1
|
Wang M, Feng N, Qin J, Wang S, Chen J, Qian S, Liu Y, Luo F. Abdominal surgery under ketamine anesthesia during second trimester impairs hippocampal learning and memory of offspring by regulating dendrite spine remodeling in rats. Neurotoxicology 2024; 101:82-92. [PMID: 38346645 DOI: 10.1016/j.neuro.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
Recent evidence showed that general anesthesia produces long-term neurotoxicity and cognitive dysfunction. However, it remains unclear whether maternal non-obstetric surgery under ketamine anesthesia during second trimester causes cognitive impairment in offspring. The present study assigned pregnant rats into three groups: 1) normal control group receiving no anesthesia and no surgery, 2) ketamine group receiving ketamine anesthesia for 2 h on the 14th day of gestation but no surgery, and 3) surgery group receiving abdominal surgery under ketamine anesthesia on the 14th day of gestation. On postnatal day 1, the offspring rats in Ketamine group and surgery group were assigned to receive intra-peritoneal injection of Senegenin (15 mg/kg), once per day for consecutive 14 days. The offspring's spatial perception, anxiety-like behavior, and learning and memory were evaluated. Then the offspring's hippocampal tissues were collected. The offspring of the surgery group were impaired in the spatial perception in the cliff avoidance test and the spatial learning and memory in the Morris water maze test. Accordingly, the activity of histone deacetylases increased, the protein levels of NEDD9, BDNF, p-TrkB, Syn and PSD-95 decreased, and the density of dendritic spines reduced in the hippocampus of the offspring of the surgery group, and such effects were not seen in the offspring of the ketamine group, neither in the offspring of control group. Senegenin alleviated the learning and memory impairment, and increased the protein levels of NEDD9, BDNF, p-TrkB, Syn and PSD-95 and the density of dendritic spines in the offspring of the surgery group. ketamine anesthesia plus surgery during second trimester impairs hippocampus-dependent learning and memory, and the deficits could be rescued by treatment with Senegenin.
Collapse
Affiliation(s)
- Mengdie Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Namin Feng
- Department of Anesthesiology, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jia Qin
- Rehabilitation Medical Center and Department of Anesthesiology, Zhejiang Provincial People's Hospital and Hangzhou Medical College Affiliated People's Hospital, Hangzhou, Zhejiang 310000, China
| | - Shengqiang Wang
- Department of Anesthesiology, Yichun People's Hospital, Yichun 336000, China
| | - Jiabao Chen
- Rehabilitation Medical Center and Department of Anesthesiology, Zhejiang Provincial People's Hospital and Hangzhou Medical College Affiliated People's Hospital, Hangzhou, Zhejiang 310000, China
| | - Shaojie Qian
- Rehabilitation Medical Center and Department of Anesthesiology, Zhejiang Provincial People's Hospital and Hangzhou Medical College Affiliated People's Hospital, Hangzhou, Zhejiang 310000, China
| | - Yulin Liu
- Department of Immunology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Foquan Luo
- Rehabilitation Medical Center and Department of Anesthesiology, Zhejiang Provincial People's Hospital and Hangzhou Medical College Affiliated People's Hospital, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
2
|
Arabi A, Karimi SA, Salehi I, Haddadi R, Komaki A. Effects of sesamin on Aβ 1-42-induced oxidative stress and LTP impairment in a rat model of Alzheimer's disease. Metab Brain Dis 2023; 38:1503-1511. [PMID: 36847969 DOI: 10.1007/s11011-023-01191-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/16/2023] [Indexed: 03/01/2023]
Abstract
The present study examined the protective effect of sesamin (Ses) on β-amyloid (Aβ)-induced long-term potentiation (LTP) impairment at the PP-DG synapses in male rats. Wistar rats were randomly assigned to seven groups: control, sham, Aβ; ICV Aβ1-42 microinjection, Ses, Aβ + Ses; first, ICV Aβ injections and then receiving Ses, Ses + Aβ: four weeks of pretreatment with Ses and then Aβ injection, and Ses + Aβ + Ses: pre (four weeks) and post (four weeks) treatment with Ses. Ses-treated groups received 30 mg/kg of Ses once a day by oral gavage for four weeks. After the treatment period, the animals were positioned in a stereotaxic device for surgery and field potential recording. The population spike (PS) amplitude and slope of excitatory postsynaptic potentials (EPSP) were evaluated in the DG region. Serum oxidative stress biomarkers (total oxidant status (TOS) and total antioxidant capacity (TAC)) were measured. Aβ impaired LTP induction at the PP-DG synapses evidenced by a decrease in EPSP slope and PS amplitude of LTP. In Aβ rats, Ses increased EPSP slope and PS amplitude of LTP in the DG granular cells. Also, an increase in TOS and a reduction in TAC caused by Aβ were significantly corrected by Ses. Ses could prevent Aβ-induced LTP impairment at the PP-DG synapses in male rats, which can be due to its preventive effects on oxidative stress.
Collapse
Affiliation(s)
- Amir Arabi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran
| | - Seyed Asaad Karimi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran
| | - Iraj Salehi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran.
| |
Collapse
|
3
|
Mehan S, Bhalla S, Siddiqui EM, Sharma N, Shandilya A, Khan A. Potential Roles of Glucagon-Like Peptide-1 and Its Analogues in Dementia Targeting Impaired Insulin Secretion and Neurodegeneration. Degener Neurol Neuromuscul Dis 2022; 12:31-59. [PMID: 35300067 PMCID: PMC8921673 DOI: 10.2147/dnnd.s247153] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Dementia is a chronic, irreversible condition marked by memory loss, cognitive decline, and mental instability. It is clinically related to various progressive neurological diseases, including Parkinson’s disease, Alzheimer’s disease, and Huntington’s. The primary cause of neurological disorders is insulin desensitization, demyelination, oxidative stress, and neuroinflammation accompanied by various aberrant proteins such as amyloid-β deposits, Lewy bodies accumulation, tau formation leading to neurofibrillary tangles. Impaired insulin signaling is directly associated with amyloid-β and α-synuclein deposition, as well as specific signaling cascades involved in neurodegenerative diseases. Insulin dysfunction may initiate various intracellular signaling cascades, including phosphoinositide 3-kinase (PI3K), c-Jun N-terminal kinases (JNK), and mitogen-activated protein kinase (MAPK). Neuronal death, inflammation, neuronal excitation, mitochondrial malfunction, and protein deposition are all influenced by insulin. Recent research has focused on GLP-1 receptor agonists as a potential therapeutic target. They increase glucose-dependent insulin secretion and are beneficial in neurodegenerative diseases by reducing oxidative stress and cytokine production. They reduce the deposition of abnormal proteins by crossing the blood-brain barrier. The purpose of this article is to discuss the role of insulin dysfunction in the pathogenesis of neurological diseases, specifically dementia. Additionally, we reviewed the therapeutic target (GLP-1) and its receptor activators as a possible treatment of dementia.
Collapse
Affiliation(s)
- Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
- Correspondence: Sidharth Mehan, Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India, Tel +91 8059889909; +91 9461322911, Email ;
| | - Sonalika Bhalla
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ehraz Mehmood Siddiqui
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Nidhi Sharma
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ambika Shandilya
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Andleeb Khan
- Department of Pharmacology & Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
4
|
Zhang Y, Jiang W, Hao X, Tan J, Wang W, Yu M, Zhang G, Zhang Y. Preparation of the Enzymatic Hydrolysates from Chlorella vulgaris Protein and Assessment of Their Antioxidant Potential Using Caenorhabditis elegans. Mol Biotechnol 2021; 63:1040-1048. [PMID: 34213689 DOI: 10.1007/s12033-021-00361-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/18/2021] [Indexed: 11/30/2022]
Abstract
This study aimed to assess the antioxidant potential of Chlorella vulgaris protein-derived enzymatic hydrolysate using Caenorhabditis elegans. Protein extraction was performed using an alkali solution after complete C. vulgaris swelling and hydrolysis using four commercial proteases (alcalase, neutrase, protamex, and flavourzyme). The results showed that the flavourzyme hydrolysates exhibited the strongest antioxidant activity both in vitro and in vivo. Under the optimum conditions of the enzymatic hydrolysis, the half-maximal effective concentration of the hydrolysates for superoxide and hydroxyl radicals was 0.323 mg/mL and 0.139 mg/mL, respectively. The hydrolysates could significantly extend the lifespan, improve the resistance to methyl viologen-induced oxidative stress, reduce the levels of reactive oxygen species, and enhance the activity of catalase and superoxide dismutase in C. elegans.
Collapse
Affiliation(s)
- Yakun Zhang
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Wen Jiang
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Xiao Hao
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Jingjing Tan
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Weimin Wang
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Mengen Yu
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Gaofan Zhang
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Yongjun Zhang
- Key Laboratory of Marine Food Quality and Hazard Controlling Technology College of Life Sciences, China Jiliang University, Hangzhou, China.
| |
Collapse
|
5
|
Effects of vanillic acid on Aβ 1-40-induced oxidative stress and learning and memory deficit in male rats. Brain Res Bull 2021; 170:264-273. [PMID: 33652070 DOI: 10.1016/j.brainresbull.2021.02.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/14/2021] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, in which the accumulation of β-amyloid (Aβ) peptide in the extracellular space causes a progressive reduction in cognitive performance. Aβ stimulates active oxygen species generation leading to oxidative stress and neural cell death. Vanillic Acid (VA) is the oxidant form of vanillin widely found in vanilla beans. VA has many properties, such as suppressing apoptosis and eliminating the harmful effects of oxidative stress in animal models. The VA effects on impaired learning and memory in Aβ rats were assessed. Forty adults male Wistar rats were assigned to the following five groups in random: the control, sham (received saline (vehicle) via intracerebroventricular (ICV) injection), Aβ (received Aβ1-40 via ICV injection), VA (50 mg/kg by oral gavage once a day through four weeks), and Aβ + VA (50 mg/kg) groups. Open field test, novel object recognition (NOR) test, Morris water maze (MWM) test, and passive avoidance learning (PAL) task were performed, and finally, we determined the malondialdehyde (MDA), total antioxidant capacity (TAC) and total oxidant status (TOS) levels. Aβ decreased the cognitive memory in NOR, spatial memory in MWM, and passive avoidance memory in PAL tests. In contrast, VA improved learning and memory in the treated group. Aβ significantly increased MDA and TOS and decreased TAC levels, whereas VA treatment significantly reversed TAC, TOS and MDA levels. In conclusion, VA decreased the Aβ effects on learning and memory by suppressing oxidative stress and can be regarded as a neuroprotective substance in AD.
Collapse
|
6
|
Kodavanti PRS, Valdez M, Richards JE, Agina-Obu DI, Phillips PM, Jarema KA, Kodavanti UP. Ozone-induced changes in oxidative stress parameters in brain regions of adult, middle-age, and senescent Brown Norway rats. Toxicol Appl Pharmacol 2021; 410:115351. [PMID: 33249117 PMCID: PMC7775355 DOI: 10.1016/j.taap.2020.115351] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022]
Abstract
A critical part of community based human health risk assessment following chemical exposure is identifying sources of susceptibility. Life stage is one such susceptibility. A prototypic air pollutant, ozone (O3) induces dysfunction of the pulmonary, cardiac, and nervous systems. Long-term exposure may cause oxidative stress (OS). The current study explored age-related and subchronic O3-induced changes in OS in brain regions of rats. To build a comprehensive assessment of OS-related effects of O3, a tripartite approach was implemented focusing on 1) the production of reactive oxygen species (ROS) [NADPH Quinone oxidoreductase 1, NADH Ubiquinone reductase] 2) antioxidant homeostasis [total antioxidant substances, superoxide dismutase, γ-glutamylcysteine synthetase] and 3) an assessment of oxidative damage [total aconitase and protein carbonyls]. Additionally, a neurobehavioral evaluation of motor activity was compared to these OS measures. Male Brown Norway rats (4, 12, and 24 months of age) were exposed to air or O3 (0.25 or 1 ppm) via inhalation for 6 h/day, 2 days per week for 13 weeks. A significant decrease in horizontal motor activity was noted only in 4-month old rats. Results on OS measures in frontal cortex (FC), cerebellum (CB), striatum (STR), and hippocampus (HIP) indicated life stage-related increases in ROS production, small decreases in antioxidant homeostatic mechanisms, a decrease in aconitase activity, and an increase in protein carbonyls. The effects of O3 exposure were brain area-specific, with the STR being more sensitive. Regarding life stage, the effects of O3 were greater in 4-month-old rats, which correlated with horizontal motor activity. These results indicate that OS may be increased in specific brain regions after subchronic O3 exposure, but the interactions between age and exposure along with their consequences on the brain require further investigation.
Collapse
Affiliation(s)
- Prasada Rao S Kodavanti
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA, ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Matthew Valdez
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA, ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Judy E Richards
- Cardiopulmonary and Immunotoxicology Branch, Public Health and Integrated Toxicology Division, CPHEA, ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Datonye I Agina-Obu
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA, ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Pamela M Phillips
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA, ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Kimberly A Jarema
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA, ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Urmila P Kodavanti
- Cardiopulmonary and Immunotoxicology Branch, Public Health and Integrated Toxicology Division, CPHEA, ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
7
|
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: 45] [Impact Index Per Article: 11.3] [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
|
8
|
Foret MK, Lincoln R, Do Carmo S, Cuello AC, Cosa G. Connecting the "Dots": From Free Radical Lipid Autoxidation to Cell Pathology and Disease. Chem Rev 2020; 120:12757-12787. [PMID: 33211489 DOI: 10.1021/acs.chemrev.0c00761] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Our understanding of lipid peroxidation in biology and medicine is rapidly evolving, as it is increasingly implicated in various diseases but also recognized as a key part of normal cell function, signaling, and death (ferroptosis). Not surprisingly, the root and consequences of lipid peroxidation have garnered increasing attention from multiple disciplines in recent years. Here we "connect the dots" between the fundamental chemistry underpinning the cascade reactions of lipid peroxidation (enzymatic or free radical), the reactive nature of the products formed (lipid-derived electrophiles), and the biological targets and mechanisms associated with these products that culminate in cellular responses. We additionally bring light to the use of highly sensitive, fluorescence-based methodologies. Stemming from the foundational concepts in chemistry and biology, these methodologies enable visualizing and quantifying each reaction in the cascade in a cellular and ultimately tissue context, toward deciphering the connections between the chemistry and physiology of lipid peroxidation. The review offers a platform in which the chemistry and biomedical research communities can access a comprehensive summary of fundamental concepts regarding lipid peroxidation, experimental tools for the study of such processes, as well as the recent discoveries by leading investigators with an emphasis on significant open questions.
Collapse
Affiliation(s)
- Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 1Y6
| | - Richard Lincoln
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A 0B8
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 1Y6
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada H3G 1Y6.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada H3A 0C7.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Gonzalo Cosa
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, Canada H3A 0B8
| |
Collapse
|
9
|
van Dam L, Dansen TB. Cross-talk between redox signalling and protein aggregation. Biochem Soc Trans 2020; 48:379-397. [PMID: 32311028 PMCID: PMC7200635 DOI: 10.1042/bst20190054] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
It is well established that both an increase in reactive oxygen species (ROS: i.e. O2•-, H2O2 and OH•), as well as protein aggregation, accompany ageing and proteinopathies such as Parkinson's and Alzheimer's disease. However, it is far from clear whether there is a causal relation between the two. This review describes how protein aggregation can be affected both by redox signalling (downstream of H2O2), as well as by ROS-induced damage, and aims to give an overview of the current knowledge of how redox signalling affects protein aggregation and vice versa. Redox signalling has been shown to play roles in almost every step of protein aggregation and amyloid formation, from aggregation initiation to the rapid oligomerization of large amyloids, which tend to be less toxic than oligomeric prefibrillar aggregates. We explore the hypothesis that age-associated elevated ROS production could be part of a redox signalling-dependent-stress response in an attempt to curb protein aggregation and minimize toxicity.
Collapse
Affiliation(s)
- Loes van Dam
- Center for Molecular Medicine, Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands
| | - Tobias B. Dansen
- Center for Molecular Medicine, Molecular Cancer Research, University Medical Center Utrecht, Universiteitsweg 100, 3584CG Utrecht, The Netherlands
| |
Collapse
|
10
|
Ojo JO, Crynen G, Algamal M, Vallabhaneni P, Leary P, Mouzon B, Reed JM, Mullan M, Crawford F. Unbiased Proteomic Approach Identifies Pathobiological Profiles in the Brains of Preclinical Models of Repetitive Mild Traumatic Brain Injury, Tauopathy, and Amyloidosis. ASN Neuro 2020; 12:1759091420914768. [PMID: 32241177 PMCID: PMC7132820 DOI: 10.1177/1759091420914768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
No concerted investigation has been conducted to explore overlapping and distinct
pathobiological mechanisms between repetitive mild traumatic brain injury
(r-mTBI) and tau/amyloid proteinopathies considering the long history of
association between TBI and Alzheimer’s disease. We address this problem by
using unbiased proteomic approaches to generate detailed time-dependent brain
molecular profiles of response to repetitive mTBI in C57BL/6 mice and in mouse
models of amyloidosis (with amyloid precursor protein KM670/671NL (Swedish) and
Presenilin 1 M146L mutations [PSAPP]) and tauopathy (hTau). Brain tissues from
animals were collected at different timepoints after injuries (24 hr–12 months
post-injury) and at different ages for tau or amyloid transgenic models (3, 9,
and 15 months old), encompassing the pre-, peri-, and post-“onset” of cognitive
and pathological phenotypes. We identified 30 hippocampal and 47 cortical
proteins that were significantly modulated over time in the r-mTBI compared with
sham mice. These proteins identified TBI-dependent modulation of
phosphatidylinositol-3-kinase/AKT signaling, protein kinase A signaling, and
PPARα/RXRα activation in the hippocampus and protein kinase A signaling,
gonadotropin-releasing hormone signaling, and B cell receptor signaling in the
cortex. Previously published neuropathological studies of our mTBI model showed
a lack of amyloid and tau pathology. In PSAPP mice, we identified 19 proteins
significantly changing in the cortex and only 7 proteins in hTau mice versus
wild-type littermates. When we explored the overlap between our r-mTBI model and
the PSAPP/hTau models, a fairly small coincidental change was observed involving
only eight significantly regulated proteins. This work suggests a very distinct
TBI neurodegeneration and also that other factors are needed to drive
pathologies such as amyloidosis and tauopathy postinjury.
Collapse
Affiliation(s)
- Joseph O Ojo
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,James A. Haley Veterans' Hospital, Tampa, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Gogce Crynen
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Moustafa Algamal
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Prashanti Vallabhaneni
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States
| | - Paige Leary
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States
| | - Benoit Mouzon
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,James A. Haley Veterans' Hospital, Tampa, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Jon M Reed
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, United States
| | - Michael Mullan
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Fiona Crawford
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,James A. Haley Veterans' Hospital, Tampa, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
11
|
Butterfield DA, Boyd-Kimball D. Redox proteomics and amyloid β-peptide: insights into Alzheimer disease. J Neurochem 2019; 151:459-487. [PMID: 30216447 PMCID: PMC6417976 DOI: 10.1111/jnc.14589] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/15/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disorder associated with aging and characterized pathologically by the presence of senile plaques, neurofibrillary tangles, and neurite and synapse loss. Amyloid beta-peptide (1-42) [Aβ(1-42)], a major component of senile plaques, is neurotoxic and induces oxidative stress in vitro and in vivo. Redox proteomics has been used to identify proteins oxidatively modified by Aβ(1-42) in vitro and in vivo. In this review, we discuss these proteins in the context of those identified to be oxidatively modified in animal models of AD, and human studies including familial AD, pre-clinical AD (PCAD), mild cognitive impairment (MCI), early AD, late AD, Down syndrome (DS), and DS with AD (DS/AD). These redox proteomics studies indicate that Aβ(1-42)-mediated oxidative stress occurs early in AD pathogenesis and results in altered antioxidant and cellular detoxification defenses, decreased energy yielding metabolism and mitochondrial dysfunction, excitotoxicity, loss of synaptic plasticity and cell structure, neuroinflammation, impaired protein folding and degradation, and altered signal transduction. Improved access to biomarker imaging and the identification of lifestyle interventions or treatments to reduce Aβ production could be beneficial in preventing or delaying the progression of AD. This article is part of the special issue "Proteomics".
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH 44601
| |
Collapse
|
12
|
Tian Y, Wang S, Jiao F, Kong Q, Liu C, Wu Y. Telomere Length: A Potential Biomarker for the Risk and Prognosis of Stroke. Front Neurol 2019; 10:624. [PMID: 31263449 PMCID: PMC6585102 DOI: 10.3389/fneur.2019.00624] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 05/28/2019] [Indexed: 12/22/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide. Age is associated with increased risk of stroke, while telomere length shortening plays a pivotal role in the process of aging. Moreover, telomere length shortening is associated with many risk factors of stroke in addition to age. Accumulated evidence shows that short leukocyte telomere length is not only associated with stroke occurrence but also associated with post-stroke recovery in the elderly population. In this review, we aimed to summarize the association between leukocyte telomere length and stroke, and discuss that telomere length might serve as a potential biomarker to predict the risk and prognosis of stroke.
Collapse
Affiliation(s)
- Yanjun Tian
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
| | - Shuai Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Fengjuan Jiao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Qingsheng Kong
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
| | - Chuanxin Liu
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
| | - Yili Wu
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China.,Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| |
Collapse
|
13
|
Yu Q, Feng N, Hu Y, Luo F, Zhao W, Zhao W, Liu Z, Li M, Xu L, Wu L, Liu Y. Suberoylanilide hydroxamic acid (SAHA) alleviates the learning and memory impairment in rat offspring caused by maternal sevoflurane exposure during late gestation. J Toxicol Sci 2019; 44:177-189. [PMID: 30842370 DOI: 10.2131/jts.44.177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Recent studies have shown that sevoflurane can cause long-term neurotoxicity and learning and memory impairment in developing and progressively neurodegenerative brains. Sevoflurane is a widely used volatile anesthetic in clinical practice. Late gestation is a rapidly developing period in the fetal brain, but whether sevoflurane anesthesia during late gestation affects learning and memory of offspring is not fully elucidated. Histone deacetylase 2 (HDAC2) plays an important regulatory role in learning and memory. This study examined the effect of maternal sevoflurane exposure on learning and memory in offspring and the underlying role of HDAC2. The Morris water maze (MWM) test was used to evaluate learning and memory function. Q-PCR and immunofluorescence staining were used to measure the expression levels of genes related to learning and memory. The results showed that sevoflurane anesthesia during late gestation impaired learning and memory in offspring rats (e.g., showing increase of the escape latency and decrease of the platform-crossing times and target quadrant traveling time in behavior tests) and upregulated the expression of HDAC2, while downregulating the expression of the cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) and the N-methyl-D-aspartate receptor 2 subunit B (NR2B) mRNA and protein in the hippocampus of offspring in a time-dependent manner. HDAC2 inhibitor suberoylanilide hydroxamic acid (SAHA) treatment alleviated all of these changes in offspring rats. Therefore, the present study indicates that sevoflurane exposure during late gestation impairs offspring rat's learning and memory via upregulation of the expression of HDAC2 and downregulation of the expression of CREB and NR2B. SAHA can alleviate these impairments.
Collapse
Affiliation(s)
- Qi Yu
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Namin Feng
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Yan Hu
- Department of Anesthesiology, Jiangxi Province Traditional Chinese Medicine Hospital, China
| | - Foquan Luo
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Weihong Zhao
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Weilu Zhao
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Zhiyi Liu
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Mengyuan Li
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Lin Xu
- Department of Anesthesiology, the First Affiliated Hospital, Nanchang University, China
| | - Liuqing Wu
- Department of Anesthesiology, Jiangxi Province Tumor Hospital, China
| | - Yulin Liu
- Department of Immunology, Jiangxi Medical College, Nanchang University, China
| |
Collapse
|
14
|
Raefsky SM, Furman R, Milne G, Pollock E, Axelsen P, Mattson MP, Shchepinov MS. Deuterated polyunsaturated fatty acids reduce brain lipid peroxidation and hippocampal amyloid β-peptide levels, without discernable behavioral effects in an APP/PS1 mutant transgenic mouse model of Alzheimer's disease. Neurobiol Aging 2018; 66:165-176. [PMID: 29579687 PMCID: PMC5924637 DOI: 10.1016/j.neurobiolaging.2018.02.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/23/2018] [Accepted: 02/24/2018] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) involves progressive deposition of amyloid β-peptide (Aβ), synapse loss, and neuronal death, which occur in brain regions critical for learning and memory. Considerable evidence suggests that lipid peroxidation contributes to synaptic dysfunction and neuronal degeneration, both upstream and downstream of Aβ pathology. Recent findings suggest that lipid peroxidation can be inhibited by replacement of polyunsaturated fatty acids (PUFA) with isotope-reinforced (deuterated) PUFA (D-PUFA), and that D-PUFA can protect neurons in experimental models of Parkinson's disease. Here, we determined whether dietary D-PUFA would ameliorate Aβ pathology and/or cognitive deficits in a mouse model of AD (amyloid precursor protein/presenilin 1 double mutant transgenic mice). The D-PUFA diet did not ameliorate spatial learning and memory deficits in the AD mice. Compared to mice fed an hydrogenated-PUFA control diet, those fed D-PUFA for 5 months exhibited high levels of incorporation of deuterium into arachidonic acid and docosahexaenoic acid, and reduced concentrations of lipid peroxidation products (F2 isoprostanes and neuroprostanes), in the brain tissues. Concentrations of Aβ40 and Aβ38 in the hippocampus were significantly lower, with a trend to reduced concentrations of Aβ42, in mice fed D-PUFA compared to those fed hydrogenated-PUFA. We conclude that a D-PUFA diet reduces the brain tissue concentrations of both arachidonic acid and docosahexaenoic acid oxidation products, as well as the concentration of Aβs.
Collapse
Affiliation(s)
- Sophia M Raefsky
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, MD
| | - Ran Furman
- Departments of Pharmacology, Biochemistry and Biophysics, and Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Ginger Milne
- Eicosanoid Core Laboratory, Vanderbilt University Medical Center, Nashville, TN
| | - Erik Pollock
- Stable Isotope Laboratory, University of Arkansas, Fayetteville, AR
| | - Paul Axelsen
- Departments of Pharmacology, Biochemistry and Biophysics, and Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, MD; Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD.
| | | |
Collapse
|
15
|
Butterfield DA, Boyd-Kimball D. Oxidative Stress, Amyloid-β Peptide, and Altered Key Molecular Pathways in the Pathogenesis and Progression of Alzheimer's Disease. J Alzheimers Dis 2018; 62:1345-1367. [PMID: 29562527 PMCID: PMC5870019 DOI: 10.3233/jad-170543] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2017] [Indexed: 12/12/2022]
Abstract
Oxidative stress is implicated in the pathogenesis and progression of Alzheimer's disease (AD) and its earlier stage, amnestic mild cognitive impairment (aMCI). One source of oxidative stress in AD and aMCI brains is that associated with amyloid-β peptide, Aβ1-42 oligomers. Our laboratory first showed in AD elevated oxidative stress occurred in brain regions rich in Aβ1-42, but not in Aβ1-42-poor regions, and was among the first to demonstrate Aβ peptides led to lipid peroxidation (indexed by HNE) in AD and aMCI brains. Oxidatively modified proteins have decreased function and contribute to damaged key biochemical and metabolic pathways in which these proteins normally play a role. Identification of oxidatively modified brain proteins by the methods of redox proteomics was pioneered in the Butterfield laboratory. Four recurring altered pathways secondary to oxidative damage in brain from persons with AD, aMCI, or Down syndrome with AD are interrelated and contribute to neuronal death. This "Quadrilateral of Neuronal Death" includes altered: glucose metabolism, mTOR activation, proteostasis network, and protein phosphorylation. Some of these pathways are altered even in brains of persons with preclinical AD. We opine that targeting these pathways pharmacologically and with lifestyle changes potentially may provide strategies to slow or perhaps one day, prevent, progression or development of this devastating dementing disorder. This invited review outlines both in vitro and in vivo studies from the Butterfield laboratory related to Aβ1-42 and AD and discusses the importance and implications of some of the major achievements of the Butterfield laboratory in AD research.
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH, USA
| |
Collapse
|
16
|
Davis SA, Gan KA, Dowell JA, Cairns NJ, Gitcho MA. TDP-43 expression influences amyloidβ plaque deposition and tau aggregation. Neurobiol Dis 2017; 103:154-162. [PMID: 28416393 DOI: 10.1016/j.nbd.2017.04.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/31/2017] [Accepted: 04/12/2017] [Indexed: 12/20/2022] Open
Abstract
Although the main focus in Alzheimer's disease (AD) has been an investigation of mechanisms causing Aβ plaque deposition and tau tangle formation, recent studies have shown that phosphorylated TDP-43 pathology is present in up to 50% of sporadic cases. Furthermore, elevated phosphorylated TDP-43 has been associated with more severe AD pathology. Therefore, we hypothesized that TDP-43 may regulate amyloid-beta precursor protein (APP) trafficking and tau phosphorylation/aggregation. In order to examine the role of TDP-43 in AD, we developed a transgenic mouse that overexpresses hippocampal and cortical neuronal TDP-43 in a mouse expressing familial mutations (K595N and M596L) in APP and presenilin 1 (PSEN1ΔE9). In our model, increased TDP-43 was related to increased tau aggregation as evidenced by thioflavin S-positive phosphorylated tau, which may implicate TDP-43 expression in pre-tangle formation. In addition, there was increased endosomal/lysosomal localization of APP and reduced Aβ plaque formation with increased TDP-43. Furthermore, there was decreased calcineurin with elevated TDP-43 expression. Since calcineurin is a phosphatase for TDP-43, the decreased calcineurin expression may be one mechanism leading to an increase in accumulation of diffuse phosphorylated TDP-43 in the hippocampus and cortex. We further show that when TDP-43 is knocked down there is an increase in calcineurin. In our model of selective TDP-43 overexpression in an APP/PSEN1 background, we show that TDP-43 decreases Aβ plaque deposition while increasing abnormal tau aggregation. These observations indicate that TDP-43 may play a role in regulating APP trafficking and tau aggregation. Our data suggest that TDP-43 could be a putative target for therapeutic intervention in AD affecting both Aβ plaque formation and tauopathy.
Collapse
Affiliation(s)
- Stephani A Davis
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE 19901, USA
| | - Kok Ann Gan
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE 19901, USA
| | - James A Dowell
- Wisconsin Institute of Discovery, Madison, WI 53705, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael A Gitcho
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE 19901, USA.
| |
Collapse
|
17
|
Vanillic acid attenuates Aβ 1-42-induced oxidative stress and cognitive impairment in mice. Sci Rep 2017; 7:40753. [PMID: 28098243 PMCID: PMC5241654 DOI: 10.1038/srep40753] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/09/2016] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence demonstrates that β-amyloid (Aβ) elicits oxidative stress, which contributes to the pathogenesis and disease progression of Alzheimer’s disease (AD). The aims of the present study were to determine and explore the antioxidant nature and potential mechanism of vanillic acid (VA) in Aβ1-42-induced oxidative stress and neuroinflammation mediated cognitive impairment in mice. An intracerebroventricular (i.c.v.) injection of Aβ1-42 into the mouse brain triggered increased reactive oxygen species (ROS) levels, neuroinflammation, synaptic deficits, memory impairment, and neurodegeneration. In contrast, the i.p. (intraperitoneal) administration of VA (30 mg/kg, for 3 weeks) after Aβ1-42-injection enhanced glutathione levels (GSH) and abrogated ROS generation accompanied by an induction of the endogenous nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) via the activation of Akt and glycogen synthase kinase 3β (GSK-3β) in the brain mice. Additionally, VA treatment decreased Aβ1-42-induced neuronal apoptosis and neuroinflammation and improved synaptic and cognitive deficits. Moreover, VA was nontoxic to HT22 cells and increased cell viability after Aβ1-42 exposure. To our knowledge, this study is the first to reveal the neuroprotective effect of VA against Aβ1-42-induced neurotoxicity. Our findings demonstrate that VA could potentially serve as a novel, promising, and accessible neuroprotective agent against progressive neurodegenerative diseases such as AD.
Collapse
|
18
|
Gu L, Robinson RAS. Proteomic approaches to quantify cysteine reversible modifications in aging and neurodegenerative diseases. Proteomics Clin Appl 2016; 10:1159-1177. [PMID: 27666938 DOI: 10.1002/prca.201600015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/13/2016] [Accepted: 09/23/2016] [Indexed: 01/11/2023]
Abstract
Cysteine is a highly reactive amino acid and is subject to a variety of reversible post-translational modifications (PTMs), including nitrosylation, glutathionylation, palmitoylation, as well as formation of sulfenic acid and disulfides. These modifications are not only involved in normal biological activities, such as enzymatic catalysis, redox signaling, and cellular homeostasis, but can also be the result of oxidative damage. Especially in aging and neurodegenerative diseases, oxidative stress leads to aberrant cysteine oxidations that affect protein structure and function leading to neurodegeneration as well as other detrimental effects. Methods that can identify cysteine modifications by type, including the site of modification, as well as the relative stoichiometry of the modification can be very helpful for understanding the role of the thiol proteome and redox homeostasis in the context of disease. Cysteine reversible modifications however, are challenging to investigate as they are low abundant, diverse, and labile especially under endogenous conditions. Thanks to the development of redox proteomic approaches, large-scale quantification of cysteine reversible modifications is possible. These approaches cover a range of strategies to enrich, identify, and quantify cysteine reversible modifications from biological samples. This review will focus on nongel-based redox proteomics workflows that give quantitative information about cysteine PTMs and highlight how these strategies have been useful for investigating the redox thiol proteome in aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Liqing Gu
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Renã A S Robinson
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Prasad KN. Simultaneous activation of Nrf2 and elevation of antioxidant compounds for reducing oxidative stress and chronic inflammation in human Alzheimer's disease. Mech Ageing Dev 2016; 153:41-7. [PMID: 26811881 DOI: 10.1016/j.mad.2016.01.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/10/2016] [Accepted: 01/14/2016] [Indexed: 11/18/2022]
Abstract
Despite extensive research, neither the incidence nor the rate of progression of Alzheimer's disease (AD) has significantly changed. Some biochemical and genetic defects that initiate and promote AD include: (a) increased oxidative stress, (b) chronic inflammation (c) mitochondrial dysfunction, (d) Aß1-42 peptides generated from the amyloid precursor protein (APP), (e) proteasome inhibition, and (f) mutations in APP, presenilin-1 and presenilin-2 genes. Increased oxidative stress appears to precede other biochemical and genetic defects. Oxidative damage induces chronic inflammation. Therefore, reducing these defects simultaneously may reduce the development and progression of AD. Previous studies with individual antioxidants produced consistent benefits in animal models of AD; however, a similar approach produced inconsistent results in human AD. This review proposes a hypothesis that simultaneous elevation of the levels of antioxidant enzymes and antioxidant compounds is necessary for optimally reducing oxidative stress and chronic inflammation in human AD. Supplementation can enhance the levels of antioxidant compounds; but elevation of antioxidant enzymes requires activation of Nrf2. This review discusses activation and regulation of Nrf2. The need for multi- antioxidants that can affect multi-targets has been proposed without specific recommendations. This review proposes a micronutrient mixture that would simultaneously enhance the levels of antioxidant enzymes and antioxidant compounds in human AD.
Collapse
|
20
|
Gu L, Robinson RAS. A simple isotopic labeling method to study cysteine oxidation in Alzheimer's disease: oxidized cysteine-selective dimethylation (OxcysDML). Anal Bioanal Chem 2016; 408:2993-3004. [PMID: 26800981 DOI: 10.1007/s00216-016-9307-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/14/2015] [Accepted: 01/04/2016] [Indexed: 02/01/2023]
Abstract
Cysteine is widely involved in redox signaling pathways through a number of reversible and irreversible modifications. Reversible modifications (e.g., S-glutathionylation, S-nitrosylation, disulfide bonds, and sulfenic acid) are used to protect proteins from oxidative attack and maintain cellular homeostasis, while irreversible oxidations (e.g., sulfinic acid and sulfonic acid) serve as hallmarks of oxidative stress. Proteomic analysis of cysteine-enriched peptides coupled with reduction of oxidized thiols can be used to measure the oxidation states of cysteine, which is helpful for elucidating the role that oxidative stress plays in biology and disease. As an extension of our previously reported cysDML method, we have developed oxidized cysteine-selective dimethylation (OxcysDML), to investigate the site-specific total oxidation of cysteine residues in biologically relevant samples. OxcysDML employs (1) blocking of free thiols by a cysteine-reactive reagent, (2) enrichment of peptides containing reversibly oxidized cysteine by a solid phase resin, and (3) isotopic labeling of peptide amino groups to quantify cysteine modifications arising from different biological conditions. On-resin enrichment and labeling minimizes sample handing time and improves efficiency in comparison with other redox proteomic methods. OxcysDML is also inexpensive and flexible, as it can accommodate the exploration of various cysteine modifications. Here, we applied the method to liver tissues from a late-stage Alzheimer's disease (AD) mouse model and wild-type (WT) controls. Because we have previously characterized this proteome using the cysDML approach, we are able here to probe deeper into the redox status of cysteine in AD. OxcysDML identified 1129 cysteine sites (from 527 proteins), among which 828 cysteine sites underwent oxidative modifications. Nineteen oxidized cysteine sites had significant alteration levels in AD and represent proteins involved in metabolic processes. Overall, we have demonstrated OxcysDML as a simple, rapid, robust, and inexpensive redox proteomic approach that is useful for gaining deeper insight into the proteome of AD.
Collapse
Affiliation(s)
- Liqing Gu
- Department of Chemistry, University of Pittsburgh, 111 Eberly Hall, 200 University Drive, Pittsburgh, PA, 15260, USA
| | - Renã A S Robinson
- Department of Chemistry, University of Pittsburgh, 111 Eberly Hall, 200 University Drive, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
21
|
Li H, Ruberu K, Muñoz SS, Jenner AM, Spiro A, Zhao H, Rassart E, Sanchez D, Ganfornina MD, Karl T, Garner B. Apolipoprotein D modulates amyloid pathology in APP/PS1 Alzheimer's disease mice. Neurobiol Aging 2015; 36:1820-33. [PMID: 25784209 DOI: 10.1016/j.neurobiolaging.2015.02.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 01/22/2015] [Accepted: 02/10/2015] [Indexed: 11/24/2022]
Abstract
Apolipoprotein D (apoD) is expressed in the brain and levels are increased in affected brain regions in Alzheimer's disease (AD). The role that apoD may play in regulating AD pathology has not been addressed. Here, we crossed both apoD-null mice and Thy-1 human apoD transgenic mice with APP-PS1 amyloidogenic AD mice. Loss of apoD resulted in a nearly 2-fold increase in hippocampal amyloid plaque load, as assessed by immunohistochemical staining. Conversely, transgenic expression of neuronal apoD reduced hippocampal plaque load by approximately 35%. This latter finding was associated with a 60% decrease in amyloid β 1-40 peptide levels, and a 34% decrease in insoluble amyloid β 1-42 peptide. Assessment of β-site amyloid precursor protein cleaving enzyme-1 (BACE1) levels and proteolytic products of amyloid precursor protein and neuregulin-1 point toward a possible association of altered BACE1 activity in association with altered apoD levels. In conclusion, the current studies provide clear evidence that apoD regulates amyloid plaque pathology in a mouse model of AD.
Collapse
Affiliation(s)
- Hongyun Li
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, NSW, Australia
| | - Kalani Ruberu
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, NSW, Australia
| | - Sonia Sanz Muñoz
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, NSW, Australia
| | - Andrew M Jenner
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, NSW, Australia
| | - Adena Spiro
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, NSW, Australia
| | - Hua Zhao
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, NSW, Australia
| | - Eric Rassart
- Laboratoire de biologie moléculaire, Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, Canada; BioMed, centre de recherches biomédicales, Université du Québec à Montréal, Montréal, Canada
| | - Diego Sanchez
- Departamento de Bioquímica y Biología Molecular y Fisiología - Instituto de Biología y Genética Molecular, Universidad de Valladolid - CSIC, Valladolid, Spain
| | - Maria D Ganfornina
- Departamento de Bioquímica y Biología Molecular y Fisiología - Instituto de Biología y Genética Molecular, Universidad de Valladolid - CSIC, Valladolid, Spain
| | - Tim Karl
- Neuroscience Research Australia, Randwick, NSW, Australia; School of Medical Sciences, University of New South Wales, NSW, Australia; Schizophrenia Research Institute, Darlinghurst, NSW, Australia
| | - Brett Garner
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, NSW, Australia.
| |
Collapse
|
22
|
Gao Y, Hu YZ, Li RS, Han ZT, Geng Y, Xia Z, Du WJ, Liu LX, Zhang HH, Wang LN. Cattle encephalon glycoside and ignotin injection improves cognitive impairment in APPswe/PS1dE9 mice used as multitarget anti-Alzheimer's drug candidates. Neuropsychiatr Dis Treat 2015; 11:537-48. [PMID: 25784809 PMCID: PMC4356454 DOI: 10.2147/ndt.s78025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Cattle encephalon glycoside and ignotin injection (CEGI), a multitargeted neurotrophic drug, has been widely used in the treatment of central and peripheral nerve injuries, such as stroke, hypoxic ischemic encephalopathy, and diabetic neuropathy in the People's Republic of China. However, data regarding the effect of CEGI on Alzheimer's disease (AD) remain scarce. The present study aimed to investigate the effect of CEGI on learning and memory in an APPswe/PS1dE9 double-transgenic mouse model, a suitable animal model of AD, and elucidate its possible mechanisms. MATERIALS AND METHODS Five-month-old APP/PS1 mice were intraperitoneally administered 6.6 mL/kg or 13.2 mL/kg of CEGI for 1 month. After 1 month of administration, all mice received Morris water maze training and a probe test. Mouse brain sections were detected by standard biochemical and immunohistochemical measures. RESULTS CEGI treatment significantly improved the spatial learning and memory deficits and decreased cerebral amyloid-β42 levels in brain homogenates of APP/PS1 mice. CEGI treatment elevated the activities of superoxide dismutase, and reduced the levels of malondialdehyde. CEGI attenuated neuronal damage in the hippocampus of APP/PS1 mice and upregulated protein and gene expression of Bcl-2 and the ratio of Bcl-2/Bax. CEGI treatment decreased the number of Iba1(+) activated microglia in the cortex of the APP/PS1 mice. CONCLUSION Our results showed that CEGI prevents memory impairment, possibly by decreasing the amyloid-β42 levels in APP/PS1 mice and inhibiting oxidative stress, apoptosis, and inflammation, making CEGI a promising therapeutic agent for AD.
Collapse
Affiliation(s)
- Ya Gao
- Institute of Geriatrics, Chinese PLA General Hospital, Beijing Key Lab of Normal Aging and Geriatrics, Beijing, People's Republic of China
| | - Ya-Zhuo Hu
- Institute of Geriatrics, Chinese PLA General Hospital, Beijing Key Lab of Normal Aging and Geriatrics, Beijing, People's Republic of China
| | - Rui-Sheng Li
- Research and Technology Service Center, PLA 302 Hospital, Beijing, People's Republic of China
| | - Zhi-Tao Han
- Institute of Geriatrics, Chinese PLA General Hospital, Beijing Key Lab of Normal Aging and Geriatrics, Beijing, People's Republic of China
| | - Yan Geng
- Institute of Geriatrics, Chinese PLA General Hospital, Beijing Key Lab of Normal Aging and Geriatrics, Beijing, People's Republic of China
| | - Zheng Xia
- Institute of Geriatrics, Chinese PLA General Hospital, Beijing Key Lab of Normal Aging and Geriatrics, Beijing, People's Republic of China
| | - Wen-Jin Du
- Department of Neurology, Air Force General Hospital, Beijing, People's Republic of China
| | - Li-Xin Liu
- Department of Neurology, Beijing Geriatric Hospital, Beijing, People's Republic of China
| | - Hong-Hong Zhang
- Institute of Geriatrics, Chinese PLA General Hospital, Beijing Key Lab of Normal Aging and Geriatrics, Beijing, People's Republic of China
| | - Lu-Ning Wang
- Department of Geriatric Neurology, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
23
|
Liu A, Zhao X, Li H, Liu Z, Liu B, Mao X, Guo L, Bi K, Jia Y. 5-Hydroxymethylfurfural, an antioxidant agent from Alpinia oxyphylla Miq. improves cognitive impairment in Aβ1–42 mouse model of Alzheimer's disease. Int Immunopharmacol 2014; 23:719-25. [DOI: 10.1016/j.intimp.2014.10.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/11/2014] [Accepted: 10/28/2014] [Indexed: 11/25/2022]
|
24
|
Butterfield DA. The 2013 SFRBM discovery award: selected discoveries from the butterfield laboratory of oxidative stress and its sequela in brain in cognitive disorders exemplified by Alzheimer disease and chemotherapy induced cognitive impairment. Free Radic Biol Med 2014; 74:157-74. [PMID: 24996204 PMCID: PMC4146642 DOI: 10.1016/j.freeradbiomed.2014.06.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/05/2014] [Accepted: 06/10/2014] [Indexed: 12/21/2022]
Abstract
This retrospective review on discoveries of the roles of oxidative stress in brain of subjects with Alzheimer disease (AD) and animal models thereof as well as brain from animal models of chemotherapy-induced cognitive impairment (CICI) results from the author receiving the 2013 Discovery Award from the Society for Free Radical Biology and Medicine. The paper reviews our laboratory's discovery of protein oxidation and lipid peroxidation in AD brain regions rich in amyloid β-peptide (Aβ) but not in Aβ-poor cerebellum; redox proteomics as a means to identify oxidatively modified brain proteins in AD and its earlier forms that are consistent with the pathology, biochemistry, and clinical presentation of these disorders; how Aβ in in vivo, ex vivo, and in vitro studies can lead to oxidative modification of key proteins that also are oxidatively modified in AD brain; the role of the single methionine residue of Aβ(1-42) in these processes; and some of the potential mechanisms in the pathogenesis and progression of AD. CICI affects a significant fraction of the 14 million American cancer survivors, and due to diminished cognitive function, reduced quality of life of the persons with CICI (called "chemobrain" by patients) often results. A proposed mechanism for CICI employed the prototypical ROS-generating and non-blood brain barrier (BBB)-penetrating chemotherapeutic agent doxorubicin (Dox, also called adriamycin, ADR). Because of the quinone moiety within the structure of Dox, this agent undergoes redox cycling to produce superoxide free radical peripherally. This, in turn, leads to oxidative modification of the key plasma protein, apolipoprotein A1 (ApoA1). Oxidized ApoA1 leads to elevated peripheral TNFα, a proinflammatory cytokine that crosses the BBB to induce oxidative stress in brain parenchyma that affects negatively brain mitochondria. This subsequently leads to apoptotic cell death resulting in CICI. This review outlines aspects of CICI consistent with the clinical presentation, biochemistry, and pathology of this disorder. To the author's knowledge this is the only plausible and self-consistent mechanism to explain CICI. These two different disorders of the CNS affect millions of persons worldwide. Both AD and CICI share free radical-mediated oxidative stress in brain, but the source of oxidative stress is not the same. Continued research is necessary to better understand both AD and CICI. The discoveries about these disorders from the Butterfield Laboratory that led to the 2013 Discovery Award from the Society of Free Radical and Medicine provide a significant foundation from which this future research can be launched.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, Free Radical Biology in Cancer, Shared Resource Facility of the Markey Cancer Center, Spinal Cord and Brain Injury Research Center, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
25
|
Swomley AM, Förster S, Keeney JT, Triplett J, Zhang Z, Sultana R, Butterfield DA. Abeta, oxidative stress in Alzheimer disease: evidence based on proteomics studies. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1248-57. [PMID: 24120836 PMCID: PMC3981962 DOI: 10.1016/j.bbadis.2013.09.015] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 01/01/2023]
Abstract
The initiation and progression of Alzheimer disease (AD) is a complex process not yet fully understood. While many hypotheses have been provided as to the cause of the disease, the exact mechanisms remain elusive and difficult to verify. Proteomic applications in disease models of AD have provided valuable insights into the molecular basis of this disorder, demonstrating that on a protein level, disease progression impacts numerous cellular processes such as energy production, cellular structure, signal transduction, synaptic function, mitochondrial function, cell cycle progression, and proteasome function. Each of these cellular functions contributes to the overall health of the cell, and the dysregulation of one or more could contribute to the pathology and clinical presentation in AD. In this review, foci reside primarily on the amyloid β-peptide (Aβ) induced oxidative stress hypothesis and the proteomic studies that have been conducted by our laboratory and others that contribute to the overall understanding of this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- Aaron M Swomley
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Sarah Förster
- Department of Biochemistry, Institute of Animal Sciences, University of Bonn, Bonn, Germany
| | - Jierel T Keeney
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Judy Triplett
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Zhaoshu Zhang
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
26
|
Puerarin alleviates cognitive impairment and oxidative stress in APP/PS1 transgenic mice. Int J Neuropsychopharmacol 2014; 17:635-44. [PMID: 24345484 DOI: 10.1017/s146114571300148x] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Increasing evidence demonstrates that β-amyloid (Aβ) elicits oxidative stress, which contributes to the pathogenesis and disease progression of Alzheimer's disease (AD). Thus, there is interest in developing antioxidant therapies for the prevention/treatment of cognitive decline during AD. We reported previously that puerarin has antioxidative properties in vitro. Therefore, the aim of the present study was to determine whether puerarin improves cognitive function and reduces oxidative stress in amyloid precursor protein/presenilin-1 (APP/PS1) mice, a well established AD mouse model, and explore its potential mechanism. Our results show that oral administration of puerarin significantly ameliorates cognitive impairment in APP/PS1 mice assessed by the Morris water maze (MWM) test. This was accompanied by a significant decrease in the levels of lipid peroxidation (LPO) through, at least in part, induction of nuclear factor erythroid 2-related factor 2 (Nrf2) target gene heme oxygenase 1 (HO-1) in the hippocampus of APP/PS1 transgenic mice at 9 months of age, but without altering brain Aβ burden. Furthermore, puerarin significantly activated Akt, reduced activation of glycogen synthase kinase 3β (GSK-3β), and induced nuclear translocation of Nrf2 in the hippocampus of APP/PS1 mice but did not alter ERK1/2 phosphorylation. Thus, puerarin may improve cognitive performance in APP/PS1 mice through activation of the Akt/GSK-3β signaling pathway. These findings suggest that puerarin might be an attractive agent for prevention and treatment of cognitive impairment and dementia.
Collapse
|
27
|
Cao Z, Robinson RAS. Proteome characterization of splenocytes from an A
βpp/ps-
1 Alzheimer's disease model. Proteomics 2014; 14:291-7. [DOI: 10.1002/pmic.201300130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 10/22/2013] [Accepted: 11/21/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Zhiyun Cao
- Department of Chemistry; University of Pittsburgh; Pittsburgh PA USA
| | | |
Collapse
|
28
|
Koivisto H, Grimm MO, Rothhaar TL, Berkecz R, Lütjohann D D, Giniatullina R, Takalo M, Miettinen PO, Lahtinen HM, Giniatullin R, Penke B, Janáky T, Broersen LM, Hartmann T, Tanila H. Special lipid-based diets alleviate cognitive deficits in the APPswe/PS1dE9 transgenic mouse model of Alzheimer's disease independent of brain amyloid deposition. J Nutr Biochem 2013; 25:157-69. [PMID: 24445040 DOI: 10.1016/j.jnutbio.2013.09.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 08/30/2013] [Accepted: 09/11/2013] [Indexed: 11/15/2022]
Abstract
Dietary fish oil, providing n3 polyunsaturated fatty acids like eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), associates with reduced dementia risk in epidemiological studies and reduced amyloid accumulation in Alzheimer mouse models. We now studied whether additional nutrients can improve the efficacy of fish oil in alleviating cognitive deficits and amyloid pathology in APPswe/PS1dE9 transgenic and wild-type mice. We compared four isocaloric (5% fat) diets. The fish oil diet differed from the control diet only by substituted fish oil. Besides fish oil, the plant sterol diet was supplemented with phytosterols, while the Fortasyn diet contained as supplements precursors and cofactors for membrane synthesis, viz. uridine-monophosphate; DHA and EPA; choline; folate; vitamins B6, B12, C and E; phospholipids and selenium. Mice began the special diets at 5 months and were sacrificed at 14 months after behavioral testing. Transgenic mice, fed with control chow, showed poor spatial learning, hyperactivity in exploring a novel cage and reduced preference to explore novel odors. All fish-oil-containing diets increased exploration of a novel odor over a familiar one. Only the Fortasyn diet alleviated the spatial learning deficit. None of the diets influenced hyperactivity in a new environment. Fish-oil-containing diets strongly inhibited β- and γ-secretase activity, and the plant sterol diet additionally reduced amyloid-β 1-42 levels. These data indicate that beneficial effects of fish oil on cognition in Alzheimer model mice can be enhanced by adding other specific nutrients, but this effect is not necessarily mediated via reduction of amyloid accumulation.
Collapse
Affiliation(s)
- Hennariikka Koivisto
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio FI-70211, Finland
| | - Marcus O Grimm
- Experimental Neurology, Saarland University, Homburg/Saar 66123, Germany; Deutsches Institut für Demenz Prävention, Saarland University, Homburg/Saar 66421, Germany
| | - Tatjana L Rothhaar
- Experimental Neurology, Saarland University, Homburg/Saar 66123, Germany
| | - Róbert Berkecz
- Department Medical Chemistry, University of Szeged, Szeged 6720, Hungary
| | - Dieter Lütjohann D
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn 53113, Germany
| | - Rajsa Giniatullina
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio FI-70211, Finland
| | - Mari Takalo
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio FI-70211, Finland
| | - Pasi O Miettinen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio FI-70211, Finland
| | - Hanna-Maija Lahtinen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio FI-70211, Finland
| | - Rashid Giniatullin
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio FI-70211, Finland
| | - Botond Penke
- Department Medical Chemistry, University of Szeged, Szeged 6720, Hungary
| | - Tamás Janáky
- Department Medical Chemistry, University of Szeged, Szeged 6720, Hungary
| | - Laus M Broersen
- Danone Research Centre for Specialised Nutrition, Wageningen 6700 CA, The Netherlands
| | - Tobias Hartmann
- Deutsches Institut für Demenz Prävention, Saarland University, Homburg/Saar 66421, Germany
| | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio FI-70211, Finland; Department of Neurology, Kuopio University Hospital, Kuopio FI-70211, Finland.
| |
Collapse
|
29
|
Webster SJ, Bachstetter AD, Van Eldik LJ. Comprehensive behavioral characterization of an APP/PS-1 double knock-in mouse model of Alzheimer's disease. Alzheimers Res Ther 2013; 5:28. [PMID: 23705774 PMCID: PMC3706792 DOI: 10.1186/alzrt182] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/29/2013] [Accepted: 05/24/2013] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Despite the extensive mechanistic and pathological characterization of the amyloid precursor protein (APP)/presenilin-1 (PS-1) knock-in mouse model of Alzheimer's disease (AD), very little is known about the AD-relevant behavioral deficits in this model. Characterization of the baseline behavioral performance in a variety of functional tasks and identification of the temporal onset of behavioral impairments are important to provide a foundation for future preclinical testing of AD therapeutics. Here we perform a comprehensive behavioral characterization of this model, discuss how the observed behavior correlates with the mechanistic and pathological observations of others, and compare this model with other commonly used AD mouse models. METHODS FOUR DIFFERENT GROUPS OF MICE RANGING ACROSS THE LIFESPAN OF THIS MODEL (TEST GROUPS: 7, 11, 15, and 24 months old) were run in a behavioral test battery consisting of tasks to assess motor function (grip strength, rotor rod, beam walk, open field ambulatory movement), anxiety-related behavior (open field time spent in peripheral zone vs. center zone, elevated plus maze), and cognitive function (novel object recognition, radial arm water maze). RESULTS There were no differences in motor function or anxiety-related behavior between APP/PS-1 knock-in mice and wild-type counterpart mice for any age group. Cognitive deficits in both recognition memory (novel object recognition) and spatial reference memory (radial arm water maze) became apparent for the knock-in animals as the disease progressed. CONCLUSION This is the first reported comprehensive behavioral analysis of the APP/PS1 knock-in mouse model of AD. The lack of motor/coordination deficits or abnormal anxiety levels, coupled with the age/disease-related cognitive decline and high physiological relevance of this model, make it well suited for utilization in preclinical testing of AD-relevant therapeutics.
Collapse
Affiliation(s)
- Scott J Webster
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
- Department of Anatomy and Neurobiology, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
30
|
Wang CM, Liu MY, Wang F, Wei MJ, Wang S, Wu CF, Yang JY. Anti-amnesic effect of pseudoginsenoside-F11 in two mouse models of Alzheimer's disease. Pharmacol Biochem Behav 2013; 106:57-67. [DOI: 10.1016/j.pbb.2013.03.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 03/12/2013] [Accepted: 03/16/2013] [Indexed: 11/15/2022]
|
31
|
Evidence for premature lipid raft aging in APP/PS1 double-transgenic mice, a model of familial Alzheimer disease. J Neuropathol Exp Neurol 2012; 71:868-81. [PMID: 22975585 DOI: 10.1097/nen.0b013e31826be03c] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Altered lipid raft homeostasis has been considered to contribute to cellular deregulation, leading to neuronal loss in Alzheimer disease. Alterations in these microdomains affect amyloid precursor protein (APP) processing, resulting in neurotoxic conditions, but modifications of the molecular structure of lipid rafts in Alzheimer disease model mice have not been characterized. Using a lipidomic approach, we investigated frontal cortex lipid rafts inwild-type mice and in double-transgenic APP/presenilin 1 (PS1) mice. Lipid rafts in wild-type mice undergo age-dependent modifications, that is, decreased cholesterol and sterol esters levels, augmented sphingomyelin and saturated fatty acid content, and increased phospholipids/cholesterol ratio. These age-dependent changes were more dramatic and occurred earlier in APP/PS1 mice; other lipid classes (e.g. sulfatides) and essential long-chain polyunsaturated fatty acids (including docosahexaenoic and arachidonic acids) were also affected. Steady state anisotropy measurements demonstrated that APP/PS1 animals exhibit more viscous (membrane-ordered) lipid rafts and that this is mainly attributable to reduced unsaturation of phospholipids and increased sphingomyelin levels rather than to changes in cholesterol. In summary, we demonstrate that aging is accompanied by alteration of the physicochemical structure of lipid raft microdomains. This "lipid raft aging," a metaphenomenon, is considerably exacerbated by the induced amyloid burden in APP/PS1 genotype.
Collapse
|
32
|
Testa G, Gamba P, Di Scipio F, Sprio AE, Salamone P, Gargiulo S, Sottero B, Biasi F, Berta GN, Poli G, Leonarduzzi G. Potentiation of amyloid-β peptide neurotoxicity in human dental-pulp neuron-like cells by the membrane lipid peroxidation product 4-hydroxynonenal. Free Radic Biol Med 2012; 53:1708-17. [PMID: 22981873 DOI: 10.1016/j.freeradbiomed.2012.08.581] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 07/24/2012] [Accepted: 08/21/2012] [Indexed: 12/30/2022]
Abstract
Lipid peroxidation is generally considered as primarily implicated in the pathogenesis of Alzheimer's disease (AD); one of its more reactive end products, 4-hydroxynonenal (HNE), has been shown to cause neuron dysfunction and degeneration. HNE production in the brain is stimulated by the amyloid-β peptide (Aβ), whose excessive accumulation in specific brain areas is a hallmark of AD. Conversely, Aβ production is up-regulated by this multifunctional aldehyde. Findings reported here point to the ability of HNE and Aβ to interact, with consequent potentiation of Aβ's cytotoxicity as determined in vitro using neuron-like cells derived from human dental-pulp progenitor cells. Preincubation of cells with the aldehyde markedly up-regulated Aβ uptake and intracellular accumulation, by overexpressing two of the three components of the plasma membrane multireceptor complex CD36/CD47/β1-integrin: experimental and clinical data indicate that intraneuronal accumulation of Aβ is an early event possibly playing a primary role in AD pathogenesis. That HNE-mediated overexpression of CD36 and β1-integrin, which plays a key role in HNE's potentiating Aβ neurotoxicity, in terms of necrosis, was confirmed when this effect was prevented by specific antibodies against the two receptors.
Collapse
Affiliation(s)
- Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Tsirulnikov K, Abuladze N, Bragin A, Faull K, Cascio D, Damoiseaux R, Schibler MJ, Pushkin A. Inhibition of aminoacylase 3 protects rat brain cortex neuronal cells from the toxicity of 4-hydroxy-2-nonenal mercapturate and 4-hydroxy-2-nonenal. Toxicol Appl Pharmacol 2012; 263:303-14. [PMID: 22819785 DOI: 10.1016/j.taap.2012.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 06/11/2012] [Accepted: 07/02/2012] [Indexed: 12/21/2022]
Abstract
4-Hydroxy-2-nonenal (4HNE) and acrolein (ACR) are highly reactive neurotoxic products of lipid peroxidation that are implicated in the pathogenesis and progression of Alzheimer's and Parkinson's diseases. Conjugation with glutathione (GSH) initiates the 4HNE and ACR detoxification pathway, which generates the mercapturates of 4HNE and ACR that can be excreted. Prior work has shown that the efficiency of the GSH-dependent renal detoxification of haloalkene derived mercapturates is significantly decreased upon their deacetylation because of rapid transformation of the deacetylated products into toxic compounds mediated by β-lyase. The enzymes of the GSH-conjugation pathway and β-lyases are expressed in the brain, and we hypothesized that a similar toxicity mechanism may be initiated in the brain by the deacetylation of 4HNE- and ACR-mercapturate. The present study was performed to identify an enzyme(s) involved in 4HNE- and ACR-mercapturate deacetylation, characterize the brain expression of this enzyme and determine whether its inhibition decreases 4HNE and 4HNE-mercapturate neurotoxicity. We demonstrated that of two candidate deacetylases, aminoacylases 1 (AA1) and 3 (AA3), only AA3 efficiently deacetylates both 4HNE- and ACR-mercapturate. AA3 was further localized to neurons and blood vessels. Using a small molecule screen we generated high-affinity AA3 inhibitors. Two of them completely protected rat brain cortex neurons expressing AA3 from the toxicity of 4HNE-mercapturate. 4HNE-cysteine (4HNE-Cys) was also neurotoxic and its toxicity was mostly prevented by a β-lyase inhibitor, aminooxyacetate. The results suggest that the AA3 mediated deacetylation of 4HNE-mercapturate may be involved in the neurotoxicity of 4HNE.
Collapse
Affiliation(s)
- Kirill Tsirulnikov
- Department of Medicine, University of California at Los Angeles, CA 90095-1689, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Zhang W, Bai M, Xi Y, Hao J, Liu L, Mao N, Su C, Miao J, Li Z. Early memory deficits precede plaque deposition in APPswe/PS1dE9 mice: involvement of oxidative stress and cholinergic dysfunction. Free Radic Biol Med 2012; 52:1443-52. [PMID: 22342520 DOI: 10.1016/j.freeradbiomed.2012.01.023] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Revised: 01/08/2012] [Accepted: 01/26/2012] [Indexed: 12/23/2022]
Abstract
A large body of evidence has shown that cognitive deficits occur early, before amyloid plaque deposition, suggesting that soluble amyloid-β protein (Aβ) contributes to the development of early cognitive dysfunction in Alzheimer disease (AD). However, the underlying mechanism(s) through which soluble Aβ exerts its neurotoxicity responsible for cognitive dysfunction in the early stage of AD remains unclear so far. In this study, we used preplaque APPswe/PS1dE9 mice ages 2.5 and 3.5 months to examine alterations in cognitive function, oxidative stress, and cholinergic function. We found that only soluble Aβ, not insoluble Aβ, was detected in these preplaque APPswe/PS1dE9 mice. APPswe/PS1dE9 mice 2.5 months of age did not show any significant changes in the measures of cognitive function, oxidative stress, and cholinergic function, whereas 3.5-month-old APPswe/PS1dE9 mice exhibited spatial memory impairment in the Morris water maze, accompanied by significantly decreased acetylcholine (ACh), choline acetyltransferase (ChAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-px) as well as increased malondialdehyde (MDA) and protein carbonyls. In 3.5-month-old preplaque APPswe/PS1dE9 mice, correlational analyses revealed that the performance of impaired spatial memory was inversely correlated with soluble Aβ, MDA, and protein carbonyls, as well as being positively correlated with ACh, ChAT, SOD, and GSH-px; soluble Aβ level was inversely correlated with ACh, ChAT, SOD, and GSH-px, as well as being positively correlated with MDA and protein carbonyls; ACh level showed a significant positive correlation with ChAT, SOD, and GSH-px, as well as a significant inverse correlation with MDA and protein carbonyls. Collectively, this study provides direct evidence that increased oxidative damage and cholinergic dysfunction may be early pathological responses to soluble Aβ and involved in early memory deficits in the preplaque stage of AD. These findings suggest that early antioxidant therapy and improving cholinergic function may be a promising strategy to prevent or delay the onset and progression of AD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shaanxi Province 710038, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Belkacemi A, Ramassamy C. Time sequence of oxidative stress in the brain from transgenic mouse models of Alzheimer's disease related to the amyloid-β cascade. Free Radic Biol Med 2012; 52:593-600. [PMID: 22172527 DOI: 10.1016/j.freeradbiomed.2011.11.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 11/16/2011] [Accepted: 11/16/2011] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial disorder characterized by the presence of amyloid plaques and neurofibrillary tangles (NFTs). Rare early-onset forms of AD are associated with autosomal dominant mutations in the amyloid precursor protein gene, presenilin 1 gene, or presenilin 2 gene. The late-onset form of the disease (LOAD) is the most common form. The causes of LOAD are not yet clarified, but several environmental and genetic risk factors have been identified. Numerous studies have highlighted a role for free radical-mediated injury to brain regions of this illness. In addition, studies from mild cognitive impairment patients suggest that oxidative stress is an early event in the pathogenesis of AD. The associations between these markers of free radical damage and the pathogenic cascades involved in AD are complex. Over the past 2 decades, a number of mouse models have been created to recapitulate the major neuropathological hallmarks of AD, namely amyloid plaques and NFTs. These mice recapitulate many, although not all, of the key features of AD. Some strains of transgenic mice develop amyloid plaques, some accumulate NFTs, and some do both. Here we review the evidence for increased free radical-mediated damage to the brain with particular attention to the stage of the disease in various transgenic models of AD related to the amyloid-β cascade.
Collapse
Affiliation(s)
| | - Charles Ramassamy
- INRS-Institut Armand-Frappier, H7V 1B7 Laval, QC, Canada; Faculté de Médecine, Université Laval, Laval, QC, Canada.
| |
Collapse
|
36
|
Lott IT. Antioxidants in Down syndrome. Biochim Biophys Acta Mol Basis Dis 2011; 1822:657-63. [PMID: 22206998 DOI: 10.1016/j.bbadis.2011.12.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/13/2011] [Accepted: 12/14/2011] [Indexed: 01/09/2023]
Abstract
Individuals with Down syndrome (DS) have high levels of oxidative stress throughout the lifespan. Mouse models of DS share some structural and functional abnormalities that parallel findings seen in the human phenotype. Several of the mouse models show evidence of cellular oxidative stress and have provided a platform for antioxidant intervention. Genes that are overexpressed on chromosome 21 are associated with oxidative stress and neuronal apoptosis. The lack of balance in the metabolism of free radicals generated during processes related to oxidative stress may have a direct role in producing the neuropathology of DS including the tendency to Alzheimer disease (AD). Mitochondria are often a target for oxidative stress and are considered to be a trigger for the onset of the AD process in DS. Biomarkers for oxidative stress have been described in DS and in AD in the general population. However, intervention trials using standard antioxidant supplements or diets have failed to produce uniform therapeutic effect. This chapter will examine the biological role of oxidative stress in DS and its relationship to abnormalities in both development and aging within the disorder. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.
Collapse
Affiliation(s)
- Ira T Lott
- Department of Pediatrics and Neurology, School of Medicine, University of California Irvine (UCI), Orange, CA 92868, USA.
| |
Collapse
|
37
|
Sultana R, Robinson RAS, Di Domenico F, Mohmmad Abdul H, St. Clair DK, Markesbery WR, Cai J, Pierce WM, Butterfield DA. Proteomic identification of specifically carbonylated brain proteins in APP(NLh)/APP(NLh) × PS-1(P264L)/PS-1(P264L) human double mutant knock-in mice model of Alzheimer disease as a function of age. J Proteomics 2011; 74:2430-40. [PMID: 21726674 PMCID: PMC3199338 DOI: 10.1016/j.jprot.2011.06.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/10/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
Abstract
Alzheimer disease (AD) is the most common type of dementia and is characterized pathologically by the presence of neurofibrillary tangles (NFTs), senile plaques (SPs), and loss of synapses. The main component of SP is amyloid-beta peptide (Aβ), a 39 to 43 amino acid peptide, generated by the proteolytic cleavage of amyloid precursor protein (APP) by the action of beta- and gamma-secretases. The presenilins (PS) are components of the γ-secretase, which contains the protease active center. Mutations in PS enhance the production of the Aβ42 peptide. To date, more than 160 mutations in PS1 have been identified. Many PS mutations increase the production of the β-secretase-mediated C-terminal (CT) 99 amino acid-long fragment (CT99), which is subsequently cleaved by γ-secretase to yield Aβ peptides. Aβ has been proposed to induce oxidative stress and neurotoxicity. Previous studies from our laboratory and others showed an age-dependent increase in oxidative stress markers, loss of lipid asymmetry, and Aβ production and amyloid deposition in the brain of APP/PS1 mice. In the present study, we used APP (NLh)/APP(NLh) × PS-1(P246L)/PS-1(P246L) human double mutant knock-in APP/PS-1 mice to identify specific targets of brain protein carbonylation in an age-dependent manner. We found a number of proteins that are oxidatively modified in APP/PS1 mice compared to age-matched controls. The relevance of the identified proteins to the progression and pathogenesis of AD is discussed.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Renã A. S. Robinson
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Hafiz Mohmmad Abdul
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Daret K. St. Clair
- Graduate Center of Toxicology, University of Kentucky, Lexington, KY 40536, USA
| | | | - Jian Cai
- Department of Pharmacology, University of Louisville, Louisville, Kentucky 40292
| | - William M. Pierce
- Department of Pharmacology, University of Louisville, Louisville, Kentucky 40292
| | - D. Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| |
Collapse
|
38
|
Robinson RAS, Joshi G, Huang Q, Sultana R, Baker AS, Cai J, Pierce W, St Clair DK, Markesbery WR, Butterfield DA. Proteomic analysis of brain proteins in APP/PS-1 human double mutant knock-in mice with increasing amyloid β-peptide deposition: insights into the effects of in vivo treatment with N-acetylcysteine as a potential therapeutic intervention in mild cognitive impairment and Alzheimer's disease. Proteomics 2011; 11:4243-56. [PMID: 21954051 DOI: 10.1002/pmic.201000523] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 07/21/2011] [Accepted: 08/18/2011] [Indexed: 12/14/2022]
Abstract
Proteomics analyses were performed on the brains of wild-type (WT) controls and an Alzheimer's disease (AD) mouse model, APP/PS-1 human double mutant knock-in mice. Mice were given either drinking water or water supplemented with N-acetylcysteine (NAC) (2 mg/kg body weight) for a period of five months. The time periods of treatment correspond to ages prior to Aβ deposition (i.e. 4-9 months), resembling human mild cognitive impairment (MCI), and after Aβ deposition (i.e. 7-12 months), more closely resembling advancing stages of AD. Substantial differences exist between the proteomes of WT and APP/PS-1 mice at 9 or 12 months, indicating that Aβ deposition and oxidative stress lead to downstream changes in protein expression. Altered proteins are involved in energy-related pathways, excitotoxicity, cell cycle signaling, synaptic abnormalities, and cellular defense and structure. Overall, the proteomic results support the notion that NAC may be beneficial for increasing cellular stress responses in WT mice and for influencing the levels of energy- and mitochondria-related proteins in APP/PS-1 mice.
Collapse
Affiliation(s)
- Renã A S Robinson
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dumont M, Beal MF. Neuroprotective strategies involving ROS in Alzheimer disease. Free Radic Biol Med 2011; 51:1014-26. [PMID: 21130159 PMCID: PMC3070183 DOI: 10.1016/j.freeradbiomed.2010.11.026] [Citation(s) in RCA: 268] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 10/29/2010] [Accepted: 11/22/2010] [Indexed: 12/14/2022]
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder in which oxidative stress is a key hallmark. It occurs early in disease pathogenesis and can exacerbate its progression. Several causes of oxidative stress have been determined over the years. First, mitochondria play an important role in the generation and accumulation of free radicals. In addition to mitochondria, inflammation can also induce oxidative damage, especially via microglia, and microglia are also important for Aβ clearance. In AD, both mitochondrial function and inflammatory response are affected, leading to increased ROS formation and oxidative damage to lipid, proteins, and nucleic acids. Some other sources have also been identified. From these findings, various neuroprotective strategies against ROS-mediated damages have been elaborated in AD research. This review recapitulates some of the major strategies used to prevent oxidative stress and disease progression. Outcomes from in vitro and in vivo studies using models of AD are encouraging. However, only a few clinical trials have provided positive results in terms of slowing down cognitive decline. Nonetheless, there is still hope for improved compounds that would better target pathways implicated in ROS production. In fact, facilitating the endogenous antioxidant system by modulating transcription has great promise for AD therapy.
Collapse
Affiliation(s)
- Magali Dumont
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA.
| | | |
Collapse
|
40
|
Koltai E, Zhao Z, Lacza Z, Cselenyak A, Vacz G, Nyakas C, Boldogh I, Ichinoseki-Sekine N, Radak Z. Combined exercise and insulin-like growth factor-1 supplementation induces neurogenesis in old rats, but do not attenuate age-associated DNA damage. Rejuvenation Res 2011; 14:585-96. [PMID: 21867412 DOI: 10.1089/rej.2011.1178] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We have investigated the effects of 2 weeks of insulin-like growth factor-1 (IGF-1) supplementation (5 μg/kg per day) and 6 weeks of exercise training (60% of the maximal oxygen consumption [VO₂ max]) on neurogenesis, DNA damage/repair, and sirtuin content in the hippocampus of young (3 months old) and old (26 months old) rats. Exercise improved the spatial memory of the old group, but IGF-1 supplementation eliminated this effect. An age-associated decrease in neurogenesis was attenuated by exercise and IGF-1 treatment. Aging increased the levels of 8-oxo-7,8-dihydroguanine (8-oxoG) and the protein Ku70, indicating the role of DNA damage in age-related neuropathology. Acetylation of 8-oxoguanine DNA glycosylase (OGG1) was detected in vivo, and this decreased with aging. However, in young animals, exercise and IGF-1 treatment increased acetylated (ac) OGG1 levels. Sirtuin 1 (SIRT1) and SIRT3, as DNA damage-associated lysine deacetylases, were measured, and SIRT1 decreased with aging, resulting in a large increase in acetylated lysine residues in the hippocampus. On the other hand, SIRT3 increased with aging. Exercise-induced neurogenesis might not be a causative factor of increased spatial memory, because IGF-1 plus exercise can induce neurogenesis in the hippocampus of older rats. Data revealed that the age-associated increase in 8-oxoG levels is due to decreased acetylation of OGG1. Age-associated decreases in SIRT1 and the associated increase in lysine acetylation, in the hippocampus, could have significant impact on function and thus, could suggest a therapeutic target.
Collapse
Affiliation(s)
- Erika Koltai
- Semmelweis University , Research Institute of Sport Science, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bruce-Keller AJ, Gupta S, Knight AG, Beckett TL, McMullen JM, Davis PR, Murphy MP, Van Eldik LJ, St Clair D, Keller JN. Cognitive impairment in humanized APP×PS1 mice is linked to Aβ(1-42) and NOX activation. Neurobiol Dis 2011; 44:317-26. [PMID: 21798347 DOI: 10.1016/j.nbd.2011.07.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 06/11/2011] [Accepted: 07/11/2011] [Indexed: 02/08/2023] Open
Abstract
Cognitive impairment in Alzheimer's disease (AD) is strongly associated with both extensive deposition of amyloid β peptides and oxidative stress, but the exact role of these indices in the development of dementia is not clear. This study was designed to determine the relationship between cognitive impairment, activation of the free radical producing enzyme NADPH oxidase (NOX), and progressive changes in Aβ deposition and solubility in humanized APP×PS1 knock-in mice of increasing age. Data show that cognitive performance and expression of key synaptic proteins were progressively decreased in aging APP×PS1 mice. Likewise, NOX activity and expression of the specific NOX subunit NOX4 were significantly increased in APP×PS1 mice in an age-dependent manner, and NOX activity and cognitive impairment shared a significant linear relationship. Data further show that age-dependent increases in Aβ(1-42) had a significant linear relationship with both NOX activity and cognitive performance in APP×PS1 knock-in mice. Collectively, these data show that NOX expression and activity are significantly upregulated with age in this humanized model of Aβ pathogenesis, and suggest that NOX-associated redox pathways are intimately linked to both the loss of cognitive function and the deposition of Aβ(1-42).
Collapse
Affiliation(s)
- Annadora J Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee HG, Won SM, Gwag BJ, Lee YB. Microglial P2X₇ receptor expression is accompanied by neuronal damage in the cerebral cortex of the APPswe/PS1dE9 mouse model of Alzheimer's disease. Exp Mol Med 2011; 43:7-14. [PMID: 21088470 DOI: 10.3858/emm.2011.43.1.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The possibility that P2X₇ receptor (P2X₇R) expression in microglia would mediate neuronal damage via reactive oxygen species (ROS) production was examined in the APPswe/PS1dE9 mouse model of Alzheimer's disease (AD). P2X7R was predominantly expressed in CD11b-immunopositive microglia from 3 months of age before Abeta plaque formation. In addition, gp91phox, a catalytic subunit of NADPH oxidase, and ethidium fluorescence were detected in P2X₇R-positive microglial cells of animals at 6 months of age, indicating that P2X₇R-positive microglia could produce ROS. Postsynaptic density 95-positive dendrites showed significant damage in regions positive for P2X₇R in the cerebral cortex of 6 month-old mice. Taken together, up-regulation of P2X₇R activation and ROS production in microglia are parallel with Aβ increase and correlate with synaptotoxicity in AD.
Collapse
Affiliation(s)
- Hwan Goo Lee
- Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Korea
| | | | | | | |
Collapse
|
43
|
Obulesu M, Somashekhar R, Venu R. Genetics of Alzheimer's Disease: An Insight Into Presenilins and Apolipoprotein E Instigated Neurodegeneration. Int J Neurosci 2011; 121:229-36. [DOI: 10.3109/00207454.2010.551432] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
44
|
Toluene effects on oxidative stress in brain regions of young-adult, middle-age, and senescent Brown Norway rats. Toxicol Appl Pharmacol 2011; 256:386-98. [PMID: 21549141 DOI: 10.1016/j.taap.2011.04.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/12/2011] [Accepted: 04/17/2011] [Indexed: 01/24/2023]
Abstract
The influence of aging on susceptibility to environmental contaminants is not well understood. To extend knowledge in this area, we examined effects in rat brain of the volatile organic compound, toluene. The objective was to test whether oxidative stress (OS) plays a role in the adverse effects caused by toluene exposure, and if so, if effects are age-dependent. OS parameters were selected to measure the production of reactive oxygen species (NADPH Quinone oxidoreductase 1 (NQO1), NADH Ubiquinone reductase (UBIQ-RD)), antioxidant homeostasis (total antioxidant substances (TAS), superoxide dismutase (SOD), γ-glutamylcysteine synthetase (γ-GCS), glutathione transferase (GST), glutathione peroxidase (GPX), glutathione reductase (GRD)), and oxidative damage (total aconitase and protein carbonyls). In this study, Brown Norway rats (4, 12, and 24 months) were dosed orally with toluene (0, 0.65 or 1g/kg) in corn oil. Four hours later, frontal cortex, cerebellum, striatum, and hippocampus were dissected, quick frozen on dry ice, and stored at -80°C until analysis. Some parameters of OS were found to increase with age in select brain regions. Toluene exposure also resulted in increased OS in select brain regions. For example, an increase in NQO1 activity was seen in frontal cortex and cerebellum of 4 and 12 month old rats following toluene exposure, but only in the hippocampus of 24 month old rats. Similarly, age and toluene effects on glutathione enzymes were varied and brain-region specific. Markers of oxidative damage reflected changes in oxidative stress. Total aconitase activity was increased by toluene in frontal cortex and cerebellum at 12 and 24 months, respectively. Protein carbonyls in both brain regions and in all age groups were increased by toluene, but step-down analyses indicated toluene effects were statistically significant only in 12month old rats. These results indicate changes in OS parameters with age and toluene exposure resulted in oxidative damage in frontal cortex and cerebellum of 12 month old rats. Although increases in oxidative damage are associated with increases in horizontal motor activity in older rats, further research is warranted to determine if these changes in OS parameters are related to neurobehavioral and neurophysiological effects of toluene in animal models of aging.
Collapse
|
45
|
Huang Q, Aluise CD, Joshi G, Sultana R, St Clair DK, Markesbery WR, Butterfield DA. Potential in vivo amelioration by N-acetyl-L-cysteine of oxidative stress in brain in human double mutant APP/PS-1 knock-in mice: toward therapeutic modulation of mild cognitive impairment. J Neurosci Res 2011; 88:2618-29. [PMID: 20648652 DOI: 10.1002/jnr.22422] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia among the elderly. Although the underlying cause has yet to be established, numerous data have shown that oxidative stress is implicated in AD as well as in preclinical stages of AD, such as mild cognitive impairment (MCI). The oxidative stress observed in brains of subjects with AD and MCI may be due, either fully or in part, to increased free radicals mediated by amyloid-beta peptide (Abeta). By using double human mutant APP/PS-1 knock-in mice as the AD model, the present work demonstrates that the APP/PS-1 double mutation results in elevated protein oxidation (as indexed by protein carbonyls), protein nitration (as indexed by 3-nitrotyrosine), as well as lipid peroxidation (as indexed by protein-bound 4-hydroxy-2-nonenal) in brains of mice aged 9 months and 12 months. APP/PS-1 mice also exhibited lower levels of brain glutathione peroxidase (GPx) in both age groups studied, whereas glutathione reductase (GR) levels in brain were unaffected by the mutation. The activities of both of these antioxidant enzymes were significantly decreased in APP/PS-1 mouse brains, whereas the activity of glucose-6-phosphate dehydrogenase (G6PDH) was increased relative to controls in both age groups. Levels of peptidyl prolyl isomerase 1 (Pin1) were significantly decreased in APP/PS-1 mouse brain aged 9 and 12 months. Administration of N-acetyl-L-cysteine (NAC), a glutathione precursor, to APP/PS-1 mice via drinking water suppressed increased protein oxidation and nitration and also significantly augmented levels and activity of GPx in brain from both age groups. Oral administration of NAC also increased the diminished activity of GR and protected against lipid peroxidation in brains of 9-month-old APP/PS-1 mice only. Pin1 levels, GR levels, and G6PDH activity in brain were unaffected by oral administration of NAC in both age groups. These results are discussed with reference to the therapeutic potential of this brain-accessible glutathione precursor in the treatment of MCI and AD.
Collapse
Affiliation(s)
- Quanzhen Huang
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
The plasma membrane redox system in Alzheimer's disease. Exp Neurol 2011; 228:9-14. [DOI: 10.1016/j.expneurol.2010.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 11/16/2010] [Accepted: 12/09/2010] [Indexed: 01/05/2023]
|
47
|
Differential expression and redox proteomics analyses of an Alzheimer disease transgenic mouse model: effects of the amyloid-β peptide of amyloid precursor protein. Neuroscience 2011; 177:207-22. [PMID: 21223993 DOI: 10.1016/j.neuroscience.2011.01.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 12/06/2010] [Accepted: 01/03/2011] [Indexed: 11/24/2022]
Abstract
Among the pathological factors known to be associated with Alzheimer disease (AD), oxidative stress induced by the amyloid-β peptide (Aβ) has been demonstrated to play a key role in human brain and animal models of AD. Recently, we reported elevated levels of oxidative damage in the brain of a transgenic (Tg) AD mouse model with Swedish and Indiana familial AD mutations in human amyloid precursor protein (APP) [PDAPP mice, line J20], as evidenced by increased levels of protein carbonyls, 3-nitrotyrosine, and protein-bound 4-hydroxy-2-nonenal. This oxidative damage was dependent on the methionine 35 residue within the Aβ peptide. Further insight into the molecular pathways affected in this Tg model of AD may be gained with discovery-based proteomics studies; therefore, two-dimensional gel-based expression proteomics was performed to compare differences in brain protein levels of J20 Tg mice with non-transgenic (NTg) littermate controls. Based on our studies, we identified six proteins that had significantly increased levels in J20 Tg relative to NTg mice: calcineurin subunit B type 1, ρ GDP-dissociation inhibitor 1, T-complex protein 1 subunit α A, α-enolase, peptidyl-prolyl cis-trans isomerase (Pin-1), and ATP synthase subunit α mitochondrial. Several of these proteins have previously been implicated in in vitro and in vivo models and subjects with AD. Additionally, using redox proteomics analyses we identified two oxidatively-modified proteins: phosphatidylethanolamine-binding protein 1 and Pin-1 with decreased levels of protein 3-nitrotyrosine in J20 Tg mice relative to NTg. Western blotting and immunoprecipitation analyses were used to validate proteomics results. Overall, these studies provide information about changes in the brain proteome as a result of Aβ deposition and clues with which to further direct studies on elucidating AD pathogenesis.
Collapse
|
48
|
Lee YJ, Choi IS, Park MH, Lee YM, Song JK, Kim YH, Kim KH, Hwang DY, Jeong JH, Yun YP, Oh KW, Jung JK, Han SB, Hong JT. 4-O-Methylhonokiol attenuates memory impairment in presenilin 2 mutant mice through reduction of oxidative damage and inactivation of astrocytes and the ERK pathway. Free Radic Biol Med 2011; 50:66-77. [PMID: 20974250 DOI: 10.1016/j.freeradbiomed.2010.10.698] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 09/10/2010] [Accepted: 10/15/2010] [Indexed: 12/26/2022]
Abstract
Presenilin 2 (PS2) mutation increases Aβ generation and neuronal cell death in the brains of Alzheimer disease (AD) patients. In a previous study, we showed that increased oxidative damage and activation of extracellular signal-regulated kinase (ERK) were associated with Aβ generation and neuronal cell death in neuronal cells expressing mutant PS2. In this study, we show that oral treatment with 4-O-methylhonokiol, a novel compound isolated from Magnolia officinalis, for 3 months (1.0mg/kg) prevented PS2 mutation-induced memory impairment and neuronal cell death accompanied by a reduction in Aβ(1-42) accumulation. We also found that 4-O-methylhonokiol inhibited PS2 mutation-induced activation of ERK and β-secretase, and oxidative protein and lipid damage, but recovered glutathione levels in the cortex and hippocampus of PS2 mutant mice. Additionally, 4-O-methylhonokiol prevented PS2 mutation-induced activation of astrocytes as well as production of TNF-α, IL-1β, reactive oxygen species (ROS), and nitric oxide (NO) in neurons. Generation of TNF-α, IL-1β, ROS, and NO and ERK activation in cultured astrocytes treated with lipopolysaccharide (1μg/ml) were also prevented by 4-O-methylhonokiol in a dose-dependent manner. These results suggest that the improving effects of 4-O-methylhonokiol on memory function may be associated with a suppression of the activation of ERK and astrocytes as well as a reduction in oxidative damage. Thus, 4-O-methylhonokiol may be useful in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Young Jung Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 361-763, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Spencer WA, Jeyabalan J, Kichambre S, Gupta RC. Oxidatively generated DNA damage after Cu(II) catalysis of dopamine and related catecholamine neurotransmitters and neurotoxins: Role of reactive oxygen species. Free Radic Biol Med 2011; 50:139-47. [PMID: 21075203 PMCID: PMC3353411 DOI: 10.1016/j.freeradbiomed.2010.10.693] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 09/20/2010] [Accepted: 10/12/2010] [Indexed: 11/28/2022]
Abstract
There is increasing evidence supporting a causal role for oxidatively damaged DNA in neurodegeneration during the natural aging process and in neurodegenerative diseases such as Parkinson and Alzheimer. The presence of redox-active catecholamine neurotransmitters coupled with the localization of catalytic copper to DNA suggests a plausible role for these agents in the induction of oxidatively generated DNA damage. In this study we have investigated the role of Cu(II)-catalyzed oxidation of several catecholamine neurotransmitters and related neurotoxins in inducing oxidatively generated DNA damage. Autoxidation of all catechol neurotransmitters and related congeners tested resulted in the formation of nearly a dozen oxidation DNA products resulting in a decomposition pattern that was essentially identical for all agents tested. The presence of Cu(II), and to a lesser extent Fe(III), had no effect on the decomposition pattern but substantially enhanced the DNA product levels by up to 75-fold, with dopamine producing the highest levels of unidentified oxidation DNA products (383±46 adducts/10(6) nucleotides), nearly 3-fold greater than 8-oxo-7,8-dihydro-2'-deoxyguanosine (122±19 adducts/10(6) nucleotides) under the same conditions. The addition of sodium azide, 2,2,6,6-tetramethyl-4-piperidone, tiron, catalase, bathocuproine, or methional to the dopamine/Cu(II) reaction mixture resulted in a substantial decrease (>90%) in oxidation DNA product levels, indicating a role for singlet oxygen, superoxide, H(2)O(2), Cu(I), and Cu(I)OOH in their formation. Whereas the addition of N-tert-butyl-α-phenylnitrone significantly decreased (67%) dopamine-mediated oxidatively damaged DNA, three other hydroxyl radical scavengers, ascorbic acid, sodium benzoate, and mannitol, had little to no effect on these oxidation DNA product levels, suggesting that free hydroxyl radicals may have limited involvement in this dopamine/Cu(II)-mediated oxidatively generated DNA damage. These studies suggest a possible contributory role of oxidatively generated DNA damage by dopamine and related catechol neurotransmitters/neurotoxins in neurodegeneration and cell death. We also found that a naturally occurring broad-spectrum antioxidant, ellagic acid, was substantially effective (nearly 50% inhibition) at low doses (1μM) at preventing this dopamine/Cu(II)-mediated oxidatively generated DNA damage. Because dietary ellagic acid has been found to reduce oxidative stress in rat brains, a neuroprotective role of this polyphenol is plausible.
Collapse
Affiliation(s)
- Wendy A. Spencer
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202
| | | | | | - Ramesh C. Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
- Correspondence to Dr. Ramesh Gupta, 304 E Delia Baxter II, 580 Preston Street, University of Louisville Medical School, Louisville, KY 40202. Telephone: 502-852-3682;
| |
Collapse
|
50
|
Camandola S, Mattson MP. Aberrant subcellular neuronal calcium regulation in aging and Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:965-73. [PMID: 20950656 DOI: 10.1016/j.bbamcr.2010.10.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/01/2010] [Accepted: 10/03/2010] [Indexed: 02/06/2023]
Abstract
In this mini-review/opinion article we describe evidence that multiple cellular and molecular alterations in Alzheimer's disease (AD) pathogenesis involve perturbed cellular calcium regulation, and that alterations in synaptic calcium handling may be early and pivotal events in the disease process. With advancing age neurons encounter increased oxidative stress and impaired energy metabolism, which compromise the function of proteins that control membrane excitability and subcellular calcium dynamics. Altered proteolytic cleavage of the β-amyloid precursor protein (APP) in response to the aging process in combination with genetic and environmental factors results in the production and accumulation of neurotoxic forms of amyloid β-peptide (Aβ). Aβ undergoes a self-aggregation process and concomitantly generates reactive oxygen species that can trigger membrane-associated oxidative stress which, in turn, impairs the functions of ion-motive ATPases and glutamate and glucose transporters thereby rendering neurons vulnerable to excitotoxicity and apoptosis. Mutations in presenilin-1 that cause early-onset AD increase Aβ production, but also result in an abnormal increase in the size of endoplasmic reticulum calcium stores. Some of the events in the neurodegenerative cascade can be counteracted in animal models by manipulations that stabilize neuronal calcium homeostasis including dietary energy restriction, agonists of glucagon-like peptide 1 receptors and drugs that activate mitochondrial potassium channels. Emerging knowledge of the actions of calcium upstream and downstream of Aβ provides opportunities to develop novel preventative and therapeutic interventions for AD. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.
Collapse
Affiliation(s)
- Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | | |
Collapse
|