101
|
Stefanova NA, Muraleva NA, Korbolina EE, Kiseleva E, Maksimova KY, Kolosova NG. Amyloid accumulation is a late event in sporadic Alzheimer's disease-like pathology in nontransgenic rats. Oncotarget 2015; 6:1396-413. [PMID: 25595891 PMCID: PMC4359302 DOI: 10.18632/oncotarget.2751] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/15/2014] [Indexed: 01/04/2023] Open
Abstract
The amyloid cascade hypothesis posits that deposition of the amyloid β (Aβ) peptide in the brain is a key event in the initiation of Alzheimer's disease (AD). Nonetheless, it now seems increasingly unlikely that amyloid toxicity is the cause of sporadic AD, which leads to cognitive decline. Here, using accelerated-senescence nontransgenic OXYS rats, we confirmed that aggregation of Aβ is a later event in AD-like pathology. We showed that an age-dependent increase in the levels of Aβ1–42 and extracellular Aβ deposits in the brain of OXYS rats occur later than do synaptic losses, neuronal cell death, mitochondrial structural abnormalities, and hyperphosphorylation of the tau protein. We identified the variants of the genes that are strongly associated with the risk of either late-onset or early-onset AD, including App, Apoe4, Bace1, Psen1, Psen2, and Picalm. We found that in OXYS rats nonsynonymous SNPs were located only in the genes Casp3 and Sorl1. Thus, we present proof that OXYS rats may be a model of sporadic AD. It is possible that multiple age-associated pathological processes may precede the toxic amyloid accumulation, which in turn triggers the final stage of the sporadic form of AD and becomes a hallmark event of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Nataliya G Kolosova
- Institute of Cytology and Genetics, Novosibirsk, Russia.,Institute of Mitoengineering, Moscow, Russia.,Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
102
|
TRPC6 specifically interacts with APP to inhibit its cleavage by γ-secretase and reduce Aβ production. Nat Commun 2015; 6:8876. [PMID: 26581893 PMCID: PMC4696454 DOI: 10.1038/ncomms9876] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/12/2015] [Indexed: 01/12/2023] Open
Abstract
Generation of β-amyloid (Aβ) peptide in Alzheimer's disease involves cleavage of amyloid precursor protein (APP) by γ-secretase, a protease known to cleave several substrates, including Notch. Finding specific modulators for γ-secretase could be a potential avenue to treat the disease. Here, we report that transient receptor potential canonical (TRPC) 6 specifically interacts with APP leading to inhibition of its cleavage by γ-secretase and reduction in Aβ production. TRPC6 interacts with APP (C99), but not with Notch, and prevents C99 interaction with presenilin 1 (PS1). A fusion peptide derived from TRPC6 also reduces Aβ levels without effect on Notch cleavage. Crossing APP/PS1 mice with TRPC6 transgenic mice leads to a marked reduction in both plaque load and Aβ levels, and improvement in structural and behavioural impairment. Thus, TRPC6 specifically modulates γ-secretase cleavage of APP and preventing APP (C99) interaction with PS1 via TRPC6 could be a novel strategy to reduce Aβ formation. Attempts to treat Alzheimer's disease by targeting γ-secretase cleavage of APP into Aß have been unsuccessful, partially due to off-target effects. Here, the authors identify TRPC6 as a novel γ-secretase modulator, showing that it interacts with APP to regulate Aß levels while sparing Notch cleavage.
Collapse
|
103
|
Puzzo D, Gulisano W, Arancio O, Palmeri A. The keystone of Alzheimer pathogenesis might be sought in Aβ physiology. Neuroscience 2015; 307:26-36. [PMID: 26314631 PMCID: PMC4591241 DOI: 10.1016/j.neuroscience.2015.08.039] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/15/2015] [Accepted: 08/18/2015] [Indexed: 01/17/2023]
Abstract
For several years Amyloid-beta peptide (Aβ) has been considered the main pathogenetic factor of Alzheimer's disease (AD). According to the so called Amyloid Cascade Hypothesis the increase of Aβ triggers a series of events leading to synaptic dysfunction and memory loss as well as to the structural brain damage in the later stage of the disease. However, several evidences suggest that this hypothesis is not sufficient to explain AD pathogenesis, especially considering that most of the clinical trials aimed to decrease Aβ levels have been unsuccessful. Moreover, Aβ is physiologically produced in the healthy brain during neuronal activity and it is needed for synaptic plasticity and memory. Here we propose a model interpreting AD pathogenesis as an alteration of the negative feedback loop between Aβ and its physiological receptors, focusing on alpha7 nicotinic acetylcholine receptors (α7-nAchRs). According to this vision, when Aβ cannot exert its physiological function a negative feedback mechanism would induce a compensatory increase of its production leading to an abnormal accumulation that reduces α7-nAchR function, leading to synaptic dysfunction and memory loss. In this perspective, the indiscriminate Aβ removal might worsen neuronal homeostasis, causing a further impoverishment of learning and memory. Even if further studies are needed to better understand and validate these mechanisms, we believe that to deepen the role of Aβ in physiological conditions might represent the keystone to elucidate important aspects of AD pathogenesis.
Collapse
Affiliation(s)
- D Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy.
| | - W Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy
| | - O Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, 630 West 168th Street, Columbia University, New York, NY 10032, USA
| | - A Palmeri
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy
| |
Collapse
|
104
|
Neurofilament light gene deletion exacerbates amyloid, dystrophic neurite, and synaptic pathology in the APP/PS1 transgenic model of Alzheimer's disease. Neurobiol Aging 2015; 36:2757-67. [DOI: 10.1016/j.neurobiolaging.2015.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 07/02/2015] [Accepted: 07/02/2015] [Indexed: 01/10/2023]
|
105
|
Omega-3 Fatty Acids Augment the Actions of Nuclear Receptor Agonists in a Mouse Model of Alzheimer's Disease. J Neurosci 2015; 35:9173-81. [PMID: 26085639 DOI: 10.1523/jneurosci.1000-15.2015] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a highly prevalent disorder for which there are no effective therapies. Accumulation of amyloid β (Aβ) peptides in the brain is associated with impaired cognition and memory, pronounced inflammatory dysregulation, and subsequent amyloid plaque deposition. Thus, drugs that promote the clearance of Aβ peptides and resolution of inflammation may represent viable therapeutic approaches. Agonists of nuclear receptors LXR:RXR and PPAR:RXR act to ameliorate AD-related cognitive impairment and amyloid accumulation in murine models of AD. The use of an agonist to the nuclear receptor RXR, bexarotene, as monotherapy against AD, presents potential challenges due to the metabolic perturbations it induces in the periphery, most prominently hypertriglyceridemia. We report that the ω-3 fatty acid docosahexaenoic acid (DHA), in combination with bexarotene, enhances LXR:RXR target gene expression of Abca1 and ApoE, reduces soluble forms of Aβ, and abrogates release of pro-inflammatory cytokines and mediators both in vitro and in a mouse model of AD. Moreover, DHA abrogates bexarotene-induced hypertriglyceridemia in vivo. Importantly, dual therapy promotes reductions in AD pathology and resultant amelioration of cognitive deficits. While monotherapy with either bexarotene or DHA resulted in modest effects in vitro and in vivo, combined treatment with both agents produced a significant additive benefit on associated AD-related phenotypes, suggesting that targeted combinatorial agents may be beneficial over single agents alone in treating AD.
Collapse
|
106
|
Snir JA, Suchy M, Lawrence KS, Hudson RH, Pasternak S, Bartha R. Prolonged In Vivo Retention of a Cathepsin D Targeted Optical Contrast Agent in a Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2015; 48:73-87. [DOI: 10.3233/jad-150123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jonatan A. Snir
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Mojmir Suchy
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Chemistry, University of Western Ontario, London, Ontario, Canada
| | - Keith St. Lawrence
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Medical Imaging, Lawson Health Research Institute, London, Ontario, Canada
| | - Robert H.E. Hudson
- Department of Chemistry, University of Western Ontario, London, Ontario, Canada
| | - Stephen H. Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Robert Bartha
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
107
|
Béliveau E, Tremblay C, Aubry-Lafontaine É, Paris-Robidas S, Delay C, Robinson C, Ferguson L, Rajput AH, Rajput A, Calon F. Accumulation of amyloid-β in the cerebellar cortex of essential tremor patients. Neurobiol Dis 2015; 82:397-408. [PMID: 26253607 DOI: 10.1016/j.nbd.2015.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/24/2015] [Accepted: 07/31/2015] [Indexed: 12/31/2022] Open
Abstract
The accumulation of insoluble amyloid-beta (Aβ) peptides is associated with neurodegenerative disorders, such as Alzheimer's disease (AD). As essential tremor (ET) could involve neurodegenerative processes in the cerebellum, we quantified soluble and insoluble Aβ in cerebellar cortices from patients diagnosed with ET (n=9), compared to Controls (n=16) or individuals with Parkinson's disease (n=10). Although ante-mortem cognitive performance was not documented, all individuals included had the diagnosis of AD ruled out by a neuropathologist. ELISA-determined concentrations of insoluble Aβ42 in ET patients displayed a bimodal distribution, with a median 246-fold higher than in Controls (P<0.01, Kruskal-Wallis). Higher Aβ42 concentrations were measured in the parietal cortex of the same ET patients, compared to Controls (107-fold median increase, P<0.01, Kruskal-Wallis), but similar phosphorylated tau levels were detected. The rise in cerebellar insoluble Aβ42 concentrations is not associated to APP expression and processing or the ApoE4 status. However, Aβ42 levels in ET individuals were correlated with cerebellar insoluble phosphorylated tau (r(2)=0.71, P=0.005), unphosphorylated neurofilament heavy chain (NF-H; r(2)=0.50, P=0.030) and Lingo-1 (r(2)=0.73, P=0.007), indicative of a generalized neurodegenerative process involving the cerebellum. Our results suggest prevalent accumulations of insoluble Aβ42 in the cerebellum of ET, but not in age-matched PD. Whether this anomaly plays a role in ET symptoms warrants further investigations.
Collapse
Affiliation(s)
- Eric Béliveau
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada; Neurosciences Axis, Centre de recherche du CHU de Québec, Québec, QC, Canada
| | - Cyntia Tremblay
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada; Neurosciences Axis, Centre de recherche du CHU de Québec, Québec, QC, Canada
| | - Émilie Aubry-Lafontaine
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada; Neurosciences Axis, Centre de recherche du CHU de Québec, Québec, QC, Canada
| | - Sarah Paris-Robidas
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada; Neurosciences Axis, Centre de recherche du CHU de Québec, Québec, QC, Canada
| | | | - Chris Robinson
- Division of Neurology, Royal University Hospital, University of Saskatchewan, Saskatoon, SK, Canada
| | - Les Ferguson
- Division of Neurology, Royal University Hospital, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ali H Rajput
- Division of Neurology, Royal University Hospital, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alex Rajput
- Division of Neurology, Royal University Hospital, University of Saskatchewan, Saskatoon, SK, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada; Neurosciences Axis, Centre de recherche du CHU de Québec, Québec, QC, Canada.
| |
Collapse
|
108
|
Norvin D, Kim G, Baker-Nigh A, Geula C. Accumulation and age-related elevation of amyloid-β within basal forebrain cholinergic neurons in the rhesus monkey. Neuroscience 2015; 298:102-11. [DOI: 10.1016/j.neuroscience.2015.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 03/31/2015] [Accepted: 04/06/2015] [Indexed: 11/29/2022]
|
109
|
Collins JM, King AE, Woodhouse A, Kirkcaldie MTK, Vickers JC. The effect of focal brain injury on beta-amyloid plaque deposition, inflammation and synapses in the APP/PS1 mouse model of Alzheimer's disease. Exp Neurol 2015; 267:219-29. [PMID: 25747037 DOI: 10.1016/j.expneurol.2015.02.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 01/11/2015] [Accepted: 02/27/2015] [Indexed: 11/26/2022]
Abstract
Traumatic brain injury is a risk factor for Alzheimer's disease (AD), however the effect of such neural damage on the onset and progression of beta-amyloid (Aβ) plaque pathology is not well understood. This study utilized an in vivo model of focal brain injury to examine how localized damage may acutely affect the onset and progression of Aβ plaque deposition as well as inflammatory and synaptic changes, in the APP/PS1 (APPSWE, PSEN1dE9) transgenic model of AD relative to wild-type (Wt) mice. Acute focal brain injury in 3- and 9-month-old APP/PS1 and Wt mice was induced by insertion of a needle into the somatosensory neocortex, as compared to sham surgery, and examined at 24h and 7d post-injury (PI). Focal brain injury did not induce thioflavine-S stained or (pan-Aβ antibody) MOAB-2-labeled plaques at either 24h or 7d PI in 3-month-old APP/PS1 mice or Wt mice. Nine-month-old APP/PS1 mice demonstrate cortical Aβ plaques but focal injury had no statistically significant (p>0.05) effect on thioflavine-S or MOAB-2 plaque load surrounding the injury site at 24h PI or 7d PI. There was a significant (p<0.001) increase in cross-sectional cortical area occupied by Iba-1 positive microglia in injured mice compared to sham animals, however this response did not differ between APP/PS1 and Wt mice (p>0.05). For both Wt and APP/PS1 mice alike, synaptophysin puncta near the injury site were significantly reduced 24h PI (compared to sites distant to the injury and the corresponding area in sham mice; p<0.01), but not after 7d PI (p>0.05). There was no significant effect of genotype on this response (p>0.05). These results indicate that focal brain injury and the associated microglial response do not acutely alter Aβ plaque deposition in the APP/PS1 mouse model. Furthermore the current study demonstrated that the brains of both Wt and APP/PS1 mice are capable of recovering lost synaptophysin immunoreactivity post-injury, the latter in the presence of Aβ plaque pathology that causes synaptic degeneration.
Collapse
Affiliation(s)
- Jessica M Collins
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia.
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia
| | - Matthew T K Kirkcaldie
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia; School of Medicine, University of Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia; School of Medicine, University of Tasmania, Australia
| |
Collapse
|
110
|
Baker-Nigh A, Vahedi S, Davis EG, Weintraub S, Bigio EH, Klein WL, Geula C. Neuronal amyloid-β accumulation within cholinergic basal forebrain in ageing and Alzheimer's disease. Brain 2015; 138:1722-37. [PMID: 25732182 DOI: 10.1093/brain/awv024] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/14/2014] [Indexed: 11/14/2022] Open
Abstract
The mechanisms that contribute to selective vulnerability of the magnocellular basal forebrain cholinergic neurons in neurodegenerative diseases, such as Alzheimer's disease, are not fully understood. Because age is the primary risk factor for Alzheimer's disease, mechanisms of interest must include age-related alterations in protein expression, cell type-specific markers and pathology. The present study explored the extent and characteristics of intraneuronal amyloid-β accumulation, particularly of the fibrillogenic 42-amino acid isoform, within basal forebrain cholinergic neurons in normal young, normal aged and Alzheimer's disease brains as a potential contributor to the selective vulnerability of these neurons using immunohistochemistry and western blot analysis. Amyloid-β1-42 immunoreactivity was observed in the entire cholinergic neuronal population regardless of age or Alzheimer's disease diagnosis. The magnitude of this accumulation as revealed by optical density measures was significantly greater than that in cortical pyramidal neurons, and magnocellular neurons in the globus pallidus did not demonstrate a similar extent of amyloid immunoreactivity. Immunoblot analysis with a panel of amyloid-β antibodies confirmed accumulation of high concentration of amyloid-β in basal forebrain early in adult life. There was no age- or Alzheimer-related alteration in total amyloid-β content within this region. In contrast, an increase in the large molecular weight soluble oligomer species was observed with a highly oligomer-specific antibody in aged and Alzheimer brains when compared with the young. Similarly, intermediate molecular weight oligomeric species displayed an increase in aged and Alzheimer brains when compared with the young using two amyloid-β42 antibodies. Compared to cortical homogenates, small molecular weight oligomeric species were lower and intermediate species were enriched in basal forebrain in ageing and Alzheimer's disease. Regional and age-related differences in accumulation were not the result of alterations in expression of the amyloid precursor protein, as confirmed by both immunostaining and western blot. Our results demonstrate that intraneuronal amyloid-β accumulation is a relatively selective trait of basal forebrain cholinergic neurons early in adult life, and increases in the prevalence of intermediate and large oligomeric assembly states are associated with both ageing and Alzheimer's disease. Selective intraneuronal amyloid-β accumulation in adult life and oligomerization during the ageing process are potential contributors to the degeneration of basal forebrain cholinergic neurons in Alzheimer's disease.
Collapse
Affiliation(s)
- Alaina Baker-Nigh
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Shahrooz Vahedi
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Elena Goetz Davis
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sandra Weintraub
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Eileen H Bigio
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - William L Klein
- 2 Neurobiology, Northwestern University, Evanston, IL 60201, USA
| | - Changiz Geula
- 1 Cognitive Neurology and Alzheimer's Disease Centre, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
111
|
González-Marrero I, Giménez-Llort L, Johanson CE, Carmona-Calero EM, Castañeyra-Ruiz L, Brito-Armas JM, Castañeyra-Perdomo A, Castro-Fuentes R. Choroid plexus dysfunction impairs beta-amyloid clearance in a triple transgenic mouse model of Alzheimer's disease. Front Cell Neurosci 2015; 9:17. [PMID: 25705176 PMCID: PMC4319477 DOI: 10.3389/fncel.2015.00017] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/12/2015] [Indexed: 01/10/2023] Open
Abstract
Compromised secretory function of choroid plexus (CP) and defective cerebrospinal fluid (CSF) production, along with accumulation of beta-amyloid (Aβ) peptides at the blood-CSF barrier (BCSFB), contribute to complications of Alzheimer’s disease (AD). The AD triple transgenic mouse model (3xTg-AD) at 16 month-old mimics critical hallmarks of the human disease: β-amyloid (Aβ) plaques and neurofibrillary tangles (NFT) with a temporal- and regional- specific profile. Currently, little is known about transport and metabolic responses by CP to the disrupted homeostasis of CNS Aβ in AD. This study analyzed the effects of highly-expressed AD-linked human transgenes (APP, PS1 and tau) on lateral ventricle CP function. Confocal imaging and immunohistochemistry revealed an increase only of Aβ42 isoform in epithelial cytosol and in stroma surrounding choroidal capillaries; this buildup may reflect insufficient clearance transport from CSF to blood. Still, there was increased expression, presumably compensatory, of the choroidal Aβ transporters: the low density lipoprotein receptor-related protein 1 (LRP1) and the receptor for advanced glycation end product (RAGE). A thickening of the epithelial basal membrane and greater collagen-IV deposition occurred around capillaries in CP, probably curtailing solute exchanges. Moreover, there was attenuated expression of epithelial aquaporin-1 and transthyretin (TTR) protein compared to Non-Tg mice. Collectively these findings indicate CP dysfunction hypothetically linked to increasing Aβ burden resulting in less efficient ion transport, concurrently with reduced production of CSF (less sink action on brain Aβ) and diminished secretion of TTR (less neuroprotection against cortical Aβ toxicity). The putative effects of a disabled CP-CSF system on CNS functions are discussed in the context of AD.
Collapse
Affiliation(s)
| | - Lydia Giménez-Llort
- Institute of Neurosciences and Department of Psychiatry and Forensic Medicine, Autonomous University of Barcelona Barcelona, Spain
| | - Conrad E Johanson
- Department of Neurosurgery, Alpert Medical School at Brown University Providence, Rhode Island, USA
| | | | | | | | | | - Rafael Castro-Fuentes
- Department of Physiology, School of Medicine, University of La Laguna Tenerife, Spain
| |
Collapse
|
112
|
Scala F, Fusco S, Ripoli C, Piacentini R, Li Puma DD, Spinelli M, Laezza F, Grassi C, D'Ascenzo M. Intraneuronal Aβ accumulation induces hippocampal neuron hyperexcitability through A-type K(+) current inhibition mediated by activation of caspases and GSK-3. Neurobiol Aging 2015; 36:886-900. [PMID: 25541422 PMCID: PMC4801354 DOI: 10.1016/j.neurobiolaging.2014.10.034] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 10/14/2014] [Accepted: 10/24/2014] [Indexed: 11/20/2022]
Abstract
Amyloid β-protein (Aβ) pathologies have been linked to dysfunction of excitability in neurons of the hippocampal circuit, but the molecular mechanisms underlying this process are still poorly understood. Here, we applied whole-cell patch-clamp electrophysiology to primary hippocampal neurons and show that intracellular Aβ42 delivery leads to increased spike discharge and action potential broadening through downregulation of A-type K(+) currents. Pharmacologic studies showed that caspases and glycogen synthase kinase 3 (GSK-3) activation are required for these Aβ42-induced effects. Extracellular perfusion and subsequent internalization of Aβ42 increase spike discharge and promote GSK-3-dependent phosphorylation of the Kv4.2 α-subunit, a molecular determinant of A-type K(+) currents, at Ser-616. In acute hippocampal slices derived from an adult triple-transgenic Alzheimer's mouse model, characterized by endogenous intracellular accumulation of Aβ42, CA1 pyramidal neurons exhibit hyperexcitability accompanied by increased phosphorylation of Kv4.2 at Ser-616. Collectively, these data suggest that intraneuronal Aβ42 accumulation leads to an intracellular cascade culminating into caspases activation and GSK-3-dependent phosphorylation of Kv4.2 channels. These findings provide new insights into the toxic mechanisms triggered by intracellular Aβ42 and offer potentially new therapeutic targets for Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Federico Scala
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Salvatore Fusco
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Cristian Ripoli
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | | | - Matteo Spinelli
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy.
| | - Marcello D'Ascenzo
- Institute of Human Physiology, Medical School, Università Cattolica, Rome, Italy.
| |
Collapse
|
113
|
Ayyadevara S, Balasubramaniam M, Gao Y, Yu L, Alla R, Shmookler Reis R. Proteins in aggregates functionally impact multiple neurodegenerative disease models by forming proteasome-blocking complexes. Aging Cell 2015; 14:35-48. [PMID: 25510159 PMCID: PMC4326912 DOI: 10.1111/acel.12296] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2014] [Indexed: 01/18/2023] Open
Abstract
Age-dependent neurodegenerative diseases progressively form aggregates containing both shared components (e.g., TDP-43, phosphorylated tau) and proteins specific to each disease. We investigated whether diverse neuropathies might have additional aggregation-prone proteins in common, discoverable by proteomics. Caenorhabditis elegans expressing unc-54p/Q40::YFP, a model of polyglutamine array diseases such as Huntington's, accrues aggregates in muscle 2–6 days posthatch. These foci, isolated on antibody-coupled magnetic beads, were characterized by high-resolution mass spectrometry. Three Q40::YFP-associated proteins were inferred to promote aggregation and cytotoxicity, traits reduced or delayed by their RNA interference knockdown. These RNAi treatments also retarded aggregation/cytotoxicity in Alzheimer's disease models, nematodes with muscle or pan-neuronal Aβ1–42 expression and behavioral phenotypes. The most abundant aggregated proteins are glutamine/asparagine-rich, favoring hydrophobic interactions with other random-coil domains. A particularly potent modulator of aggregation, CRAM-1/HYPK, contributed < 1% of protein aggregate peptides, yet its knockdown reduced Q40::YFP aggregates 72–86% (P < 10−6). In worms expressing Aβ1–42, knockdown of cram-1 reduced β-amyloid 60% (P < 0.002) and slowed age-dependent paralysis > 30% (P < 10−6). In wild-type worms, cram-1 knockdown reduced aggregation and extended lifespan, but impaired early reproduction. Protection against seeded aggregates requires proteasome function, implying that normal CRAM-1 levels promote aggregation by interfering with proteasomal degradation of misfolded proteins. Molecular dynamic modeling predicts spontaneous and stable interactions of CRAM-1 (or human orthologs) with ubiquitin, and we verified that CRAM-1 reduces degradation of a tagged-ubiquitin reporter. We propose that CRAM-1 exemplifies a class of primitive chaperones that are initially protective and highly beneficial for early reproduction, but ultimately impair aggregate clearance and limit longevity.
Collapse
Affiliation(s)
- Srinivas Ayyadevara
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205USA
| | - Meenakshisundaram Balasubramaniam
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205USA
- BioInformatics Program University of Arkansas for Medical Sciences and University of Arkansas at Little Rock Little Rock AR 72205 USA
| | - Yuan Gao
- National Center for Toxicological Research Food & Drug Administration Jefferson AR 72079USA
| | - Li‐Rong Yu
- National Center for Toxicological Research Food & Drug Administration Jefferson AR 72079USA
| | - Ramani Alla
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205USA
| | - Robert Shmookler Reis
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205USA
- Department of Biochemistry & Molecular Biology University of Arkansas for Medical Sciences Little Rock AR 72205USA
| |
Collapse
|
114
|
Esteves-Villanueva JO, Trzeciakiewicz H, Loeffler DA, Martić S. Effects of tau domain-specific antibodies and intravenous immunoglobulin on tau aggregation and aggregate degradation. Biochemistry 2015; 54:293-302. [PMID: 25545358 DOI: 10.1021/bi501272x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tau pathology, including neurofibrillary tangles, develops in Alzheimer's disease (AD). The aggregation and hyperphosphorylation of tau are potential therapeutic targets for AD. Administration of anti-tau antibodies reduces tau pathology in transgenic "tauopathy" mice; however, the optimal tau epitopes and conformations to target are unclear. Also unknown is whether intravenous immunoglobulin (IVIG) products, currently being evaluated in AD trials, exert effects on pathological tau. This study examined the effects of anti-tau antibodies targeting different tau epitopes and the IVIG Gammagard on tau aggregation and preformed tau aggregates. Tau aggregation was assessed by transmission electron microscopy and fluorescence spectroscopy, and the binding affinity of the anti-tau antibodies for tau was evaluated by enzyme-linked immunosorbent assays. Antibodies used were anti-tau 1-150 ("D-8"), anti-tau 259-266 ("Paired-262"), anti-tau 341-360 ("A-10"), and anti-tau 404-441 ("Tau-46"), which bind to tau's N-terminus, microtubule binding domain (MBD) repeat sequences R1 and R4, and the C-terminus, respectively. The antibodies Paired-262 and A-10, but not D-8 and Tau-46, reduced tau fibrillization and degraded preformed tau aggregates, whereas the IVIG reduced tau aggregation but did not alter preformed aggregates. The binding affinities of the antibodies for the epitope for which they were specific did not appear to be related to their effects on tau aggregation. These results confirm that antibody binding to tau's MBD repeat sequences may inhibit tau aggregation and indicate that such antibodies may also degrade preformed tau aggregates. In the presence of anti-tau antibodies, the resulting tau morphologies were antigen-dependent. The results also suggested the possibility of different pathways regulating antibody-mediated inhibition of tau aggregation and antibody-mediated degradation of preformed tau aggregates.
Collapse
Affiliation(s)
- Jose O Esteves-Villanueva
- Department of Chemistry, Oakland University , 2200 North Squirrel Road, Rochester, Michigan 48309, United States
| | | | | | | |
Collapse
|
115
|
Stargardt A, Swaab DF, Bossers K. The storm before the quiet: neuronal hyperactivity and Aβ in the presymptomatic stages of Alzheimer's disease. Neurobiol Aging 2015; 36:1-11. [DOI: 10.1016/j.neurobiolaging.2014.08.014] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/29/2014] [Accepted: 08/12/2014] [Indexed: 12/27/2022]
|
116
|
Zhang X, Li Y, Xu H, Zhang YW. The γ-secretase complex: from structure to function. Front Cell Neurosci 2014; 8:427. [PMID: 25565961 PMCID: PMC4263104 DOI: 10.3389/fncel.2014.00427] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 11/27/2014] [Indexed: 12/21/2022] Open
Abstract
One of the most critical pathological features of Alzheimer’s disease (AD) is the accumulation of β-amyloid (Aβ) peptides that form extracellular senile plaques in the brain. Aβ is derived from β-amyloid precursor protein (APP) through sequential cleavage by β- and γ-secretases. γ-secretase is a high molecular weight complex minimally composed of four components: presenilins (PS), nicastrin, anterior pharynx defective 1 (APH-1), and presenilin enhancer 2 (PEN-2). In addition to APP, γ-secretase also cleaves many other type I transmembrane (TM) protein substrates. As a crucial enzyme for Aβ production, γ-secretase is an appealing therapeutic target for AD. Here, we summarize current knowledge on the structure and function of γ-secretase, as well as recent progress in developing γ-secretase targeting drugs for AD treatment.
Collapse
Affiliation(s)
- Xian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| | - Yanfang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China ; Degenerative Disease Research Program, Sanford-Burnham Medical Research Institute La Jolla, CA, USA
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| |
Collapse
|
117
|
Katsouri L, Ashraf A, Birch AM, Lee KKL, Mirzaei N, Sastre M. Systemic administration of fibroblast growth factor-2 (FGF2) reduces BACE1 expression and amyloid pathology in APP23 mice. Neurobiol Aging 2014; 36:821-31. [PMID: 25457554 DOI: 10.1016/j.neurobiolaging.2014.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/18/2014] [Accepted: 10/07/2014] [Indexed: 01/15/2023]
Abstract
There is an emerging evidence that growth factors may have a potential beneficial use in the treatment of Alzheimer's disease (AD) because of their neuroprotective properties and effects on neuronal proliferation. Basic fibroblast growth factor or fibroblast growth factor-2 (FGF2) is an anti-inflammatory, angiogenic, and neurotrophic factor that is expressed in many cell types, including neurons and glial cells. Here, we explored whether subcutaneous administration of FGF2 could have therapeutic effects in the APP 23 transgenic mouse, a model of amyloid pathology. FGF2 treatment attenuated spatial memory deficits, reduced amyloid-β (Aβ) and tau pathologies, decreased inducible nitric oxide synthase expression, and increased the number of astrocytes in the dentate gyrus in APP 23 mice compared with the vehicle-treated controls. The decrease in Aβ deposition was associated with a reduction in the expression of BACE1, the main enzyme responsible for Aβ generation. These results were confirmed in a neuroblastoma cell line, which demonstrated that incubation with FGF2 regulates BACE1 transcription. In addition, and in contrast with what has been previously published, the levels of FGF2 were reduced in postmortem brains from AD patients compared with controls. These data, therefore, suggest that systemic administration of FGF2 could have a potential therapeutic application in AD.
Collapse
Affiliation(s)
- Loukia Katsouri
- Division of Brain Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Azhaar Ashraf
- Division of Brain Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Amy M Birch
- Division of Brain Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Kevin K L Lee
- Division of Brain Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Nazanin Mirzaei
- Division of Brain Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Magdalena Sastre
- Division of Brain Sciences, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
118
|
Zhao W, Zhang J, Davis EG, Rebeck GW. Aging reduces glial uptake and promotes extracellular accumulation of Aβ from a lentiviral vector. Front Aging Neurosci 2014; 6:210. [PMID: 25177293 PMCID: PMC4133689 DOI: 10.3389/fnagi.2014.00210] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/28/2014] [Indexed: 01/27/2023] Open
Abstract
We used a lentiviral system for expressing secreted human Aβ in the brains of young and old APOE knock-in mice. This system allowed us to examine Aβ metabolism in vivo, and test the effects of both aging and APOE genotype, two of the strongest risk factors for Alzheimer’s disease. We injected the Aβ1-42 lentivirus into the motor cortex of young (2 month old) and old (20–22 month old) APOE3 and APOE4 mice. After 2 weeks of lentiviral expression, we analyzed the pattern of Aβ accumulation, glial activation, and phosphor-tau. In young mice, Aβ accumulated mainly within neurons with no evidence of extracellular Aβ. Significantly higher levels of intraneuronal Aβ were observed in APOE4 mice compared to APOE3 mice. In old mice, APOE4 predisposed again to higher levels of Aβ accumulation, but the Aβ was mainly in extracellular spaces. In younger mice, we also observed Aβ in microglia but not astrocytes. The numbers of microglia containing Aβ were significantly higher in APOE3 mice compared to APOE4 mice, and were significantly lower in both genetic backgrounds with aging. The astrocytes in old mice were activated to a greater extent in the brain regions where Aβ was introduced, an effect that was again increased by the presence of APOE4. Finally, phospho-tau accumulated in the region of Aβ expression, with evidence of extracellular phospho-tau increasing with aging. These data suggest that APOE4 predisposes to less microglial clearance of Aβ, leading to more intraneuronal accumulation. In older brains, decreased clearance leads to more extracellular Aβ, and more downstream consequences relating to astrocyte activation and phospho-tau accumulation. We conclude that both aging and APOE genotype affect pathways related to Aβ metabolism by microglia.
Collapse
Affiliation(s)
- Wenjuan Zhao
- School of Pharmacy, Shanghai Jiao Tong University Shanghai, China
| | - Jiguo Zhang
- Department of Pharmacology, School of Pharmacy, Taishan Medical University Taian, China
| | - Elizabeth G Davis
- Department of Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Medical Center Washington, DC, USA
| |
Collapse
|
119
|
Rodriguez GA, Tai LM, LaDu MJ, Rebeck GW. Human APOE4 increases microglia reactivity at Aβ plaques in a mouse model of Aβ deposition. J Neuroinflammation 2014; 11:111. [PMID: 24948358 PMCID: PMC4077554 DOI: 10.1186/1742-2094-11-111] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/29/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Having the apolipoprotein E4 (APOE-ϵ4) allele is the strongest genetic risk factor for the development of Alzheimer's disease (AD). Accumulation of amyloid beta (Aβ) in the brain is influenced by APOE genotype. Transgenic mice co-expressing five familial AD mutations (5xFAD) in the presence of human APOE alleles (ϵ2, ϵ3 or ϵ4) exhibit APOE genotype-specific differences in early Aβ accumulation, suggesting an interaction between APOE and AD pathology. Whether APOE genotype affects Aβ-plaque-associated neuroinflammation remains unclear. In the current study, we address the role of APOE genotype on Aβ-associated microglial reactivity in the EFAD transgenic mouse model. METHODS We analyzed Aβ-induced glial activation in the brains of 6-month-old EFAD transgenic mice (E2FAD, E3FAD and E4FAD). Region-specific morphological profiles of Aβ plaques in EFAD brain sections were compared using immunofluorescence staining. We then determined the degree of glial activation in sites of Aβ deposition while comparing levels of the inflammatory cytokine Interleukin-1β (IL-1β) by ELISA. Finally, we quantified parameters of Aβ-associated microglial reactivity using double-stained EFAD brain sections. RESULTS Characterization of Aβ plaques revealed there were larger and more intensely stained plaques in E4FAD mice relative to E2FAD and E3FAD mice. E4FAD mice also had a greater percentage of compact plaques in the subiculum than E3FAD mice. Reactive microglia and dystrophic astrocytes were prominent in EFAD brains, and primarily localized to two sites of significant Aβ deposition: the subiculum and deep layers of the cortex. Cortical levels of IL-1β were nearly twofold greater in E4FAD mice relative to E3FAD mice. To control for differences in levels of Aβ in the different EFAD mice, we analyzed the microglia within domains of specific Aβ deposits. Morphometric analyses revealed increased measures of microglial reactivity in E4FAD mice, including greater dystrophy, increased fluorescence intensity and a higher density of reactive cells surrounding cortical plaques, than in E3FAD mice. CONCLUSIONS In addition to altering morphological profiles of Aβ deposition, APOE genotype influences Aβ-induced glial activation in the adult EFAD cortex. These data support a role for APOE in modulating Aβ-induced neuroinflammatory responses in AD progression, and support the use of EFAD mice as a suitable model for mechanistic studies of Aβ-associated neuroinflammation.
Collapse
Affiliation(s)
- Gustavo A Rodriguez
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road, NW Washington, DC 20057, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road, NW Washington, DC 20057, USA
| |
Collapse
|
120
|
Iulita MF, Allard S, Richter L, Munter LM, Ducatenzeiler A, Weise C, Do Carmo S, Klein WL, Multhaup G, Cuello AC. Intracellular Aβ pathology and early cognitive impairments in a transgenic rat overexpressing human amyloid precursor protein: a multidimensional study. Acta Neuropathol Commun 2014; 2:61. [PMID: 24903713 PMCID: PMC4229908 DOI: 10.1186/2051-5960-2-61] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 05/07/2014] [Indexed: 01/09/2023] Open
Abstract
Numerous studies have implicated the abnormal accumulation of intraneuronal amyloid-β (Aβ) as an important contributor to Alzheimer's disease (AD) pathology, capable of triggering neuroinflammation, tau hyperphosphorylation and cognitive deficits. However, the occurrence and pathological relevance of intracellular Aβ remain a matter of controversial debate. In this study, we have used a multidimensional approach including high-magnification and super-resolution microscopy, cerebro-spinal fluid (CSF) mass spectrometry analysis and ELISA to investigate the Aβ pathology and its associated cognitive impairments, in a novel transgenic rat model overexpressing human APP. Our microscopy studies with quantitative co-localization analysis revealed the presence of intraneuronal Aβ in transgenic rats, with an immunological signal that was clearly distinguished from that of the amyloid precursor protein (APP) and its C-terminal fragments (CTFs). The early intraneuronal pathology was accompanied by a significant elevation of soluble Aβ42 peptides that paralleled the presence and progression of early cognitive deficits, several months prior to amyloid plaque deposition. Aβ38, Aβ39, Aβ40 and Aβ42 peptides were detected in the rat CSF by MALDI-MS analysis even at the plaque-free stages; suggesting that a combination of intracellular and soluble extracellular Aβ may be responsible for impairing cognition at early time points. Taken together, our results demonstrate that the intraneuronal development of AD-like amyloid pathology includes a mixture of molecular species (Aβ, APP and CTFs) of which a considerable component is Aβ; and that the early presence of these species within neurons has deleterious effects in the CNS, even before the development of full-blown AD-like pathology.
Collapse
Affiliation(s)
- M Florencia Iulita
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
| | - Simon Allard
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
| | - Luise Richter
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
| | - Lisa-Marie Munter
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
| | - Adriana Ducatenzeiler
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
| | - Christoph Weise
- />Intitutes of Chemistry and Biochemistry, Freie Universität, Berlin, Germany
| | - Sonia Do Carmo
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
| | - William L Klein
- />Cognitive Neurology and Alzheimer’s Disease Center, Northwestern University Institute for Neuroscience, Chicago, USA
| | - Gerhard Multhaup
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
| | - A Claudio Cuello
- />Department of Pharmacology and Therapeutics, McGill University, 3655 Sir-William-Osler Promenade, Room 1210, Montreal, Quebec Canada
- />Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
- />Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| |
Collapse
|
121
|
Development of a novel cellular model of Alzheimer's disease utilizing neurosphere cultures derived from B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J embryonic mouse brain. SPRINGERPLUS 2014; 3:161. [PMID: 25140287 PMCID: PMC4137416 DOI: 10.1186/2193-1801-3-161] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 03/07/2014] [Indexed: 12/29/2022]
Abstract
Increased production, oligomerization and aggregation of amyloid-β (Aβ) peptides are hallmark pathologies of Alzheimer’s disease (AD). Expressing familial AD mutations (amyloid precursor protein and/or presenilins mutations), the Aβ-pathologies of AD has been recapitulated in animal models of AD. Very few primary cell culture-based models of AD are available and they exhibit very weak Aβ-pathologies compared to what is seen in AD patients and animal models of AD. CNS stem/progenitor cells are present in both embryonic and adult brains. They can be isolated, grown as neurospheres and differentiated into neurons, astrocytes and oligodendrocytes. It is not yet known whether CNS stem/progenitor cells can support the production of Aβ peptides in culture. In this report, we have established Aβ-pathologies such as production, secretion, oligomerization and aggregation of Aβ peptides utilizing neurosphere cultures to create a new cellular model of AD. These cultures were developed from E15 embryonic brains of transgenic mice carrying the Swedish mutations in humanized mouse APP cDNA and the exon-9 deleted human presenilin 1 cDNA both regulated by mouse prion protein gene (Prnp) promoter. Results demonstrated the expression of transgene transcripts, APPswe protein and its processed products only in transgene positive neurosphere cultures. These cultures generate and secrete both Aβ40 and Aβ42 peptides into culture medium at levels comparable to the Aβ load in the brain of AD patients and animal models of AD, and produce pathogenic oligomers of Aβ peptides. The Aβ42/Aβ40 ratio in the medium of transgene positive neurosphere cultures is higher than any known cellular models of AD. Conformation dependent immunocytochemistry demonstrated the possible presence of intracellular and extracellular aggregation of Aβ peptides in neurosphere cultures, which are also seen in AD brain and animal models of AD. Collectively, our neurosphere cultures provide robust Aβ-pathologies of AD better than existing cellular model of Alzheimer’s disease.
Collapse
|
122
|
Ari C, Borysov SI, Wu J, Padmanabhan J, Potter H. Alzheimer amyloid beta inhibition of Eg5/kinesin 5 reduces neurotrophin and/or transmitter receptor function. Neurobiol Aging 2014; 35:1839-49. [PMID: 24636920 DOI: 10.1016/j.neurobiolaging.2014.02.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 12/23/2013] [Accepted: 02/05/2014] [Indexed: 12/29/2022]
Abstract
The mechanism by which amyloid beta (Aβ) causes neuronal dysfunction and/or death in Alzheimer's disease (AD) is unclear. Previously, we showed that Aβ inhibits several microtubule-dependent kinesin motors essential for mitosis and also present in mature neurons. Here, we show that inhibition of kinesin 5 (Eg5) by Aβ blocks neuronal function by reducing transport of neurotrophin and neurotransmitter receptors to the cell surface. Specifically, cell-surface NGF/NTR(p75) and NMDA receptors decline in cells treated with Aβ or the kinesin 5 inhibitor monastrol, or expressing APP. Aβ and monastrol also inhibit NGF-dependent neurite outgrowth from PC12 cells and glutamate-dependent Ca++ entry into primary neurons. Like Aβ, monastrol inhibits long-term potentiation, a cellular model of NMDA-dependent learning and memory, and kinesin 5 activity is absent from APP/PS transgenic mice brain or neurons treated with Aβ. These data imply that cognitive deficits in AD may derive in part from inhibition of neuronal Eg5 by Aβ, resulting in impaired neuronal function and/or survival through receptor mislocalization. Preventing inhibition of Eg5 or other motors by Aβ may represent a novel approach to AD therapy.
Collapse
Affiliation(s)
- Csilla Ari
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, College of Medicine, University of South Florida, Tampa, FL, USA
| | - Sergiy I Borysov
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, College of Medicine, University of South Florida, Tampa, FL, USA; Eric Pfeiffer Suncoast Alzheimer's Center, University of South Florida, Tampa, FL, USA; Department of Oncology, H. Lee Moffitt Cancer and Research Center, Tampa, FL, USA
| | - Jiashin Wu
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jaya Padmanabhan
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, College of Medicine, University of South Florida, Tampa, FL, USA
| | - Huntington Potter
- USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, College of Medicine, University of South Florida, Tampa, FL, USA; Eric Pfeiffer Suncoast Alzheimer's Center, University of South Florida, Tampa, FL, USA; Department of Neurology and Linda Crnic Institute for Down Syndrome, Anschutz Medical Campus, University of Colorado, Denver, Aurora, CO, USA.
| |
Collapse
|
123
|
Perez SE, Raghanti MA, Hof PR, Kramer L, Ikonomovic MD, Lacor PN, Erwin JM, Sherwood CC, Mufson EJ. Alzheimer's disease pathology in the neocortex and hippocampus of the western lowland gorilla (Gorilla gorilla gorilla). J Comp Neurol 2013; 521:4318-38. [PMID: 23881733 PMCID: PMC6317365 DOI: 10.1002/cne.23428] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 06/28/2013] [Accepted: 07/10/2013] [Indexed: 12/11/2022]
Abstract
The two major histopathologic hallmarks of Alzheimer's disease (AD) are amyloid beta protein (Aβ) plaques and neurofibrillary tangles (NFT). Aβ pathology is a common feature in the aged nonhuman primate brain, whereas NFT are found almost exclusively in humans. Few studies have examined AD-related pathology in great apes, which are the closest phylogenetic relatives of humans. In the present study, we examined Aβ and tau-like lesions in the neocortex and hippocampus of aged male and female western lowland gorillas using immunohistochemistry and histochemistry. Analysis revealed an age-related increase in Aβ-immunoreactive plaques and vasculature in the gorilla brain. Aβ plaques were more abundant in the neocortex and hippocampus of females, whereas Aβ-positive blood vessels were more widespread in male gorillas. Plaques were also Aβ40-, Aβ42-, and Aβ oligomer-immunoreactive, but only weakly thioflavine S- or 6-CN-PiB-positive in both sexes, indicative of the less fibrillar (diffuse) nature of Aβ plaques in gorillas. Although phosphorylated neurofilament immunostaining revealed a few dystrophic neurites and neurons, choline acetyltransferase-immunoreactive fibers were not dystrophic. Neurons stained for the tau marker Alz50 were found in the neocortex and hippocampus of gorillas at all ages. Occasional Alz50-, MC1-, and AT8-immunoreactive astrocyte and oligodendrocyte coiled bodies and neuritic clusters were seen in the neocortex and hippocampus of the oldest gorillas. This study demonstrates the spontaneous presence of both Aβ plaques and tau-like lesions in the neocortex and hippocampus in old male and female western lowland gorillas, placing this species at relevance in the context of AD research.
Collapse
Affiliation(s)
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, Ohio 44242
- Cleveland Metroparks Zoo, Cleveland, Ohio 44109
| | - Patrick R. Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | | | - Milos D. Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pennsylvania 15213
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pennsylvania 15213
| | - Pascale N. Lacor
- Neurobiology Department and Cognitive Neurology and Alzheimer’s Disease Center, Northwestern University, Evanston, Illinois 60208
| | - Joseph M. Erwin
- Department of Anthropology, The George Washington University, Washington, DC 20052
| | - Chet C. Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20052
| | | |
Collapse
|
124
|
Maarouf CL, Kokjohn TA, Whiteside CM, Macias MP, Kalback WM, Sabbagh MN, Beach TG, Vassar R, Roher AE. Molecular Differences and Similarities Between Alzheimer's Disease and the 5XFAD Transgenic Mouse Model of Amyloidosis. BIOCHEMISTRY INSIGHTS 2013; 6:1-10. [PMID: 25210460 PMCID: PMC4154482 DOI: 10.4137/bci.s13025] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Transgenic (Tg) mouse models of Alzheimer’s disease (AD) have been extensively used to study the pathophysiology of this dementia and to test the efficacy of drugs to treat AD. The 5XFAD Tg mouse, which contains two presenilin-1 and three amyloid precursor protein (APP) mutations, was designed to rapidly recapitulate a portion of the pathologic alterations present in human AD. APP and its proteolytic peptides, as well as apolipoprotein E and endogenous mouse tau, were investigated in the 5XFAD mice at 3 months, 6 months, and 9 months. AD and nondemented subjects were used as a frame of reference. APP, amyloid-beta (Aβ) peptides, APP C-terminal fragments (CT99, CT83, AICD), β-site APP-cleaving enzyme, and APLP1 substantially increased with age in the brains of 5XFAD mice. Endogenous mouse tau did not show age-related differences. The rapid synthesis of Aβ and its impact on neuronal loss and neuroinflammation make the 5XFAD mice a desirable paradigm to model AD.
Collapse
Affiliation(s)
- Chera L Maarouf
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute Sun City, AZ, USA
| | - Tyler A Kokjohn
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute Sun City, AZ, USA. ; Department of Microbiology, Midwestern University School of Medicine, Glendale, AZ, USA
| | - Charisse M Whiteside
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute Sun City, AZ, USA
| | - MiMi P Macias
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute Sun City, AZ, USA
| | - Walter M Kalback
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute Sun City, AZ, USA
| | - Marwan N Sabbagh
- Roberts Clinical Center, Banner Sun Health Research Institute Sun City, AZ, USA. ; University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Thomas G Beach
- Harold Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Robert Vassar
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alex E Roher
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute Sun City, AZ, USA
| |
Collapse
|
125
|
Zhao W, Dumanis SB, Tamboli IY, Rodriguez GA, Jo Ladu M, Moussa CEH, William Rebeck G. Human APOE genotype affects intraneuronal Aβ1-42 accumulation in a lentiviral gene transfer model. Hum Mol Genet 2013; 23:1365-75. [PMID: 24154541 DOI: 10.1093/hmg/ddt525] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intraneuronal accumulation of β-amyloid (Aβ)42 is one of the earliest pathological events in humans and in animal models of Alzheimer's disease (AD). Apolipoprotein E 4 (APOE4) is the major identified genetic risk factor for late-onset AD, with Aβ deposition beginning earlier in apoE4-positive subjects. To directly determine the effects of APOE genotype on intraneuronal accumulation of Aβ1-42 at the onset of AD pathogenesis, we introduced lentiviral Aβ1-42 into the cortex of APOE targeted replacement (TR) mice at the age of 8-9 months. We demonstrated a significant isoform-dependent effect of human APOE, with dramatically enhanced intracellular Aβ1-42 deposits in the cerebral cortex of APOE4-TR mice 2 weeks after injection. Double-immunofluorescent staining showed that intracellular accumulation of lentiviral Aβ1-42 was mainly present in neurons, localized to late endosomes/lysosomes. This intraneuronal accumulation of Aβ1-42 correlated with increased tau phosphorylation and cell death in the ipsilateral cortex around the injection site. Aβ1-42 was also observed in microglia, but not in astrocytes. Quantitative analysis revealed more neurons with Aβ1-42 while less microglia with Aβ1-42 nearest to the injection site of Aβ1-42 lentivirus in APOE4-TR mice. Finally, apoE was present in neurons of the ipsilateral cortex of APOE-TR mice at 2 weeks after lentivirus injection, in addition to astrocytes and microglia in both the ipsilateral and contralateral cerebral cortex. Taken together, these results demonstrate that apoE4 tips the balance of the glial and neuronal Aβ toward the intraneuronal accumulation of Aβ.
Collapse
Affiliation(s)
- Wenjuan Zhao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | | | | | | | | | | | | |
Collapse
|
126
|
Stargardt A, Gillis J, Kamphuis W, Wiemhoefer A, Kooijman L, Raspe M, Benckhuijsen W, Drijfhout JW, M. Hol E, Reits E. Reduced amyloid-β degradation in early Alzheimer's disease but not in the APPswePS1dE9 and 3xTg-AD mouse models. Aging Cell 2013; 12:499-507. [PMID: 23534431 DOI: 10.1111/acel.12074] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2013] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is hallmarked by amyloid-β (Aβ) peptides accumulation and aggregation in extracellular plaques, preceded by intracellular accumulation. We examined whether intracellular Aβ can be cleared by cytosolic peptidases and whether this capacity is affected during progression of sporadic AD (sAD) in humans and in the commonly used APPswePS1dE9 and 3xTg-AD mouse models. A quenched Aβ peptide that becomes fluorescent upon degradation was used to screen for Aβ-degrading cytoplasmic peptidases cleaving the aggregation-prone KLVFF region of the peptide. In addition, this quenched peptide was used to analyze Aβ-degrading capacity in the hippocampus of sAD patients with different Braak stages as well as APPswePS1dE9 and 3xTg-AD mice. Insulin-degrading enzyme (IDE) was found to be the main peptidase that degrades cytoplasmic, monomeric Aβ. Oligomerization of Aβ prevents its clearance by IDE. Intriguingly, the Aβ-degrading capacity decreases already during the earliest Braak stages of sAD, and this decline correlates with IDE protein levels, but not with mRNA levels. This suggests that decreased IDE levels could contribute to early sAD. In contrast to the human data, the commonly used APPswePS1dE9 and 3xTg-AD mouse models do not show altered Aβ degradation and IDE levels with AD progression, raising doubts whether mouse models that overproduce Aβ peptides are representative for human sAD.
Collapse
Affiliation(s)
- Anita Stargardt
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Judith Gillis
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Willem Kamphuis
- Department of Astrocyte Biology and Neurodegeneration Netherlands Institute for Neuroscience – an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW) Meibergdreef 47, 1105 BA Amsterdam The Netherlands
| | - Anne Wiemhoefer
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Lieneke Kooijman
- Department of Astrocyte Biology and Neurodegeneration Netherlands Institute for Neuroscience – an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW) Meibergdreef 47, 1105 BA Amsterdam The Netherlands
| | - Marcel Raspe
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Willemien Benckhuijsen
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Albinusdreef 2, 2333 ZA Leiden The Netherlands
| | - Jan W. Drijfhout
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Albinusdreef 2, 2333 ZA Leiden The Netherlands
| | - Elly M. Hol
- Department of Astrocyte Biology and Neurodegeneration Netherlands Institute for Neuroscience – an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW) Meibergdreef 47, 1105 BA Amsterdam The Netherlands
- Swammerdam Institute for Life Sciences Center for Neuroscience University of Amsterdam Sciencepark 904 1098 XH AmsterdamThe Netherlands
| | - Eric Reits
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| |
Collapse
|
127
|
Kumar S, Wirths O, Theil S, Gerth J, Bayer TA, Walter J. Early intraneuronal accumulation and increased aggregation of phosphorylated Abeta in a mouse model of Alzheimer's disease. Acta Neuropathol 2013; 125:699-709. [PMID: 23525537 DOI: 10.1007/s00401-013-1107-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/06/2013] [Accepted: 03/09/2013] [Indexed: 12/13/2022]
Abstract
The progressive accumulation of extracellular amyloid plaques in the brain is a common hallmark of Alzheimer's disease (AD). We recently identified a novel species of Aβ phosphorylated at serine residue 8 with increased propensity to form toxic aggregates as compared to non-phosphorylated species. The age-dependent analysis of Aβ depositions using novel monoclonal phosphorylation-state specific antibodies revealed that phosphorylated Aβ variants accumulate first inside of neurons in a mouse model of AD already at 2 month of age. At higher ages, phosphorylated Aβ is also abundantly detected in extracellular plaques. Besides a large overlap in the spatiotemporal deposition of phosphorylated and non-phosphorylated Aβ species, fractionized extraction of Aβ from brains revealed an increased accumulation of phosphorylated Aβ in oligomeric assemblies as compared to non-phosphorylated Aβ in vivo. Thus, phosphorylated Aβ could represent an important species in the formation and stabilization of neurotoxic aggregates, and might be targeted for AD therapy and diagnosis.
Collapse
Affiliation(s)
- Sathish Kumar
- Department of Neurology, University of Bonn, 53127, Bonn, Germany
| | | | | | | | | | | |
Collapse
|
128
|
Eimer WA, Vassar R. Neuron loss in the 5XFAD mouse model of Alzheimer's disease correlates with intraneuronal Aβ42 accumulation and Caspase-3 activation. Mol Neurodegener 2013; 8:2. [PMID: 23316765 PMCID: PMC3552866 DOI: 10.1186/1750-1326-8-2] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/09/2013] [Indexed: 01/23/2023] Open
Abstract
Background Although the mechanism of neuron loss in Alzheimer’s disease (AD) is enigmatic, it is associated with cerebral accumulation of Aβ42. The 5XFAD mouse model of amyloid deposition expresses five familial AD (FAD) mutations that are additive in driving Aβ42 overproduction. 5XFAD mice exhibit intraneuronal Aβ42 accumulation at 1.5 months, amyloid deposition at 2 months, and memory deficits by 4 months of age. Results Here, we demonstrate by unbiased stereology that statistically significant neuron loss occurs by 9 months of age in 5XFAD mice. We validated two Aβ42-selective antibodies by immunostaining 5XFAD; BACE1−/− bigenic brain sections and then used these antibodies to show that intraneuronal Aβ42 and amyloid deposition develop in the same regions where neuron loss is observed in 5XFAD brain. In 5XFAD neuronal soma, intraneuronal Aβ42 accumulates in puncta that co-label for Transferrin receptor and LAMP-1, indicating endosomal and lysosomal localization, respectively. In addition, in young 5XFAD brains, we observed activated Caspase-3 in the soma and proximal dendrites of intraneuronal Aβ42-labeled neurons. In older 5XFAD brains, we found activated Caspase-3-positive punctate accumulations that co-localize with the neuronal marker class III β-tubulin, suggesting neuron loss by apoptosis. Conclusions Together, our results indicate a temporal sequence of intraneuronal Aβ42 accumulation, Caspase-3 activation, and neuron loss that implies a potential apoptotic mechanism of neuron death in the 5XFAD mouse.
Collapse
Affiliation(s)
- William A Eimer
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | | |
Collapse
|
129
|
Tai LM, Bilousova T, Jungbauer L, Roeske SK, Youmans KL, Yu C, Poon WW, Cornwell LB, Miller CA, Vinters HV, Van Eldik LJ, Fardo DW, Estus S, Bu G, Gylys KH, Ladu MJ. Levels of soluble apolipoprotein E/amyloid-β (Aβ) complex are reduced and oligomeric Aβ increased with APOE4 and Alzheimer disease in a transgenic mouse model and human samples. J Biol Chem 2013; 288:5914-26. [PMID: 23293020 DOI: 10.1074/jbc.m112.442103] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human apolipoprotein E (apoE) isoforms may differentially modulate amyloid-β (Aβ) levels. Evidence suggests physical interactions between apoE and Aβ are partially responsible for these functional effects. However, the apoE/Aβ complex is not a single static structure; rather, it is defined by detection methods. Thus, literature results are inconsistent and difficult to interpret. An ELISA was developed to measure soluble apoE/Aβ in a single, quantitative method and was used to address the hypothesis that reduced levels of soluble apoE/Aβ and an increase in soluble Aβ, specifically oligomeric Aβ (oAβ), are associated with APOE4 and AD. Previously, soluble Aβ42 and oAβ levels were greater with APOE4 compared with APOE2/APOE3 in hippocampal homogenates from EFAD transgenic mice (expressing five familial AD mutations and human apoE isoforms). In this study, soluble apoE/Aβ levels were lower in E4FAD mice compared with E2FAD and E3FAD mice, thus providing evidence that apoE/Aβ levels isoform-specifically modulate soluble oAβ clearance. Similar results were observed in soluble preparations of human cortical synaptosomes; apoE/Aβ levels were lower in AD patients compared with controls and lower with APOE4 in the AD cohort. In human CSF, apoE/Aβ levels were also lower in AD patients and with APOE4 in the AD cohort. Importantly, although total Aβ42 levels decreased in AD patients compared with controls, oAβ levels increased and were greater with APOE4 in the AD cohort. Overall, apoE isoform-specific formation of soluble apoE/Aβ modulates oAβ levels, suggesting a basis for APOE4-induced AD risk and a mechanistic approach to AD biomarkers.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Youmans KL, Tai LM, Nwabuisi-Heath E, Jungbauer L, Kanekiyo T, Gan M, Kim J, Eimer WA, Estus S, Rebeck GW, Weeber EJ, Bu G, Yu C, LaDu MJ. APOE4-specific changes in Aβ accumulation in a new transgenic mouse model of Alzheimer disease. J Biol Chem 2012; 287:41774-86. [PMID: 23060451 PMCID: PMC3516726 DOI: 10.1074/jbc.m112.407957] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/02/2012] [Indexed: 11/06/2022] Open
Abstract
APOE4 is the greatest risk factor for Alzheimer disease (AD) and synergistic effects with amyloid-β peptide (Aβ) suggest interactions among apoE isoforms and different forms of Aβ accumulation. However, it remains unclear how the APOE genotype affects plaque morphology, intraneuronal Aβ, soluble Aβ42, and oligomeric Aβ (oAβ), particularly in vivo. As the introduction of human APOE significantly delays amyloid deposition in transgenic mice expressing familial AD (FAD) mutations (FAD-Tg), 5xFAD-Tg mice, which exhibit amyloid deposition by age 2 months, were crossed with apoE-targeted replacement mice to produce the new EFAD-Tg mice. Compared with 5xFAD mice, Aβ deposition was delayed by ∼4 months in the EFAD mice, allowing detection of early changes in Aβ accumulation from 2-6 months. Although plaque deposition is generally greater in E4FAD mice, E2/E3FAD mice have significantly more diffuse and E4FAD more compact plaques. As a first report in FAD-Tg mice, the APOE genotypes had no effect on intraneuronal Aβ accumulation in EFAD mice. In E4FAD mice, total apoE levels were lower and total Aβ levels higher than in E2FAD and E3FAD mice. Profiles from sequential three-step extractions (TBS, detergent, and formic acid) demonstrated that the lower level of total apoE4 is reflected only in the detergent-soluble fraction, indicating that less apoE4 is lipoprotein-associated, and perhaps less lipidated, compared with apoE2 and apoE3. Soluble Aβ42 and oAβ levels were highest in E4FAD mice, although soluble apoE2, apoE3, and apoE4 levels were comparable, suggesting that the differences in soluble Aβ42 and oAβ result from functional differences among the apoE isoforms. Thus, APOE differentially regulates multiple aspects of Aβ accumulation.
Collapse
Affiliation(s)
- Katherine L. Youmans
- From the Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Leon M. Tai
- From the Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Evelyn Nwabuisi-Heath
- From the Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Lisa Jungbauer
- From the Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Takahisa Kanekiyo
- the Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Ming Gan
- the Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Jungsu Kim
- the Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - William A. Eimer
- the Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Steve Estus
- the Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky 40536
| | - G. William Rebeck
- the Department of Neuroscience, Georgetown University, Washington, D. C. 20057, and
| | - Edwin J. Weeber
- the Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida 33613
| | - Guojun Bu
- the Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224
| | - Chunjiang Yu
- From the Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Mary Jo LaDu
- From the Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| |
Collapse
|
131
|
Youmans KL, Wolozin B. TDP-43: a new player on the AD field? Exp Neurol 2012; 237:90-5. [PMID: 22691390 DOI: 10.1016/j.expneurol.2012.05.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 05/17/2012] [Accepted: 05/27/2012] [Indexed: 12/20/2022]
|