101
|
Villette V, Poindessous-Jazat F, Bellessort B, Roullot E, Peterschmitt Y, Epelbaum J, Stéphan A, Dutar P. A new neuronal target for beta-amyloid peptide in the rat hippocampus. Neurobiol Aging 2012; 33:1126.e1-14. [DOI: 10.1016/j.neurobiolaging.2011.11.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/10/2011] [Accepted: 11/20/2011] [Indexed: 02/03/2023]
|
102
|
Gibson GE, Chen HL, Xu H, Qiu L, Xu Z, Denton TT, Shi Q. Deficits in the mitochondrial enzyme α-ketoglutarate dehydrogenase lead to Alzheimer's disease-like calcium dysregulation. Neurobiol Aging 2012; 33:1121.e13-24. [PMID: 22169199 PMCID: PMC3321099 DOI: 10.1016/j.neurobiolaging.2011.11.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 10/27/2011] [Accepted: 11/03/2011] [Indexed: 01/08/2023]
Abstract
Understanding the molecular sequence of events that culminate in multiple abnormalities in brains from patients that died with Alzheimer's disease (AD) will help to reveal the mechanisms of the disease and identify upstream events as therapeutic targets. The activity of the mitochondrial α-ketoglutarate dehydrogenase complex (KGDHC) in homogenates from autopsy brain declines with AD. Experimental reductions in KGDHC in mouse models of AD promote plaque and tangle formation, the hallmark pathologies of AD. We hypothesize that deficits in KGDHC also lead to the abnormalities in endoplasmic reticulum (ER) calcium stores and cytosolic calcium following K(+) depolarization that occurs in cells from AD patients and transgenic models of AD. The activity of the mitochondrial enzyme KGDHC was diminished acutely (minutes), long-term (days), or chronically (weeks). Acute inhibition of KGDHC produced effects on calcium opposite to those in AD, while the chronic or long-term inhibition of KGDHC mimicked the AD-related changes in calcium. Divergent changes in proteins released from the mitochondria that affect endoplasmic reticulum calcium channels may underlie the selective cellular consequences of acute versus longer term inhibition of KGDHC. The results suggest that the mitochondrial abnormalities in AD can be upstream of those in calcium.
Collapse
Affiliation(s)
- Gary E Gibson
- Department of Neurology and Neuroscience, Weill Cornell Medical College, Burke Medical Research Institute, White Plains, NY 10605, USA.
| | | | | | | | | | | | | |
Collapse
|
103
|
Lalonde R, Fukuchi K, Strazielle C. APP transgenic mice for modelling behavioural and psychological symptoms of dementia (BPSD). Neurosci Biobehav Rev 2012; 36:1357-75. [PMID: 22373961 PMCID: PMC3340431 DOI: 10.1016/j.neubiorev.2012.02.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 02/10/2012] [Accepted: 02/13/2012] [Indexed: 12/17/2022]
Abstract
The discovery of gene mutations responsible for autosomal dominant Alzheimer's disease has enabled researchers to reproduce in transgenic mice several hallmarks of this disorder, notably Aβ accumulation, though in most cases without neurofibrillary tangles. Mice expressing mutated and wild-type APP as well as C-terminal fragments of APP exhibit variations in exploratory activity reminiscent of behavioural and psychological symptoms of Alzheimer dementia (BPSD). In particular, open-field, spontaneous alternation, and elevated plus-maze tasks as well as aggression are modified in several APP transgenic mice relative to non-transgenic controls. However, depending on the precise murine models, changes in open-field and elevated plus-maze exploration occur in either direction, either increased or decreased relative to controls. It remains to be determined which neurotransmitter changes are responsible for this variability, in particular with respect to GABA, 5HT, and dopamine.
Collapse
Affiliation(s)
- R Lalonde
- Département de Psychologie, Faculté des Sciences, Université de Rouen, 76821 Mont-Saint-Aignan Cedex, France.
| | | | | |
Collapse
|
104
|
Hall AM, Roberson ED. Mouse models of Alzheimer's disease. Brain Res Bull 2012; 88:3-12. [PMID: 22142973 PMCID: PMC3546481 DOI: 10.1016/j.brainresbull.2011.11.017] [Citation(s) in RCA: 228] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 11/11/2011] [Accepted: 11/21/2011] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting 35 million people today. The search for new treatments is made ever more urgent by prospects for increasing prevalence due to population aging. Mouse models are one of the most important research tools for finding new treatments for AD. Here, we review those models. We begin by briefly reviewing the AD genetics on which mouse models are based and then consider the most common mouse models of AD, including mice transgenic for human amyloid precursor protein (hAPP) and beta-amyloid (Aβ), mice expressing mutant presenilin genes, mice modeling tau's role in AD, and apolipoprotein E models. The discussion highlights key features and important differences between these mouse models. We conclude with a discussion about the role of AD mouse models in the translational pipeline.
Collapse
Affiliation(s)
- Alicia M Hall
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
105
|
Kawamoto EM, Vivar C, Camandola S. Physiology and pathology of calcium signaling in the brain. Front Pharmacol 2012; 3:61. [PMID: 22518105 PMCID: PMC3325487 DOI: 10.3389/fphar.2012.00061] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/26/2012] [Indexed: 12/31/2022] Open
Abstract
Calcium (Ca(2+)) plays fundamental and diversified roles in neuronal plasticity. As second messenger of many signaling pathways, Ca(2+) as been shown to regulate neuronal gene expression, energy production, membrane excitability, synaptogenesis, synaptic transmission, and other processes underlying learning and memory and cell survival. The flexibility of Ca(2+) signaling is achieved by modifying cytosolic Ca(2+) concentrations via regulated opening of plasma membrane and subcellular Ca(2+) sensitive channels. The spatiotemporal patterns of intracellular Ca(2+) signals, and the ultimate cellular biological outcome, are also dependent upon termination mechanism, such as Ca(2+) buffering, extracellular extrusion, and intra-organelle sequestration. Because of the central role played by Ca(2+) in neuronal physiology, it is not surprising that even modest impairments of Ca(2+) homeostasis result in profound functional alterations. Despite their heterogeneous etiology neurodegenerative disorders, as well as the healthy aging process, are all characterized by disruption of Ca(2+) homeostasis and signaling. In this review we provide an overview of the main types of neuronal Ca(2+) channels and their role in neuronal plasticity. We will also discuss the participation of Ca(2+) signaling in neuronal aging and degeneration.
Collapse
Affiliation(s)
- Elisa Mitiko Kawamoto
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research ProgramBaltimore, MD, USA
| | - Carmen Vivar
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research ProgramBaltimore, MD, USA
| | - Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research ProgramBaltimore, MD, USA
| |
Collapse
|
106
|
Peng J, Liang G, Inan S, Wu Z, Joseph DJ, Meng Q, Peng Y, Eckenhoff MF, Wei H. Dantrolene ameliorates cognitive decline and neuropathology in Alzheimer triple transgenic mice. Neurosci Lett 2012; 516:274-9. [PMID: 22516463 DOI: 10.1016/j.neulet.2012.04.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 04/02/2012] [Accepted: 04/03/2012] [Indexed: 11/29/2022]
Abstract
Disruption of intracellular calcium homeostasis via abnormal and excessive activation of ryanodine receptors plays an important role in the neuropathology of Alzheimer's disease. We investigated the therapeutic effect of dantrolene, a ryanodine receptor antagonist, on cognitive dysfunction and neuropathology in the triple transgenic Alzheimer mouse model (3xTg-AD). 3xTg-AD mice were treated with dantrolene from 2 to 13 months of age. Learning and memory were measured with the Morris Water Maze at 6, 10, and 13 months of age. Amyloid and tau neuropathology and biomarkers for synaptic dysfunction and neurodegeneration were examined in the brain using immunoblotting or immunohistochemistry. Dantrolene treatment for 11 months significantly reduced both memory deficits and amyloid plaque load in the hippocampus in 13-month-old 3xTg-AD mice. Dantrolene treatment, however, had no effect on phosphorylated tau, phosphorylated or total GSK-3β, synaptic markers, or mitochondrial or cytosolic cytochrome C. Our results suggest that dantrolene significantly improves cognition in a murine model of Alzheimer's disease and is associated with a reduction in amyloid plaque burden, forming the basis for a novel therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Jun Peng
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
107
|
MOCA is an integrator of the neuronal death signals that are activated by familial Alzheimer's disease-related mutants of amyloid β precursor protein and presenilins. Biochem J 2012; 442:413-22. [PMID: 22115042 DOI: 10.1042/bj20100993] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The death of cholinergic neurons in the cerebral cortex and certain subcortical regions is linked to irreversible dementia relevant to AD (Alzheimer's disease). Although multiple studies have shown that expression of a FAD (familial AD)-linked APP (amyloid β precursor protein) or a PS (presenilin) mutant, but not that of wild-type APP or PS, induced neuronal death by activating intracellular death signals, it remains to be addressed how these signals are interrelated and what the key molecule involved in this process is. In the present study, we show that the PS1-mediated (or possibly the PS2-mediated) signal is essential for the APP-mediated death in a γ-secretase-independent manner and vice versa. MOCA (modifier of cell adhesion), which was originally identified as being a PS- and Rac1-binding protein, is a common downstream constituent of these neuronal death signals. Detailed molecular analysis indicates that MOCA is a key molecule of the AD-relevant neuronal death signals that links the PS-mediated death signal with the APP-mediated death signal at a point between Rac1 [or Cdc42 (cell division cycle 42)] and ASK1 (apoptosis signal-regulating kinase 1).
Collapse
|
108
|
Guo Q, Zheng H, Justice NJ. Central CRF system perturbation in an Alzheimer's disease knockin mouse model. Neurobiol Aging 2012; 33:2678-91. [PMID: 22336193 DOI: 10.1016/j.neurobiolaging.2012.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 12/23/2011] [Accepted: 01/07/2012] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is often accompanied by changes in mood as well as increases in circulating cortisol levels, suggesting that regulation of the stress responsive hypothalamic-pituitary-adrenal (HPA) axis is disturbed. Here, we show that amyloid precursor protein (APP) is endogenously expressed in important limbic, hypothalamic, and midbrain nuclei that regulate hypothalamic-pituitary-adrenal axis activity. Furthermore, in a knockin mouse model of AD that expresses familial AD (FAD) mutations of both APP with humanized amyloid beta (hAβ), and presenilin 1 (PS1), in their endogenous patterns (APP/hAβ/PS1 animals), corticotropin releasing factor (CRF) levels are increased in key stress-related nuclei, resting corticosteroid levels are elevated, and animals display increased anxiety-related behavior. Endocrine and behavioral phenotypes can be normalized by loss of 1 copy of CRF receptor type-1 (Crfr1), consistent with a perturbation of central CRF signaling in APP/hAβ/PS1 animals. However, reductions in anxiety and corticosteroid levels conferred by heterozygosity of CRF receptor type-1 do not improve a deficit in working memory observed in APP/hAβ/PS1 mice, suggesting that perturbations of the CRF system are not the primary cause of decreased cognitive performance.
Collapse
Affiliation(s)
- Qinxi Guo
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
109
|
Wang Y, Cheng Z, Qin W, Jia J. Val97Leu mutant presenilin-1 induces tau hyperphosphorylation and spatial memory deficit in mice and the underlying mechanisms. J Neurochem 2012; 121:135-45. [PMID: 21929538 DOI: 10.1111/j.1471-4159.2011.07489.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although the pathological role of presenilin-1 mutation in early onset familial Alzheimer's disease has been widely studied, few focused on how the presenilin-1 mutations result in memory impairment and tau hyperphosphorylation. In the present study, we expressed human Val97Leu mutant presenilin-1, which is reported in Chinese pedigrees by our group, in transgenic mice and found that the mutant presenilin-1 induced spatial memory deficit and tau hyperphosphorylation at PHF-1, pS199/202, pT231 and pS396 epitopes, but not at pS214 and pS422 epitopes. Pearson analysis showed that the memory deficit was only significantly correlated with tau phosphorylation level at PHF-1, pS199/202, pT231 and pS396 epitopes. Additionally, the hyperphosphorylated tau and tangle-like argentophilic structures were detected at CA3 and CA4, but not CA1, region of hippocampus, and we also found tangle-like structure and wizened degenerative neurons in frontal cortex. We demonstrated the tau hyperphosphorylation at the same epitopes in N2a cells expressing the mutant presenilin-1, which is caused by inhibition of phosphoinositol-3 kinase/Akt and activation of glycogen synthase kinase-3 specifically. Our data demonstrated that human Val97Leu mutant presenilin-1 causes spatial memory deficit in mice and increases tau phosphorylation level in glycogen synthase kinase-3-dependent manner.
Collapse
Affiliation(s)
- Yue Wang
- Department of Neurology, Xuan Wu Hospital of the Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
110
|
Shilling D, Mak DOD, Kang DE, Foskett JK. Lack of evidence for presenilins as endoplasmic reticulum Ca2+ leak channels. J Biol Chem 2012; 287:10933-44. [PMID: 22311977 DOI: 10.1074/jbc.m111.300491] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Familial Alzheimer disease (FAD) is linked to mutations in the presenilin (PS) homologs. FAD mutant PS expression has several cellular consequences, including exaggerated intracellular Ca(2+) ([Ca(2+)](i)) signaling due to enhanced agonist sensitivity and increased magnitude of [Ca(2+)](i) signals. The mechanisms underlying these phenomena remain controversial. It has been proposed that PSs are constitutively active, passive endoplasmic reticulum (ER) Ca(2+) leak channels and that FAD PS mutations disrupt this function resulting in ER store overfilling that increases the driving force for release upon ER Ca(2+) release channel opening. To investigate this hypothesis, we employed multiple Ca(2+) imaging protocols and indicators to directly measure ER Ca(2+) dynamics in several cell systems. However, we did not observe consistent evidence that PSs act as ER Ca(2+) leak channels. Nevertheless, we confirmed observations made using indirect measurements employed in previous reports that proposed this hypothesis. Specifically, cells lacking PS or expressing a FAD-linked PS mutation displayed increased area under the ionomycin-induced [Ca(2+)](i) versus time curve (AI) compared with cells expressing WT PS. However, an ER-targeted Ca(2+) indicator revealed that this did not reflect overloaded ER stores. Monensin pretreatment selectively attenuated the AI in cells lacking PS or expressing a FAD PS allele. These findings contradict the hypothesis that PSs form ER Ca(2+) leak channels and highlight the need to use ER-targeted Ca(2+) indicators when studying ER Ca(2+) dynamics.
Collapse
Affiliation(s)
- Dustin Shilling
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
111
|
Stranahan AM, Mattson MP. Recruiting adaptive cellular stress responses for successful brain ageing. Nat Rev Neurosci 2012; 13:209-16. [PMID: 22251954 DOI: 10.1038/nrn3151] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Successful ageing is determined in part by genetic background, but also by experiential factors associated with lifestyle and culture. Dietary, behavioural and pharmacological interventions have been identified as potential means to slow brain ageing and forestall neurodegenerative disease. Many of these interventions recruit adaptive cellular stress responses to strengthen neuronal networks and enhance plasticity. In this Science and Society article, we describe several determinants of healthy and pathological brain ageing, with insights into how these processes are accelerated or prevented. We also describe the mechanisms underlying the neuroprotective actions of exercise and nutritional interventions, with the goal of recruiting these molecular targets for the treatment and prevention of neurodegenerative disease.
Collapse
Affiliation(s)
- Alexis M Stranahan
- Physiology Department, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | |
Collapse
|
112
|
Ryanodine Receptor Physiology and Its Role in Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:217-34. [DOI: 10.1007/978-94-007-2888-2_9] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
113
|
Guo Q, Li H, Gaddam SSK, Justice NJ, Robertson CS, Zheng H. Amyloid precursor protein revisited: neuron-specific expression and highly stable nature of soluble derivatives. J Biol Chem 2011; 287:2437-45. [PMID: 22144675 DOI: 10.1074/jbc.m111.315051] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
APP processing and amyloid-β production play a central role in Alzheimer disease pathogenesis. APP has been considered a ubiquitously expressed protein. In addition to amyloid-β, α- or β-secretase-dependent cleavage of APP also generates soluble secreted APP (APPsα or APPsβ, respectively). Interestingly, APPsβ has been shown to be subject to further cleavage to create an N-APP fragment that binds to the DR6 death receptor and mediates axon pruning and degeneration under trophic factor withdrawal conditions. By performing APP immunocytochemical staining, we found that, unexpectedly, many antibodies yielded nonspecific staining in APP-null samples. Screening of a series of antibodies allowed us to identify a rabbit monoclonal antibody Y188 that is highly specific for APP and prompted us to re-examine the expression, localization, and stability of endogenous APP and APPsβ in wild-type and in APPsβ knock-in mice, respectively. In contrast to earlier studies, we found that APP is specifically expressed in neurons and that its expression cannot be detected in major types of glial cells under basal or neuroinflammatory conditions. Both APPsα and APPsβ are highly stable in the central nervous system (CNS) and do not undergo further cleavage with or without trophic factor support. Our results clarify several key questions with regard to the fundamental properties of APP and offer critical cellular insights into the pathophysiology of APP.
Collapse
Affiliation(s)
- Qinxi Guo
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
114
|
Chen X, Wu J, Lvovskaya S, Herndon E, Supnet C, Bezprozvanny I. Dantrolene is neuroprotective in Huntington's disease transgenic mouse model. Mol Neurodegener 2011; 6:81. [PMID: 22118545 PMCID: PMC3235068 DOI: 10.1186/1750-1326-6-81] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 11/25/2011] [Indexed: 11/21/2022] Open
Abstract
Background Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a polyglutamine expansion in the Huntingtin protein which results in the selective degeneration of striatal medium spiny neurons (MSNs). Our group has previously demonstrated that calcium (Ca2+) signaling is abnormal in MSNs from the yeast artificial chromosome transgenic mouse model of HD (YAC128). Moreover, we demonstrated that deranged intracellular Ca2+ signaling sensitizes YAC128 MSNs to glutamate-induced excitotoxicity when compared to wild type (WT) MSNs. In previous studies we also observed abnormal neuronal Ca2+ signaling in neurons from spinocerebellar ataxia 2 (SCA2) and spinocerebellar ataxia 3 (SCA3) mouse models and demonstrated that treatment with dantrolene, a ryanodine receptor antagonist and clinically relevant Ca2+ signaling stabilizer, was neuroprotective in experiments with these mouse models. The aim of the current study was to evaluate potential beneficial effects of dantrolene in experiments with YAC128 HD mouse model. Results The application of caffeine and glutamate resulted in increased Ca2+ release from intracellular stores in YAC128 MSN cultures when compared to WT MSN cultures. Pre-treatment with dantrolene protected YAC128 MSNs from glutamate excitotoxicty, with an effective concentration of 100 nM and above. Feeding dantrolene (5 mg/kg) twice a week to YAC128 mice between 2 months and 11.5 months of age resulted in significantly improved performance in the beam-walking and gait-walking assays. Neuropathological analysis revealed that long-term dantrolene feeding to YAC128 mice significantly reduced the loss of NeuN-positive striatal neurons and reduced formation of Httexp nuclear aggregates. Conclusions Our results support the hypothesis that deranged Ca2+ signaling plays an important role in HD pathology. Our data also implicate the RyanRs as a potential therapeutic target for the treatment of HD and demonstrate that RyanR inhibitors and Ca2+ signaling stabilizers such as dantrolene should be considered as potential therapeutics for the treatment of HD and other polyQ-expansion disorders.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| | | | | | | | | | | |
Collapse
|
115
|
Doyle KM, Kennedy D, Gorman AM, Gupta S, Healy SJM, Samali A. Unfolded proteins and endoplasmic reticulum stress in neurodegenerative disorders. J Cell Mol Med 2011; 15:2025-39. [PMID: 21722302 PMCID: PMC4394214 DOI: 10.1111/j.1582-4934.2011.01374.x] [Citation(s) in RCA: 259] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 06/16/2011] [Indexed: 12/11/2022] Open
Abstract
The stimuli for neuronal cell death in neurodegenerative disorders are multi-factorial and may include genetic predisposition, environmental factors, cellular stressors such as oxidative stress and free radical production, bioenergy failure, glutamate-induced excitotoxicity, neuroinflammation, disruption of Ca(2+) -regulating systems, mitochondrial dysfunction and misfolded protein accumulation. Cellular stress disrupts functioning of the endoplasmic reticulum (ER), a critical organelle for protein quality control, leading to induction of the unfolded protein response (UPR). ER stress may contribute to neurodegeneration in a range of neurodegenerative disorders. This review summarizes the molecular events occurring during ER stress and the unfolded protein response and it specifically evaluates the evidence suggesting the ER stress response plays a role in neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Donna Kennedy
- School of Natural Sciences, NUI GalwayGalway, Ireland
- Apoptosis Research Center, NUI GalwayGalway, Ireland
| | - Adrienne M Gorman
- School of Natural Sciences, NUI GalwayGalway, Ireland
- Apoptosis Research Center, NUI GalwayGalway, Ireland
| | - Sanjeev Gupta
- School of Medicine, NUI GalwayGalway, Ireland
- Apoptosis Research Center, NUI GalwayGalway, Ireland
| | - Sandra J M Healy
- School of Natural Sciences, NUI GalwayGalway, Ireland
- Apoptosis Research Center, NUI GalwayGalway, Ireland
| | - Afshin Samali
- School of Medicine, NUI GalwayGalway, Ireland
- School of Natural Sciences, NUI GalwayGalway, Ireland
| |
Collapse
|
116
|
Huang W, Xing W, Ryskamp DA, Punzo C, Križaj D. Localization and phenotype-specific expression of ryanodine calcium release channels in C57BL6 and DBA/2J mouse strains. Exp Eye Res 2011; 93:700-9. [PMID: 21933672 DOI: 10.1016/j.exer.2011.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/06/2011] [Accepted: 09/04/2011] [Indexed: 11/27/2022]
Abstract
The DBA/2J (D2) and C57BL6 (B6) mouse strains are widely used in research as models for anxiety, addiction and chronic glaucoma. D2, but not B6, animals develop elevated intraocular pressure (IOP) that leads to progressive degeneration of retinal ganglion cell (RGC) axons and perikarya. Here we compare the expression and localization of intracellular ryanodine receptor (RyR) Ca(2+) store mechanisms in retinas from D2 and B6 animals. A subset of experiments included retinas from D2-Gpnmb(+) mice as strain-specific controls for D2s. RT-PCR analysis showed 6-8 -fold upregulation RyR1, but not RyR2 or RyR3 transcripts, in D2 retinas. The upregulation was more pronounced in D2 retinas categorized as exhibiting moderate or severe glaucoma eyes compared to eyes with no/little glaucoma. In B6 retinas, RyR1 was expressed in neuronal perikarya/processes across all three retinal layers whereas little labeling was observed in astrocyte, microglial or Müller cell processes. In contrast, RyR1 antibodies strongly labeled radial processes of in D2 Müller glia, in which the staining colocalized with the activated glial stress marker GFAP. RyR1 staining in 1 month-old D2-Gpnmb(+) strain resembled expression in B6 retinas whereas moderate RyR1, but not GFAP, localization to Müller glia was observed in 10-12 months - old D2-Gpnmb(+) eyes. Both RyR1-ir and GFAP-ir were augmented in the microbead injection model of acute experimental glaucoma. We conclude that RyR1 exhibits differential expression and localization in two ubiquitously used mouse lines. While RyR1 signals can be regulated in a strain-specific manner, our data also suggest that RyR1 transcription is induced by early glial activation and/or elevation in intraocular pressure.
Collapse
Affiliation(s)
- Wei Huang
- Department of Ophthalmology & Visual Sciences, John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | | | |
Collapse
|
117
|
Oxidative Stress and β-Amyloid Protein in Alzheimer’s Disease. Neuromolecular Med 2011; 13:223-50. [DOI: 10.1007/s12017-011-8155-9] [Citation(s) in RCA: 218] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 08/26/2011] [Indexed: 12/21/2022]
|
118
|
Stutzmann GE, Mattson MP. Endoplasmic reticulum Ca(2+) handling in excitable cells in health and disease. Pharmacol Rev 2011; 63:700-27. [PMID: 21737534 PMCID: PMC3141879 DOI: 10.1124/pr.110.003814] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum (ER) is a morphologically and functionally diverse organelle capable of integrating multiple extracellular and internal signals and generating adaptive cellular responses. It plays fundamental roles in protein synthesis and folding and in cellular responses to metabolic and proteotoxic stress. In addition, the ER stores and releases Ca(2+) in sophisticated scenarios that regulate a range of processes in excitable cells throughout the body, including muscle contraction and relaxation, endocrine regulation of metabolism, learning and memory, and cell death. One or more Ca(2+) ATPases and two types of ER membrane Ca(2+) channels (inositol trisphosphate and ryanodine receptors) are the major proteins involved in ER Ca(2+) uptake and release, respectively. There are also direct and indirect interactions of ER Ca(2+) stores with plasma membrane and mitochondrial Ca(2+)-regulating systems. Pharmacological agents that selectively modify ER Ca(2+) release or uptake have enabled studies that revealed many different physiological roles for ER Ca(2+) signaling. Several inherited diseases are caused by mutations in ER Ca(2+)-regulating proteins, and perturbed ER Ca(2+) homeostasis is implicated in a range of acquired disorders. Preclinical investigations suggest a therapeutic potential for use of agents that target ER Ca(2+) handling systems of excitable cells in disorders ranging from cardiac arrhythmias and skeletal muscle myopathies to Alzheimer disease.
Collapse
Affiliation(s)
- Grace E Stutzmann
- Department of Neuroscience, Rosalind Franklin University/The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL 60064, USA.
| | | |
Collapse
|
119
|
Presenilin 1 regulates homeostatic synaptic scaling through Akt signaling. Nat Neurosci 2011; 14:1112-4. [PMID: 21841774 PMCID: PMC3164917 DOI: 10.1038/nn.2893] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 06/27/2011] [Indexed: 11/08/2022]
Abstract
Neurons adapt to long-lasting changes in network activity, both in vivo and in vitro, by adjusting their synaptic strengths to stabilize firing rates. We found that homeostatic scaling of excitatory synapses was impaired in hippocampal neurons derived from mice lacking presenilin 1 (Psen1(-/-) mice) or expressing a familial Alzheimer's disease-linked Psen1 mutation (Psen1(M146V)). These findings suggest that deficits in synaptic homeostasis may contribute to brain dysfunction in Alzheimer's disease.
Collapse
|
120
|
Camandola S, Mattson MP. Aberrant subcellular neuronal calcium regulation in aging and Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1813:965-73. [PMID: 20950656 PMCID: PMC3032815 DOI: 10.1016/j.bbamcr.2010.10.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/01/2010] [Accepted: 10/03/2010] [Indexed: 02/06/2023]
Abstract
In this mini-review/opinion article we describe evidence that multiple cellular and molecular alterations in Alzheimer's disease (AD) pathogenesis involve perturbed cellular calcium regulation, and that alterations in synaptic calcium handling may be early and pivotal events in the disease process. With advancing age neurons encounter increased oxidative stress and impaired energy metabolism, which compromise the function of proteins that control membrane excitability and subcellular calcium dynamics. Altered proteolytic cleavage of the β-amyloid precursor protein (APP) in response to the aging process in combination with genetic and environmental factors results in the production and accumulation of neurotoxic forms of amyloid β-peptide (Aβ). Aβ undergoes a self-aggregation process and concomitantly generates reactive oxygen species that can trigger membrane-associated oxidative stress which, in turn, impairs the functions of ion-motive ATPases and glutamate and glucose transporters thereby rendering neurons vulnerable to excitotoxicity and apoptosis. Mutations in presenilin-1 that cause early-onset AD increase Aβ production, but also result in an abnormal increase in the size of endoplasmic reticulum calcium stores. Some of the events in the neurodegenerative cascade can be counteracted in animal models by manipulations that stabilize neuronal calcium homeostasis including dietary energy restriction, agonists of glucagon-like peptide 1 receptors and drugs that activate mitochondrial potassium channels. Emerging knowledge of the actions of calcium upstream and downstream of Aβ provides opportunities to develop novel preventative and therapeutic interventions for AD. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.
Collapse
Affiliation(s)
- Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | | |
Collapse
|
121
|
Casas-Tinto S, Zhang Y, Sanchez-Garcia J, Gomez-Velazquez M, Rincon-Limas DE, Fernandez-Funez P. The ER stress factor XBP1s prevents amyloid-beta neurotoxicity. Hum Mol Genet 2011; 20:2144-60. [PMID: 21389082 DOI: 10.1093/hmg/ddr100] [Citation(s) in RCA: 227] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disorder clinically characterized by progressive cognitive impairment. A prominent pathologic hallmark in the AD brain is the abnormal accumulation of the amyloid-β 1-42 peptide (Aβ), but the exact pathways mediating Aβ neurotoxicity remain enigmatic. Endoplasmic reticulum (ER) stress is induced during AD, and has been indirectly implicated as a mediator of Aβ neurotoxicity. We report here that Aβ activates the ER stress response factor X-box binding protein 1 (XBP1) in transgenic flies and in mammalian cultured neurons, yielding its active form, the transcription factor XBP1s. XBP1s shows neuroprotective activity in two different AD models, flies expressing Aβ and mammalian cultured neurons treated with Aβ oligomers. Trying to identify the mechanisms mediating XBP1s neuroprotection, we found that in PC12 cells treated with Aβ oligomers, XBP1s prevents the accumulation of free calcium (Ca(2+)) in the cytosol. This protective activity can be mediated by the downregulation of a specific isoform of the ryanodine Ca(2+) channel, RyR3. In support of this observation, a mutation in the only ryanodine receptor (RyR) in flies also suppresses Aβ neurotoxicity, indicating the conserved mechanisms between the two AD models. These results underscore the functional relevance of XBP1s in Aβ toxicity, and uncover the potential of XBP1 and RyR as targets for AD therapeutics.
Collapse
Affiliation(s)
- Sergio Casas-Tinto
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | | | | | |
Collapse
|
122
|
Alzheimer disease-related presenilin-1 variants exert distinct effects on monoamine oxidase-A activity in vitro. J Neural Transm (Vienna) 2011; 118:987-95. [PMID: 21373759 DOI: 10.1007/s00702-011-0616-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 02/17/2011] [Indexed: 10/18/2022]
Abstract
Monoamine oxidase-A (MAO-A) has been associated with both depression and Alzheimer disease (AD). Recently, carriers of AD-related presenilin-1 (PS-1) alleles have been found to be at higher risk for developing clinical depression. We chose to examine whether PS-1 could influence MAO-A function in vitro. Overexpression of selected AD-related PS-1 variants (wildtype, Y115H, ΔEx9 and M146V) in mouse hippocampal HT-22 cells affects MAO-A catalytic activity in a variant-specific manner. The ability of the PS-1 substrate-competitor DAPT to induce MAO-A activity in cells expressing either PS-1 wildtype or PS-1(M146V) suggests the potential for a direct influence of PS-1 on MAO-A function. In support of this, we were able to co-immunoprecipitate MAO-A with FLAG-tagged PS-1 wildtype and M146V proteins. This potential for a direct protein-protein interaction between PS-1 and MAO-A is not specific for HT-22 cells as we were also able to co-immunoprecipitate MAO-A with FLAG-PS-1 variants in N2a mouse neuroblastoma cells and in HEK293 human embryonic kidney cells. Finally, we demonstrate that the two PS-1 variants reported to be associated with an increased incidence of clinical depression [e.g., A431E and L235V] both induce MAO-A activity in HT-22 cells. A direct influence of PS-1 variants on MAO-A function could provide an explanation for the changes in monoaminergic tone observed in several neurodegenerative processes including AD. The ability to induce MAO-A catalytic activity with a PS-1/γ-secretase inhibitor should also be considered when designing secretase inhibitor-based therapeutics.
Collapse
|
123
|
Trancikova A, Ramonet D, Moore DJ. Genetic Mouse Models of Neurodegenerative Diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:419-82. [DOI: 10.1016/b978-0-12-384878-9.00012-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
124
|
Tamayev R, Matsuda S, Fà M, Arancio O, D’Adamio L. Danish dementia mice suggest that loss of function and not the amyloid cascade causes synaptic plasticity and memory deficits. Proc Natl Acad Sci U S A 2010; 107:20822-7. [PMID: 21098268 PMCID: PMC2996452 DOI: 10.1073/pnas.1011689107] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
According to the prevailing "amyloid cascade hypothesis," genetic dementias such as Alzheimer's disease and familial Danish dementia (FDD) are caused by amyloid deposits that trigger tauopathy, neurodegeneration, and behavioral/cognitive alterations. To efficiently reproduce amyloid lesions, murine models of human dementias invariably use transgenic expression systems. However, recent FDD transgenic models showed that Danish amyloidosis does not cause memory defects, suggesting that other mechanisms cause Danish dementia. We studied an animal knock-in model of FDD (FDD(KI/+)) genetically congruous with human cases. FDD(KI/+) mice present reduced Bri2 levels, impaired synaptic plasticity and severe hippocampal memory deficits. These animals show no cerebral lesions that are reputed characteristics of human dementia, such as tangles or amyloid plaques. Bri2(+/-) mice exhibit synaptic and memory deficits similar to FDD(KI/+) mice, and memory loss of FDD(KI/+) mice is prevented by expression of WT BRI2, indicating that Danish dementia is caused by loss of BRI2 function. Together, the data suggest that clinical dementia in Danish patients occurs via a loss of function mechanism and not as a result of amyloidosis and tauopathy.
Collapse
Affiliation(s)
- Robert Tamayev
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; and
| | - Shuji Matsuda
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; and
| | - Mauro Fà
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032
| | - Luciano D’Adamio
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; and
| |
Collapse
|
125
|
van Tijn P, Kamphuis W, Marlatt MW, Hol EM, Lucassen PJ. Presenilin mouse and zebrafish models for dementia: focus on neurogenesis. Prog Neurobiol 2010; 93:149-64. [PMID: 21056616 DOI: 10.1016/j.pneurobio.2010.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 10/27/2010] [Accepted: 10/31/2010] [Indexed: 01/18/2023]
Abstract
Autosomal dominant mutations in the presenilin gene PSEN cause familial Alzheimer's disease (AD), a neurological disorder pathologically characterized by intraneuronal accumulation and extracellular deposition of amyloid-β in plaques and intraneuronal, hyperphosphorylated tau aggregation in neurofibrillary tangles. Presenilins (PS/PSENs) are part of the proteolytic γ-secretase complex, which cleaves substrate proteins within the membrane. Cleavage of the amyloid precursor protein (APP) by γ-secretase releases amyloid-β peptides. Besides its role in the processing of APP and other transmembrane proteins, presenilin plays an important role in neural progenitor cell maintenance and neurogenesis. In this review, we discuss the role of presenilin in relation to neurogenesis and neurodegeneration and review the currently available presenilin animal models. In addition to established mouse models, zebrafish are emerging as an attractive vertebrate model organism to study the role of presenilin during the development of the nervous system and in neurodegenerative disorders involving presenilin. Zebrafish is a suitable model organism for large-scale drug screening, making this a valuable model to identify novel therapeutic targets for AD.
Collapse
Affiliation(s)
- Paula van Tijn
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
126
|
Barron AM, Verdile G, Taddei K, Bates KA, Martins RN. Effect of chronic hCG administration on Alzheimer's-related cognition and A beta accumulation in PS1KI mice. Endocrinology 2010; 151:5380-8. [PMID: 20844010 DOI: 10.1210/en.2009-1168] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Age-associated changes in the reproductive hormones-the gonadal steroid hormones and the gonadotropins-have been identified as potential risk factors for Alzheimer's disease (AD). However, levels of gonadotropins and estrogens are closely linked in vivo, and it has proven difficult to separate the effects of gonadotropins from the well-documented estrogenic effects on AD-related neuropathology in experimental models of menopause. To assess the effects of gonadotropins on cognition and AD biochemical markers independent of estrogenic effects, a potent analog of luteinizing hormone [human chorionic gonadotropin (hCG)] was administered to ovariectomized presenilin1 knock-in mice (PS1KI). Gonadotropin administration was found to induce hyperactivity and anxiety (Open Field Maze and Taste Neophobia Task) and working memory dysfunction, without altering reference memory (Morris Water Maze). Although gonadotropin administration modestly altered β amyloid (Aβ40) levels, levels of the longer more toxic form (Aβ42) were unaffected. Furthermore, altered Aβ40 levels were not associated with observed behavioral and cognitive impairments. These findings provide proof, in principle, that the gonadotropin hormones play a role in the modulation of AD-related behavior, cognition, and neuropathology.
Collapse
Affiliation(s)
- A M Barron
- School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Nedlands, 6009, Australia
| | | | | | | | | |
Collapse
|
127
|
Zhou J, Wang H, Xiong Y, Li Z, Feng Y, Chen J. Puerarin attenuates glutamate-induced neurofilament axonal transport impairment. JOURNAL OF ETHNOPHARMACOLOGY 2010; 132:150-156. [PMID: 20727960 DOI: 10.1016/j.jep.2010.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 07/09/2010] [Accepted: 08/06/2010] [Indexed: 05/29/2023]
Abstract
AIM OF THE STUDY Puerarin (Pur) is a primary component of the most functional extracts of Pueraria lobata used in traditional Chinese medicine for centuries. Since it has been postulated that Pur protects the brain against glutamate (Glu) neurotoxicity, we investigated the effects of Pur on Glu-induced axonal transport impairment in primary hippocampal neurons in this study. MATERIALS AND METHODS Primary hippocampal cultures were prepared from 2-day-old Sprague-Dawley rats. Intracellular calcium concentration [Ca(2+)](i), neurofilament (NF) phosphorylation and protein kinase activity for Cdk5 were measured. Time-lapse imaging technology was used to capture the NF axonal transport in the cultured neurons with transiently transfected fluorescence protein linked to the N-terminus of NF-M (EGFP-NFM). RESULTS The results showed that Pur significantly diminished the Glu-induced elevation of [Ca(2+)](i) in dose-dependent manner and antagonized the Glu-evoked increases in NF phosphorylation at protein levels. The neurons under the Glu treatment displayed the accumulation of immobile NF clusters in the cell body and the reduced rates of axonal transport of NFs by 72.8% compared to the control neurons. Intriguingly, Pur reversed the slowed rate of the axonal transport by 35.6%. Pur also remarkably attenuated Glu-evoked activation of Cdk5. CONCLUSIONS Pur may play a role in protecting against Glu-induced NF axonal transport impairment in rat primary hippocampal neurons by inhibiting the increased [Ca(2+)](i) and by impeding the activation of Cdk5.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | | | | | | | |
Collapse
|
128
|
Li H, Wang Z, Wang B, Guo Q, Dolios G, Tabuchi K, Hammer RE, Südhof TC, Wang R, Zheng H. Genetic dissection of the amyloid precursor protein in developmental function and amyloid pathogenesis. J Biol Chem 2010; 285:30598-605. [PMID: 20693289 DOI: 10.1074/jbc.m110.137729] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Proteolytic processing of the amyloid precursor protein (APP) generates large soluble APP derivatives, β-amyloid (Aβ) peptides, and APP intracellular domain. Expression of the extracellular sequences of APP or its Caenorhabditis elegans counterpart has been shown to be sufficient in partially rescuing the CNS phenotypes of the APP-deficient mice and the lethality of the apl-1 null C. elegans, respectively, leaving open the question as what is the role of the highly conserved APP intracellular domain? To address this question, we created an APP knock-in allele in which the mouse Aβ sequence was replaced by the human Aβ. A frameshift mutation was introduced that replaced the last 39 residues of the APP sequence. We demonstrate that the C-terminal mutation does not overtly affect APP processing and amyloid pathology. In contrast, crossing the mutant allele with APP-like protein 2 (APLP2)-null mice results in similar neuromuscular synapse defects and early postnatal lethality as compared with mice doubly deficient in APP and APLP2, demonstrating an indispensable role of the APP C-terminal domain in these development activities. Our results establish an essential function of the conserved APP intracellular domain in developmental regulation, and this activity can be genetically uncoupled from APP processing and Aβ pathogenesis.
Collapse
Affiliation(s)
- Hongmei Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Evidence for the involvement of calbindin D28k in the presenilin 1 model of Alzheimer's disease. Neuroscience 2010; 169:532-43. [DOI: 10.1016/j.neuroscience.2010.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 03/26/2010] [Accepted: 04/01/2010] [Indexed: 11/23/2022]
|
130
|
Parent AT, Thinakaran G. Modeling presenilin-dependent familial Alzheimer's disease: emphasis on presenilin substrate-mediated signaling and synaptic function. Int J Alzheimers Dis 2010; 2010:825918. [PMID: 20798900 PMCID: PMC2925324 DOI: 10.4061/2010/825918] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 06/17/2010] [Indexed: 11/24/2022] Open
Abstract
Mutations in PSEN genes, which encode presenilin proteins, cause familial early-onset Alzheimer's disease (AD). Transgenic mouse models based on coexpression of familial AD-associated presenilin and amyloid precursor protein variants successfully mimic characteristic pathological features of AD, including plaque formation, synaptic dysfunction, and loss of memory. Presenilins function as the catalytic subunit of gamma-secretase, the enzyme that catalyzes intramembraneous proteolysis of amyloid precursor protein to release beta-amyloid peptides. Familial AD-associated mutations in presenilins alter the site of gamma-secretase cleavage in a manner that increases the generation of longer and highly fibrillogenic beta-amyloid peptides. In addition to amyloid precursor protein, gamma-secretase catalyzes intramembrane proteolysis of many other substrates known to be important for synaptic function. This paper focuses on how various animal models have enabled us to elucidate the physiological importance of diverse gamma-secretase substrates, including amyloid precursor protein and discusses their roles in the context of cellular signaling and synaptic function.
Collapse
Affiliation(s)
- Angèle T. Parent
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Gopal Thinakaran
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
- Department of Neurology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| |
Collapse
|
131
|
Zhang H, Sun S, Herreman A, De Strooper B, Bezprozvanny I. Role of presenilins in neuronal calcium homeostasis. J Neurosci 2010; 30:8566-80. [PMID: 20573903 PMCID: PMC2906098 DOI: 10.1523/jneurosci.1554-10.2010] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 04/30/2010] [Accepted: 05/09/2010] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disorder. Familial AD (FAD) mutations in presenilins have been linked to calcium (Ca(2+)) signaling abnormalities. To explain these results, we previously proposed that presenilins function as endoplasmic reticulum (ER) passive Ca(2+) leak channels. To directly investigate the role of presenilins in neuronal ER Ca(2+) homeostasis, we here performed a series of Ca(2+) imaging experiments with primary neuronal cultures from conditional presenilin double-knock-out mice (PS1(dTAG/dTAG), PS2(-/-)) and from triple-transgenic AD mice (KI-PS1(M146V), Thy1-APP(KM670/671NL), Thy1-tau(P301L)). Obtained results provided additional support to the hypothesis that presenilins function as ER Ca(2+) leak channels in neurons. Interestingly, we discovered that presenilins play a major role in ER Ca(2+) leak function in hippocampal but not in striatal neurons. We further discovered that, in hippocampal neurons, loss of presenilin-mediated ER Ca(2+) leak function was compensated by an increase in expression and function of ryanodine receptors (RyanRs). Long-term feeding of the RyanR inhibitor dantrolene to amyloid precursor protein-presenilin-1 mice (Thy1-APP(KM670/671NL), Thy1-PS1(L166P)) resulted in an increased amyloid load, loss of synaptic markers, and neuronal atrophy in hippocampal and cortical regions. These results indicate that disruption of ER Ca(2+) leak function of presenilins may play an important role in AD pathogenesis.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Suya Sun
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - An Herreman
- Center for Human Genetics, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium, and
- Department for Molecular and Developmental Genetics, VIB, Flanders Institute for Biotechnology, B-3000 Leuven, Belgium
| | - Bart De Strooper
- Center for Human Genetics, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium, and
- Department for Molecular and Developmental Genetics, VIB, Flanders Institute for Biotechnology, B-3000 Leuven, Belgium
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| |
Collapse
|
132
|
Jo DG, Arumugam TV, Woo HN, Park JS, Tang SC, Mughal M, Hyun DH, Park JH, Choi YH, Gwon AR, Camandola S, Cheng A, Cai H, Song W, Markesbery WR, Mattson MP. Evidence that gamma-secretase mediates oxidative stress-induced beta-secretase expression in Alzheimer's disease. Neurobiol Aging 2010; 31:917-25. [PMID: 18687504 PMCID: PMC2858254 DOI: 10.1016/j.neurobiolaging.2008.07.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2007] [Revised: 06/18/2008] [Accepted: 07/03/2008] [Indexed: 01/16/2023]
Abstract
Beta-secretase (BACE1), an enzyme responsible for the production of amyloid beta-peptide (Abeta), is increased by oxidative stress and is elevated in the brains of patients with sporadic Alzheimer's disease (AD). Here, we show that oxidative stress fails to induce BACE1 expression in presenilin-1 (gamma-secretase)-deficient cells and in normal cells treated with gamma-secretase inhibitors. Oxidative stress-induced beta-secretase activity and sAPPbeta levels were suppressed by gamma-secretase inhibitors. Levels of gamma- and beta-secretase activities were greater in brain tissue samples from AD patients compared to non-demented control subjects, and the elevated BACE1 level in the brains of 3xTgAD mice was reduced by treatment with a gamma-secretase inhibitor. Our findings suggest that gamma-secretase mediates oxidative stress-induced expression of BACE1 resulting in excessive Abeta production in AD.
Collapse
Affiliation(s)
- Dong-Gyu Jo
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Thiruma V. Arumugam
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
| | - Ha-Na Woo
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Jong-Sung Park
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Sung-Chun Tang
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
- Stroke Center, Department of Neurology, National Taiwan University Hospital, Taiwan
| | - Mohamed Mughal
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
| | - Dong-Hoon Hyun
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
- Division of Molecular Life Science, Ewha Women's University, Seoul, Korea
| | - Jun-Hyung Park
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Yun-Hyung Choi
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - A-Ryeong Gwon
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
| | - Aiwu Cheng
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
| | - Huaibin Cai
- Department of Psychiatry, Brain Research Center, and Graduate Program in Neuroscience, University of British Columbia, BC, Canada
| | - Weihong Song
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD
| | | | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
133
|
Abstract
The calcium ion (Ca(2+)) plays fundamental roles in orchestrating dynamic changes in the function and structure of nerve cell circuits in the brain. The endoplasmic reticulum (ER), an organelle that actively removes Ca(2+) from the cytoplasm, can release stored Ca(2+) through ER membrane receptor channels responsive either to the lipid messenger inositol trisphosphate (IP(3)) or to cytosolic Ca(2+). Emerging findings suggest that perturbed ER Ca(2+) homeostasis contributes to the dysfunction and degeneration of neurons that occurs in Alzheimer's disease (AD). Presenilin-1 (PS1) is an integral membrane protein in the ER; mutations in PS1 that cause early-onset inherited AD increase the pool of ER Ca(2+) available for release and also enhance Ca(2+) release through ER IP(3)- and ryanodine-sensitive channels. By enhancing Ca(2+) flux across the ER membrane, PS1 mutations may exaggerate Ca(2+) signaling in synaptic terminals and thereby render them vulnerable to dysfunction and degeneration in the settings of aging and amyloid accumulation in AD.
Collapse
Affiliation(s)
- Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
134
|
Nifosì F, Martinuzzi A, Toffanin T, Costanzo R, Vestri A, Battaglia M, Bertagnoni GE, Lupi A, Amistà P, Carollo C, Perini G. Hippocampal remodelling after MDMA neurotoxicity: a single case study. World J Biol Psychiatry 2010; 10:961-8. [PMID: 18609419 DOI: 10.1080/15622970701870933] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Acute ingestion of MDMA (ecstasy) causes a transient marked increase in serotonin and dopamine at central synapses. Recent studies demonstrated that MDMA induces damage of serotonergic nerve terminals and alters hippocampal processing. Pronounced cognitive deficits in MDMA users affect learning and memory abilities. This pattern of predominant and long-lasting memory dysfunction suggests that the functioning of the hippocampus might be affected by the neurotoxic effects of MDMA. We present the case of a 16-year-old girl who developed an acute organic and psychotic syndrome caused by occasional use of low to moderate dose of MDMA. Serial neuroimaging ((18)F-FDG-PET and brain MRI) were correlated with her neurocognitive performance and clinical evolution. The structural and metabolic changes correlated with a severe cognitive impairment. After 16 months of intensive neuropsychological rehabilitation she showed significant improvement in hippocampal-related memory cognitive functions, which correlated with normalization of her (18)F-FDG-PET and remarkable hippocampal remodelling. This case report indicates that even non-chronic MDMA use may cause subacute toxic encephalopathy in which the clinical evolution is paralleled by neuroimaging changes in specific cerebral areas. The most relevant aspect is the reversibility of the volumetric changes, which may be the structural correlate of an ongoing hippocampal remodelling.
Collapse
Affiliation(s)
- Francesco Nifosì
- Department of Neurosciences, Psychiatric Clinic, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Randall AD, Witton J, Booth C, Hynes-Allen A, Brown JT. The functional neurophysiology of the amyloid precursor protein (APP) processing pathway. Neuropharmacology 2010; 59:243-67. [PMID: 20167227 DOI: 10.1016/j.neuropharm.2010.02.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 02/11/2010] [Indexed: 01/12/2023]
Abstract
Amyloid beta (Abeta) peptides derived from proteolytic cleavage of amyloid precursor protein (APP) are thought to be a pivotal toxic species in the pathogenesis of Alzheimer's disease (AD). Furthermore, evidence has been accumulating that components of APP processing pathway are involved in non-pathological normal function of the CNS. In this review we aim to cover the extensive body of research aimed at understanding how components of this pathway contribute to neurophysiological function of the CNS in health and disease. We briefly outline changes to clinical neurophysiology seen in AD patients before discussing functional changes in mouse models of AD which range from changes to basal synaptic transmission and synaptic plasticity through to abnormal synchronous network activity. We then describe the various neurophysiological actions that are produced by application of exogenous Abeta in various forms, and finally discuss a number or other neurophysiological aspects of the APP pathway, including functional activities of components of secretase complexes other than Abeta production.
Collapse
Affiliation(s)
- A D Randall
- MRC Centre for Synaptic Plasticity, Department of Anatomy, University of Bristol School of Medical Sciences, Bristol, UK.
| | | | | | | | | |
Collapse
|
136
|
Supnet C, Bezprozvanny I. The dysregulation of intracellular calcium in Alzheimer disease. Cell Calcium 2010; 47:183-9. [PMID: 20080301 PMCID: PMC2834825 DOI: 10.1016/j.ceca.2009.12.014] [Citation(s) in RCA: 237] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Accepted: 12/29/2009] [Indexed: 11/24/2022]
Abstract
Alzheimer disease (AD) is the most common neurodegenerative disorder worldwide and is at present, incurable. The accumulation of toxic amyloid-beta (Abeta) peptide aggregates in AD brain are thought to trigger the extensive synaptic loss and neurodegeneration linked to cognitive decline, an idea that underlies the 'amyloid hypothesis' of AD etiology in both the familal (FAD) and sporadic forms of the disease. Mutations causing FAD also result in the dysregulation of neuronal calcium (Ca2+) handling and may contribute to AD pathogenesis, an idea termed the 'calcium hypothesis' of AD. In particular, Ca2+ dysregulation by the endoplasmic reticulum (ER) in AD mouse models results in augmented cytosolic Ca2+ levels which can trigger signalling cascades that are detrimental to neuronal function and health. However, there is growing evidence to suggest that not all forms of Ca2+ dysregulation in AD neurons are harmful and some of them instead may be compensatory. These changes may help modulate neuronal excitability and slow AD pathology, especially in the early stages of the disease. Clearly, a better understanding of how dysregulation of neuronal Ca2+ handling contributes to neurodegeneration and neuroprotection in AD is needed as Ca2+ signalling modulators are targets of great interest as potential AD therapeutics.
Collapse
Affiliation(s)
- Charlene Supnet
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX
| |
Collapse
|
137
|
Auffret A, Mariani J, Rovira C. Age-Related Progressive Synaptic Dysfunction: The Critical Role of Presenilin 1. Rev Neurosci 2010; 21:239-50. [DOI: 10.1515/revneuro.2010.21.4.239] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
138
|
Sacca R, Engle SJ, Qin W, Stock JL, McNeish JD. Genetically engineered mouse models in drug discovery research. Methods Mol Biol 2010; 602:37-54. [PMID: 20012391 DOI: 10.1007/978-1-60761-058-8_3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetically modified mouse models have been proven to be a powerful tool in drug discovery. The ability to genetically modify the mouse genome by removing or replacing a specific gene has enhanced our ability to identify and validate target genes of interest. In addition, many human diseases can be mimicked in the mouse and signaling pathways have been shown to be conserved. In spite of these advantages the technology has limitations. In transgenic animals there may be significant heterogeneity among different founders. In knock-out animals the predicted phenotypes are not always readily observed and occasionally a completely novel and unexpected phenotype emerges. To address the latter and ensure that a deep knowledge of the target of interest is obtained, we have developed a comprehensive phenotyping program which has identified novel phenotypes as well as any potential safety concerns which may be associated with a particular target. Finally we continue to explore innovative technologies as they become available such as RNAi for temporal and spatial gene knock-down and humanized models that may better simulate human disease states.
Collapse
Affiliation(s)
- Rosalba Sacca
- Genetically Modified Models Center of Emphasis, Pfizer Global Research and Development, Pfizer Inc., Groton, CT, USA
| | | | | | | | | |
Collapse
|
139
|
Auffret A, Gautheron V, Mattson MP, Mariani J, Rovira C. Progressive age-related impairment of the late long-term potentiation in Alzheimer's disease presenilin-1 mutant knock-in mice. J Alzheimers Dis 2010; 19:1021-33. [PMID: 20157256 PMCID: PMC2891870 DOI: 10.3233/jad-2010-1302] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Presenilin 1 (PS1) mutations are responsible for many early-onset familial Alzheimer's disease (FAD) cases. While increasing evidence points to impaired synaptic plasticity as an early event in AD, PS1 mutant mice exhibit a paradoxical increase in hippocampal long-term potentiation (LTP). Among PS1 mouse models, PS1 M146V mutant knock-in mice (PS1KI) are particularly interesting in that they exhibit memory impairment in spatial tasks. Here we investigated the effects of aging on two forms of LTP in PS1KI mice, the widely-studied early phase of LTP (E-LTP) and a particular form of LTP called late-LTP (L-LTP) which requires transcription and protein synthesis. L-LTP is thought to be critical for long-term memory. We found a lower L-LTP maintenance phase in PS1KI mice compared to wild type littermates at 3 months of age. As the mice age, they exhibit impairment of both the induction and maintenance phases of LTP. When E-LTP and NMDA receptor-mediated transmission were analyzed, PS1KI mice displayed an increase at 3 months compared to wild type littermates; this difference did not persist at older ages and finally decreased at 12 months. These results reveal an L-LTP decrease in PS1 mutant mice at an early stage, which occurs coincidently with a paradoxical enhancement of E-LTP. The observation of a decrease in both forms of LTP during aging supports the view that PS1KI mice are a valuable model for the study of age-dependent synaptic dysfunction and cognitive decline in AD.
Collapse
Affiliation(s)
- Alexandra Auffret
- Université Pierre et Marie Curie-Paris6, Centre National de la Recherche Scientifique, UMR 7102-NPA, Paris, France.
| | | | | | | | | |
Collapse
|
140
|
Abstract
More than two decades ago, dysregulation of the intracellular Ca2+ homeostasis was suggested to underlie the development of Alzheimer’s disease (AD). This hypothesis was tested in numerous in vitro studies, which revealed multiple Ca2+ signalling pathways able to contribute to AD pathology. It remained, however, unclear whether these pathways are also activated in vivo, in cells involved in signal processing in the living brain. Here we review recent data analysing intracellular Ca2+ signalling in vivo in the context of previous in vitro findings. We particularly focus on the processes taking place in the immediate vicinity of amyloid plaques and on their possible role for AD-mediated brain dysfunction.
Collapse
Affiliation(s)
- Marina Hermes
- University of Tübingen, Institute of Physiology II, Tübingen, Germany
| | | | | |
Collapse
|
141
|
Presenilin transgenic mice as models of Alzheimer's disease. Brain Struct Funct 2009; 214:127-43. [PMID: 19921519 DOI: 10.1007/s00429-009-0227-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2009] [Accepted: 10/20/2009] [Indexed: 12/21/2022]
Abstract
Mutations in presenilin-1 (PS1) and presenilin-2 (PS2) cause familial Alzheimer's disease (FAD). Presenilins influence multiple molecular pathways and are best known for their role in the gamma-secretase cleavage of type I transmembrane proteins including the amyloid precursor protein (APP). PS1 and PS2 FAD mutant transgenic mice have been generated using a variety of promoters. PS1-associated FAD mutations have also been knocked into the endogenous mouse gene. PS FAD mutant mice consistently show elevations of Abeta42 with little if any effect on Abeta40. When crossed with plaque forming APP FAD mutant lines, the PS1 FAD mutants cause earlier and more extensive plaque deposition. Although single transgenic PS1 or PS2 mice do not form plaques, they exhibit a number of pathological features including age-related neuronal and synaptic loss as well as vascular pathology. They also exhibit increased susceptibility to excitotoxic injury most likely on the basis of exaggerated calcium release from the endoplasmic reticulum. Electrophysiologically long-term potentiation in the hippocampus is increased in young PS1 FAD mutant mice but this effect appears to be lost with aging. In most studies neurogenesis in the adult hippocampus is also impaired by PS1 FAD mutants. Mice in which PS1 has been conditionally knocked out in adult forebrain on a PS2 null background (PS1/2 cDKO) develop a striking neurodegeneration that mimics AD neuropathology in being associated with neuronal and synaptic loss, astrogliosis and hyperphosphorylation of tau, although it is not accompanied by plaque deposits. The relevance of PS transgenic mice as models of AD is discussed.
Collapse
|
142
|
Christensen DZ, Bayer TA, Wirths O. Formic acid is essential for immunohistochemical detection of aggregated intraneuronal Abeta peptides in mouse models of Alzheimer's disease. Brain Res 2009; 1301:116-25. [PMID: 19751708 DOI: 10.1016/j.brainres.2009.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 09/03/2009] [Accepted: 09/04/2009] [Indexed: 11/27/2022]
Abstract
The staining protocols so far applied to study intracellular Abeta accumulation in human tissue have been inconsistent with varying use of heat and formic acid (FA) for antigen retrieval. Microwave heat treatment has been reported to enhance the staining of intraneuronal Abeta as compared to no or enzymatic pretreatment. FA is widely used to increase the staining of plaque pathology in AD, yet the effect of FA on intraneuronal Abeta staining has been reported to be low and similar to the effect of heat or even to counteract the enhancing effect of heat pretreatment on intraneuronal Abeta immunohistochemical detection. To overcome these inconsistencies, there is a need for optimization of the staining protocol for intraneuronal Abeta detection and more knowledge is required concerning the effects of the different antigen retrieval methods. In the present work, we optimized the staining protocol for intraneuronal Abeta in paraffin-embedded sections in relation to heat and FA using four different mouse models known to accumulate intraneuronal Abeta peptides. It was found that FA is essential for the staining of highly aggregated intraneuronal Abeta peptides in AD transgenic mouse tissue.
Collapse
|
143
|
Strazielle C, Jazi R, Verdier Y, Qian S, Lalonde R. Regional brain metabolism with cytochrome c oxidase histochemistry in a PS1/A246E mouse model of autosomal dominant Alzheimer's disease: correlations with behavior and oxidative stress. Neurochem Int 2009; 55:806-14. [PMID: 19682525 DOI: 10.1016/j.neuint.2009.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 08/01/2009] [Accepted: 08/05/2009] [Indexed: 01/08/2023]
Abstract
Mitochondrial dysfunction and brain metabolic alteration are early neurofunctional aspects in Alzheimer's disease (AD). Regional brain metabolism was analyzed by cytochrome c oxidase (COX) histochemistry in PS1-A246E mouse mutants, a model of autosomal dominant AD overexpressing beta-amyloid (Abeta) peptide without amyloidosis or cell degeneration. Immunohistochemical samples were analyzed on adjacent sections for regional Abeta1-42 levels, as well as DNA oxidative damage with 8-hydroxy-2-deoxyguanosine (8-OHdG). COX activity increased in the basal forebrain nuclear complex, specific parts of the amygdala and hippocampus, as well as in striatum and connected regions. On the contrary, a hypometabolism was observed in midline thalamic, interpeduncular, and pedonculopontine nuclei. The integration of these regions in circuitries subserving emotions, arousal, and cognitive functions may explain why neurochemical alterations in specific brain regions were linearly correlated with psychomotor slowing and disinhibition previously reported in the mutant. As the PS1-A246E model appears to mimick prodromal AD, the results support the existence of mitochondrial abnormalities prior to AD-related cognitive deficits. However, since affected PS1-A246E brain regions were not primarily those altered in AD-associated histopathological features and did not systematically display either Abeta overexpression or higher 8-OHdG immunolabelling, the hypermetabolism observed seems to comprise a compensatory reaction to early mitochondrial abnormalities; furthermore, neuronal synaptic function should be considered as particularly relevant in COX activity changes.
Collapse
|
144
|
Gautheron V, Auffret A, Mattson MP, Mariani J, Vernet-der Garabedian B. A new and simple approach for genotyping Alzheimer's disease presenilin-1 mutant knock-in mice. J Neurosci Methods 2009; 181:235-40. [PMID: 19465058 PMCID: PMC2789286 DOI: 10.1016/j.jneumeth.2009.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 05/14/2009] [Accepted: 05/15/2009] [Indexed: 12/21/2022]
Abstract
The use of transgenic mice expressing point mutations demands that the detection of the different alleles is efficient and reliable. In addition, the multiplication of transgenes included in mouse models of human disease underlines the importance of correct controls and the fact that investigators need an accurate and rapid genotyping of the littermates generated. In this study, we demonstrate a powerful alternative for genotyping using presenilin-1 mutant knock-in (PS1M146KI) mice as an example. Mutations in the presenilin-1 (PS1) gene are causally linked to many cases of early-onset inherited Alzheimer's disease (AD). PS1M146VKI mice that express the PS1M146V targeted allele at normal physiological levels and triple-transgenic model (3 x Tg-AD) derived from homozygous PS1M146VKI mice were generated to study the pathogenesis of AD. Genotyping PS1M146VKI line requires many steps and thus a large quantity of DNA. In PS1M146VKI mice, only three nucleotides are modified in the gene. Here we show that this small mutated DNA sequence can affect its secondary structure resulting in altered mobility that can be easily detected on a polyacrylamide gel, by the single-strand conformation polymorphism (SSCP) technique. Our results demonstrate that SSCP is a simple, accurate, repeatable and efficient method for the routine genotyping of this current AD model. This method could be easily applied to other transgenic mice.
Collapse
Affiliation(s)
- Vanessa Gautheron
- Université Pierre et Marie Curie-Paris6, Unité Mixte de Recherche (UMR) 7102-Neurobiologie des Processus Adaptatifs (NPA), Centre National de la Recherche Scientifique (CNRS), UMR 7102-NPA, F-75005 Paris, France.
| | | | | | | | | |
Collapse
|
145
|
Liu J, Tang TS, Tu H, Nelson O, Herndon E, Huynh DP, Pulst SM, Bezprozvanny I. Deranged calcium signaling and neurodegeneration in spinocerebellar ataxia type 2. J Neurosci 2009; 29:9148-62. [PMID: 19625506 PMCID: PMC2749883 DOI: 10.1523/jneurosci.0660-09.2009] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Revised: 03/18/2009] [Accepted: 06/07/2009] [Indexed: 01/31/2023] Open
Abstract
Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominantly inherited, neurodegenerative disease caused by an expansion of polyglutamine tracts in the cytosolic protein ataxin-2 (Atx2). Cerebellar Purkinje cells (PCs) are predominantly affected in SCA2. The cause of PC degeneration in SCA2 is unknown. Here we demonstrate that mutant Atx2-58Q, but not wild-type (WT) Atx2-22Q, specifically associates with the cytosolic C-terminal region of type 1 inositol 1,4,5-trisphosphate receptor (InsP(3)R1), an intracellular calcium (Ca(2+)) release channel. Association with Atx2-58Q increased the sensitivity of InsP(3)R1 to activation by InsP(3) in planar lipid bilayer reconstitution experiments. To validate physiological significance of these findings, we performed a series of experiments with an SCA2-58Q transgenic mouse model that expresses human full-length Atx2-58Q protein under the control of a PC-specific promoter. In Ca(2+) imaging experiments, we demonstrated that stimulation with 3,5-dihydroxyphenylglycine (DHPG) resulted in higher Ca(2+) responses in 58Q PC cultures than in WT PC cultures. DHPG-induced Ca(2+) responses in 58Q PC cultures were blocked by the addition of ryanodine, an inhibitor of the ryanodine receptor (RyanR). We further demonstrated that application of glutamate induced more pronounced cell death in 58Q PC cultures than in WT PC cultures. Glutamate-induced cell death of 58Q PC cultures was attenuated by dantrolene, a clinically relevant RyanR inhibitor and Ca(2+) stabilizer. In whole animal experiments, we demonstrated that long-term feeding of SCA1-58Q mice with dantrolene alleviated age-dependent motor deficits (quantified in beam-walk and rotarod assays) and reduced PC loss observed in untreated SCA2-58Q mice by 12 months of age (quantified by stereology). Results of our studies indicate that disturbed neuronal Ca(2+) signaling may play an important role in SCA2 pathology and also suggest that the RyanR constitutes a potential therapeutic target for treatment of SCA2 patients.
Collapse
Affiliation(s)
| | | | | | | | - Emily Herndon
- Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, and
| | | | - Stefan M. Pulst
- Department of Neurology and
- Utah Brain Institute, University of Utah, Salt Lake City, Utah 84112
| | | |
Collapse
|
146
|
Fu YJ, Xiong S, Lovell MA, Lynn BC. Quantitative proteomic analysis of mitochondria in aging PS-1 transgenic mice. Cell Mol Neurobiol 2009; 29:649-64. [PMID: 19241155 PMCID: PMC2880535 DOI: 10.1007/s10571-009-9359-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/03/2009] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests mitochondrial alterations are intimately associated with the pathogenesis of Alzheimer's disease (AD). In order to determine if mutations of presenilin-1 (PS-1) affect levels of mitochondrial proteins at different ages we enriched mitochondrial fractions from 3-, 6-, 12-month-old knock-in mice expressing the M146V PS-1 mutation and identified, and quantified proteins using cleavable isotope-coded affinity tag labeling and two-dimensional liquid chromatography/tandem mass spectrometry (2D-LC/MS/MS). Using this approach, 165 non-redundant proteins were identified with 80 of them present in all three age groups. Specifically, at young ages (3 and 6 months), Na(+)/K(+) ATPase and several signal transduction proteins exhibited elevated levels, but dropped dramatically at 12 months. In contrast, components of the oxidative phosporylation pathway (OXPHOS), the mitochondrial permeability transition pore (MPTP), and energy metabolism proteins remained unchanged at 3 months but significantly increased with age. We propose that alterations in calcium homeostasis induced by the PS-1 mutation have a major impact in young animals by inhibiting the function of relevant proteins and inducing compensatory changes. However, in older mice combination of the PS-1 mutation and accumulated oxidative damage results in a functional suppression of OXPHOS and MPTP proteins requiring a compensatory increase in expression levels. In contrast, signal transduction proteins showed decreased levels due to a break down in the compensatory mechanisms. The dysfunction of Na(+)/K(+) ATPase and signal transduction proteins may induce impaired cognition and memory before neurodegeneration occurs.
Collapse
Affiliation(s)
- You-Jun Fu
- Department of Chemistry, University of Kentucky, Lexington, KY 40506 USA
| | - Shuling Xiong
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA
| | - Mark A. Lovell
- Department of Chemistry, University of Kentucky, Lexington, KY 40506 USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA
| | - Bert C. Lynn
- Department of Chemistry, University of Kentucky, Lexington, KY 40506 USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA
| |
Collapse
|
147
|
Wang Y, Greig NH, Yu QS, Mattson MP. Presenilin-1 mutation impairs cholinergic modulation of synaptic plasticity and suppresses NMDA currents in hippocampus slices. Neurobiol Aging 2009; 30:1061-8. [PMID: 18068871 PMCID: PMC2717610 DOI: 10.1016/j.neurobiolaging.2007.10.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Revised: 10/10/2007] [Accepted: 10/18/2007] [Indexed: 11/25/2022]
Abstract
Presenilin-1 (PS1) mutations cause many cases of early-onset inherited Alzheimer's disease, in part, by increasing the production of neurotoxic forms of amyloid beta-peptide (Abeta). However, Abeta-independent effects of mutant PS1 on neuronal Ca(2+) homeostasis and sensitivity to excitatory neurotransmitters have been reported. Here we show that cholinergic modulation of hippocampal synaptic plasticity is impaired in PS1 mutant knockin (PS1KI) mice. Whereas activation of muscarinic receptors enhances LTP at CA1 synapses of normal mice, it impairs LTP in PS1KI mice. Similarly, mutant PS1 impairs the ability of the cholinesterase inhibitor phenserine to enhance LTP. The NMDA current is decreased in CA1 neurons of PS1KI mice and is restored by intracellular Ca(2+)chelation. Similar alterations in acetylcholine and NMDA receptor-mediated components of synaptic plasticity are evident in 3xTgAD mice with PS1, amyloid precursor protein and tau mutations, suggesting that the adverse effects of mutant PS1 on synaptic plasticity can occur in the absence or presence of amyloid and tau pathologies.
Collapse
Affiliation(s)
- Yue Wang
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Gerontology Research Center, Baltimore, MD 21224, USA.
| | | | | | | |
Collapse
|
148
|
Chihara K, Saito A, Murakami T, Hino SI, Aoki Y, Sekiya H, Aikawa Y, Wanaka A, Imaizumi K. Increased vulnerability of hippocampal pyramidal neurons to the toxicity of kainic acid in OASIS-deficient mice. J Neurochem 2009; 110:956-65. [PMID: 19549009 DOI: 10.1111/j.1471-4159.2009.06188.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The endoplasmic reticulum (ER) stress response is a defense system for dealing with the accumulation of unfolded proteins in the ER lumen. Old astrocyte specifically induced substance (OASIS) is known to be expressed in astrocytes and involved in the ER stress response; however the function of OASIS in the injured brain has remained unclear. In this study, we examined the roles of OASIS in neuronal degeneration in the hippocampi of mice intraperitoneally injected with kainic acid (KA). OASIS mRNA was strongly induced in response to KA injection, with a similar time course to the induction of ER molecular chaperone immunoglobulin heavy chain binding protein mRNA. In situ hybridization showed that KA injection causes induction of immunoglobulin heavy chain binding protein mRNA in glial fibrillary acidic protein-positive astrocytes as well as in pyramidal neurons, although up-regulation of OASIS mRNA was only detected in glial fibrillary acidic protein-positive astrocytes. Primary cultured astrocytes, but not the neurons of OASIS-/- mice, revealed reduced vulnerability to ER stress. Furthermore, pyramidal neurons in the hippocampi of OASIS-/- mice were more susceptible to the toxicity induced by KA than those of wild-type mice. Taken together, these data suggest that OASIS expressed in astrocytes plays important roles in protection against the neuronal damage induced by KA.
Collapse
Affiliation(s)
- Kazuyasu Chihara
- Division of Molecular and Cellular Biology, Department of Anatomy, Faculty of Medicine, University of Miyazaki, Kihara, Kiyotake, Miyazaki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Blocking Abeta42 accumulation delays the onset and progression of tau pathology via the C terminus of heat shock protein70-interacting protein: a mechanistic link between Abeta and tau pathology. J Neurosci 2009; 28:12163-75. [PMID: 19020010 DOI: 10.1523/jneurosci.2464-08.2008] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The molecular alterations that induce tau pathology in Alzheimer disease (AD) are not known, particularly whether this is an amyloid-beta (Abeta)-dependent or -independent event. We addressed this issue in the 3xTg-AD mice using both genetic and immunological approaches and show that a selective decrease in Abeta(42) markedly delays the progression of tau pathology. The mechanism underlying this effect involves alterations in the levels of C terminus of heat shock protein70-interacting protein (CHIP) as we show that Abeta accumulation decreases CHIP expression and increases tau levels. We show that the Abeta-induced effects on tau were rescued by restoring CHIP levels. Our findings have profound clinical implications as they indicate that preventing Abeta accumulation will significantly alter AD progression. These data highlight the critical role CHIP plays as a link between Abeta and tau and identify CHIP as a new potential target not only for AD but for other neurodegenerative disorders characterized by tau accumulation.
Collapse
|
150
|
Hashimoto Y, Kurita M, Aiso S, Nishimoto I, Matsuoka M. Humanin inhibits neuronal cell death by interacting with a cytokine receptor complex or complexes involving CNTF receptor alpha/WSX-1/gp130. Mol Biol Cell 2009; 20:2864-73. [PMID: 19386761 DOI: 10.1091/mbc.e09-02-0168] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Humanin (HN) inhibits neuronal death induced by various Alzheimer's disease (AD)-related insults via an unknown receptor on cell membranes. Our earlier study indicated that the activation of STAT3 was essential for HN-induced neuroprotection, suggesting that the HN receptor may belong to the cytokine receptor family. In this study, a series of loss-of-function tests indicated that gp130, the common subunit of receptors belonging to the IL-6 receptor family, was essential for HN-induced neuroprotection. Overexpression of ciliary neurotrophic factor receptor alpha (CNTFR) and/or the IL-27 receptor subunit, WSX-1, but not that of any other tested gp130-related receptor subunit, up-regulated HN binding to neuronal cells, whereas siRNA-mediated knockdown of endogenous CNTFR and/or WSX-1 reduced it. These results suggest that both CNTFR and WSX-1 may be also involved in HN binding to cells. Consistent with these results, loss-of-functions of CNTFR or WSX-1 in neuronal cells nullified their responsiveness to HN-mediated protection. In vitro-reconstituted binding assays showed that HN, but not the other control peptide, induced the hetero-oligomerization of CNTFR, WSX-1, and gp130. Together, these results indicate that HN protects neurons by binding to a complex or complexes involving CNTFR/WSX-1/gp130.
Collapse
Affiliation(s)
- Yuichi Hashimoto
- Department of Pharmacology and Neuroscience, Tokyo Medical University, Shinjuku-ku, Tokyo 160-8402, Japan
| | | | | | | | | |
Collapse
|