Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and the leading cause of age‐related dementia worldwide. Several models for AD have been developed to provide information regarding the initial changes that lead to degeneration. Transgenic mouse models recapitulate many, but not all, of the features of AD, most likely because of the high complexity of the pathology. In this context, the validation of a wild‐type animal model of AD that mimics the neuropathological and behavioral abnormalities is necessary. In previous studies, we have reported that the Chilean rodent O ctodon degus could represent a natural model for AD. In the present work, we further describe the age‐related neurodegeneration observed in the O . degus brain. We report some histopathological markers associated with the onset progression of AD, such as glial activation, increase in oxidative stress markers, neuronal apoptosis and the expression of the peroxisome proliferative‐activated receptor γ coactivator‐1α (PGC‐1α). With these results, we suggest that the O . degus could represent a new model for AD research and a powerful tool in the search for therapeutic strategies against AD.
Keywords: Alzheimer' disease, glial activation, oxidative stress, metabolic sensors, neuronal apoptosis, natural model
Introduction
Alzheimer's disease (AD) is the most common form of dementia and affects nearly 10% of individuals over the age of 65 and nearly 50% of individuals over the age of 85. The increased longevity in the population, combined with the high incidence of AD in older adults, will only exacerbate the global impact on public health 13, 80. AD was first described over 100 years ago, but the etiology of AD is still not well understood, which limits the pharmacological treatment of the disease 66, 81. Different cellular and histopathological biomarkers have been described, including amyloid plaques, neurofibrillary tangles (NFTs), increased production of reactive oxygen species (ROS), mitochondrial dysfunction, decreased cerebral glucose consumption and altered autophagy processes 13, 29, 36, 40, 54, 75.
Transgenic mice have been the most useful tool in studying the pathological mechanisms of AD. However, these models do not recapitulate the entire spectrum of lesions present in human AD brains 17, 42, 84. Moreover, the overexpression of human transgenes in a nonphysiological scenario strongly influences the onset of histopathological features and cognitive decline observed in AD 16, 58. The poor reliability of the currently available AD models limits our understanding of AD pathophysiology and compromises the translation of preclinical data into human clinical trials. Thus, the identification and validation of a natural, wild‐type AD model that can mimic the pathological hallmarks observed in AD patients would be highly useful to unravel the mechanisms of AD and validate potential therapeutic targets. We have previously suggested that the Chilean rodent O. degus could represent a natural model to study the onset and progression of AD 4, 10, 60. We have described that the O. degus naturally develops extracellular amyloid plaques, NFT, failure in cholinergic transmission and hippocampal disconnection in an age‐related manner 5, 8, 34.
In this study, we used the brains of O. degus derived from several different aged (12–72 months) individuals to evaluate the progression of histopathological markers related to AD progression. We observed significant age‐dependent increase in astrocyte activation together with neuroinflammation, microglial markers and several oxidative stress markers, apoptotic signaling in critical brain areas, and alterations in cellular bioenergy regulators, such as peroxisome proliferative‐activated receptor γ coactivator‐1α (PGC‐1α). Taken together these results, we conclude that the spontaneous age‐related neurodegeneration reported in O. degus mimics the cellular and molecular events observed during the onset and progression of human AD.
Materials and Methods
Animals
O. degus were obtained from a breeding colony at the animal facility of the University of Valparaiso, Chile; the animals were maintained in a controlled room temperature (23 ± 1°C) under a 12:12 h light/dark cycle with water and food ad libitum 4, 5. At the time of behavioral characterization, O. degus of either sex or different ages were evaluated considering 12, 36, 60 and 72 months old. Animals were separated in two groups: young (12, 24 and 36 moths) and old (48, 60 and 72 months). The Ethics and Animal Care and Use Committee of the Pontifical Catholic University of Chile approved all animal work.
Glial activation
To study astrocyte and microglial activation, we used immunocytochemical staining and biochemical analysis with anti‐glial fibrillary acidic protein (anti‐GFAP) (DAKO, Carpentaria, CA, USA) and anti‐differentiation molecule 11B (anti‐CD11B) antibody (Serotec, Oxford, UK). A free‐floating immunocytochemical procedure was performed as previously described. Briefly, the brain sections were pretreated with 0.5% hydrogen peroxide (H2O2) for 30 minutes to reduce endogenous peroxidase activity followed by treatment with 3% bovine serum albumin at room temperature for 1 h to prevent nonspecific binding. Hippocampal tissue was homogenized in Radioimmunoprecipitation assay (RIPA) buffer [50 mM, Tris‐Cl, pH 7.5, 150 mM NaCl, 1% NP‐40, 0.5% sodium deoxycholate and 1% sodium dodecyl sulfate (SDS)] supplemented with a protease inhibitor cocktail (P8340, Sigma‐Aldrich, St. Louis, MO, USA) and phosphatase inhibitors (50 mM NaF, 1 mM Na3VO4 and 30 μM Na4P2O7) using a Potter homogenizer and then sequentially passed through decreasing caliber syringes from 25G × 5/8" to 28G × 1/2". Protein samples were centrifuged at 14 000 rpm at 4°C twice for 15 minutes. The protein concentration was determined using the BCA protein assay kit (Pierce, N Meridian Rd, Rockford, IL, USA). Twenty micrograms of hippocampal samples was resolved by 10% SDS‐polyacrylamide gel electrophoresis and transferred to a polyvinylidene difluoride membrane followed by incubation with a primary antibody, a secondary anti‐goat peroxidase conjugated antibody (Pierce) and ECL development according to the manufacturer's instructions (ECL, PerkinElmer, Waltham, MA, USA).
Oxidative stress markers
To study the progression of age‐related oxidative stress in the brain of O. degus, we used immunofluorescence and immunocytochemical techniques to measure the expression and distribution of two well‐described oxidative markers: 4‐hydroxynonenal (4‐HNE) and nitrotyrosine (N‐TYR). 4‐HNE is a marker of lipid peroxidation; for these experiments, we used a specific polyclonal antibody (AbD Serotec, Oxford, UK). N‐TYR is a marker of the oxidation of proteins generated by an increase in the production of nitric oxide (NO); we used a polyclonal antibody for detection (AbD Serotec, Oxford, UK). Our protocol has previously been described by our laboratory 19, 33, 90.
Interleukin‐6 measurement
To study the effects of age on inflammation in the brain of O. degus, we examined the expression and localization of the inflammatory marker interleukin 6 (IL‐6) using a specific antibody (Abcam, Cambridge, MA, USA). The protocol was performed as previously described by Ansar et al 3.
Metabolic sensors and autophagy rate
To study the effects of aging on the expression of metabolic sensors in the brain of O. degus, we studied the expression of PGC‐1α, AMPK, activated AMPK and the autophagy marker LC3B via immunofluorescence and Western blot. For Western blot analysis, we used an anti‐phospho‐Thr172‐AMPK antibody, an anti‐total AMPK antibody (Cell Signalling, Danvers, PA, USA), an anti‐LC3B (Cell Signalling) for detection of band I (16 kDa) and lipidate band II (14 kDa). For complementary autophagy studies, we used an anti‐p62 antibody (Cell Signalling). Our Western blot protocol was previously described 5. Immunofluorescence was performed with the same antibodies used in the Western blot analysis with the previously described protocol.
Neuronal cell death
Neuronal cell death measured by microscopic analysis with Hoechst staining, which enabled the number of condensed/pyknotic nuclei in the brain slices to be quantified, and the anti‐caspase‐3 (casp‐3)‐active antibody for an immunofluorescence assay (Chemicon International, Temecula, CA, USA). Both protocols were carried out as previously described 90.
Image analysis
The stained brain sections photographed using an Olympus BX51 microscope coupled to a Micro‐publisher 3.3 RTV camera (QImaging Surrey, BC, Canada). The images were analyzed with ImageJ v.1.40g software (NIH, Bethesda, MD). The selection of the measurement areas was performed by manual threshold adjustment or by direct manual selection of regions of interest (ROIs) in heterogeneous stains. Images of neuron immunofluorescence were captured with a Zeiss LSM 5 Pascal confocal microscope (Jena, Germany). We typically examined a series of 15–20 confocal layers, which represented the fluorescence data from the ROI. The relative intensities of the protein bands were analyzed using ImageJ software. Statistical analysis was performed using GraphPad InStat version 3.00 (GraphPad Software, San Diego, CA, USA).
Statistical analysis
Results expressed as the mean ± standard error of the mean. The data were analyzed by one‐way analysis of variance followed by Bonferroni's post hoc test. Asterisks indicate the statistical significance of the observed differences (*P < 0.05; **P < 0.01). The statistical analyses performed using Prism software (GraphPad Software).
Results
Astrocytes and microglia are activated in the brains of aging O . degus
Increased astrogliosis and microgliosis is an undisputed feature of AD brains with increased Aβ loads 49, 65. Brain sections obtained from O. degus of different ages (12–72 months) were stained with a specific antibody against GFAP in two regions of the brain: the hippocampus and the cortex. Representative micrographs are shown in Figure 1A; detailed images are shown for GFAP in the inset. The graph (Figure 1B) represents the staining intensity in the two regions. Total levels of GFAP in hippocampus were significantly increased in eightfold old animals (60 and 72 months old) compared with the young animals. However, we do not observed an increase in the cortex region (Figure 1B). Total protein levels of GFAP and the microglial marker CD11b were evaluated in the hippocampus and cortex (Figure 1C). The levels of CD11b in the hippocampus increased approximately twofold in the old animals compared with the young animals, whereas no changes were observed in GFAP (Figure 1C). We do not observe an increase in CD11b and GFAP in the cortex region. These results indicate that glial activation is observed in old animals at different brain regions of O. degus, and this trend is similar to the one observed in aging humans and during the progression of AD‐like pathology.
Figure 1.

Levels of glial fibrillary acidic protein ( GFAP ) and CD11b in the brains of Octodon degus at different ages. A . Representative images of the cortex and hippocampus of young and old O . degus stained with GFAP antibody. Inset shows a magnification of the immunostaining in individual cells. B . Graph shows the quantification of GFAP levels in the brains of O . degus in young and old animals. C. Western blot shows the levels of CD11b and GFAP proteins in the hippocampus. D . Graph shows the quantification of both protein levels. E . Blot shows the levels of CD11b and GFAP in the cortex. F . Graph shows the quantification of both protein levels. Data are presented as the mean ± standard error of the mean. Significant differences were calculated using analysis of variance followed by a post hoc Bonferroni test. Asterisks indicate statistical significance (*P < 0.05; ***P < 0.001).
Brains of aging O . degus present high levels of oxidative stress markers
Oxidative stress is triggered by an imbalance between oxidants and antioxidants in favor of the formation of oxidized compounds and results in damage to membranes, proteins and DNA 6. Extensive and early work demonstrated that oxidative stress plays a role in the onset of AD 7, 20, 43, 89. Tissue oxidative stress markers, including lipid peroxidation, protein oxidation, DNA oxidation, and structurally and functionally damaged mitochondria are major sources of ROS 53, 83. We analyzed two markers of oxidative stress in the brain of O. degus, including an aldehyde product of lipid peroxidation, 4‐HNE (Figure 2A,B) and N‐TYR, a marker of NO damage (Figure 2C,D). Both markers were analyzed in the brain at different ages. As expected, older O. degus showed increased levels of 4‐HNE. 4‐HNE was increased over ninefold compared with the young animals in the hippocampal region (Figure 2B). The N‐TYR levels were determined using immunohistochemistry. We analyzed the intensity of the signal in the neuronal soma of the young and older animals. We found a fourfold increase of tyrosine nitration in old animals compared with the young animals in the hippocampus (Figure 2D). These data support that the aging in O. degus trigger an increase in the oxidative stress in the hippocampus.
Figure 2.

Levels of 4‐hydroxynonenal (4‐ HNE ) and nitrotyrosine ( N ‐ TYR ) in the brains of Octodon degus at different ages. A . Representative images showing immunodetection of 4‐HNE in the brains of young and old O . degus. B . Graph showing the quantification of the proteins in the hippocampus and cortex. C and D. Images of N‐TYR staining in the sections of cortex and hippocampus of young and old animals, and a graph showing the levels of N‐TYR species in the hippocampus and cortex. Data are presented as the mean ± standard error of the mean. Significant differences were calculated using analysis of variance followed by a post hoc Bonferroni test. Asterisks indicate statistical significance (*P < 0.05; ***P < 0.001).
Brains of O . degus present high levels of IL‐6
Neuroinflammation represents one of several cellular and neurochemical processes that may underlie neurodegeneration 1, 27, 59. As a prototypical cytokine with roles in the control of inflammation, IL‐6 is altered in many central nervous system diseases. IL‐6 is overexpressed in the brains of AD patients and increase around amyloid plaques and in the cerebrospinal fluid 2, 32. We measured IL‐6 in the hippocampus and cortex of O. degus at different ages using immunofluorescence (Figure 3A) and quantified the results by densitometric analysis (Figure 3B). We found a two‐ to threefold increase in the levels of IL‐6 in the hippocampus and cortex in the older animals compared with the younger animals.
Figure 3.

Levels of interleukin‐6 ( IL ‐6) in the brains of Octodon degus at different ages. A . Representative images showing immunodetection of IL‐6 in the brains of young and older O . degus. The sections were counter stained with Hoechst. B . Graph showing the quantification of IL‐6 in the hippocampus and cortex. Data are represented as the mean ± standard error of the mean. Significant differences were calculated using analysis of variance followed by a post hoc Bonferroni test. Asterisks indicate statistical significance (*P < 0.05; **P < 0.01).
Metabolic sensors in the brains of aging O . degus
To detect the active form of AMPK in the hippocampus and cortex of young and old O. degus, we conducted both immunofluorescence and Western blot analyses. We observed only a tendency to increase in phosphorylated AMPK in hippocampus in the old animals compared with the young animals by immunofluorescence (Figure 4A,B), whereas no changes were observed in the total levels of active AMPK measured by Western blot in the hippocampus (Figure 4C,D).
Figure 4.

AMPK and autophagy markers, two associated metabolic elements, in the brains of Octodon degus. A and B. Representative images of cortical and hippocampal sections immunostained to detect pThr172‐AMPK in young and old animals. The graph shows the levels of active AMPK in both areas. C and D. Blots show the phosphorylated and total AMPK in the hippocampus. The graph shows the levels of phosphorylated AMPK in young and old animals. E and F. Autophagy markers were studied in the hippocampus of young and old animals. The graph shows the ratio of LC3‐II/LC3‐I and p62 protein in young and old animals. Data are represented as the mean ± standard error of the mean. Significant differences were calculated using analysis of variance followed by a post hoc Bonferroni test.
We also investigated the LC3‐I to LC3‐II protein conversion rate and p62 protein levels in the brains of the young and old animals. These proteins are critical in pre‐autophagosomal membrane formation and autophagy is deregulated in AD 55, 78. We found that the ratio of LC3‐II/LC3‐I does not change in the hippocampal tissues of old O. degus. Protein p62 plays an essential role in canonical autophagy by selectively detecting damaged cargo for degradation; it also plays a putative role in the NFTs observed in AD brains 41. Although the levels of p62 have the tendency to increase, we observed that these changes were not significantly different (Figure 4E,F).
PGC‐1α response in the brains of aging O . degus
PGC‐1α is a potent stimulator of mitochondrial biogenesis that enhances the transcription of several key ROS‐detoxifying enzymes (glutathione peroxidase‐1 and superoxide dismutase 2) 86. Recently, several studies have reported that the amyloid precursor protein (APP) increases PGC‐1α expression in the murine brain 69. Thus, we evaluated the levels of PGC‐1α using immunofluorescence in the hippocampus and cortex. The levels of PGC‐1α were significantly higher (twofold) in the hippocampus of the old animals than young animals (Figure 5A,B).
Figure 5.

Peroxisome proliferative‐activated receptor γ coactivator‐1α ( PGC ‐1α) increased in the brains of Octodon degus. A and B. Representative images of PGC‐1α immunoreactivity in young and old O . degus. The graph shows the quantification in the cortex and hippocampus. Data are represented as the mean ± standard error of the mean. Significant differences were calculated using analysis of variance followed by a post hoc Bonferroni test. Asterisks indicate statistical significance (*P < 0.05).
Markers of apoptotic neurons in the brains of aging O . degus
Apoptosis has been associated with several neurodegenerative pathologies 26. We performed nuclear staining using Hoechst and studied the expression of activated casp‐3 in the hippocampus and cortex at different ages. The number of condensed nuclei did not change in the cortex of animals of different ages. However, in the hippocampus, we observed a notable increase in apoptotic nuclei, which progressed with age. The increase was four times greater in the older animals compared with the younger animals (Figure 6A,B). Based on this observation, we measured the expression of the apoptotic marker‐activated casp‐3 as observed in AD and other neurological disorders 9, 71, 72. Activated casp‐3 increased in the old animals compared with the young animals particularly in the hippocampus (Figure 6C,D).
Figure 6.

Apoptosis markers in the brains of Octodon degus. A and B . Representative images of Hoechst staining in the young and old O . degus. The graph shows the quantification of condensed nuclei in the cortex and hippocampus. B and D. Images show active caspase‐3 immunodetection in the hippocampus and cortex of the young and older O . degus. The graph shows that the levels of active caspase‐3 in the hippocampus and cortex are in young and old animals. Data represented as the mean ± standard error of the mean. Significant differences were calculated using analysis of variance followed by a post hoc Bonferroni test. Asterisks indicate statistical significance (*P < 0.05; ***P < 0.001).
Discussion
Research on neurodegenerative disorders as well as other pathologies was limited by the reliability of available disease models. In several cases disease models are based on the introduction and/or expression of foreign genes in a specific organism, such as the transgenic models of AD. Even though these models are widely used, the rate of success of new therapeutic intervention strategies or therapeutic alternatives derived from the models is quite poor 12, 23, 38, 42, 46. AD and other neurodegenerative disorders are highly complex pathologies, with several critical factors that interact during the establishment and progression of the disease. Unfortunately, none of the current models are able to mimic the full range of alterations that occur during AD 66. A natural model of AD could provide a powerful tool to understand the complexity of AD neuropathology. Previously, Inestrosa et al 34 demonstrated that the Aβ peptide shares the same amino acid sequence in humans and O. degus, with only one amino acid difference 34. Subsequently, we demonstrated neural plasticity impairments prior to the appearance of amyloid and tau deposition 5. van Groen et al 82 have demonstrated that Aβ deposits in the blood vessel walls precede the deposits of Aβ and tau in the hippocampus of 3‐year‐old animals 82.
In the present work, we used a comprehensive approach to identify several age‐related neurocytopathological alterations related to the onset and progression of AD. Moreover, our findings demonstrate that the brains of aged O. degus exhibit a wide range of differential biomarkers related to aging and human AD neuropathology. Our findings provide further support to hypothesis that O. degus represents a natural wild‐type rodent model of AD 8, 34, 42.
We observed age‐related glial cell activation. In young animals, we identified normal expression of the astrocyte marker GFAP and the microglial marker CD11b. However, in old subjects, both markers were increased. The activation of astrocytes and microglia has been described in several models of neurodegenerative disease and has been postulated to represent a common alteration in AD in response to neuronal damage 37, 56. Neuronal damage in AD induced by Aβ aggregates (amyloid fibrils and Aβ oligomers) could be an important triggering factor for the activation of glial cells, which is an important step in the pathology of the disease. The activation of glial cells is a well‐recognized trigger of an inflammatory response in the progression to brain damage 31, 44, 56, 73, 74. A direct relationship between glial and microglia activation is mediated by systematic inflammation and IL cross talk. The activation of glial cells induces the production of IL‐6 by blood cells, including T‐cell and monocytes. In brain tissue, IL‐6 stimulates several process related to protection against brain injury, including tissue remodeling, reduction of oxidative stress and inhibition of apoptosis induced by an increase in intracellular Ca2+ 21, 39, 57. However, the molecular mechanisms responsible for the effects of IL‐6 remain unclear. In the O. degus model, we observed an increase in the levels of IL‐6 in old animals. This increase could represent a response against the oxidative stress and massive neuronal death observed in the final stages of the animal's life. The relative importance of the inflammatory process in age‐related AD in O. degus deserves further studies.
Glial activation and inflammatory processes are present in several neurodegenerative diseases, including AD. Both processes have also been correlated with oxidative stress 30, 45, 51. In the same line, the administration of an anti‐oxidant, such as vitamin E, or an anti‐inflammatory drug, such as indomethacin, causes an additive anti‐amyloidogenic effect in some mouse models of AD 87. In our model, we observed an increase in the expression of two markers of oxidative stress: 4‐HNE and N‐TYR. 4‐HNE is a marker of lipid peroxidation. N‐TYR is a product of tyrosine nitration mediated by reactive nitrogen species, such as peroxy‐nitrite anion and nitrogen dioxide, which are markers of inflammation and NO production 77. The increase in both markers was primarily observed in the hippocampus, which is similar to the changes described in AD pathology 47, 48, 85. In fact, the damage induced by oxidative stress in the hippocampus has been suggested as critical to the failure of cognitive performance in AD patients 62, 64, 79, 89. Moreover, we also observed increased levels of additional cell death markers, such as activated casp‐3, which is typically considered the terminal step in the biochemical cascade that leads to apoptosis, which has been for a long time a clear trend in AD 35, 50, 67, 70. The role of casp‐3 activation in AD has been recently emphasized in vitro in Aβ‐challenged neurons and the brains of individuals with AD 76.
Oxidative stress affects several processes in the brain, including DNA homeostasis, protein oxidation and mitochondrial dysfunction. Several groups have described that mitochondrial failure is a critical step in the progression of AD because of its fundamental role in neuronal metabolism 24, 83, 89. Altered cell metabolism and energy imbalances represent additional variables that explain some reported biochemical changes in AD 22, 36. In the clinic, diabetes and some cardiometabolic disorders have been correlated with an increased risk of dementia and AD development because of an altered energy balance 52, 68. Some authors have even defined AD as a “diabetic brain” primarily because of a molecular link between diabetes and AD, which includes brain resistance to insulin 11, 14, 15, 18 and results in the deregulation of memory processes. This “cognitive–metabolic syndrome” might include altered cellular energy sensors, such as AMPK and PGC‐1α 25. PGC‐1α has been associated with the mitochondrial health status and has been proposed to play a major role in the modulation of mitochondrial fusion–fission events, which provide protection against Aβ‐derived oxidative damage 69, 91. An altered expression of the active form of AMPK (pThr172‐AMPK) has been observed in AD transgenic mice models, such as the APPswe‐PSEN1dE9 28, 63. We evaluated these markers in the brains of O. degus and found increased expression only in the case of PGC‐1α, which suggests a compensatory mechanism to overcome Aβ‐induced damage and prevent neuronal metabolic stress 22.
A general hypothesis is that the augmentation of autophagy flux capacity could represent a potential mechanism of the clearance of tau intracellular deposits 61. LC3 is involved in membrane recruitment, and p62 is involved in auto‐phagosomal elongation 41. In our model, both markers (mainlyp62) exhibited similar increases, which replicates the pathology in other animal models of AD (APP/PS1) and in the human post‐mortem brain 88. We were not able to find changes in LC3 and p62 markers suggesting a difference with the pathology in other models 88. In conclusion, we measured several tissue biomarkers in O. degus of different ages that have been described as characteristic of AD, including the classic hallmarks and emerging markers for inflammation (gliosis and microgliosis), oxidative stress (4‐HNE and N‐TYR), neuronal death and the metabolic stress marker PGC‐1α. A summary of the observed changes is in Figure 7. We present evidence for a high correlation between the pathological changes that occur during the aging process in O. degus and the changes described in human AD. Taken together, the previously reported evidence and the research presented here provide a strong platform to consider O. degus as a reliable “natural” model to investigate the pathobiology of AD. The potential uses of this model are wide: it could increase our knowledge regarding aging and neurodegeneration, enable the development of more efficient staging, and provide novel therapies to slow down or ameliorate AD progression.
Figure 7.

Old O ctodon degus brains recapitulated the main pathophysiological alterations observed in Alzheimer's disease (AD) individuals. In this diagram, we summarize the most important phenomena observed during aging of O . degus. These mechanisms are responsible for the neuropathological lesions previously described in our studies. In the top left panel, we show two representative animals, young (12–36 months) and old (60–72 months), with an emphasis on the eye of the animal that indicates the presence of cataracts, which is a classic finding in both wild and captive old O . degus. The insets indicate the underlying molecular disturbances that occur in the O . degus hippocampus (green). In the top right panel (neuroinflammation), activated glial cells surrounded by the two classical neuropathological AD hallmarks, amyloid plaques and neurofibrillary tangles. Cytokines (interleukin 6) mediate the cross talk between glial and microglial cells. In the bottom left panel (oxidative stress), mitochondrial damage is a source of reactive oxygen species, which enhance the formation of 4‐hydroxynonenal and nitrotyrosine. The bottom right panel (metabolic stress and neuronal death) emphasizes how an increase in peroxisome proliferative‐activated receptor γ coactivator‐1α and possible p62 might protect against the neuronal damage, and in parallel, it shows the apoptotic hallmarks of AD (condensed nuclei and activation of the caspase pathway).
Acknowledgments
This work was supported by grants PFB 12/2007 from the Basal Centre for Excellence in Science and Technology, Fondecyt 1120156 to NCI, predoctoral fellowships to JAR and CT‐R from CONICYT and postdoctoral fellowships to DSR and AOA from Fondecyt and to PC from CARE. FJC was a pre‐doctoral fellow from the VRI, Catholic University of Chile. NB is a recipient of the Alzheimer's Australia and NHMRC Early Career Postdoctoral Research Fellowship at the University of New South Wales, Sydney, Australia. We also thank the Sociedad Química y Minera de Chile (SQM) for a special grant to study “The role of potassium in hypertension and cognition.” Graphic work was performed with Graphique‐Science (http://www.illustrative‐science.com/).
References
- 1. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM et al (2000) Inflammation and Alzheimer's disease. Neurobiol Aging 21:383–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Angelis P, Scharf S, Mander A, Vajda F, Christophidis N (1998) Serum interleukin‐6 and interleukin‐6 soluble receptor in Alzheimer's disease. Neurosci Lett 244:106–108. [DOI] [PubMed] [Google Scholar]
- 3. Ansar S, Chatzikonstantinou E, Thiagarajah R, Tritschler L, Fatar M, Hennerici MG, Meairs S (2014) Pro‐inflammatory mediators and apoptosis correlate to rt‐PA response in a novel mouse model of thromboembolic stroke. PLoS ONE 9:e85849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Ardiles AO, Ewer J, Acosta ML, Kirkwood A, Martinez AD, Ebensperger LA et al (2013) Octodon degus (Molina 1782): a model in comparative biology and biomedicine. Cold Spring Harb Protoc 2013:312–318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Ardiles AO, Tapia‐Rojas CC, Mandal M, Alexandre F, Kirkwood A, Inestrosa NC, Palacios AG (2012) Postsynaptic dysfunction is associated with spatial and object recognition memory loss in a natural model of Alzheimer's disease. Proc Natl Acad Sci U S A 109:13835–13840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Barnham KJ, Masters CL, Bush AI (2004) Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov 3:205–214. [DOI] [PubMed] [Google Scholar]
- 7. Bonda DJ, Wang X, Perry G, Nunomura A, Tabaton M, Zhu X, Smith MA (2010) Oxidative stress in Alzheimer disease: a possibility for prevention. Neuropharmacology 59:290–294. [DOI] [PubMed] [Google Scholar]
- 8. Braidy N, Munoz P, Palacios AG, Castellano‐Gonzalez G, Inestrosa NC, Chung RS et al (2012) Recent rodent models for Alzheimer's disease: clinical implications and basic research. J Neural Transm 119:173–195. [DOI] [PubMed] [Google Scholar]
- 9. Bredesen DE (2009) Neurodegeneration in Alzheimer's disease: caspases and synaptic element interdependence. Mol Neurodegener 4:27. doi: 10.1186/1750-1326-4-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Brown C, Donnelly TM (2001) Cataracts and reduced fertility in degus (Octodon degus). Contracts secondary to spontaneous diabetes mellitus. Lab Anim 30:25–26. [PubMed] [Google Scholar]
- 11. Butterfield DA, Di Domenico F, Barone E (2014) Elevated risk of type 2 diabetes for development of Alzheimer disease: a key role for oxidative stress in brain. Biochim Biophys Acta 1842:1693–1706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Casadesus G, Arendash G, Laferla F, McDonald M (2010) Animal models of Alzheimer's disease. Int J Alzheimer's Dis 2010:606357. doi: 10.4061/2010/606357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Chen Z, Zhong C (2013) Decoding Alzheimer's disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies. Prog Neurobiol 108:21–43. [DOI] [PubMed] [Google Scholar]
- 14. Cole GM, Frautschy SA (2007) The role of insulin and neurotrophic factor signaling in brain aging and Alzheimer's disease. Exp Gerontol 42:10–21. [DOI] [PubMed] [Google Scholar]
- 15. Craft S, Cholerton B, Baker LD (2013) Insulin and Alzheimer's disease: untangling the web. J Alzheimer's Dis 33(Suppl. 1):S263–S275. [DOI] [PubMed] [Google Scholar]
- 16. Crews L, Masliah E (2010) Molecular mechanisms of neurodegeneration in Alzheimer's disease. Hum Mol Genet 19(R1):R12–R20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Crews L, Rockenstein E, Masliah E (2008) Biological transgenic mouse models of Alzheimer's disease. Handb Clin Neurol 89:291–301. [DOI] [PubMed] [Google Scholar]
- 18. de la Monte SM, Tong M (2014) Brain metabolic dysfunction at the core of Alzheimer's disease. Biochem Pharmacol 88:548–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Dinamarca MC, Arrazola M, Toledo E, Cerpa WF, Hancke J, Inestrosa NC (2008) Release of acetylcholinesterase (AChE) from beta‐amyloid plaques assemblies improves the spatial memory impairments in APP‐transgenic mice. Chem Biol Interact 175:142–149. [DOI] [PubMed] [Google Scholar]
- 20. Dumont M, Beal MF (2011) Neuroprotective strategies involving ROS in Alzheimer disease. Free Radic Biol Med 51:1014–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Erta M, Quintana A, Hidalgo J (2012) Interleukin‐6, a major cytokine in the central nervous system. Int J Biol Sci 8:1254–1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Ferreira IL, Resende R, Ferreiro E, Rego AC, Pereira CF (2010) Multiple defects in energy metabolism in Alzheimer's disease. Curr Drug Targets 11:1193–1206. [DOI] [PubMed] [Google Scholar]
- 23. Franco R, Cedazo‐Minguez A (2014) Successful therapies for Alzheimer's disease: why so many in animal models and none in humans? Front Pharmacol 5:146. doi: 10.3389/fphar.2014.00146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Garcia‐Escudero V, Martin‐Maestro P, Perry G, Avila J (2013) Deconstructing mitochondrial dysfunction in Alzheimer disease. Oxid Med Cell Longev 2013:162152. doi: 10.1155/2013/162152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Godoy JA, Rios JA, Zolezzi JM, Braidy N, Inestrosa NC (2014) Signaling pathway cross talk in Alzheimer's disease. Cell Commun Signal 12:23. doi: 10.1186/1478-811X-12-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Gorman AM (2008) Neuronal cell death in neurodegenerative diseases: recurring themes around protein handling. J Cell Mol Med 12(6A):2263–2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Griffin WS (2013) Neuroinflammatory cytokine signaling and Alzheimer's disease. N Engl J Med 368:770–771. [DOI] [PubMed] [Google Scholar]
- 28. Guo XL, Li D, Hu F, Song JR, Zhang SS, Deng WJ et al (2012) Targeting autophagy potentiates chemotherapy‐induced apoptosis and proliferation inhibition in hepatocarcinoma cells. Cancer Lett 320:171–179. [DOI] [PubMed] [Google Scholar]
- 29. Gustaw‐Rothenberg K, Lerner A, Bonda DJ, Lee HG, Zhu X, Perry G, Smith MA (2010) Biomarkers in Alzheimer's disease: past, present and future. Biomark Med 4:15–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Haass C, Selkoe DJ (2007) Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta‐peptide. Nat Rev Mol Cell Biol 8:101–112. [DOI] [PubMed] [Google Scholar]
- 31. Hayward JH, Lee SJ (2014) A decade of research on TLR2 discovering its pivotal role in glial activation and neuroinflammation in neurodegenerative diseases. Exp Neurobiol 23:138–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Hull M, Fiebich BL, Lieb K, Strauss S, Berger SS, Volk B, Bauer J (1996) Interleukin‐6‐associated inflammatory processes in Alzheimer's disease: new therapeutic options. Neurobiol Aging 17:795–800. [DOI] [PubMed] [Google Scholar]
- 33. Ill‐Raga G, Ramos‐Fernandez E, Guix FX, Tajes M, Bosch‐Morato M, Palomer E et al (2010) Amyloid‐beta peptide fibrils induce nitro‐oxidative stress in neuronal cells. J Alzheimer's Dis 22:641–652. [DOI] [PubMed] [Google Scholar]
- 34. Inestrosa NC, Reyes AE, Chacon MA, Cerpa W, Villalon A, Montiel J et al (2005) Human‐like rodent amyloid‐beta‐peptide determines Alzheimer pathology in aged wild‐type Octodon degu . Neurobiol Aging 26:1023–1028. [DOI] [PubMed] [Google Scholar]
- 35. Ji C, Amarnath V, Pietenpol JA, Marnett LJ (2001) 4‐hydroxynonenal induces apoptosis via caspase‐3 activation and cytochrome c release. Chem Res Toxicol 14:1090–1096. [DOI] [PubMed] [Google Scholar]
- 36. Kapogiannis D, Mattson MP (2011) Disrupted energy metabolism and neuronal circuit dysfunction in cognitive impairment and Alzheimer's disease. Lancet Neurol 10:187–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Kaushik DK, Basu A (2013) A friend in need may not be a friend indeed: role of microglia in neurodegenerative diseases. CNS Neurol Disord Drug Targets 12:726–740. [DOI] [PubMed] [Google Scholar]
- 38. Kitazawa M, Medeiros R, Laferla FM (2012) Transgenic mouse models of Alzheimer disease: developing a better model as a tool for therapeutic interventions. Curr Pharm Des 18:1131–1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Kossmann T, Hans VH, Imhof HG, Stocker R, Grob P, Trentz O, Morganti‐Kossmann C (1995) Intrathecal and serum interleukin‐6 and the acute‐phase response in patients with severe traumatic brain injuries. Shock 4:311–317. [DOI] [PubMed] [Google Scholar]
- 40. Kragh CL, Ubhi K, Wyss‐Coray T, Masliah E (2012) Autophagy in dementias. Brain Pathol 22:99–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Kuusisto E, Salminen A, Alafuzoff I (2002) Early accumulation of p62 in neurofibrillary tangles in Alzheimer's disease: possible role in tangle formation. Neuropathol Appl Neurobiol 28:228–237. [DOI] [PubMed] [Google Scholar]
- 42. LaFerla FM, Green KN (2012) Animal models of Alzheimer disease. Cold Spring Harb Perspect Med 2(11). pii: a006320. doi: 10.1101/cshperspect.a006320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Lee HP, Zhu X, Casadesus G, Castellani RJ, Nunomura A, Smith MA et al (2010) Antioxidant approaches for the treatment of Alzheimer's disease. Expert Rev Neurother 10:1201–1208. [DOI] [PubMed] [Google Scholar]
- 44. Lee JE, Han PL (2013) An update of animal models of Alzheimer disease with a reevaluation of plaque depositions. Exp Neurobiol 22:84–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Lesne SE, Sherman MA, Grant M, Kuskowski M, Schneider JA, Bennett DA, Ashe KH (2013) Brain amyloid‐beta oligomers in ageing and Alzheimer's disease. Brain 136(Pt 5):1383–1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Lithner CU, Hedberg MM, Nordberg A (2011) Transgenic mice as a model for Alzheimer's disease. Curr Alzheimer Res 8:818–831. [DOI] [PubMed] [Google Scholar]
- 47. Liu Q, Raina AK, Smith MA, Sayre LM, Perry G (2003) Hydroxynonenal, toxic carbonyls, and Alzheimer disease. Mol Aspects Med 24:305–313. [DOI] [PubMed] [Google Scholar]
- 48. Mark RJ, Lovell MA, Markesbery WR, Uchida K, Mattson MP (1997) A role for 4‐hydroxynonenal, an aldehydic product of lipid peroxidation, in disruption of ion homeostasis and neuronal death induced by amyloid beta‐peptide. J Neurochem 68:255–264. [DOI] [PubMed] [Google Scholar]
- 49. Medeiros R, LaFerla FM (2013) Astrocytes: conductors of the Alzheimer disease neuroinflammatory symphony. Exp Neurol 239:133–138. [DOI] [PubMed] [Google Scholar]
- 50. Migheli A, Cavalla P, Piva R, Giordana MT, Schiffer D (1994) Bcl‐2 protein expression in aged brain and neurodegenerative diseases. Neuroreport 5:1906–1908. [DOI] [PubMed] [Google Scholar]
- 51. Miranda S, Opazo C, Larrondo LF, Munoz FJ, Ruiz F, Leighton F, Inestrosa NC (2000) The role of oxidative stress in the toxicity induced by amyloid beta‐peptide in Alzheimer's disease. Prog Neurobiol 62:633–648. [DOI] [PubMed] [Google Scholar]
- 52. Misiak B, Leszek J, Kiejna A (2012) Metabolic syndrome, mild cognitive impairment and Alzheimer's disease—the emerging role of systemic low‐grade inflammation and adiposity. Brain Res Bull 89:144–149. [DOI] [PubMed] [Google Scholar]
- 53. Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G (2010) Mitochondrial dysfunction is a trigger of Alzheimer's disease pathophysiology. Biochim Biophys Acta 1802:2–10. [DOI] [PubMed] [Google Scholar]
- 54. Moreira PI, Honda K, Liu Q, Santos MS, Oliveira CR, Aliev G et al (2005) Oxidative stress: the old enemy in Alzheimer's disease pathophysiology. Curr Alzheimer Res 2:403–408. [DOI] [PubMed] [Google Scholar]
- 55. Moreira PI, Santos RX, Zhu X, Lee HG, Smith MA, Casadesus G, Perry G (2010) Autophagy in Alzheimer's disease. Expert Rev Neurother 10:1209–1218. [DOI] [PubMed] [Google Scholar]
- 56. Mosher KI, Wyss‐Coray T (2014) Microglial dysfunction in brain aging and Alzheimer's disease. Biochem Pharmacol 88:594–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Murakami M, Hirano T (2012) The pathological and physiological roles of IL‐6 amplifier activation. Int J Biol Sci 8:1267–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H, Cairns NJ et al (2012) Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol 71:362–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Niranjan R (2013) Molecular basis of etiological implications in Alzheimer's disease: focus on neuroinflammation. Mol Neurobiol 48:412–428. [DOI] [PubMed] [Google Scholar]
- 60. Nishi M, Steiner DF (1990) Cloning of complementary DNAs encoding islet amyloid polypeptide, insulin, and glucagon precursors from a New World rodent, the degu, Octodon degus . Mol Endocrinol 4:1192–1198. [DOI] [PubMed] [Google Scholar]
- 61. Nixon RA (2013) The role of autophagy in neurodegenerative disease. Nat Med 19:983–997. [DOI] [PubMed] [Google Scholar]
- 62. Padurariu M, Ciobica A, Lefter R, Serban IL, Stefanescu C, Chirita R (2013) The oxidative stress hypothesis in Alzheimer's disease. Psychiatr Danub 25:401–409. [PubMed] [Google Scholar]
- 63. Pedros I, Petrov D, Allgaier M, Sureda F, Barroso E, Beas‐Zarate C et al (2014) Early alterations in energy metabolism in the hippocampus of APPswe/PS1dE9 mouse model of Alzheimer's disease. Biochim Biophys Acta 1842:1556–1566. [DOI] [PubMed] [Google Scholar]
- 64. Pratico D (2008) Oxidative stress hypothesis in Alzheimer's disease: a reappraisal. Trends Pharmacol Sci 29:609–615. [DOI] [PubMed] [Google Scholar]
- 65. Prokop S, Miller KR, Heppner FL (2013) Microglia actions in Alzheimer's disease. Acta Neuropathol 126:461–477. [DOI] [PubMed] [Google Scholar]
- 66. Querfurth HW, LaFerla FM (2010) Alzheimer's disease. N Engl J Med 362:329–344. [DOI] [PubMed] [Google Scholar]
- 67. Raina AK, Zhu X, Shimohama S, Perry G, Smith MA (2003) Tipping the apoptotic balance in Alzheimer's disease: the abortosis concept. Cell Biochem Biophys 39:249–255. [DOI] [PubMed] [Google Scholar]
- 68. Rios JA, Cisternas P, Arrese M, Barja S, Inestrosa NC (2014) Is Alzheimer's disease related to metabolic syndrome? A Wnt signaling conundrum. Prog Neurobiol 121:125–146. [DOI] [PubMed] [Google Scholar]
- 69. Robinson A, Grosgen S, Mett J, Zimmer VC, Haupenthal VJ, Hundsdorfer B et al (2014) Upregulation of PGC‐1alpha expression by Alzheimer's disease‐associated pathway: presenilin 1/amyloid precursor protein (APP)/intracellular domain of APP. Aging Cell 13:263–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Rohn TT (2010) The role of caspases in Alzheimer's disease; potential novel therapeutic opportunities. Apoptosis 15:1403–1409. [DOI] [PubMed] [Google Scholar]
- 71. Rohn TT, Head E (2009) Caspases as therapeutic targets in Alzheimer's disease: is it time to “cut” to the chase? Int J Clin Exp Pathol 2:108–118. [PMC free article] [PubMed] [Google Scholar]
- 72. Rohn TT, Kokoulina P, Eaton CR, Poon WW (2009) Caspase activation in transgenic mice with Alzheimer‐like pathology: results from a pilot study utilizing the caspase inhibitor, Q‐VD‐OPh. Int J Clin Exp Med 2:300–308. [PMC free article] [PubMed] [Google Scholar]
- 73. Rowe CC, Villemagne VL (2013) Amyloid imaging with PET in early Alzheimer disease diagnosis. Med Clin North Am 97:377–398. [DOI] [PubMed] [Google Scholar]
- 74. Rowe CC, Villemagne VL (2013) Brain amyloid imaging. J Nucl Med Technol 41:11–18. [DOI] [PubMed] [Google Scholar]
- 75. Serrano‐Pozo A, Frosch MP, Masliah E, Hyman BT (2011) Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med 1:a006189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Sheng M, Sabatini BL, Sudhof TC (2012) Synapses and Alzheimer's disease. Cold Spring Harb Perspect Biol 4(5). pii: a005777. doi: 10.1101/cshperspect.a005777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Simon DI, Mullins ME, Jia L, Gaston B, Singel DJ, Stamler JS (1996) Polynitrosylated proteins: characterization, bioactivity, and functional consequences. Proc Natl Acad Sci U S A 93:4736–4741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Spilman P, Podlutskaya N, Hart MJ, Debnath J, Gorostiza O, Bredesen D et al (2010) Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid‐beta levels in a mouse model of Alzheimer's disease. PLoS ONE 5:e9979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Swomley AM, Forster S, Keeney JT, Triplett J, Zhang Z, Sultana R, Butterfield DA (2014) Abeta, oxidative stress in Alzheimer disease: evidence based on proteomics studies. Biochim Biophys Acta 1842:1248–1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Thies W, Bleiler L, Alzheimer's A (2013) 2013 Alzheimer's disease facts and figures. Alzheimer's Dement 9:208–245. [DOI] [PubMed] [Google Scholar]
- 81. Ubhi K, Masliah E (2013) Alzheimer's disease: recent advances and future perspectives. J Alzheimer's Dis 33(Suppl. 1):S185–S194. [DOI] [PubMed] [Google Scholar]
- 82. van Groen T, Kadish I, Popovic N, Popovic M, Caballero‐Bleda M, Bano‐Otalora B et al (2011) Age‐related brain pathology in Octodon degu: blood vessel, white matter and Alzheimer‐like pathology. Neurobiol Aging 32:1651–1661. [DOI] [PubMed] [Google Scholar]
- 83. Wang X, Wang W, Li L, Perry G, Lee HG, Zhu X (2014) Oxidative stress and mitochondrial dysfunction in Alzheimer's disease. Biochim Biophys Acta 1842:1240–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Webster SJ, Bachstetter AD, Nelson PT, Schmitt FA, Van Eldik LJ (2014) Using mice to model Alzheimer's dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models. Front Genet 5:88. doi: 10.3389/fgene.2014.00088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Williams TI, Lynn BC, Markesbery WR, Lovell MA (2006) Increased levels of 4‐hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in mild cognitive impairment and early Alzheimer's disease. Neurobiol Aging 27:1094–1099. [DOI] [PubMed] [Google Scholar]
- 86. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V et al (1999) Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC‐1. Cell 98:115–124. [DOI] [PubMed] [Google Scholar]
- 87. Yao Y, Chinnici C, Tang H, Trojanowski JQ, Lee VM, Pratico D (2004) Brain inflammation and oxidative stress in a transgenic mouse model of Alzheimer‐like brain amyloidosis. J Neuroinflammation 1:21. doi: 10.1186/1742-2094-1-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Yu WH, Cuervo AM, Kumar A, Peterhoff CM, Schmidt SD, Lee JH et al (2005) Macroautophagy—a novel beta‐amyloid peptide‐generating pathway activated in Alzheimer's disease. J Cell Biol 171:87–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Zhao Y, Zhao B (2013) Oxidative stress and the pathogenesis of Alzheimer's disease. Oxid Med Cell Longev 2013:316523. doi: 10.1155/2013/316523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Zolezzi JM, Carvajal FJ, Rios JA, Ordenes D, Silva‐Alvarez C, Godoy JA, Inestrosa NC (2013) Tetrahydrohyperforin induces mitochondrial dynamics and prevents mitochondrial Ca2 + overload after Abeta and Abeta‐AChE complex challenge in rat hippocampal neurons. J Alzheimer's Dis 37:735–746. [DOI] [PubMed] [Google Scholar]
- 91. Zolezzi JM, Silva‐Alvarez C, Ordenes D, Godoy JA, Carvajal FJ, Santos MJ, Inestrosa NC (2013) Peroxisome proliferator‐activated receptor (PPAR) gamma and PPARalpha agonists modulate mitochondrial fusion‐fission dynamics: relevance to reactive oxygen species (ROS)‐related neurodegenerative disorders? PLoS ONE 8:e64019. [DOI] [PMC free article] [PubMed] [Google Scholar]
