1
|
Ashkavand Z, Ryan KC, Laboy JT, Patel R, Geller B, Norman KR. Identification of presenilin mutations that have sufficient gamma-secretase proteolytic activity to mediate Notch signaling but disrupt organelle and neuronal health. Neurobiol Dis 2025; 212:106961. [PMID: 40404063 DOI: 10.1016/j.nbd.2025.106961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/09/2025] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Mutations that cause familial Alzheimer's disease (AD) are predominantly found in the presenilin (PSEN) encoding genes PSEN1 and PSEN2. While the association of PSEN mutations with familial AD have been known for over 20 years, the mechanism underlying the impact these mutations have on disease is not fully understood. PSENs are phylogenetically conserved proteins that are found in diverse multicellular organisms ranging from plants to humans. PSENs form the proteolytic core of gamma-secretase that is required for cleaving type I transmembrane proteins, such as Notch receptors and the amyloid precursor protein. Importantly, familial AD-associated PSEN mutations are broadly distributed and do not clearly define a specific PSEN function essential for neuronal fitness. Here, using C. elegans as a model organism to study the in vivo functions of PSENs, we confirm that C. elegans PSEN plays a pivotal role in gamma-secretase proteolytic activity as well as maintaining neuronal and organelle health. Notably, we demonstrate that these two functions can be genetically uncoupled. Our research identifies several conserved familial AD-like missense mutations in the endogenous sel-12 gene, which encodes C. elegans PSEN. These mutations preserve sufficient gamma-secretase proteolytic activity to mediate Notch signaling but abolish PSEN's role in supporting neuronal and organelle health. Furthermore, we provide evidence that these familial AD-like missense mutations disrupt mitochondrial calcium regulation, ultimately leading to neuronal dysfunction. These results indicate that C. elegans PSEN plays at least two independent roles: one that mediates gamma-secretase proteolytic activity and another that mediates organelle and neuronal health.
Collapse
Affiliation(s)
- Zahra Ashkavand
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, NY 12208, USA
| | - Kerry C Ryan
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, NY 12208, USA
| | - Jocelyn T Laboy
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, NY 12208, USA
| | - Ritika Patel
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, NY 12208, USA
| | - Brian Geller
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, NY 12208, USA
| | - Kenneth R Norman
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, NY 12208, USA.
| |
Collapse
|
2
|
Liu L, Tang L, Wang Y, Liu S, Zhang Y. Expression of ITPR2 regulated by lncRNA-NONMMUT020270.2 in LPS-stimulated HT22 cells. Heliyon 2024; 10:e33491. [PMID: 39040287 PMCID: PMC11260991 DOI: 10.1016/j.heliyon.2024.e33491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Background Long non-coding RNA (lncRNA)-NONMMUT020270.2 is downregulated and co-expressed with inositol 1,4,5-trisphosphate receptor type 2 (ITPR2) in the hippocampus of Alzheimer's disease (AD) mice. However, whether the expression of ITPR2 was regulated by lncRNA-NONMMUT020270.2 remains unclear. we aimed to investigate regulating relationship of lncRNA-NONMMUT020270.2 and ITPR2. Methods HT22 cells were firstly transfected with the pcDNA3.1-lncRNA-NONMMUT020270.2 overexpression plasmid or with the lncRNA-NONMMUT020270.2 smart silencer, and then were stimulated with lipopolysaccharide (LPS) for 24h. The mRNA expression levels of lncRNA-NONMMUT020270.2 and ITPR2 were measured by reverse transcription-quantitative PCR. Cell viability was assessed using a Cell Counting Kit 8 assay. The expression of Aβ1-42 was detected by ELISA. The expression levels of p-tau, caspase-1, and inositol trisphosphate receptor (IP3R) proteins were detected by western-blotting. Nuclear morphological changes were detected by Hoechst staining. Flow cytometry and Fluo-3/AM were carried out to determine cell apoptosis and the intracellular Ca2+. Results LPS significantly decreased cell viability, and ITPR2 mRNA and IP3R protein expression levels. While it markedly enhanced the expression levels of p-tau and Aβ1-42, cell apoptosis rate, as well as intracellular Ca2+ concentration (P < 0.05). In addition, lncRNA-NONMMUT020270.2 overexpression significantly increased the expressions levels of ITPR2 mRNA and IP3R protein (P < 0.05), and inhibited expression of p-tau and Aβ1-42, cell apoptosis rate, and reduced intracellular Ca2+ concentration (P < 0.05). By contrast, lncRNA-NONMMUT020270.2 silencing notably downregulated expressions levels of ITPR2 mRNA and IP3R protein (P < 0.05), and elevated expression levels of p-tau and Aβ1-42, cell apoptosis rate, and intracellular Ca2+ concentration (P < 0.05). Conclusion lncRNA-NONMMUT020270.2 was positively correlated with ITPR2 expression in LPS-induced cell. Downregulating the lncRNA-NONMMUT020270.2 and ITPR2 may promote cell apoptosis and increase intracellular Ca2+ concentration.
Collapse
Affiliation(s)
- Lan Liu
- Medical College, Tibet University, Lhasa, Tibet, 850000, People's Republic of China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Liang Tang
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Yan Wang
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Shanling Liu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yongcang Zhang
- Medical College, Tibet University, Lhasa, Tibet, 850000, People's Republic of China
| |
Collapse
|
3
|
Wang T, Pang L, He M, Wang Z. Small-molecule inhibitors targeting apoptosis signal-regulated kinase 1. Eur J Med Chem 2023; 262:115889. [PMID: 37883895 DOI: 10.1016/j.ejmech.2023.115889] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
Apoptosis signal regulated kinase 1 (ASK1, also known as MAP3K5) is a member of the mitogen activated protein kinase kinase kinase (MAP3K) family. Since its first isolation from a human macrophage library in 1996, its research has been ongoing for over 25 years. A large number of reports have revealed that ASK1, as a key activator of the p38 mitogen-activated protein kinase and c-Jun N-terminal kinase (JNK) signaling cascade, responds to various stressors, and its inhibitors have important potential value in the treatment of diseases such as inflammation, cancer, and the nervous system and so on. This review summarizes the recent development in this field, including the structure and signaling pathways of ASK1, with a particular focus on the structure-activity relationships, and the hit-to-lead optimization strategies.
Collapse
Affiliation(s)
- Tiantian Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Lidan Pang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China
| | - Mengni He
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China
| | - Zengtao Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| |
Collapse
|
4
|
Alexander C, Parsaee A, Vasefi M. Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer's Disease. BIOLOGY 2023; 12:1453. [PMID: 37998052 PMCID: PMC10669725 DOI: 10.3390/biology12111453] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder impairing cognition and memory in the elderly. This disorder has a complex etiology, including senile plaque and neurofibrillary tangle formation, neuroinflammation, oxidative stress, and damaged neuroplasticity. Current treatment options are limited, so alternative treatments such as herbal medicine could suppress symptoms while slowing cognitive decline. We followed PRISMA guidelines to identify potential herbal treatments, their associated medicinal phytochemicals, and the potential mechanisms of these treatments. Common herbs, including Ginkgo biloba, Camellia sinensis, Glycyrrhiza uralensis, Cyperus rotundus, and Buplerum falcatum, produced promising pre-clinical results. These herbs are rich in kaempferol and quercetin, flavonoids with a polyphenolic structure that facilitate multiple mechanisms of action. These mechanisms include the inhibition of Aβ plaque formation, a reduction in tau hyperphosphorylation, the suppression of oxidative stress, and the modulation of BDNF and PI3K/AKT pathways. Using pre-clinical findings from quercetin research and the comparatively limited data on kaempferol, we proposed that kaempferol ameliorates the neuroinflammatory state, maintains proper cellular function, and restores pro-neuroplastic signaling. In this review, we discuss the anti-AD mechanisms of quercetin and kaempferol and their limitations, and we suggest a potential alternative treatment for AD. Our findings lead us to conclude that a polyherbal kaempferol- and quercetin-rich cocktail could treat AD-related brain damage.
Collapse
Affiliation(s)
- Claire Alexander
- Department of Biology, Lamar University, Beaumont, TX 77705, USA
| | - Ali Parsaee
- Biological Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Maryam Vasefi
- Department of Biology, Lamar University, Beaumont, TX 77705, USA
| |
Collapse
|
5
|
Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer's disease: challenges, successes and future. Signal Transduct Target Ther 2023; 8:248. [PMID: 37386015 PMCID: PMC10310781 DOI: 10.1038/s41392-023-01484-7] [Citation(s) in RCA: 341] [Impact Index Per Article: 170.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
Amyloid β protein (Aβ) is the main component of neuritic plaques in Alzheimer's disease (AD), and its accumulation has been considered as the molecular driver of Alzheimer's pathogenesis and progression. Aβ has been the prime target for the development of AD therapy. However, the repeated failures of Aβ-targeted clinical trials have cast considerable doubt on the amyloid cascade hypothesis and whether the development of Alzheimer's drug has followed the correct course. However, the recent successes of Aβ targeted trials have assuaged those doubts. In this review, we discussed the evolution of the amyloid cascade hypothesis over the last 30 years and summarized its application in Alzheimer's diagnosis and modification. In particular, we extensively discussed the pitfalls, promises and important unanswered questions regarding the current anti-Aβ therapy, as well as strategies for further study and development of more feasible Aβ-targeted approaches in the optimization of AD prevention and treatment.
Collapse
Affiliation(s)
- Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huaqiu Chen
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| |
Collapse
|
6
|
Pontrello CG, McWhirt JM, Glabe CG, Brewer GJ. Age-Related Oxidative Redox and Metabolic Changes Precede Intraneuronal Amyloid-β Accumulation and Plaque Deposition in a Transgenic Alzheimer's Disease Mouse Model. J Alzheimers Dis 2022; 90:1501-1521. [PMID: 36278355 PMCID: PMC9789488 DOI: 10.3233/jad-220824] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Many identified mechanisms could be upstream of the prominent amyloid-β (Aβ) plaques in Alzheimer's disease (AD). OBJECTIVE To profile the progression of pathology in AD. METHODS We monitored metabolic signaling, redox stress, intraneuronal amyloid-β (iAβ) accumulation, and extracellular plaque deposition in the brains of 3xTg-AD mice across the lifespan. RESULTS Intracellular accumulation of aggregated Aβ in the CA1 pyramidal cells at 9 months preceded extracellular plaques that first presented in the CA1 at 16 months of age. In biochemical assays, brain glutathione (GSH) declined with age in both 3xTg-AD and non-transgenic controls, but the decline was accelerated in 3xTg-AD brains from 2 to 4 months. The decline in GSH correlated exponentially with the rise in iAβ. Integrated metabolic signaling as the ratio of phospho-Akt (pAkt) to total Akt (tAkt) in the PI3kinase and mTOR pathway declined at 6, 9, and 12 months, before rising at 16 and 20 months. These pAkt/tAkt ratios correlated with both iAβ and GSH levels in a U-shaped relationship. Selective vulnerability of age-related AD-genotype-specific pAkt changes was greatest in the CA1 pyramidal cell layer. To demonstrate redox causation, iAβ accumulation was lowered in cultured middle-age adult 3xTg-AD neurons by treatment of the oxidized redox state in the neurons with exogenous cysteine. CONCLUSION The order of pathologic progression in the 3xTg-AD mouse was loss of GSH (oxidative redox shift) followed by a pAkt/tAkt metabolic shift in CA1, iAβ accumulation in CA1, and extracellular Aβ deposition. Upstream targets may prove strategically more effective for therapy before irreversible changes.
Collapse
Affiliation(s)
- Crystal G. Pontrello
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA,
Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Joshua M. McWhirt
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Charles G. Glabe
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA,
Center for Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA,
MIND Institute, University of California Irvine, Irvine, CA, USA
| | - Gregory J. Brewer
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA,
Center for Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA,
MIND Institute, University of California Irvine, Irvine, CA, USA,Correspondence to: Gregory J. Brewer, Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697, USA. Tel.: +1 217 502 4511; E-mail:
| |
Collapse
|
7
|
Influence of Nitric Oxide-Cyclic GMP and Oxidative STRESS on Amyloid-β Peptide Induced Decrease of Na,K-ATPase Activity in Rat Hippocampal Slices. J Membr Biol 2021; 254:463-473. [PMID: 34327545 DOI: 10.1007/s00232-021-00196-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022]
Abstract
Amyloid-β peptide (Aβ) has been shown to cause synaptic dysfunction and can render neurons vulnerable to excitotoxicity and oxidative stress. Na,K-ATPase plays an important role to maintain cell ionic equilibrium and it can be modulated by N-methyl-D-aspartate (NMDA)-nitric oxide (NO)-cyclic GMP pathway. Disruption of NO synthase (NOS) activity and reactive oxygen species (ROS) production could lead to changes in Na,K-ATPase isoforms' activities that may be detrimental to the cells. Our aim was to evaluate the signaling pathways of Aβ in relation to NMDA-NOS-cyclic GMP versus oxidative stress on α1-/α2,3-Na,K-ATPase activities in rat hippocampal slices. Aβ1-40 induced a concentration-dependent increase of NOS activity and increased cyclic guanosine monophosphate (cGMP), TBARS (thiobarbituric acid reactive substances), and 3-Nitrotyrosine (3-NT)-modified protein levels in rat hippocampal slices. The increase in NOS activity and cyclic GMP levels induced by Aβ1-40 was completely blocked by MK-801 (inhibitor of NMDA receptor) and L-NAME (inhibitor of NOS) pre-treatment but changes in TBARS levels were only partially blocked by both compounds. The Aβ treatment also decreased Na,K-ATPase activity which was reverted by N-nitro-L-arginine methyl ester hydrochloride (L-NAME) but not by MK-801 pre-treatment. The decrease in enzyme activity induced by Aβ was isoform-specific since only α1-Na,K-ATPase was affected. These findings suggest that the activation of NMDA-NOS signaling cascade linked to α2,3-Na,K-ATPase activity may mediate an adaptive, neuroprotective response to Aβ in rat hippocampus.
Collapse
|
8
|
Fu CH, Han XY, Tong L, Nie PY, Hu YD, Ji LL. miR-142 downregulation alleviates the impairment of spatial learning and memory, reduces the level of apoptosis, and upregulates the expression of pCaMKII and BAI3 in the hippocampus of APP/PS1 transgenic mice. Behav Brain Res 2021; 414:113485. [PMID: 34302879 DOI: 10.1016/j.bbr.2021.113485] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/20/2021] [Accepted: 07/17/2021] [Indexed: 01/20/2023]
Abstract
MicroRNA-142-5p (miR-142-5p) has been found to be dysregulated in several neurodegenerative disorders. However, little is known about the involvement of miR-142-5p in Alzheimer's disease (AD). Brain angiogenesis inhibitor 3 (BAI3), which belongs to the adhesion-G protein-coupled receptor subgroup, contributes to a variety of neuropsychiatric disorders. Despite its very high expression in neurons, the role of BAI3 in AD remains elusive, and its mechanism at the cellular and molecular levels needs to be further elucidated. The current study sought to investigate whether miR-142-5p influenced BAI3 expression and neuronal synaptotoxicity induced by Aβ, both in APP/PS1 transgenic mice and a cellular model of Alzheimer's disease. Altered expression of miR-142-5p was found in the hippocampus of AD mice. Inhibition of miR-142 could upregulate BAI3 expression, enhance neuronal viability and prevent neurons from undergoing apoptosis. In addition, the reduction of phosphorylation of Synapsin I and calcium/calmodulin-dependent protein kinase II (CaMKII), as well as the expression of PSD-95 in the hippocampus of APP/PS1 transgenic mice, were significantly restored by inhibiting miR-142. Meanwhile, the levels of Aβ1-42, β-APP, BACE-1 and PS-1 in cultured neurons were detected, and the effects of inhibiting miR-142 on spatial learning and memory were also observed. Interestingly, we found that BAI3, an important regulator of excitatory synapses, was a potential target gene of miR-142-5p. Collectively, our findings suggest that miR-142 inhibition can alleviate the impairment of spatial learning and memory, reduce the level of apoptosis, and upregulate the expression of pCaMKII and BAI3 in the hippocampus of APP/PS1 transgenic mice; thus, appropriate interference of miR-142 may provide a potential therapeutic approach to rescue cognitive dysfunction in AD patients.
Collapse
Affiliation(s)
- Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xue-Yan Han
- Department of Neurology, Seventh People's Hospital of Jinan City, Jinan, China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Peng-Yin Nie
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yue-Dong Hu
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
9
|
Subramanian J, Savage JC, Tremblay MÈ. Synaptic Loss in Alzheimer's Disease: Mechanistic Insights Provided by Two-Photon in vivo Imaging of Transgenic Mouse Models. Front Cell Neurosci 2020; 14:592607. [PMID: 33408613 PMCID: PMC7780885 DOI: 10.3389/fncel.2020.592607] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/25/2020] [Indexed: 01/05/2023] Open
Abstract
Synapse loss is the strongest correlate for cognitive decline in Alzheimer's disease. The mechanisms underlying synapse loss have been extensively investigated using mouse models expressing genes with human familial Alzheimer's disease mutations. In this review, we summarize how multiphoton in vivo imaging has improved our understanding of synapse loss mechanisms associated with excessive amyloid in the living animal brain. We also discuss evidence obtained from these imaging studies for the role of cell-intrinsic calcium dyshomeostasis and cell-extrinsic activities of microglia, which are the immune cells of the brain, in mediating synapse loss.
Collapse
Affiliation(s)
- Jaichandar Subramanian
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, United States
| | - Julie C Savage
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Butterfield DA. Brain lipid peroxidation and alzheimer disease: Synergy between the Butterfield and Mattson laboratories. Ageing Res Rev 2020; 64:101049. [PMID: 32205035 PMCID: PMC7502429 DOI: 10.1016/j.arr.2020.101049] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/08/2020] [Accepted: 03/12/2020] [Indexed: 02/05/2023]
Abstract
Brains from persons with Alzheimer disease (AD) and its earlier stage, amnestic mild cognitive impairment (MCI), exhibit high levels of oxidative damage, including that to phospholipids. One type of oxidative damage is lipid peroxidation, the most important index of which is protein-bound 4-hydroxy-2-trans-nonenal (HNE). This highly reactive alkenal changes the conformations and lowers the activities of brain proteins to which HNE is covalently bound. Evidence exists that suggests that lipid peroxidation is the first type of oxidative damage associated with amyloid β-peptide (Aβ), a 38-42 amino acid peptide that is highly neurotoxic and critical to the pathophysiology of AD. The Butterfield laboratory is one of, if not the, first research group to show that Aβ42 oligomers led to lipid peroxidation and to demonstrate this modification in brains of subjects with AD and MCI. The Mattson laboratory, particularly when Dr. Mattson was a faculty member at the University of Kentucky, also showed evidence for lipid peroxidation associated with Aβ peptides, mostly in in vitro systems. Consequently, there is synergy between our two laboratories. Since this special tribute issue of Aging Research Reviews is dedicated to the career of Dr. Mattson, a review of some aspects of this synergy of lipid peroxidation and its relevance to AD, as well as the role of lipid peroxidation in the progression of this dementing disorder seems germane. Accordingly, this review outlines some of the individual and/or complementary research on lipid peroxidation related to AD published from our two laboratories either separately or jointly.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University Of Kentucky, Lexington, KY, 40506, United States.
| |
Collapse
|
11
|
Heckmann BL, Tummers B, Green DR. Crashing the computer: apoptosis vs. necroptosis in neuroinflammation. Cell Death Differ 2018; 26:41-52. [PMID: 30341422 DOI: 10.1038/s41418-018-0195-3] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022] Open
Abstract
Programmed cell death (PCD) plays critical roles in development, homeostasis, and both control and progression of a plethora of diseases, including cancer and neurodegenerative pathologies. Besides classical apoptosis, several different forms of PCD have now been recognized, including necroptosis. The way a cell dies determines the reaction of the surrounding environment, and immune activation in response to cell death proceeds in a manner dependent on which death pathways are activated. Apoptosis and necroptosis are major mechanisms of cell death that typically result in opposing immune responses. Apoptotic death usually leads to immunologically silent responses whereas necroptotic death releases molecules that promote inflammation, a process referred to as necroinflammation. Diseases of the nervous system, in particular neurodegenerative diseases, are characterized by neuronal death and progressive neuroinflammation. The mechanisms of neuronal death are not well defined and significant cross-talk between pathways has been suggested. Moreover, it has been proposed that the dying of neurons is a catalyst for activating immune cells in the brain and sustaining inflammatory output. In the current review we discuss the effects of apoptotis and necroptosis on inflammatory immune activation, and evaluate the roles of each cell death pathway in a variety of pathologies with specific focus on neurodegeneration. A putative model is proposed for the regulation of neuronal death and neuroinflammation that features a role for both the apoptotic and necroptotic pathways in disease establishment and progression.
Collapse
Affiliation(s)
- Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Bart Tummers
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
12
|
The Lesion Analysis of Cholinergic Neurons in 5XFAD Mouse Model in the Three-Dimensional Level of Whole Brain. Mol Neurobiol 2017; 55:4115-4125. [PMID: 28597200 DOI: 10.1007/s12035-017-0621-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/16/2017] [Indexed: 10/19/2022]
Abstract
Cholinergic system is very important for many higher brain functions, including learning and memory. Cholinergic neurons, especially those in the basal forebrain, are specifically susceptible in some neurodegenerative diseases, such as in Alzheimer's disease (AD). Here, we studied the cholinergic system lesion effects of five familial AD mutations in 5XFAD mice, a transgenic mouse model of AD. Although the cholinergic system has been studied in this mouse model, the cholinergic deficits in AD mice have never been systematically mapped in a whole-brain three-dimensional (3D) reconstruction. Using the 3D reconstruction technology combined with immunohistochemistry (3D-IHC) and design-based stereology, we comprehensively compared the differences of the cholinergic neurons and fibers between the 5XFAD mice and C57BL/6 control mice at different age. Here, we found that the lesion of cholinergic fibers occurred earlier than the cholinergic neuron loss in 5XFAD mice. The cholinergic fiber lesions in the AD mice started sequentially in amygdala, cortex, hippocampus, and then basal forebrain. However, the basal forebrain was the first brain region observed with cholinergic neuron loss at the age of 9 months in 5XFAD mice, whereas such phenomenon first occurred at the age of 15 months in C57BL/6 control mice. Moreover, using 3D reconstruction to compare the lesion of cholinergic system of aged 5XFAD and C57BL/6 control mice, it is intuitive to notice the pathologic regions and severity of lesion. Therefore, the 3D-IHC provides detailed overview of the cholinergic neurons in the whole mouse brain, which will contribute to the study of the developing and pathologic mouse brain.
Collapse
|
13
|
Hatefi M, Dastjerdi MM, Ghiasi B, Rahmani A. Association of Serum Uric Acid Level with the Severity of Brain Injury and Patient's Outcome in Severe Traumatic Brain Injury. J Clin Diagn Res 2016; 10:OC20-OC24. [PMID: 28208906 DOI: 10.7860/jcdr/2016/21918.8993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/25/2016] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The prognostic value of serum Uric Acid (UA) levels in Traumatic Brain Injury (TBI) is unclear. AIM To investigate the relationship between serum UA levels and prognosis of patients with TBI when in hospital and at six months after discharge. MATERIALS AND METHODS All patients attended our emergency department during July 2014 and December 2015 and were consecutively entered into the study and among 890 evaluated candidates based on inclusion criteria we finally investigated the serum UA levels of 725 TBI patients. Computed Tomography (CT) images of the brain were obtained within the first 24 hours of hospitalization. Outcome was assessed using the Glasgow Outcome Scale (GOS) score at discharge and at six months after discharge. RESULTS Data of 725 patients (42.89% men; mean age: 54.69±12.37 years) were analyzed. Mean±Standard Deviation (SD) of Glasgow Coma Scale (GCS) scores was 4.65±1.76. Serum levels of UA, when in hospital and at six months after discharge, among those who died were lower than those who survived (in hospital: 0.126±0.026 vs. 0.243±0.942 mmol/l, p = 0.000; 6 months post-discharge: 0.130±0.044 vs. 0.286±0.069 mmol/l, p<0.001). The mean UA plasma was significantly different between deceased and alive patients according to GOS scores (p<0.001 and p=0.030, respectively). The UA levels showed a significant relationship with GCS scores and severity of brain injury assessed using the Marshall Classification Score (p=0.005). CONCLUSION Our results showed a strong relationship between UA levels and patients' outcomes either in hospital or at six months after discharge. Serum UA level could be considered as a valuable marker for evaluating the severity of brain injury and outcomes of TBI.
Collapse
Affiliation(s)
- Masoud Hatefi
- Assistant Professor, Department of Neurosurgery, Faculty of Medicine, Ilam University of Medical Science , Ilam, Iran
| | - Masoud Moghadas Dastjerdi
- Assistant Professor, Department of Emergency Medicine, School of Medicine, Isfahan University of Medical Science , Isfahan, Iran
| | - Bahareh Ghiasi
- Assistant Professor, Department of Nephrology, Faculty of Medicine, Ilam University of Medical Science , Ilam, Iran
| | - Asghar Rahmani
- Student's Researches Committee, Ilam University of Medical Sciences , Ilam, Iran
| |
Collapse
|
14
|
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder that has no known cure, nor is there a clear mechanistic understanding of the disease process itself. Although amyloid plaques, neurofibrillary tangles, and cognitive decline are late-stage markers of the disease, it is unclear how they are initially generated, and if they represent a cause, effect, or end phase in the pathology process. Recent studies in AD models have identified marked dysregulations in calcium signaling and related downstream pathways, which occur long before the diagnostic histopathological or cognitive changes. Under normal conditions, intracellular calcium signals are coupled to effectors that maintain a healthy physiological state. Consequently, sustained up-regulation of calcium may have pathophysiological consequences. Indeed, upon reviewing the current body of literature, increased calcium levels are functionally linked to the major features and risk factors of AD: ApoE4 expression, presenilin and APP mutations, beta amyloid plaques, hyperphosphorylation of tau, apoptosis, and synaptic dysfunction. In turn, the histopathological features of AD, once formed, are capable of further increasing calcium levels, leading to a rapid feed-forward acceleration once the disease process has taken hold. The views proposed here consider that AD pathogenesis reflects long-term calcium dysregulations that ultimately serve an enabling role in the disease process. Therefore, “Calcinists” do not necessarily reject βAptist or Tauist doctrine, but rather believe that their genesis is associated with earlier calcium signaling dysregulations. NEUROSCIENTIST 13(5):546—559, 2007.
Collapse
Affiliation(s)
- Grace E Stutzmann
- Rosalind Franklin University of Medicine and Science, The Chicago Medical School, North Chicago, IL 60064, USA.
| |
Collapse
|
15
|
Puig KL, Kulas JA, Franklin W, Rakoczy SG, Taglialatela G, Brown-Borg HM, Combs CK. The Ames dwarf mutation attenuates Alzheimer's disease phenotype of APP/PS1 mice. Neurobiol Aging 2016; 40:22-40. [PMID: 26973101 DOI: 10.1016/j.neurobiolaging.2015.12.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 12/28/2015] [Accepted: 12/28/2015] [Indexed: 12/21/2022]
Abstract
APP/PS1 double transgenic mice expressing human mutant amyloid precursor protein (APP) and presenilin-1 (PS1) demonstrate robust brain amyloid beta (Aβ) peptide containing plaque deposition, increased markers of oxidative stress, behavioral dysfunction, and proinflammatory gliosis. On the other hand, lack of growth hormone, prolactin, and thyroid-stimulating hormone due to a recessive mutation in the Prop 1 gene (Prop1df) in Ames dwarf mice results in a phenotype characterized by potentiated antioxidant mechanisms, improved learning and memory, and significantly increased longevity in homozygous mice. Based on this, we hypothesized that a similar hormone deficiency might attenuate disease changes in the brains of APP/PS1 mice. To test this idea, APP/PS1 mice were crossed to the Ames dwarf mouse line. APP/PS1, wild-type, df/+, df/df, df/+/APP/PS1, and df/df/APP/PS1 mice were compared at 6 months of age through behavioral testing and assessing amyloid burden, reactive gliosis, and brain cytokine levels. df/df mice demonstrated lower brain growth hormone and insulin-like growth factor 1 concentrations. This correlated with decreased astrogliosis and microgliosis in the df/df/APP/PS1 mice and, surprisingly, reduced Aβ plaque deposition and Aβ 1-40 and Aβ 1-42 concentrations. The df/df/APP/PS1 mice also demonstrated significantly elevated brain levels of multiple cytokines in spite of the attenuated gliosis. These data indicate that the df/df/APP/PS1 line is a unique resource in which to study aging and resistance to disease and suggest that the affected pituitary hormones may have a role in regulating disease progression.
Collapse
Affiliation(s)
- Kendra L Puig
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Joshua A Kulas
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Whitney Franklin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA; Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sharlene G Rakoczy
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Giulio Taglialatela
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Holly M Brown-Borg
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Colin K Combs
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA.
| |
Collapse
|
16
|
Abstract
The most accredited (and fashionable) hypothesis of the pathogenesis of Alzheimer Disease (AD) sees accumulation of β-amyloid protein in the brain (in both soluble and insoluble forms) as a leading mechanism of neurotoxicity. How β-amyloid triggers the neurodegenerative disorder is at present unclear, but growing evidence suggests that a deregulation of Ca(2+) homeostasis and deficient Ca(2+) signalling may represent a fundamental pathogenic factor. Given that symptoms of AD are most likely linked to synaptic dysfunction (at the early stages) followed by neuronal loss (at later and terminal phases of the disease), the effects of β-amyloid have been mainly studied in neurones. Yet, it must be acknowledged that neuroglial cells, including astrocytes, contribute to pathological progression of most (if not all) neurological diseases. Here, we review the literature pertaining to changes in Ca(2+) signalling in astrocytes exposed to exogenous β-amyloid or in astrocytes from transgenic Alzheimer disease animals models, characterized by endogenous β-amyloidosis. Accumulated experimental data indicate deregulation of Ca(2+) homeostasis and signalling in astrocytes in AD, which should be given full pathogenetic consideration. Further studies are warranted to comprehend the role of deficient astroglial Ca(2+) signalling in the disease progression.
Collapse
|
17
|
Lin H, Arispe NJ. Single-cell screening of cytosolic [Ca(2+)] reveals cell-selective action by the Alzheimer's Aβ peptide ion channel. Cell Stress Chaperones 2015; 20:333-42. [PMID: 25366568 PMCID: PMC4326387 DOI: 10.1007/s12192-014-0551-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/09/2014] [Accepted: 10/21/2014] [Indexed: 01/02/2023] Open
Abstract
Interaction of the Alzheimer's Aβ peptides with the plasma membrane of cells in culture results in chronic increases in cytosolic [Ca(2+)]. Such increases can cause a variety of secondary effects leading to impaired cell growth or cell degeneration. In this investigation, we made a comprehensive study of the changes in cytosolic [Ca(2+)] in single PC12 cells and human neurons stressed by continuous exposure to a medium containing Aβ42 for several days. The differential timing and magnitude of the Aβ42-induced increase in [Ca(2+)] reveal subpopulations of cells with differential sensitivity to Aβ42. These results suggest that the effect produced by Aβ on the level of cytosolic [Ca(2+)] depends on the type of cell being monitored. Moreover, the results obtained of using potent inhibitors of Aβ cation channels such as Zn(2+) and the small peptide NA7 add further proof to the suggestion that the long-term increases in cytosolic [Ca(2+)] in cells stressed by continuous exposure to Aβ is the result of Aβ ion channel activity.
Collapse
Affiliation(s)
- Hopi Lin
- />Department of Anatomy, Physiology and Genetics, and Institute for Molecular Medicine, Uniformed Services University School of Medicine, USUHS, Bethesda, MD 20814 USA
| | - Nelson J. Arispe
- />Department of Anatomy, Physiology and Genetics, and Institute for Molecular Medicine, Uniformed Services University School of Medicine, USUHS, Bethesda, MD 20814 USA
- />Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814 USA
| |
Collapse
|
18
|
Davis KE, Fox S, Gigg J. Increased hippocampal excitability in the 3xTgAD mouse model for Alzheimer's disease in vivo. PLoS One 2014; 9:e91203. [PMID: 24621690 PMCID: PMC3951322 DOI: 10.1371/journal.pone.0091203] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 02/11/2014] [Indexed: 01/24/2023] Open
Abstract
Mouse Alzheimer's disease (AD) models develop age- and region-specific pathology throughout the hippocampal formation. One recently established pathological correlate is an increase in hippocampal excitability in vivo. Hippocampal pathology also produces episodic memory decline in human AD and we have shown a similar episodic deficit in 3xTg AD model mice aged 3–6 months. Here, we tested whether hippocampal synaptic dysfunction accompanies this cognitive deficit by probing dorsal CA1 and DG synaptic responses in anaesthetized, 4–6 month-old 3xTgAD mice. As our previous reports highlighted a decline in episodic performance in aged control mice, we included aged cohorts for comparison. CA1 and DG responses to low-frequency perforant path stimulation were comparable between 3xTgAD and controls at both age ranges. As expected, DG recordings in controls showed paired-pulse depression; however, paired-pulse facilitation was observed in DG and CA1 of young and old 3xTgAD mice. During stimulus trains both short-latency (presumably monosynaptic: ‘direct’) and long-latency (presumably polysynaptic: ‘re-entrant’) responses were observed. Facilitation of direct responses was modest in 3xTgAD animals. However, re-entrant responses in DG and CA1 of young 3xTgAD mice developed earlier in the stimulus train and with larger amplitude when compared to controls. Old mice showed less DG paired-pulse depression and no evidence for re-entrance. In summary, DG and CA1 responses to low-frequency stimulation in all groups were comparable, suggesting no loss of synaptic connectivity in 3xTgAD mice. However, higher-frequency activation revealed complex change in synaptic excitability in DG and CA1 of 3xTgAD mice. In particular, short-term plasticity in DG and CA1 was facilitated in 3xTgAD mice, most evidently in younger animals. In addition, re-entrance was facilitated in young 3xTgAD mice. Overall, these data suggest that the episodic-like memory deficit in 3xTgAD mice could be due to the development of an abnormal hyper-excitable state in the hippocampal formation.
Collapse
Affiliation(s)
- Katherine E. Davis
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Sarah Fox
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - John Gigg
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
19
|
Song J, Park KA, Lee WT, Lee JE. Apoptosis signal regulating kinase 1 (ASK1): potential as a therapeutic target for Alzheimer's disease. Int J Mol Sci 2014; 15:2119-2129. [PMID: 24481061 PMCID: PMC3958840 DOI: 10.3390/ijms15022119] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by a decline in memory and cognitive function. Clinical manifestations of AD are closely associated with the formation of senile plaques and neurofibrillary tangles, neuronal loss and cognitive decline. Apoptosis signal regulating kinase 1 (ASK1) is a mediator of the MAPK pathway, which regulates various cellular responses such as apoptosis, cell survival, and differentiation. Accumulating evidence indicates that ASK1 plays a key role in the pathogenesis of neurodegenerative disorders such as Huntington's disease and AD. Of particular interest, ASK1 is associated with many signaling pathways, which include endoplasmic reticulum (ER) stress-mediated apoptosis, Aβ-induced neurotoxicity, tau protein phosphorylation, and insulin signal transduction. Here, we review experimental evidence that links ASK1 signaling and AD pathogenesis and propose that ASK1 might be a new point of therapeutic intervention to prevent or treat AD.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea.
| | - Kyung Ah Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea.
| | - Won Taek Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea.
| |
Collapse
|
20
|
p53 in neurodegenerative diseases and brain cancers. Pharmacol Ther 2013; 142:99-113. [PMID: 24287312 DOI: 10.1016/j.pharmthera.2013.11.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 11/07/2013] [Indexed: 12/21/2022]
Abstract
More than thirty years elapsed since a protein, not yet called p53 at the time, was detected to bind SV40 during viral infection. Thousands of papers later, p53 evolved as the main tumor suppressor involved in growth arrest and apoptosis. A lot has been done but the protein has not yet revealed all its secrets. Particularly important is the observation that in totally distinct pathologies where apoptosis is either exacerbated or impaired, p53 appears to play a central role. This is exemplified for Alzheimer's and Parkinson's diseases that represent the two main causes of age-related neurodegenerative affections, where cell death enhancement appears as one of the main etiological paradigms. Conversely, in cancers, about half of the cases are linked to mutations in p53 leading to the impairment of p53-dependent apoptosis. The involvement of p53 in these pathologies has driven a huge amount of studies aimed at designing chemical tools or biological approaches to rescue p53 defects or over-activity. Here, we describe the data linking p53 to neurodegenerative diseases and brain cancers, and we document the various strategies to interfere with p53 dysfunctions in these disorders.
Collapse
|
21
|
Trafficking in neurons: Searching for new targets for Alzheimer's disease future therapies. Eur J Pharmacol 2013; 719:84-106. [DOI: 10.1016/j.ejphar.2013.07.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/11/2013] [Indexed: 11/22/2022]
|
22
|
Veeraraghavalu K, Choi SH, Zhang X, Sisodia SS. Endogenous expression of FAD-linked PS1 impairs proliferation, neuronal differentiation and survival of adult hippocampal progenitors. Mol Neurodegener 2013; 8:41. [PMID: 24138759 PMCID: PMC3853710 DOI: 10.1186/1750-1326-8-41] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/16/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by progressive memory loss and impaired cognitive function. Early-onset familial forms of the disease (FAD) are caused by inheritance of mutant genes encoding presenilin 1 (PS1) variants. We have demonstrated that prion promoter (PrP)-driven expression of human FAD-linked PS1 variants in mice leads to impairments in environmental enrichment (EE)-induced adult hippocampal neural progenitor cell (AHNPC) proliferation and neuronal differentiation, and have provided evidence that accessory cells in the hippocampal niche expressing PS1 variants may modulate AHNPC phenotypes, in vivo. While of significant interest, these latter studies relied on transgenic mice that express human PS1 variant transgenes ubiquitously and at high levels, and the consequences of wild type or mutant PS1 expressed under physiologically relevant levels on EE-mediated AHNPC phenotypes has not yet been tested. RESULTS To assess the impact of mutant PS1 on EE-induced AHNPC phenotypes when expressed under physiological levels, we exposed adult mice that constitutively express the PSEN1 M146V mutation driven by the endogenous PSEN1 promoter (PS1 M146V "knock-in" (KI) mice) to standard or EE-housed conditions. We show that in comparison to wild type PS1 mice, AHNPCs in mice carrying homozygous (PS1M146V/M146V) or heterozygous (PS1M146V/+) M146V mutant alleles fail to exhibit EE-induced proliferation and commitment towards neurogenic lineages. More importantly, we report that the survival of newborn progenitors are diminished in PS1 M146V KI mice exposed to EE-conditions compared to respective EE wild type controls. CONCLUSIONS Our findings reveal that expression at physiological levels achieved by a single PS1 M146V allele is sufficient to impair EE-induced AHNPC proliferation, survival and neuronal differentiation, in vivo. These results and our finding that microglia expressing a single PS1 M146V allele impairs the proliferation of wild type AHNPCs in vitro argue that expression of mutant PS1 in the AHNPC niche impairs AHNPCs phenotypes in a dominant, non-cell autonomous manner.
Collapse
Affiliation(s)
| | | | | | - Sangram S Sisodia
- Department of Neurobiology, The University of Chicago, 947 E 58th Street, AB 308, Chicago, Illinois 60637, USA.
| |
Collapse
|
23
|
Greenough MA, Camakaris J, Bush AI. Metal dyshomeostasis and oxidative stress in Alzheimer’s disease. Neurochem Int 2013; 62:540-55. [DOI: 10.1016/j.neuint.2012.08.014] [Citation(s) in RCA: 340] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/13/2012] [Accepted: 08/30/2012] [Indexed: 01/21/2023]
|
24
|
Inhibition of β-amyloid peptide-induced neurotoxicity by kaempferol 3-O-(6″-acetyl)-β-glucopyranoside from butterbur (Petasites japonicus) leaves in B103 cells. Food Sci Biotechnol 2012. [DOI: 10.1007/s10068-012-0109-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
25
|
Yoo SE, Chen L, Na R, Liu Y, Rios C, Remmen HV, Richardson A, Ran Q. Gpx4 ablation in adult mice results in a lethal phenotype accompanied by neuronal loss in brain. Free Radic Biol Med 2012; 52:1820-7. [PMID: 22401858 PMCID: PMC3341497 DOI: 10.1016/j.freeradbiomed.2012.02.043] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 02/16/2012] [Accepted: 02/25/2012] [Indexed: 01/12/2023]
Abstract
Glutathione peroxidase 4 (Gpx4) is an antioxidant defense enzyme important in reducing hydroperoxides in membrane lipids and lipoproteins. Gpx4 is essential for survival of embryos and neonatal mice; however, whether Gpx4 is required for adult animals remains unclear. In this study, we generated a floxed Gpx4 mouse (Gpx4(f/f)), in which exons 2-4 of Gpx4 gene are flanked by loxP sites. We then cross-bred the Gpx4(f/f) mice with a tamoxifen (tam)-inducible Cre transgenic mouse (R26CreER mice) to obtain mice in which the Gpx4 gene could be ablated by tam administration (Gpx4(f/f)/Cre mice). After treatment with tam, adult Gpx4(f/f)/Cre mice (6-9 months of age) showed a significant reduction of Gpx4 levels (a 75-85% decrease) in tissues such as brain, liver, lung, and kidney. Tam-treated Gpx4(f/f)/Cre mice lost body weight and died within 2 weeks, indicating that Gpx4 is essential for survival of adult animals. Tam-treated Gpx4(f/f)/Cre mice exhibited increased mitochondrial damage, as evidenced by the elevated 4-hydroxylnonenal (4-HNE) level, decreased activities of electron transport chain complexes I and IV, and reduced ATP production in liver. Tam treatment also significantly elevated apoptosis in Gpx4(f/f)/Cre mice. Moreover, tam-treated Gpx4(f/f)/Cre mice showed neuronal loss in the hippocampus region and had increased astrogliosis. These data indicate that Gpx4 is essential for mitochondria integrity and survival of neurons in adult animals.
Collapse
Affiliation(s)
- Si-Eun Yoo
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas 78229
- Department of Physiology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Liuji Chen
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas 78229
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Ren Na
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Yuhong Liu
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Carmen Rios
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas 78229
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Holly Van Remmen
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas 78229
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas 78229
- South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Arlan Richardson
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas 78229
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas 78229
- South Texas Veterans Health Care System, San Antonio, Texas 78229
| | - Qitao Ran
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, Texas 78229
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, Texas 78229
- South Texas Veterans Health Care System, San Antonio, Texas 78229
- Corresponding author contact: Qitao Ran, Ph.D. 15355 Lambda Drive San Antonio, TX 78245-3207 Phone: 210-562-6129 FAX: 210-562-6130
| |
Collapse
|
26
|
Chau DM, Crump CJ, Villa JC, Scheinberg DA, Li YM. Familial Alzheimer disease presenilin-1 mutations alter the active site conformation of γ-secretase. J Biol Chem 2012; 287:17288-17296. [PMID: 22461631 DOI: 10.1074/jbc.m111.300483] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Presenilin-1 (PS1) is the catalytic subunit of γ-secretase, and mutations in this protein cause familial Alzheimer Disease (FAD). However, little is known about how these mutations affect the active site of γ-secretase. Here, we show that PS1 mutations alter the S2 subsite within the active site of γ-secretase using a multiple photoaffinity probe approach called "photophore walking." Moreover, we developed a unique in vitro assay with a biotinylated recombinant Notch1 substrate and demonstrated that PS1 FAD mutations directly and significantly reduced γ-secretase activity for Notch1 cleavage. Substitution of the Notch Cys-1752 residue, which interacts with the S2 subsite, with Val, Met, or Ile has little effect on wild-type PS1 but leads to more efficient substrates for mutant PS1s. This study indicates that alteration of this S2 subsite plays an important role in determining the activity and specificity of γ-secretase for APP and Notch1 processing, which provides structural basis and insights on how certain PS1 FAD mutations lead to AD pathogenesis.
Collapse
Affiliation(s)
- De-Ming Chau
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 and the Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences of Cornell University, New York, NY 10021
| | - Christina J Crump
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 and the Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences of Cornell University, New York, NY 10021
| | - Jennifer C Villa
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 and the Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences of Cornell University, New York, NY 10021
| | - David A Scheinberg
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 and the Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences of Cornell University, New York, NY 10021
| | - Yue-Ming Li
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065 and the Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences of Cornell University, New York, NY 10021.
| |
Collapse
|
27
|
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
|
28
|
Salvemini D, Little JW, Doyle T, Neumann WL. Roles of reactive oxygen and nitrogen species in pain. Free Radic Biol Med 2011; 51:951-66. [PMID: 21277369 PMCID: PMC3134634 DOI: 10.1016/j.freeradbiomed.2011.01.026] [Citation(s) in RCA: 235] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 01/19/2011] [Accepted: 01/20/2011] [Indexed: 02/07/2023]
Abstract
Peroxynitrite (PN; ONOO⁻) and its reactive oxygen precursor superoxide (SO; O₂•⁻) are critically important in the development of pain of several etiologies including pain associated with chronic use of opiates such as morphine (also known as opiate-induced hyperalgesia and antinociceptive tolerance). This is now an emerging field in which considerable progress has been made in terms of understanding the relative contributions of SO, PN, and nitroxidative stress in pain signaling at the molecular and biochemical levels. Aggressive research in this area is poised to provide the pharmacological basis for development of novel nonnarcotic analgesics that are based upon the unique ability to selectively eliminate SO and/or PN. As we have a better understanding of the roles of SO and PN in pathophysiological settings, targeting PN may be a better therapeutic strategy than targeting SO. This is because, unlike PN, which has no currently known beneficial role, SO may play a significant role in learning and memory. Thus, the best approach may be to spare SO while directly targeting its downstream product, PN. Over the past 15 years, our team has spearheaded research concerning the roles of SO and PN in pain and these results are currently leading to the development of solid therapeutic strategies in this important area.
Collapse
Affiliation(s)
- Daniela Salvemini
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | | | | | | |
Collapse
|
29
|
Gong EJ, Park HR, Kim ME, Piao S, Lee E, Jo DG, Chung HY, Ha NC, Mattson MP, Lee J. Morin attenuates tau hyperphosphorylation by inhibiting GSK3β. Neurobiol Dis 2011; 44:223-30. [PMID: 21782947 DOI: 10.1016/j.nbd.2011.07.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 05/31/2011] [Accepted: 07/06/2011] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the major form of age-related dementia and is characterized by progressive cognitive impairment, the accumulation of extracellular amyloid β-peptide (Aβ), and intracellular hyperphosphorylated tau aggregates in affected brain regions. Tau hyperphosphorylation and accumulation in neurofibrillary tangles is strongly correlated with cognitive deficits, and is apparently a critical event in the dementia process because mutations in tau can cause a tangle-only form of dementia called frontotemporal lobe dementia. Among kinases that phosphorylate tau, glycogen synthase kinase 3β (GSK3β) is strongly implicated in AD pathogenesis. In the present study, we established an ELISA to screen for agents that inhibit GSK3β activity and found that the flavonoid morin effectively inhibited GSK3β activity and blocked GSK3β-induced tau phosphorylation in vitro. In addition, morin attenuated Aβ-induced tau phosphorylation and protected human neuroblastoma cells against Aβ cytotoxicity. Furthermore, treatment of 3xTg-AD mice with morin resulted in reductions in tau hyperphosphorylation and paired helical filament-like immunoreactivity in hippocampal neurons. Morin is a novel inhibitor of GSK3β that can reduce tau pathology in vivo and may have potential as a therapeutic agent in tauopathies.
Collapse
Affiliation(s)
- Eun Ji Gong
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug Development, Longevity Life Science and Technology Institutes, Pusan National University, Geumjeong-gu, Busan, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Desai MK, Guercio BJ, Narrow WC, Bowers WJ. An Alzheimer's disease-relevant presenilin-1 mutation augments amyloid-beta-induced oligodendrocyte dysfunction. Glia 2011; 59:627-40. [PMID: 21294162 PMCID: PMC3045399 DOI: 10.1002/glia.21131] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 11/30/2010] [Indexed: 11/10/2022]
Abstract
White matter pathology has been documented in the brains of familial Alzheimer's disease (FAD)-afflicted individuals during presymptomatic and preclinical stages of AD. How these defects in myelination integrity arise and what roles they may play in AD pathophysiology have yet to be fully elucidated. We previously demonstrated that triple-transgenic AD (3xTg-AD) mice, which harbor the human amyloid precursor Swedish mutation, presenilin-1 M146V (PS1(M146V) ) knock-in mutation, and tau(P301L) mutation, exhibit myelin abnormalities analogous to FAD patients and that Aβ(1-42) contributes to these white matter deficits. Herein, we demonstrate that the PS1(M146V) mutation predisposes mouse oligodendrocyte precursor (mOP) cells to Aβ(1-42) -induced alterations in cell differentiation in vitro. Furthermore, PS1(M146V) expression compromised mOP cell function and MBP protein distribution, a process that is further aggravated with exposure to Aβ(1-42) . We found that the myelination defect and MBP subcellular mislocalization triggered by PS1(M146V) and Aβ(1-42) can be effectively prevented by treatment with the GSK-3β inhibitor, TWS119, thereby implicating GSK-3β kinase activity in this pathogenic cascade. Overall, this work provides further mechanistic insights into PS1(M146V) and Aβ(1-42) -driven oligodendrocyte dysfunction andmyelin damage during early presymptomatic stages of AD, and provides a new target in oligodendrocytes for developing therapies designed to avert AD-related white matter pathology.
Collapse
Affiliation(s)
- Maya K. Desai
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Center for Neural Development and Disease, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Brendan J. Guercio
- Department of Center for Neural Development and Disease, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Wade C. Narrow
- Department of Center for Neural Development and Disease, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - William J. Bowers
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Center for Neural Development and Disease, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
31
|
Müller M, Cheung KH, Foskett JK. Enhanced ROS generation mediated by Alzheimer's disease presenilin regulation of InsP3R Ca2+ signaling. Antioxid Redox Signal 2011; 14:1225-35. [PMID: 20701429 PMCID: PMC3048838 DOI: 10.1089/ars.2010.3421] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Familial Alzheimer's disease (FAD) is caused by mutations in amyloid precursor protein and presenilins (PS1, PS2). Many FAD-linked PS mutations affect intracellular calcium (Ca(2+)) homeostasis by proximal mechanisms independent of amyloid production by dramatically enhancing gating of the inositol trisphosphate receptor (InsP(3)R) intracellular Ca(2+) release channel by a gain-of-function effect that mirrors genetics of FAD and is independent of secretase activity. Electrophysiological recordings of InsP(3)R in FAD patient B cells, cortical neurons of asymptomatic PS1-AD mice, and other cells revealed they have higher occupancy in a high open probability burst mode, resulting in enhanced Ca(2+) signaling. Exaggerated Ca(2+) signaling through this mechanism results in enhanced generation of reactive oxygen species, believed to be an important component in AD pathogenesis. Exaggerated Ca(2+) signaling through InsP(3)R-PS interaction is a disease specific and robust proximal mechanism in AD that may contribute to the pathology of AD by enhanced generation of reactive oxygen species.
Collapse
Affiliation(s)
- Marioly Müller
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
32
|
Greenough MA, Volitakis I, Li QX, Laughton K, Evin G, Ho M, Dalziel AH, Camakaris J, Bush AI. Presenilins promote the cellular uptake of copper and zinc and maintain copper chaperone of SOD1-dependent copper/zinc superoxide dismutase activity. J Biol Chem 2011; 286:9776-86. [PMID: 21239495 PMCID: PMC3058959 DOI: 10.1074/jbc.m110.163964] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 01/11/2011] [Indexed: 12/22/2022] Open
Abstract
Dyshomeostasis of extracellular zinc and copper has been implicated in β-amyloid aggregation, the major pathology associated with Alzheimer disease. Presenilin mediates the proteolytic cleavage of the β-amyloid precursor protein to release β-amyloid, and mutations in presenilin can cause familial Alzheimer disease. We tested whether presenilin expression affects copper and zinc transport. Studying murine embryonic fibroblasts (MEFs) from presenilin knock-out mice or RNA interference of presenilin expression in HEK293T cells, we observed a marked decrease in saturable uptake of radiolabeled copper and zinc. Measurement of basal metal levels in 6-month-old presenilin 1 heterozygous knock-out (PS1(+/-)) mice revealed significant deficiencies of copper and zinc in several tissues, including brain. Copper/zinc superoxide dismutase (SOD1) activity was significantly decreased in both presenilin knock-out MEFs and brain tissue of presenilin 1 heterozygous knock-out mice. In the MEFs and PS1(+/-) brains, copper chaperone of SOD1 (CCS) levels were decreased. Zinc-dependent alkaline phosphatase activity was not decreased in the PS null MEFs. These data indicate that presenilins are important for cellular copper and zinc turnover, influencing SOD1 activity, and having the potential to indirectly impact β-amyloid aggregation through metal ion clearance.
Collapse
Affiliation(s)
- Mark A. Greenough
- From the Mental Health Research Institute, Parkville, Victoria 3052, Australia, and
- the Departments of Genetics and
| | - Irene Volitakis
- From the Mental Health Research Institute, Parkville, Victoria 3052, Australia, and
- Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Qiao-Xin Li
- Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Katrina Laughton
- Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Genevieve Evin
- Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Michael Ho
- Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Andrew H. Dalziel
- From the Mental Health Research Institute, Parkville, Victoria 3052, Australia, and
- the Departments of Genetics and
| | | | - Ashley I. Bush
- From the Mental Health Research Institute, Parkville, Victoria 3052, Australia, and
- Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
33
|
Lattarulo C, Thyssen D, Kuchibholta KV, Hyman BT, Bacskaiq BJ. Microscopic imaging of intracellular calcium in live cells using lifetime-based ratiometric measurements of Oregon Green BAPTA-1. Methods Mol Biol 2011; 793:377-89. [PMID: 21913114 DOI: 10.1007/978-1-61779-328-8_25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Calcium is a ubiquitous intracellular messenger that has important functions in normal neuronal function. The pathology of Alzheimer's disease has been shown to alter calcium homeostasis in neurons and astrocytes. Several calcium dye indicators are available to measure intracellular calcium within cells, including Oregon Green BAPTA-1 (OGB-1). Using fluorescence lifetime imaging microscopy, we adapted this single wavelength calcium dye into a ratiometric dye to allow quantitative imaging of cellular calcium. We used this approach for in vitro calibrations, single-cell microscopy, high-throughput imaging in automated plate readers, and in single cells in the intact living brain. While OGB is a commonly used fluorescent dye for imaging calcium qualitatively, there are distinct advantages to using a ratiometric approach, which allows quantitative determinations of calcium that are independent of dye concentration. Taking advantage of the distinct lifetime contrast of the calcium-free and calcium-bound forms of OGB, we used time-domain lifetime measurements to generate calibration curves for OGB lifetime ratios as a function of calcium concentration. In summary, we demonstrate approaches using commercially available tools to measure calcium concentrations in live cells at multiple scales using lifetime contrast. These approaches are broadly applicable to other fluorescent readouts that exhibit lifetime contrast and serve as powerful alternatives to spectral or intensity readouts in multiplexing experiments.
Collapse
Affiliation(s)
- Carli Lattarulo
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | | | | | | | | |
Collapse
|
34
|
Desai MK, Mastrangelo MA, Ryan DA, Sudol KL, Narrow WC, Bowers WJ. Early oligodendrocyte/myelin pathology in Alzheimer's disease mice constitutes a novel therapeutic target. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1422-35. [PMID: 20696774 DOI: 10.2353/ajpath.2010.100087] [Citation(s) in RCA: 173] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The detection of myelin disruptions in Alzheimer's disease (AD)-affected brain raises the possibility that oligodendrocytes undergo pathophysiological assault over the protracted course of this neurodegenerative disease. Oligodendrocyte compromise arising from direct toxic effects imparted by pathological amyloid-beta peptides and/or through signals derived from degenerating neurons could play an important role in the disease process. We previously demonstrated that 3xTg-AD mice, which harbor the human amyloid precursor protein Swedish mutant transgene, presenilin knock-in mutation, and tau P301L mutant transgene, exhibit significant alterations in overall myelination patterns and oligodendrocyte status at time points preceding the appearance of amyloid and tau pathology. Herein, we demonstrate that Abeta(1-42) leads to increased caspase-3 expression and apoptotic cell death of both nondifferentiated and differentiated mouse oligodendrocyte precursor (mOP) cells in vitro. Through use of a recombinant adeno-associated virus serotype-2 (rAAV2) vector expressing an Abeta(1-42)-specific intracellular antibody (intrabody), oligodendrocyte and myelin marker expression, as well as myelin integrity, were restored in the vector-infused brain regions of 3xTg-AD mice. Overall, this work provides further insights into the impact of Abeta(1-42)-mediated toxicity on the temporal and spatial progression of subtle myelin disruption during the early presymptomatic stages of AD and may help to validate new therapeutic options designed to avert these early impairments.
Collapse
Affiliation(s)
- Maya K Desai
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | | | | | |
Collapse
|
35
|
Elder GA, Gama Sosa MA, De Gasperi R. Transgenic mouse models of Alzheimer's disease. ACTA ACUST UNITED AC 2010; 77:69-81. [PMID: 20101721 DOI: 10.1002/msj.20159] [Citation(s) in RCA: 225] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease is the most common cause of senile dementia in the United States and Europe. At present, there is no effective treatment. Given the disease's prevalence and poor prognosis, the development of animal models has been a high research priority. Transgenic modeling has been pursued on the basis of the amyloid hypothesis and has taken advantage of mutations in the amyloid precursor protein and the presenilins that cause familial forms of Alzheimer's disease. Modeling has been most aggressively pursued in mice, for which the techniques of genetic modification are well developed. Transgenic mouse models now exist that mimic a range of Alzheimer's disease-related pathologies. Although none of the models fully replicates the human disease, the models have contributed significant insights into the pathophysiology of beta-amyloid toxicity, particularly with respect to the effects of different beta-amyloid species and the possible pathogenic role of beta-amyloid oligomers. They have also been widely used in the preclinical testing of potential therapeutic modalities and have played a pivotal role in the development of immunotherapies for Alzheimer's disease that are currently in clinical trials. These models will, without a doubt, continue to play central roles in preclinical testing and be used as tools for developing insights into the biological basis of Alzheimer's disease.
Collapse
Affiliation(s)
- Gregory A Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA.
| | | | | |
Collapse
|
36
|
Supnet C, Noonan C, Richard K, Bradley J, Mayne M. Up-regulation of the type 3 ryanodine receptor is neuroprotective in the TgCRND8 mouse model of Alzheimer's disease. J Neurochem 2009; 112:356-65. [PMID: 19903243 DOI: 10.1111/j.1471-4159.2009.06487.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cellular pathology of Alzheimer's disease is progressive and protracted leading eventually to considerable neuronal death. The underlying mechanisms of the pathology are complex but changes in the control of intracellular Ca2+ are believed to contribute to the demise of neurons. In this study, we investigated the functional consequences of an increase in the expression of the type 3 isoform of the ryanodine receptor (RyR3). We found that although cortical neurons from TgCRND8 mice secreted significantly more amyloid beta protein and showed significantly increased RyR3 expression, they were no more sensitive to cell stress than non-transgenic neurons. Furthermore, despite increased intracellular Ca2+ release in response to ryanodine, we found that basal Ca2+, K+-evoked Ca2+ responses, and capacitative Ca2+ entry were no different in TgCRND8 neurons compared with non-transgenic neurons. Therefore, as RyR3 up-regulation did not affect neuronal health or global Ca2+ homeostasis, we investigated the effect of reducing RyR3 expression using small interfering RNA. Surprisingly, a reduction of RyR3 expression in TgCRND8, but not in non-transgenic, neurons increased neuronal death. These data reveal a new role for RyR3 and indicate a novel potential therapeutic target to delay or prevent the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Charlene Supnet
- Department of Biomedical Sciences, University of Prince Edward Island, Charlottetown, PE, Canada
| | | | | | | | | |
Collapse
|
37
|
Röskam S, Neff F, Schwarting R, Bacher M, Dodel R. APP transgenic mice: the effect of active and passive immunotherapy in cognitive tasks. Neurosci Biobehav Rev 2009; 34:487-99. [PMID: 19857518 DOI: 10.1016/j.neubiorev.2009.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 10/11/2009] [Accepted: 10/18/2009] [Indexed: 12/24/2022]
Abstract
Various immunotherapy strategies for APP transgenic mice have emerged in recent years. Specifically, active immunization with beta-amyloid (A beta) or passive immunization with anti-A beta-antibodies in APP transgenic mice has appeared most promising. Recent studies have shown that treatment of APP transgenic mice either with A beta(40/42) or A beta-specific antibodies can have beneficial effects in cognitive tasks. Active as well as passive immunization have been shown to affect spatial, non-spatial, emotional and object-related learning and memory. Such effects can be observed when treatments are applied prophylactically (before apparent A beta pathology) or therapeutically (after the development of A beta pathology) in APP transgenic mice. This review focuses on such cognitive outcomes of different active and passive immunization strategies in APP transgenic mice.
Collapse
Affiliation(s)
- Stephan Röskam
- Department of Neurology, Research Group for Neurological Therapeutics, Biomedical Research Centre, Philipps-University Marburg, Hans-Meerwein-Strasse, 35043 Marburg, Germany
| | | | | | | | | |
Collapse
|
38
|
The in vitro effects of superoxide, some commercially available antioxidants and red palm oil on sperm motility. Asian J Androl 2009; 11:695-702. [PMID: 19802000 DOI: 10.1038/aja.2009.55] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In this study, two commercially available superoxide scavengers, tetrakis (1-methyl-4-pyridyl) porphyrin (Mn[III]TMPyP) and superoxide dismutase (SOD), as well as red palm oil (RPO), a natural vegetable oil, had been used to investigate their possible in vitro effects against the toxic effects of superoxide (O(2).) on human sperm motility. Semen samples were obtained from 12 normozoospermic healthy volunteer donors aged between 19 and 23 years. The O(2). donor 2,3-dimetoxyl-1,4-naphthoquinone (DMNQ) (2.5 micromol L(-1)-100 micromol L(-1)) was added to normozoospermic post-swim-up sperm in the presence or absence of Mn(III)TMPyP (50 micromol L(-1)), SOD (50 IU) or RPO (0.1% or 0.5%). Computer-assisted semen analysis was used to analyze various motility parameters. The parameters of interest were percentage of motile cells, progressive motility, rapid cells and static cells. Concentrations of higher than 25 micromol L(-1) DMNQ were detrimental to sperm motility. Mn(III)TMPyP was able to attenuate the effect of O(2). on the motility parameters. In vitro addition of SOD and RPO showed harmful effects on sperm motility.
Collapse
|
39
|
Yu JT, Chang RCC, Tan L. Calcium dysregulation in Alzheimer's disease: from mechanisms to therapeutic opportunities. Prog Neurobiol 2009; 89:240-55. [PMID: 19664678 DOI: 10.1016/j.pneurobio.2009.07.009] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2009] [Revised: 07/28/2009] [Accepted: 07/31/2009] [Indexed: 11/28/2022]
Abstract
Calcium is involved in many facets of neuronal physiology, including activity, growth and differentiation, synaptic plasticity, and learning and memory, as well as pathophysiology, including necrosis, apoptosis, and degeneration. Though disturbances in calcium homeostasis in cells from Alzheimer's disease (AD) patients have been observed for many years, much more attention was focused on amyloid-beta (Abeta) and tau as key causative factors for the disease. Nevertheless, increasing lines of evidence have recently reported that calcium dysregulation plays a central role in AD pathogenesis. Systemic calcium changes accompany almost the whole brain pathology process that is observed in AD, including synaptic dysfunction, mitochondrial dysfunction, presenilins mutation, Abeta production and Tau phosphorylation. Given the early and ubiquitous involvement of calcium dysregulation in AD pathogenesis, it logically presents a variety of potential therapeutic targets for AD prevention and treatment, such as calcium channels in the plasma membrane, calcium channels in the endoplasmic reticulum membrane, Abeta-formed calcium channels, calcium-related proteins. The review aims to provide an overview of the current understanding of the molecular mechanisms involved in calcium dysregulation in AD, and an insight on how to exploit calcium regulation as therapeutic opportunities in AD.
Collapse
Affiliation(s)
- Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 5 Donghai Middle Road, Qingdao, Shandong Province 266071, China
| | | | | |
Collapse
|
40
|
Hass MR, Sato C, Kopan R, Zhao G. Presenilin: RIP and beyond. Semin Cell Dev Biol 2009; 20:201-10. [PMID: 19073272 PMCID: PMC2693421 DOI: 10.1016/j.semcdb.2008.11.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/19/2008] [Accepted: 11/19/2008] [Indexed: 12/22/2022]
Abstract
Over the years the presenilins (PSENs), a family of multi-transmembrane domain proteins, have been ascribed a number of diverse potential functions. Recent in vivo evidence has supported the existence of PSEN functions beyond its well-established role in regulated intramembrane proteolysis. In this review, we will briefly discuss the ability of PSEN to modulate cellular signaling pathways through gamma-secretase cleavage of transmembrane proteins. Additionally, we will critically examine the proposed roles of PSEN in the regulation of beta-catenin function, protein trafficking, calcium regulation, and apoptosis.
Collapse
Affiliation(s)
- Matthew R. Hass
- Department of Developmental Biology, Washington University School of Medicine, Box 8103, 4566 Scott Avenue, Saint Louis, Missouri, MO 63110, United States of America
| | - Chihiro Sato
- Department of Developmental Biology, Washington University School of Medicine, Box 8103, 4566 Scott Avenue, Saint Louis, Missouri, MO 63110, United States of America
| | - Raphael Kopan
- Department of Developmental Biology, Washington University School of Medicine, Box 8103, 4566 Scott Avenue, Saint Louis, Missouri, MO 63110, United States of America
| | - Guojun Zhao
- Department of Developmental Biology, Washington University School of Medicine, Box 8103, 4566 Scott Avenue, Saint Louis, Missouri, MO 63110, United States of America
| |
Collapse
|
41
|
Castellani RJ, Zhu X, Lee HG, Smith MA, Perry G. Molecular pathogenesis of Alzheimer's disease: reductionist versus expansionist approaches. Int J Mol Sci 2009; 10:1386-1406. [PMID: 19399255 PMCID: PMC2672036 DOI: 10.3390/ijms10031386] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 03/20/2009] [Accepted: 03/23/2009] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is characterized clinically by dementia and pathologically by two hallmark lesions, senile plaques and neurofibrillary tangles. About a quarter century ago these hallmark lesions were purified and their protein constituents identified, precipitating an avalanche of molecular studies as well as substantial optimism about successful therapeutic intervention. In 2009, we now have copious knowledge on the biochemical cascades that produce these proteins, the different modifications and forms in which these proteins exist, and the ability to selectively target these proteins for therapeutic intervention on an experimental basis. At the same time, there has been no discernible alteration in the natural course of AD in humans. While it may be that the complexity of AD will exceed our capacity to make significant treatment progress for decades or more, a paradigm shift from the reductionism that defines amyloid-beta and tau hypotheses, to one that more accurately reflects the meaning of neuropathological changes, may be warranted. We and others have demonstrated that AD pathology is a manifestation of cellular adaptation, specifically as a defense against oxidative injury. As such, AD pathology is therefore a host response rather than a manifestation of cytotoxic protein injury, and is unlikely to be a fruitful target for therapeutic intervention. An "expansionist" view of the disease, we believe, with oxidative stress as a pleiotropic and upstream process, more aptly describes the relationship between various and numerous molecular alterations and clinical disease.
Collapse
Affiliation(s)
- Rudy J. Castellani
- Division of Neuropathology, University of Maryland, Baltimore, Maryland, USA
- Author to whom correspondence should be addressed; E-Mail:
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hyoung-Gon Lee
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark A. Smith
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - George Perry
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- College of Sciences, University of Texas, San Antonio, Texas, USA
| |
Collapse
|
42
|
Lu Y, Ansar S, Michaelis ML, Blagg BSJ. Neuroprotective activity and evaluation of Hsp90 inhibitors in an immortalized neuronal cell line. Bioorg Med Chem 2009; 17:1709-15. [PMID: 19138859 PMCID: PMC2729088 DOI: 10.1016/j.bmc.2008.12.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 12/17/2008] [Accepted: 12/19/2008] [Indexed: 01/31/2023]
Abstract
Alzheimer's disease (AD) neuropathology is characterized by loss of synapses and neurons, neuritic plaques consisting of beta-amyloid (Abeta) peptides, and neurofibrillary tangles consisting of intracellular aggregates of hyperphosphorylated tau protein in susceptible brain regions. Abeta oligomers trigger a cascade of pathogenic events including tau hyperphosphorylation and aggregation, inflammatory reactions, and excitotoxicity that contribute to the progression of AD. The molecular chaperone Hsp90 facilitates the folding of newly synthesized and denatured proteins and is believed to play a role in neurodegenerative disorders in which the defining pathology results in misfolded proteins and the accumulation of protein aggregates. Some agents that inhibit Hsp90 protect neurons against Abeta toxicity and tau aggregation, and assays for rapidly screening potential Hsp90 inhibitors are of interest. We used the release of the soluble cytosolic enzyme lactate dehydrogenase (LDH) as an indicator of the loss of cell membrane integrity and cytotoxicity resulting from exposure to Abeta peptides to evaluate the neuroprotective properties of novel novobiocin analogues and established Hsp90 inhibitors. Compounds were assessed for potency in protecting proliferating and differentiated SH-SY5Y neuronal cells against Abeta-induced cell death; the potential toxicity of each agent alone was also determined. The data indicated that several of the compounds decreased Abeta toxicity even at low nanomolar concentrations and, unexpectedly, were more potent in protecting the undifferentiated cells against Abeta. The novobiocin analogues alone were not toxic even up to 10 microM concentrations whereas GDA and the parent compound, novobiocin, were toxic at 1 and 10 microM, respectively. The results suggest that novobiocin analogues may provide novel leads for the development of neuroprotective drugs.
Collapse
Affiliation(s)
- Yuanming Lu
- The Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Drive, Malott 4070, Lawrence, KS 66045-7563 USA
| | - Sabah Ansar
- Department of Pharmacology and Toxicology, The University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS 66045 USA
| | - Mary L. Michaelis
- Department of Pharmacology and Toxicology, The University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS 66045 USA
| | - Brian S. J. Blagg
- The Department of Medicinal Chemistry, The University of Kansas, 1251 Wescoe Hall Drive, Malott 4070, Lawrence, KS 66045-7563 USA
| |
Collapse
|
43
|
Jazi R, Lalonde R, Qian S, Strazielle C. Regional brain evaluation of acetylcholinesterase activity in PS1/A246E transgenic mice. Neurosci Res 2009; 63:106-14. [DOI: 10.1016/j.neures.2008.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Revised: 10/31/2008] [Accepted: 11/06/2008] [Indexed: 10/21/2022]
|
44
|
Morgan GA, Guo Q, Chan SL, Gary DS, Osborne BA, Mattson MP. Defects of immune regulation in the presenilin-1 mutant knockin mouse. Neuromolecular Med 2008; 9:35-45. [PMID: 17114823 DOI: 10.1385/nmm:9:1:35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Revised: 04/17/2006] [Accepted: 04/19/2006] [Indexed: 11/11/2022]
Abstract
Mutations in the presenilin-1 (PS1) gene are causally linked to early-onset Alzheimer's disease (AD). Studies of neurons suggest that PS1 mutations result in a gain-of-function, which perturbs calcium regulation and increases cell vulnerability to apoptosis. Alterations in immune cell function have also been demonstrated in AD, and a role for PS1 in immune regulation has been suggested recently. We now report that splenocytes from PS1-mutant (M146V) knockin mice exhibit increased caspase activity, abnormal calcium regulation and aberrant mitochondrial function. Isolated splenic T cells from PS1-mutant mice respond poorly to proliferative signals and have downregulated cluster designation 3 and interleukin (IL)- 2-receptor expression necessary for a normal T-cell immune response. Thus, adverse effects of a mutation that causes AD on immune function that involves perturbed calcium regulation and cytokine signaling in lymphocytes, and associated sensitivity of lymphocytes to apoptosis are demonstrated. These findings suggest that abnormalities in immune function might play major roles in the pathogenesis of AD.
Collapse
Affiliation(s)
- Grant A Morgan
- Department of Veterinary and Animal Sciences, University of Massachusetts, 311 Paige Lab, Amherst, MA 01003, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Alzheimer's disease (AD) is characterized by the deposition of beta-amyloid peptides (Abeta) and a progressive loss of neurons leading to dementia. Because hippocampal neurogenesis is linked to functions such as learning, memory and mood, there has been great interest in examining the effects of AD on hippocampal neurogenesis. This article reviews the pertinent studies and tries to unite them in one possible disease model. Early in the disease, oligomeric Abeta may transiently promote the generation of immature neurons from neural stem cells (NSCs). However, reduced concentrations of multiple neurotrophic factors and higher levels of fibroblast growth factor-2 seem to induce a developmental arrest of newly generated neurons. Furthermore, fibrillary Abeta and down-regulation of oligodendrocyte-lineage transcription factor-2 (OLIG2) may cause the death of these nonfunctional neurons. Therefore, altering the brain microenvironment for fostering apt maturation of graft-derived neurons may be critical for improving the efficacy of NSC transplantation therapy for AD.
Collapse
Affiliation(s)
- B. Waldau
- Department of Surgery (Neurosurgery), Duke University Medical Center, Durham, NC 27710 USA
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC 27705 USA
| | - A. K. Shetty
- Department of Surgery (Neurosurgery), Duke University Medical Center, Durham, NC 27710 USA
- Medical Research and Surgery Services, Veterans Affairs Medical Center, Durham, NC 27705 USA
| |
Collapse
|
46
|
Kuchibhotla KV, Goldman ST, Lattarulo CR, Wu HY, Hyman BT, Bacskai BJ. Abeta plaques lead to aberrant regulation of calcium homeostasis in vivo resulting in structural and functional disruption of neuronal networks. Neuron 2008; 59:214-25. [PMID: 18667150 PMCID: PMC2578820 DOI: 10.1016/j.neuron.2008.06.008] [Citation(s) in RCA: 504] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2007] [Revised: 04/14/2008] [Accepted: 06/03/2008] [Indexed: 01/19/2023]
Abstract
Alzheimer's disease is characterized by the deposition of senile plaques and progressive dementia. The molecular mechanisms that couple plaque deposition to neural system failure, however, are unknown. Using transgenic mouse models of AD together with multiphoton imaging, we measured neuronal calcium in individual neurites and spines in vivo using the genetically encoded calcium indicator Yellow Cameleon 3.6. Quantitative imaging revealed elevated [Ca(2+)]i (calcium overload) in approximately 20% of neurites in APP mice with cortical plaques, compared to less than 5% in wild-type mice, PS1 mutant mice, or young APP mice (animals without cortical plaques). Calcium overload depended on the existence and proximity to plaques. The downstream consequences included the loss of spinodendritic calcium compartmentalization (critical for synaptic integration) and a distortion of neuritic morphologies mediated, in part, by the phosphatase calcineurin. Together, these data demonstrate that senile plaques impair neuritic calcium homeostasis in vivo and result in the structural and functional disruption of neuronal networks.
Collapse
Affiliation(s)
- Kishore V Kuchibhotla
- Massachusetts General Hospital, Department of Neurology/Alzheimer's Disease Research Laboratory, 114 16th Street, Charlestown, MA 02129, USA
| | | | | | | | | | | |
Collapse
|
47
|
Troy CM, Rabacchi SA, Xu Z, Maroney AC, Connors TJ, Shelanski ML, Greene LA. β-Amyloid-induced neuronal apoptosis requires c-Jun N-terminal kinase activation. J Neurochem 2008. [DOI: 10.1046/j.1471-4159.2001.00218.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Reddy PH. Mitochondrial medicine for aging and neurodegenerative diseases. Neuromolecular Med 2008; 10:291-315. [PMID: 18566920 PMCID: PMC3235551 DOI: 10.1007/s12017-008-8044-z] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 05/22/2008] [Indexed: 12/22/2022]
Abstract
Mitochondria are key cytoplasmic organelles, responsible for generating cellular energy, regulating intracellular calcium levels, altering the reduction-oxidation potential of cells, and regulating cell death. Increasing evidence suggests that mitochondria play a central role in aging and in neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Freidriech ataxia. Further, several lines of evidence suggest that mitochondrial dysfunction is an early event in most late-onset neurodegenerative diseases. Biochemical and animal model studies of inherited neurodegenerative diseases have revealed that mutant proteins of these diseases are associated with mitochondria. Mutant proteins are reported to block the transport of nuclear-encoded mitochondrial proteins to mitochondria, interact with mitochondrial proteins and disrupt the electron transport chain, induce free radicals, cause mitochondrial dysfunction, and, ultimately, damage neurons. This article discusses critical issues of mitochondria causing dysfunction in aging and neurodegenerative diseases, and discusses the potential of developing mitochondrial medicine, particularly mitochondrially targeted antioxidants, to treat aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- P Hemachandra Reddy
- Neurogenetics Laboratory, Neurological Sciences Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
| |
Collapse
|
49
|
Wang G, Silva J, Dasgupta S, Bieberich E. Long-chain ceramide is elevated in presenilin 1 (PS1M146V) mouse brain and induces apoptosis in PS1 astrocytes. Glia 2008; 56:449-56. [PMID: 18205190 DOI: 10.1002/glia.20626] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pro-apoptotic sphingolipid ceramide plays an emergent role in the etiology of Alzheimer's disease (AD), although its function for neurodegeneration is not known. We determined the concentration and composition of ceramide in hippocampal tissue from newborn presenilin 1 (PS1) knock-in (PS1M146V) mice, a mouse model for early-onset familial AD. We found that PS1 tissue contains 3.1 (+/-0.5)-fold more total ceramide than wild-type tissue. In particular, the proportion of C20 and C24 ceramide is increased by 4.0- or 8.5-fold, respectively. The ceramide elevation in PS1 brain is consistent with a 3.7 (+/-0.5)-fold increase of the protein level of the neurotrophin receptor p75NTR, which has been suggested to stimulate the hydrolysis of sphingomyelin to generate ceramide. The predominance of C20 and C24 ceramide is concurrent with the elevated gene expression of lass 2 and lass 4, two isoforms of ceramide synthase that generate dihydroceramide with long-chain fatty acid. Our study indicates that primary cultured astrocytes but not neurons from PS1 mice undergo apoptosis when incubated with C20 ceramide. In contrast, wild-type astrocytes remain unaffected. The sensitivity of PS1 astrocytes is most likely due to the 9.5 (+/-0.4)-fold elevated expression of PAR-4 (prostate apoptosis response-4), a protein that inhibits atypical PKC zeta/lambda in the presence of ceramide. Our results suggest that astroglial death due to ceramide/PAR-4-induced apoptosis may critically contribute to the etiology of AD.
Collapse
Affiliation(s)
- Guanghu Wang
- Program in Developmental Neurobiology, Institute of Molecular Medicine and Genetics, School of Medicine, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | |
Collapse
|
50
|
Damjanac M, Rioux Bilan A, Paccalin M, Pontcharraud R, Fauconneau B, Hugon J, Page G. Dissociation of Akt/PKB and ribosomal S6 kinase signaling markers in a transgenic mouse model of Alzheimer’s disease. Neurobiol Dis 2008; 29:354-67. [DOI: 10.1016/j.nbd.2007.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Revised: 09/05/2007] [Accepted: 09/23/2007] [Indexed: 01/15/2023] Open
|