jungwirth S, Fischer P, Berger J: Peroxisomal alterations in Alzheimer’s disease. Acta Neuropathol. 2011, 122: 271-283. 10.1007/s00401-011-0836-9.PubMedCentralCrossRefPubMed Kou J, Kovacs GG, Höftberger R, Kulik W, Brodde A, Forss-Petter S, Hönigschnabl S, Gleiss A, Brugger B, Wanders R, Just W, Budka H, Jungwirth S, Fischer P, Berger J: Peroxisomal alterations in Alzheimer’s disease. Acta Neuropathol. 2011, 122: 271-283. 10.1007/s00401-011-0836-9.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Santos MJ, Quintanilla RA, Toro A, Grandy R, Dinamarca MC, Godoy JA, Inestrosa NC: Peroxisomal proliferation protects from beta-amyloid neurodegeneration. J Biol Chem. 2005, 280: 41057-41068. 10.1074/jbc.M505160200.CrossRefPubMed Santos MJ, Quintanilla RA, Toro A, Grandy R, Dinamarca MC, Godoy JA, Inestrosa NC: Peroxisomal proliferation protects from beta-amyloid neurodegeneration. J Biol Chem. 2005, 280: 41057-41068. 10.1074/jbc.M505160200.CrossRefPubMed
18.
Zurück zum Zitat Cimini A, Moreno S, D’Amelio M, Cristiano L, D’Angelo B, Falone S, Benedetti E, Carrara P, Fanelli F, Cecconi F, Amicarelli F, Cerù MP: Early biochemical and morphological modifications in the brain of a transgenic mouse model of Alzheimer’s disease: a role for peroxisomes. J Alzheimer Dis. 2009, 18: 935-952. Cimini A, Moreno S, D’Amelio M, Cristiano L, D’Angelo B, Falone S, Benedetti E, Carrara P, Fanelli F, Cecconi F, Amicarelli F, Cerù MP: Early biochemical and morphological modifications in the brain of a transgenic mouse model of Alzheimer’s disease: a role for peroxisomes. J Alzheimer Dis. 2009, 18: 935-952.
19.
Zurück zum Zitat Cimini A, Benedetti E, D’Angelo B, Cristiano L, Falone S, Di Loreto S, Amicarelli F, Cerù MP: Neuronal response of peroxisomal and peroxisome-related proteins to chronic and acute Abeta injury. Curr Alzheimer Res. 2009, 6: 238-251. 10.2174/156720509788486518.CrossRefPubMed Cimini A, Benedetti E, D’Angelo B, Cristiano L, Falone S, Di Loreto S, Amicarelli F, Cerù MP: Neuronal response of peroxisomal and peroxisome-related proteins to chronic and acute Abeta injury. Curr Alzheimer Res. 2009, 6: 238-251. 10.2174/156720509788486518.CrossRefPubMed
20.
Zurück zum Zitat Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G: Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996, 274: 99-102. 10.1126/science.274.5284.99.CrossRefPubMed Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G: Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996, 274: 99-102. 10.1126/science.274.5284.99.CrossRefPubMed
21.
Zurück zum Zitat Jacobsen JS, Wu CC, Redwine JM, Comery TA, Arias R, Bowlby M, Martone R, Morrison JH, Pangalos MN, Reinhart PH, Bloom FE: Early-onset behavioral and synaptic deficits in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2006, 103: 5161-5166. 10.1073/pnas.0600948103.PubMedCentralCrossRefPubMed Jacobsen JS, Wu CC, Redwine JM, Comery TA, Arias R, Bowlby M, Martone R, Morrison JH, Pangalos MN, Reinhart PH, Bloom FE: Early-onset behavioral and synaptic deficits in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2006, 103: 5161-5166. 10.1073/pnas.0600948103.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat D’Amelio M, Cavallucci V, Middei S, Marchetti C, Pacioni S, Ferri A, Diamantini A, De Zio D, Carrara P, Battistini L, Moreno S, Bacci A, Ammassari-Teule M, Marie H, Cecconi F: Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nat Neurosci. 2011, 14: 69-76. 10.1038/nn.2709.CrossRefPubMed D’Amelio M, Cavallucci V, Middei S, Marchetti C, Pacioni S, Ferri A, Diamantini A, De Zio D, Carrara P, Battistini L, Moreno S, Bacci A, Ammassari-Teule M, Marie H, Cecconi F: Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nat Neurosci. 2011, 14: 69-76. 10.1038/nn.2709.CrossRefPubMed
23.
Zurück zum Zitat Lee SH, Kim KR, Ryu SY, Son S, Hong HS, Mook-Jung I, Lee SH, Ho WK: Impaired short-term plasticity in mossy fiber synapses caused by mitochondrial dysfunction of dentate granule cells is the earliest synaptic deficit in a mouse model of Alzheimer’s disease. J Neurosci. 2012, 32: 5953-5963. 10.1523/JNEUROSCI.0465-12.2012.CrossRefPubMed Lee SH, Kim KR, Ryu SY, Son S, Hong HS, Mook-Jung I, Lee SH, Ho WK: Impaired short-term plasticity in mossy fiber synapses caused by mitochondrial dysfunction of dentate granule cells is the earliest synaptic deficit in a mouse model of Alzheimer’s disease. J Neurosci. 2012, 32: 5953-5963. 10.1523/JNEUROSCI.0465-12.2012.CrossRefPubMed
24.
Zurück zum Zitat Scheff SW, Price DA, Schmitt FA, DeKosky ST, Mufson EJ: Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology. 2007, 68: 1501-1508. 10.1212/01.wnl.0000260698.46517.8f.CrossRefPubMed Scheff SW, Price DA, Schmitt FA, DeKosky ST, Mufson EJ: Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology. 2007, 68: 1501-1508. 10.1212/01.wnl.0000260698.46517.8f.CrossRefPubMed
25.
Zurück zum Zitat Schrader M, Fahimi HD: Peroxisomes and oxidative stress. Biochim Biophys Acta. 2006, 1763: 1755-1766. 10.1016/j.bbamcr.2006.09.006.CrossRefPubMed Schrader M, Fahimi HD: Peroxisomes and oxidative stress. Biochim Biophys Acta. 2006, 1763: 1755-1766. 10.1016/j.bbamcr.2006.09.006.CrossRefPubMed
26.
Zurück zum Zitat Montagner A, Rando G, Degueurce G, Leuenberger N, Michalik L, Wahli W: New insights into the role of PPARs. Prostaglandins Leukot Essent Fatty Acids. 2011, 85: 235-243. 10.1016/j.plefa.2011.04.016.CrossRefPubMed Montagner A, Rando G, Degueurce G, Leuenberger N, Michalik L, Wahli W: New insights into the role of PPARs. Prostaglandins Leukot Essent Fatty Acids. 2011, 85: 235-243. 10.1016/j.plefa.2011.04.016.CrossRefPubMed
27.
Zurück zum Zitat Issemann I, Green S: Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature. 1990, 347: 645-650. 10.1038/347645a0.CrossRefPubMed Issemann I, Green S: Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature. 1990, 347: 645-650. 10.1038/347645a0.CrossRefPubMed
28.
Zurück zum Zitat Chung JH, Seo AY, Chung SW, Kim MK, Leeuwenburgh C, Yu BP, Chung HY: Molecular mechanism of PPAR in the regulation of age-related inflammation. Ageing Res Rev. 2008, 7: 126-136. 10.1016/j.arr.2008.01.001.CrossRefPubMed Chung JH, Seo AY, Chung SW, Kim MK, Leeuwenburgh C, Yu BP, Chung HY: Molecular mechanism of PPAR in the regulation of age-related inflammation. Ageing Res Rev. 2008, 7: 126-136. 10.1016/j.arr.2008.01.001.CrossRefPubMed
29.
Zurück zum Zitat Campolongo P, Roozendaal B, Trezza V, Cuomo V, Astarita G, Fu J, McGaugh JL, Piomelli D: Fat-induced satiety factor oleoylethanolamide enhances memory consolidation. PNAS. 2009, 106: 8027-8031. 10.1073/pnas.0903038106.PubMedCentralCrossRefPubMed Campolongo P, Roozendaal B, Trezza V, Cuomo V, Astarita G, Fu J, McGaugh JL, Piomelli D: Fat-induced satiety factor oleoylethanolamide enhances memory consolidation. PNAS. 2009, 106: 8027-8031. 10.1073/pnas.0903038106.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Delille HK, Alves R, Schrader M: Biogenesis of peroxisomes and mitochondria: linked by division. Histochem Cell Biol. 2009, 131: 441-446. 10.1007/s00418-009-0561-9.CrossRefPubMed Delille HK, Alves R, Schrader M: Biogenesis of peroxisomes and mitochondria: linked by division. Histochem Cell Biol. 2009, 131: 441-446. 10.1007/s00418-009-0561-9.CrossRefPubMed
31.
Zurück zum Zitat Bagattin A, Hugendubler L, Mueller E: Transcriptional coactivator PGC-1alpha promotes peroxisomal remodeling and biogenesis. Proc Natl Acad Sci USA. 2010, 107: 20376-20381. 10.1073/pnas.1009176107.PubMedCentralCrossRefPubMed Bagattin A, Hugendubler L, Mueller E: Transcriptional coactivator PGC-1alpha promotes peroxisomal remodeling and biogenesis. Proc Natl Acad Sci USA. 2010, 107: 20376-20381. 10.1073/pnas.1009176107.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Itoh R, Fujiki Y: Functional domains and dynamic assembly of the Peroxin Pex14p, the entry site of matrix proteins. J Biol Chem. 2006, 281: 10196-10205. 10.1074/jbc.M600158200.CrossRefPubMed Itoh R, Fujiki Y: Functional domains and dynamic assembly of the Peroxin Pex14p, the entry site of matrix proteins. J Biol Chem. 2006, 281: 10196-10205. 10.1074/jbc.M600158200.CrossRefPubMed
33.
Zurück zum Zitat Meinecke M, Cizmowski C, Schliebs W, Kruger V, Beck S, Wagner R, Erdmann R: The peroxisomal importomer constitutes a large and highly dynamic pore. Nat Cell Biol. 2010, 12: 273-277.PubMed Meinecke M, Cizmowski C, Schliebs W, Kruger V, Beck S, Wagner R, Erdmann R: The peroxisomal importomer constitutes a large and highly dynamic pore. Nat Cell Biol. 2010, 12: 273-277.PubMed
34.
Zurück zum Zitat van der Zand A, Gent J, Braakman I, Tabak HF: Biochemically distinct vesicles from the endoplasmic reticulum fuse to form peroxisomes. Cell. 2012, 149: 397-409. 10.1016/j.cell.2012.01.054.CrossRefPubMed van der Zand A, Gent J, Braakman I, Tabak HF: Biochemically distinct vesicles from the endoplasmic reticulum fuse to form peroxisomes. Cell. 2012, 149: 397-409. 10.1016/j.cell.2012.01.054.CrossRefPubMed
35.
Zurück zum Zitat Ahlemeyer B, Neubert I, Kovacs WJ, Baumgart-Vogt E: Differential expression of peroxisomal matrix and membrane proteins during postnatal development of mouse brain. J Comp Neurol. 2007, 505: 1-17. 10.1002/cne.21448.CrossRefPubMed Ahlemeyer B, Neubert I, Kovacs WJ, Baumgart-Vogt E: Differential expression of peroxisomal matrix and membrane proteins during postnatal development of mouse brain. J Comp Neurol. 2007, 505: 1-17. 10.1002/cne.21448.CrossRefPubMed
36.
Zurück zum Zitat Santos MJ, Kawada ME, Espeel M, Figueroa C, Alvarez A, Hidalgo U, Metz C: Characterization of human peroxisomal membrane proteins. J Biol Chem. 1994, 269: 24890-24896.PubMed Santos MJ, Kawada ME, Espeel M, Figueroa C, Alvarez A, Hidalgo U, Metz C: Characterization of human peroxisomal membrane proteins. J Biol Chem. 1994, 269: 24890-24896.PubMed
37.
Zurück zum Zitat Morita M, Imanaka T: Peroxisomal ABC transporters: Structure, function and role in disease. Biochim Biophys Acta. 2012, 1822: 1387-1396. 10.1016/j.bbadis.2012.02.009.CrossRefPubMed Morita M, Imanaka T: Peroxisomal ABC transporters: Structure, function and role in disease. Biochim Biophys Acta. 2012, 1822: 1387-1396. 10.1016/j.bbadis.2012.02.009.CrossRefPubMed
38.
Zurück zum Zitat Moreno S, Mugnaini E, Cerù MP: Immunocytochemical localization of catalase in the central nervous system of the rat. J Histochem Cytochem. 1995, 43: 1253-1267. 10.1177/43.12.8537642.CrossRefPubMed Moreno S, Mugnaini E, Cerù MP: Immunocytochemical localization of catalase in the central nervous system of the rat. J Histochem Cytochem. 1995, 43: 1253-1267. 10.1177/43.12.8537642.CrossRefPubMed
39.
Zurück zum Zitat Moreno S, Nardacci R, Cerù MP: Regional and ultrastructural immunolocalization of copper-zinc superoxide dismutase in rat central nervous system. J Histochem Cytochem. 1997, 45: 1611-1622. 10.1177/002215549704501204.CrossRefPubMed Moreno S, Nardacci R, Cerù MP: Regional and ultrastructural immunolocalization of copper-zinc superoxide dismutase in rat central nervous system. J Histochem Cytochem. 1997, 45: 1611-1622. 10.1177/002215549704501204.CrossRefPubMed
40.
Zurück zum Zitat Qin W, Haroutunian V, Katsel P, Cardozo CP, Ho L, Buxbaum JD, Pasinetti GM: PGC-1alpha expression decreases in the Alzheimer disease brain as a function of dementia. Arch Neurol. 2009, 66: 352-361. 10.1001/archneurol.2008.588.PubMedCentralCrossRefPubMed Qin W, Haroutunian V, Katsel P, Cardozo CP, Ho L, Buxbaum JD, Pasinetti GM: PGC-1alpha expression decreases in the Alzheimer disease brain as a function of dementia. Arch Neurol. 2009, 66: 352-361. 10.1001/archneurol.2008.588.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Hirata-Fukae C, Li HF, Hoe HS, Gray AJ, Minami SS, Hamada K, Niikura T, Hua F, Tsukagoshi-Nagai H, Horikoshi-Sakuraba Y, Mughal M, Rebeck GW, LaFerla FM, Mattson MP, Iwata N, Saido TC, Klein WL, Duff KE, Aisen PS, Matsuoka Y: Females exhibit more extensive amyloid, but not tau, pathology in an Alzheimer transgenic model. Brain Res. 2008, 1216: 92-103.CrossRefPubMed Hirata-Fukae C, Li HF, Hoe HS, Gray AJ, Minami SS, Hamada K, Niikura T, Hua F, Tsukagoshi-Nagai H, Horikoshi-Sakuraba Y, Mughal M, Rebeck GW, LaFerla FM, Mattson MP, Iwata N, Saido TC, Klein WL, Duff KE, Aisen PS, Matsuoka Y: Females exhibit more extensive amyloid, but not tau, pathology in an Alzheimer transgenic model. Brain Res. 2008, 1216: 92-103.CrossRefPubMed
42.
Zurück zum Zitat Craig MC, Murphy DG: Estrogen therapy and Alzheimer’s dementia. Ann N Y Acad Sci. 2010, 1205: 245-253. 10.1111/j.1749-6632.2010.05673.x.CrossRefPubMed Craig MC, Murphy DG: Estrogen therapy and Alzheimer’s dementia. Ann N Y Acad Sci. 2010, 1205: 245-253. 10.1111/j.1749-6632.2010.05673.x.CrossRefPubMed
43.
Zurück zum Zitat Small SA, Schobel SA, Buxton RB, Witter MP, Barnes CA: A pathophysiological framework of hippocampal dysfunction in ageing and disease. Nat Rev Neurosci. 2011, 12: 585-601.PubMedCentralCrossRefPubMed Small SA, Schobel SA, Buxton RB, Witter MP, Barnes CA: A pathophysiological framework of hippocampal dysfunction in ageing and disease. Nat Rev Neurosci. 2011, 12: 585-601.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Bharti P, Schliebs W, Schievelbusch T, Neuhaus A, David C, Kock K, Herrmann C, Meyer HE, Wiese S, Warscheid B, Theiss C, Erdmann R: PEX14 is required for microtubule-based peroxisome motility in human cells. J Cell Sci. 2011, 124: 1759-1768. 10.1242/jcs.079368.CrossRefPubMed Bharti P, Schliebs W, Schievelbusch T, Neuhaus A, David C, Kock K, Herrmann C, Meyer HE, Wiese S, Warscheid B, Theiss C, Erdmann R: PEX14 is required for microtubule-based peroxisome motility in human cells. J Cell Sci. 2011, 124: 1759-1768. 10.1242/jcs.079368.CrossRefPubMed
45.
Zurück zum Zitat Pagani L, Eckert A: Amyloid-Beta interaction with mitochondria. Int J Alzheimers Dis. 2011, 2011: 1-12.CrossRef Pagani L, Eckert A: Amyloid-Beta interaction with mitochondria. Int J Alzheimers Dis. 2011, 2011: 1-12.CrossRef
46.
Zurück zum Zitat Ferrer I: Altered mitochondria, energy metabolism, voltage-dependent anion channel, and lipid rafts converge to exhaust neurons in Alzheimer’s disease. J Bioenerg Biomembr. 2009, 41: 425-431. 10.1007/s10863-009-9243-5.CrossRefPubMed Ferrer I: Altered mitochondria, energy metabolism, voltage-dependent anion channel, and lipid rafts converge to exhaust neurons in Alzheimer’s disease. J Bioenerg Biomembr. 2009, 41: 425-431. 10.1007/s10863-009-9243-5.CrossRefPubMed
47.
Zurück zum Zitat Gillardon F, Rist W, Kussmaul L, Vogel J, Berg M, Danzer K, Kraut N, Hengerer B: Proteomic and functional alterations in brain mitochondria from Tg2576 mice occur before amyloid plaque deposition. Proteomics. 2007, 7: 605-616. 10.1002/pmic.200600728.CrossRefPubMed Gillardon F, Rist W, Kussmaul L, Vogel J, Berg M, Danzer K, Kraut N, Hengerer B: Proteomic and functional alterations in brain mitochondria from Tg2576 mice occur before amyloid plaque deposition. Proteomics. 2007, 7: 605-616. 10.1002/pmic.200600728.CrossRefPubMed
48.
Zurück zum Zitat Gibson GE, Shi Q: A mitocentric view of Alzheimer’s disease suggests multi-faceted treatments. J Alzheimers Dis. 2010, 2: S591-S607. Gibson GE, Shi Q: A mitocentric view of Alzheimer’s disease suggests multi-faceted treatments. J Alzheimers Dis. 2010, 2: S591-S607.
49.
Zurück zum Zitat Nicholson RM, Kusne Y, Nowak LA, LaFerla FM, Reiman EM, Valla J: Regional cerebral glucose uptake in the 3xTG model of Alzheimer’s disease highlights common regional vulnerability across AD mouse models. Brain Res. 2010, 1347: 179-185.CrossRefPubMed Nicholson RM, Kusne Y, Nowak LA, LaFerla FM, Reiman EM, Valla J: Regional cerebral glucose uptake in the 3xTG model of Alzheimer’s disease highlights common regional vulnerability across AD mouse models. Brain Res. 2010, 1347: 179-185.CrossRefPubMed
50.
Zurück zum Zitat Mosconi L, Pupi A, De Leon MJ: Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer’s disease. Ann N Y Acad Sci. 2008, 1147: 180-195. 10.1196/annals.1427.007.PubMedCentralCrossRefPubMed Mosconi L, Pupi A, De Leon MJ: Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer’s disease. Ann N Y Acad Sci. 2008, 1147: 180-195. 10.1196/annals.1427.007.PubMedCentralCrossRefPubMed
51.
Zurück zum Zitat Yao J, Rettberg JR, Klosinski LP, Cadenas E, Brinton RD: Shift in brain metabolism in late onset Alzheimer’s disease: implications for biomarkers and therapeutic interventions. Mol Aspects Med. 2011, 32: 247-257. 10.1016/j.mam.2011.10.005.PubMedCentralCrossRefPubMed Yao J, Rettberg JR, Klosinski LP, Cadenas E, Brinton RD: Shift in brain metabolism in late onset Alzheimer’s disease: implications for biomarkers and therapeutic interventions. Mol Aspects Med. 2011, 32: 247-257. 10.1016/j.mam.2011.10.005.PubMedCentralCrossRefPubMed
52.
Zurück zum Zitat Baarine M, Andréoletti P, Athias A, Nury T, Zarrouk A, Ragot K, Vejux A, Riedinger JM, Kattan Z, Bessede G, Trompier D, Savary S, Cherkaoui-Malki M, Lizard G: Evidence of oxidative stress in very long chain fatty acid - treated oligodendrocytes and potentialization of ROS production using RNA interference-directed knockdown of ABCD1 and ACOX1 peroxisomal proteins. Neuroscience. 2012, 213: 1-18.CrossRefPubMed Baarine M, Andréoletti P, Athias A, Nury T, Zarrouk A, Ragot K, Vejux A, Riedinger JM, Kattan Z, Bessede G, Trompier D, Savary S, Cherkaoui-Malki M, Lizard G: Evidence of oxidative stress in very long chain fatty acid - treated oligodendrocytes and potentialization of ROS production using RNA interference-directed knockdown of ABCD1 and ACOX1 peroxisomal proteins. Neuroscience. 2012, 213: 1-18.CrossRefPubMed
53.
Zurück zum Zitat Zarrouk A, Vejux A, Nury T, El Hajj HI, Haddad M, Cherkaoui-Malki M, Riedinger JM, Hammami M, Lizard G: Induction of mitochondrial changes associated with oxidative stress on very long chain fatty acids (C22:0, C24:0, or C26:0)-treated human neuronal cells (SK-NB-E). Oxid Med Cell Longev. 2012, 10.1155/2012/623257. Zarrouk A, Vejux A, Nury T, El Hajj HI, Haddad M, Cherkaoui-Malki M, Riedinger JM, Hammami M, Lizard G: Induction of mitochondrial changes associated with oxidative stress on very long chain fatty acids (C22:0, C24:0, or C26:0)-treated human neuronal cells (SK-NB-E). Oxid Med Cell Longev. 2012, 10.1155/2012/623257.
54.
Zurück zum Zitat Kane CD, Francone OL, Stevens KA: Differential regulation of the cynomolgus, human, and rat acyl-CoA oxidase promoters by PPARalpha. Gene. 2006, 380: 84-94. 10.1016/j.gene.2006.05.011.CrossRefPubMed Kane CD, Francone OL, Stevens KA: Differential regulation of the cynomolgus, human, and rat acyl-CoA oxidase promoters by PPARalpha. Gene. 2006, 380: 84-94. 10.1016/j.gene.2006.05.011.CrossRefPubMed
55.
Zurück zum Zitat Gray E, Ginty M, Kemp K, Scolding N, Wilkins A: Peroxisome proliferator-activated receptor-α agonists protect cortical neurons from inflammatory mediators and improve peroxisomal function. Eur J Neurosci. 2011, 33: 1421-1432. 10.1111/j.1460-9568.2011.07637.x.CrossRefPubMed Gray E, Ginty M, Kemp K, Scolding N, Wilkins A: Peroxisome proliferator-activated receptor-α agonists protect cortical neurons from inflammatory mediators and improve peroxisomal function. Eur J Neurosci. 2011, 33: 1421-1432. 10.1111/j.1460-9568.2011.07637.x.CrossRefPubMed
56.
Zurück zum Zitat Anderson EJ, Yamazaki H, Neufer PD: Induction of endogenous uncoupling protein 3 suppresses mitochondrial oxidant emission during fatty acid-supported respiration. J Biol Chem. 2007, 282: 31257-31266. 10.1074/jbc.M706129200.CrossRefPubMed Anderson EJ, Yamazaki H, Neufer PD: Induction of endogenous uncoupling protein 3 suppresses mitochondrial oxidant emission during fatty acid-supported respiration. J Biol Chem. 2007, 282: 31257-31266. 10.1074/jbc.M706129200.CrossRefPubMed
57.
Zurück zum Zitat Reddy PH, McWeeney S, Park BS, Manczak M, Gutala RV, Partovi D, Jung Y, Yau V, Searles R, Mori M, Quinn J: Gene expression profiles of transcripts in amyloid precursor protein transgenic mice: up-regulation of mitochondrial metabolism and apoptotic genes is an early cellular change in Alzheimer’s disease. Hum Mol Genet. 2004, 13: 1225-1240. 10.1093/hmg/ddh140.CrossRefPubMed Reddy PH, McWeeney S, Park BS, Manczak M, Gutala RV, Partovi D, Jung Y, Yau V, Searles R, Mori M, Quinn J: Gene expression profiles of transcripts in amyloid precursor protein transgenic mice: up-regulation of mitochondrial metabolism and apoptotic genes is an early cellular change in Alzheimer’s disease. Hum Mol Genet. 2004, 13: 1225-1240. 10.1093/hmg/ddh140.CrossRefPubMed
58.
Zurück zum Zitat Crack PJ, Cimdins K, Ali U, Hertzog PJ, Iannello RC: Lack of glutathione peroxidase-1 exacerbates αβ-mediated neurotoxicity in cortical neurons. J Neural Transm. 2006, 113: 645-657. 10.1007/s00702-005-0352-y.CrossRefPubMed Crack PJ, Cimdins K, Ali U, Hertzog PJ, Iannello RC: Lack of glutathione peroxidase-1 exacerbates αβ-mediated neurotoxicity in cortical neurons. J Neural Transm. 2006, 113: 645-657. 10.1007/s00702-005-0352-y.CrossRefPubMed
59.
Zurück zum Zitat Barkats M, Millecamps S, Abrioux P, Geoffroy MC, Mallet J: Overexpression of glutathione peroxidase increases the resistance of neuronal cells to Abeta-mediated neurotoxicity. J Neurochem. 2000, 75: 1438-1446.CrossRefPubMed Barkats M, Millecamps S, Abrioux P, Geoffroy MC, Mallet J: Overexpression of glutathione peroxidase increases the resistance of neuronal cells to Abeta-mediated neurotoxicity. J Neurochem. 2000, 75: 1438-1446.CrossRefPubMed
60.
Zurück zum Zitat Lubos E, Loscalzo J, Handy DE: Glutathione peroxidase-1 in health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 2011, 15: 1957-1997. 10.1089/ars.2010.3586.PubMedCentralCrossRefPubMed Lubos E, Loscalzo J, Handy DE: Glutathione peroxidase-1 in health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 2011, 15: 1957-1997. 10.1089/ars.2010.3586.PubMedCentralCrossRefPubMed
61.
Zurück zum Zitat Fujiwara N, Nakano M, Kato S, Yoshihara D, Ookawara T, Eguchi H, Taniguchi N, Suzuki K: Oxidative modification to cysteine sulfonic acid of Cys111 in human copper-zinc superoxide dismutase. J Biol Chem. 2007, 282: 35933-35944. 10.1074/jbc.M702941200.CrossRefPubMed Fujiwara N, Nakano M, Kato S, Yoshihara D, Ookawara T, Eguchi H, Taniguchi N, Suzuki K: Oxidative modification to cysteine sulfonic acid of Cys111 in human copper-zinc superoxide dismutase. J Biol Chem. 2007, 282: 35933-35944. 10.1074/jbc.M702941200.CrossRefPubMed
62.
Zurück zum Zitat Murakami K, Murata N, Noda Y, Tahara S, Kaneko T, Kinoshita N, Hatsuta H, Murayama S, Barnham KJ, Irie K, Shirasawa T, Shimizu T: SOD1 (copper/zinc superoxide dismutase) deficiency drives amyloid β protein oligomerization and memory loss in mouse model of Alzheimer disease. J Biol Chem. 2011, 286: 44557-44568. 10.1074/jbc.M111.279208.PubMedCentralCrossRefPubMed Murakami K, Murata N, Noda Y, Tahara S, Kaneko T, Kinoshita N, Hatsuta H, Murayama S, Barnham KJ, Irie K, Shirasawa T, Shimizu T: SOD1 (copper/zinc superoxide dismutase) deficiency drives amyloid β protein oligomerization and memory loss in mouse model of Alzheimer disease. J Biol Chem. 2011, 286: 44557-44568. 10.1074/jbc.M111.279208.PubMedCentralCrossRefPubMed
63.
Zurück zum Zitat Nunomura A, Tamaoki T, Motohashi N, Nakamura M, McKeel DW, Tabaton M, Lee HG, Smith MA, Perry G, Zhu X: The earliest stage of cognitive impairment in transition from normal aging to Alzheimer disease is marked by prominent RNA oxidation in vulnerable neurons. J Neuropathol Exp Neurol. 2012, 71: 233-241. 10.1097/NEN.0b013e318248e614.PubMedCentralCrossRefPubMed Nunomura A, Tamaoki T, Motohashi N, Nakamura M, McKeel DW, Tabaton M, Lee HG, Smith MA, Perry G, Zhu X: The earliest stage of cognitive impairment in transition from normal aging to Alzheimer disease is marked by prominent RNA oxidation in vulnerable neurons. J Neuropathol Exp Neurol. 2012, 71: 233-241. 10.1097/NEN.0b013e318248e614.PubMedCentralCrossRefPubMed
64.
Zurück zum Zitat Singh M, Nam DT, Arseneault M, Ramassamy C: Role of by-products of lipid oxidation in Alzheimer’s disease brain: a focus on acrolein. J Alzheimers Dis. 2010, 21: 741-756.PubMed Singh M, Nam DT, Arseneault M, Ramassamy C: Role of by-products of lipid oxidation in Alzheimer’s disease brain: a focus on acrolein. J Alzheimers Dis. 2010, 21: 741-756.PubMed
65.
Zurück zum Zitat Dang TN, Arseneault M, Murthy V, Ramassamy C: Potential role of acrolein in neurodegeneration and in Alzheimer’s disease. Curr Mol Pharmacol. 2010, 3: 66-78.CrossRefPubMed Dang TN, Arseneault M, Murthy V, Ramassamy C: Potential role of acrolein in neurodegeneration and in Alzheimer’s disease. Curr Mol Pharmacol. 2010, 3: 66-78.CrossRefPubMed
66.
Zurück zum Zitat Shi R, Zhang Y, Shi Y, Shi S, Jiang L: Inhibition of peroxisomal β-oxidation by thioridazine increases the amount of VLCFAs and Aβ generation in the rat brain. Neurosci Lett. 2012, 528: 6-10. 10.1016/j.neulet.2012.08.086.CrossRefPubMed Shi R, Zhang Y, Shi Y, Shi S, Jiang L: Inhibition of peroxisomal β-oxidation by thioridazine increases the amount of VLCFAs and Aβ generation in the rat brain. Neurosci Lett. 2012, 528: 6-10. 10.1016/j.neulet.2012.08.086.CrossRefPubMed
67.
Zurück zum Zitat Clark J, Simon DK: Transcribe to survive: transcriptional control of antioxidant defense programs for neuroprotection in Parkinson’s disease. Antioxid Redox Signal. 2009, 11: 509-528. 10.1089/ars.2008.2241.CrossRefPubMed Clark J, Simon DK: Transcribe to survive: transcriptional control of antioxidant defense programs for neuroprotection in Parkinson’s disease. Antioxid Redox Signal. 2009, 11: 509-528. 10.1089/ars.2008.2241.CrossRefPubMed
68.
Zurück zum Zitat Wenz T: Mitochondria and PGC-1α in aging and Age-associated diseases. J Aging Res. 2011, 2011: 1-12.CrossRef Wenz T: Mitochondria and PGC-1α in aging and Age-associated diseases. J Aging Res. 2011, 2011: 1-12.CrossRef
69.
Zurück zum Zitat Zhang C, Rodriguez C, Spaulding J, Aw TY, Feng J: Age-dependent and tissue-related glutathione redox status in a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2012, 28: 655-666.PubMedCentralPubMed Zhang C, Rodriguez C, Spaulding J, Aw TY, Feng J: Age-dependent and tissue-related glutathione redox status in a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2012, 28: 655-666.PubMedCentralPubMed
70.
Zurück zum Zitat Casado A, Encarnación López-Fernández M, Concepción Casado M, de La Torre R: Lipid peroxidation and antioxidant enzyme activities in vascular and Alzheimer dementias. Neurochem Res. 2008, 33: 450-458. 10.1007/s11064-007-9453-3.CrossRefPubMed Casado A, Encarnación López-Fernández M, Concepción Casado M, de La Torre R: Lipid peroxidation and antioxidant enzyme activities in vascular and Alzheimer dementias. Neurochem Res. 2008, 33: 450-458. 10.1007/s11064-007-9453-3.CrossRefPubMed
71.
Zurück zum Zitat Massaad CA, Washington TM, Pautler RG, Klann E: Overexpression of SOD-2 reduces hippocampal superoxide and prevents memory deficits in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2009, 106: 13576-13581. 10.1073/pnas.0902714106.PubMedCentralCrossRefPubMed Massaad CA, Washington TM, Pautler RG, Klann E: Overexpression of SOD-2 reduces hippocampal superoxide and prevents memory deficits in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2009, 106: 13576-13581. 10.1073/pnas.0902714106.PubMedCentralCrossRefPubMed
72.
Zurück zum Zitat Amenta F, Bronzetti E, Sabbatini M, Vega JA: Astrocyte changes in aging cerebral cortex and hippocampus: a quantitative immunohistochemical study. Microsc Res Tech. 1998, 43: 29-33. 10.1002/(SICI)1097-0029(19981001)43:1<29::AID-JEMT5>3.0.CO;2-H.CrossRefPubMed Amenta F, Bronzetti E, Sabbatini M, Vega JA: Astrocyte changes in aging cerebral cortex and hippocampus: a quantitative immunohistochemical study. Microsc Res Tech. 1998, 43: 29-33. 10.1002/(SICI)1097-0029(19981001)43:1<29::AID-JEMT5>3.0.CO;2-H.CrossRefPubMed
73.
Zurück zum Zitat Mandrekar-Colucci S, Landreth GE: Microglia and inflammation in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2010, 9: 156-167. 10.2174/187152710791012071.CrossRefPubMed Mandrekar-Colucci S, Landreth GE: Microglia and inflammation in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2010, 9: 156-167. 10.2174/187152710791012071.CrossRefPubMed
74.
Zurück zum Zitat Li C, Zhao R, Gao K, Wei Z, Yin MY, Lau LT, Chui D, Hoi Yu AC: Astrocytes: implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2011, 8: 67-80. 10.2174/156720511794604543.CrossRefPubMed Li C, Zhao R, Gao K, Wei Z, Yin MY, Lau LT, Chui D, Hoi Yu AC: Astrocytes: implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2011, 8: 67-80. 10.2174/156720511794604543.CrossRefPubMed
75.
Zurück zum Zitat Arnold G, Holtzman E: Microperoxisomes in the central nervous system of the postnatal rat. Brain Res. 1978, 155: 1-17. 10.1016/0006-8993(78)90300-1.CrossRefPubMed Arnold G, Holtzman E: Microperoxisomes in the central nervous system of the postnatal rat. Brain Res. 1978, 155: 1-17. 10.1016/0006-8993(78)90300-1.CrossRefPubMed
76.
Zurück zum Zitat Cimini A, Moreno S, Giorgi M, Serafini B, Cerú MP: Purification of peroxisomal fraction from rat brain. Neurochem Int. 1993, 23: 249-260. 10.1016/0197-0186(93)90116-M.CrossRefPubMed Cimini A, Moreno S, Giorgi M, Serafini B, Cerú MP: Purification of peroxisomal fraction from rat brain. Neurochem Int. 1993, 23: 249-260. 10.1016/0197-0186(93)90116-M.CrossRefPubMed
77.
Zurück zum Zitat Cimini A, Singh I, Farioli-Vecchioli S, Cristiano L, Cerú MP: Presence of heterogeneous peroxisomal populations in the rat nervous tissue. Biochim Biophys Acta. 1998, 1425: 13-26. 10.1016/S0304-4165(98)00049-X.CrossRefPubMed Cimini A, Singh I, Farioli-Vecchioli S, Cristiano L, Cerú MP: Presence of heterogeneous peroxisomal populations in the rat nervous tissue. Biochim Biophys Acta. 1998, 1425: 13-26. 10.1016/S0304-4165(98)00049-X.CrossRefPubMed
78.
Zurück zum Zitat Gemma C, Vila J, Bachstetter A, Bickford PC: Oxidative stress and the aging brain: from theory to prevention. Brain aging: models, methods, and mechanisms. Edited by: Riddle DR. 2007, Boca Raton (FL): CRC Press, Chapter 15-Frontiers in Neuroscience Gemma C, Vila J, Bachstetter A, Bickford PC: Oxidative stress and the aging brain: from theory to prevention. Brain aging: models, methods, and mechanisms. Edited by: Riddle DR. 2007, Boca Raton (FL): CRC Press, Chapter 15-Frontiers in Neuroscience
79.
Zurück zum Zitat Filomeni G, Bolaños JP, Mastroberardino PG: Redox status and bioenergetics liaison in cancer and neurodegeneration. Int J Cell Bio. 2012, 2012: 659645- Filomeni G, Bolaños JP, Mastroberardino PG: Redox status and bioenergetics liaison in cancer and neurodegeneration. Int J Cell Bio. 2012, 2012: 659645-
80.
Zurück zum Zitat Röhl C, Armbrust E, Kolbe K, Lucius R, Maser E, Venz S, Gülden M: Activated microglia modulate astroglial enzymes involved in oxidative and inflammatory stress and increase the resistance of astrocytes to oxidative stress in vitro. Glia. 2008, 56: 1114-1126. 10.1002/glia.20683.CrossRefPubMed Röhl C, Armbrust E, Kolbe K, Lucius R, Maser E, Venz S, Gülden M: Activated microglia modulate astroglial enzymes involved in oxidative and inflammatory stress and increase the resistance of astrocytes to oxidative stress in vitro. Glia. 2008, 56: 1114-1126. 10.1002/glia.20683.CrossRefPubMed
81.
Zurück zum Zitat Cuajungco MP, Goldstein LE, Nunomura A, Smith MA, Lim JT, Atwood CS, Huang X, Farrag YW, Perry G, Bush AI: Evidence that the beta-amyloid plaques of Alzheimer’s disease represent the redox-silencing and entombment of abeta by zinc. J Biol Chem. 2000, 275: 19439-19442. 10.1074/jbc.C000165200.CrossRefPubMed Cuajungco MP, Goldstein LE, Nunomura A, Smith MA, Lim JT, Atwood CS, Huang X, Farrag YW, Perry G, Bush AI: Evidence that the beta-amyloid plaques of Alzheimer’s disease represent the redox-silencing and entombment of abeta by zinc. J Biol Chem. 2000, 275: 19439-19442. 10.1074/jbc.C000165200.CrossRefPubMed
82.
Zurück zum Zitat Weidner AM, Bradley MA, Beckett TL, Niedowicz DM, Dowling AL, Matveev SV, LeVine H, Lovell MA, Murphy MP: RNA oxidation adducts 8-OHG and 8-OHA change with Aβ42 levels in late-stage Alzheimer’s disease. PLoS One. 2011, 6: 1-6.CrossRef Weidner AM, Bradley MA, Beckett TL, Niedowicz DM, Dowling AL, Matveev SV, LeVine H, Lovell MA, Murphy MP: RNA oxidation adducts 8-OHG and 8-OHA change with Aβ42 levels in late-stage Alzheimer’s disease. PLoS One. 2011, 6: 1-6.CrossRef
83.
Zurück zum Zitat Moreno S, Nardacci R, Cimini A, Cerù MP: Immunocytochemical localization of D-amino acid oxidase in rat brain. J Neurocytol. 1999, 28: 169-185. 10.1023/A:1007064504007.CrossRefPubMed Moreno S, Nardacci R, Cimini A, Cerù MP: Immunocytochemical localization of D-amino acid oxidase in rat brain. J Neurocytol. 1999, 28: 169-185. 10.1023/A:1007064504007.CrossRefPubMed
84.
Zurück zum Zitat Farioli-Vecchioli S, Moreno S, Cerù MP: Immunocytochemical localization of acyl-CoA oxidase in the rat central nervous system. J Neurocytol. 2001, 30: 21-33. 10.1023/A:1011913223541.CrossRefPubMed Farioli-Vecchioli S, Moreno S, Cerù MP: Immunocytochemical localization of acyl-CoA oxidase in the rat central nervous system. J Neurocytol. 2001, 30: 21-33. 10.1023/A:1011913223541.CrossRefPubMed
85.
86.
Zurück zum Zitat Selkoe DJ: Preventing Alzheimer’s disease. Science. 2012, 337: 1488-1492. 10.1126/science.1228541.CrossRefPubMed Selkoe DJ: Preventing Alzheimer’s disease. Science. 2012, 337: 1488-1492. 10.1126/science.1228541.CrossRefPubMed
87.
Zurück zum Zitat Bradford MM: Rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976, 72: 248-254. 10.1016/0003-2697(76)90527-3.CrossRefPubMed Bradford MM: Rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976, 72: 248-254. 10.1016/0003-2697(76)90527-3.CrossRefPubMed
Metadaten
Titel
Age-dependent roles of peroxisomes in the hippocampus of a transgenic mouse model of Alzheimer’s disease
verfasst von
Francesca Fanelli
Sara Sepe
Marcello D’Amelio
Cinzia Bernardi
Loredana Cristiano
AnnaMaria Cimini
Francesco Cecconi
Maria Paola Ceru'
Sandra Moreno
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2013
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/1750-1326-8-8

Weitere Artikel der Ausgabe 1/2013

Molecular Neurodegeneration 1/2013 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.