1
|
Sui Y, Feng X, Ma Y, Zou Y, Liu Y, Huang J, Zhu X, Wang J. BHBA attenuates endoplasmic reticulum stress-dependent neuroinflammation via the gut-brain axis in a mouse model of heat stress. CNS Neurosci Ther 2024; 30:e14840. [PMID: 38973202 PMCID: PMC11228358 DOI: 10.1111/cns.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Heat stress (HS) commonly occurs as a severe pathological response when the body's sensible temperature exceeds its thermoregulatory capacity, leading to the development of chronic brain inflammation, known as neuroinflammation. Emerging evidence suggests that HS leads to the disruption of the gut microbiota, whereas abnormalities in the gut microbiota have been demonstrated to affect neuroinflammation. However, the mechanisms underlying the effects of HS on neuroinflammation are poorly studied. Meanwhile, effective interventions have been unclear. β-Hydroxybutyric acid (BHBA) has been found to have neuroprotective and anti-inflammatory properties in previous studies. This study aims to explore the modulatory effects of BHBA on neuroinflammation induced by HS and elucidate the underlying molecular mechanisms. METHODS An in vivo and in vitro model of HS was constructed under the precondition of BHBA pretreatment. The modulatory effects of BHBA on HS-induced neuroinflammation were explored and the underlying molecular mechanisms were elucidated by flow cytometry, WB, qPCR, immunofluorescence staining, DCFH-DA fluorescent probe assay, and 16S rRNA gene sequencing of colonic contents. RESULTS Heat stress was found to cause gut microbiota disruption in HS mouse models, and TM7 and [Previotella] spp. may be the best potential biomarkers for assessing the occurrence of HS. Fecal microbiota transplantation associated with BHBA effectively reversed the disruption of gut microbiota in HS mice. Moreover, BHBA may inhibit microglia hyperactivation, suppress neuroinflammation (TNF-α, IL-1β, and IL-6), and reduce the expression of cortical endoplasmic reticulum stress (ERS) markers (GRP78 and CHOP) mainly through its modulatory effects on the gut microbiota (TM7, Lactobacillus spp., Ruminalococcus spp., and Prevotella spp.). In vitro experiments revealed that BHBA (1 mM) raised the expression of the ERS marker GRP78, enhanced cellular activity, and increased the generation of reactive oxygen species (ROS) and anti-inflammatory cytokines (IL-10), while also inhibiting HS-induced apoptosis, ROS production, and excessive release of inflammatory cytokines (TNF-α and IL-1β) in mouse BV2 cells. CONCLUSION β-Hydroxybutyric acid may be an effective agent for preventing neuroinflammation in HS mice, possibly due to its ability to inhibit ERS and subsequent microglia neuroinflammation via the gut-brain axis. These findings lay the groundwork for future research and development of BHBA as a preventive drug for HS and provide fresh insights into techniques for treating neurological illnesses by modifying the gut microbiota.
Collapse
Affiliation(s)
- Yuzhen Sui
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yimeng Zou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanli Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jian Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
2
|
Perumal N, Yurugi H, Dahm K, Rajalingam K, Grus FH, Pfeiffer N, Manicam C. Proteome landscape and interactome of voltage-gated potassium channel 1.6 (Kv1.6) of the murine ophthalmic artery and neuroretina. Int J Biol Macromol 2024; 257:128464. [PMID: 38043654 DOI: 10.1016/j.ijbiomac.2023.128464] [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: 08/29/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/05/2023]
Abstract
The voltage-gated potassium channel 1.6 (Kv1.6) plays a vital role in ocular neurovascular beds and exerts its modulatory functions via interaction with other proteins. However, the interactome and their potential roles remain unknown. Here, the global proteome landscape of the ophthalmic artery (OA) and neuroretina was mapped, followed by the determination of Kv1.6 interactome and validation of its functionality and cellular localization. Microfluorimetric analysis of intracellular [K+] and Western blot validated the native functionality and cellular expression of the recombinant Kv1.6 channel protein. A total of 54, 9 and 28 Kv1.6-interacting proteins were identified in the mouse OA and, retina of mouse and rat, respectively. The Kv1.6-protein partners in the OA, namely actin cytoplasmic 2, alpha-2-macroglobulin and apolipoprotein A-I, were implicated in the maintenance of blood vessel integrity by regulating integrin-mediated adhesion to extracellular matrix and Ca2+ flux. Many retinal protein interactors, particularly the ADP/ATP translocase 2 and cytoskeleton protein tubulin, were involved in endoplasmic reticulum stress response and cell viability. Three common interactors were found in all samples comprising heat shock cognate 71 kDa protein, Ig heavy constant gamma 1 and Kv1.6 channel. This foremost in-depth investigation enriched and identified the elusive Kv1.6 channel and, elucidated its complex interactome.
Collapse
Affiliation(s)
- Natarajan Perumal
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hajime Yurugi
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Katrin Dahm
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Franz H Grus
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
3
|
Huseby CJ, Delvaux E, Brokaw DL, Coleman PD. Blood RNA transcripts reveal similar and differential alterations in fundamental cellular processes in Alzheimer's disease and other neurodegenerative diseases. Alzheimers Dement 2023; 19:2618-2632. [PMID: 36541444 PMCID: PMC11633037 DOI: 10.1002/alz.12880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dysfunctional processes in Alzheimer's disease and other neurodegenerative diseases lead to neural degeneration in the central and peripheral nervous system. Research demonstrates that neurodegeneration of any kind is a systemic disease that may even begin outside of the region vulnerable to the disease. Neurodegenerative diseases are defined by the vulnerabilities and pathology occurring in the regions affected. METHOD A random forest machine learning analysis on whole blood transcriptomes from six neurodegenerative diseases generated unbiased disease-classifying RNA transcripts subsequently subjected to pathway analysis. RESULTS We report that transcripts of the blood transcriptome selected for each of the neurodegenerative diseases represent fundamental biological cell processes including transcription regulation, degranulation, immune response, protein synthesis, apoptosis, cytoskeletal components, ubiquitylation/proteasome, and mitochondrial complexes that are also affected in the brain and reveal common themes across six neurodegenerative diseases. CONCLUSION Neurodegenerative diseases share common dysfunctions in fundamental cellular processes. Identifying regional vulnerabilities will reveal unique disease mechanisms. HIGHLIGHTS Transcriptomics offer information about dysfunctional processes. Comparing multiple diseases will expose unique malfunctions within diseases. Blood RNA can be used ante mortem to track expression changes in neurodegenerative diseases. Protocol standardization will make public datasets compatible.
Collapse
Affiliation(s)
- Carol J. Huseby
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Elaine Delvaux
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Danielle L. Brokaw
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paul D. Coleman
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
4
|
Lim D, Tapella L, Dematteis G, Genazzani AA, Corazzari M, Verkhratsky A. The endoplasmic reticulum stress and unfolded protein response in Alzheimer's disease: a calcium dyshomeostasis perspective. Ageing Res Rev 2023; 87:101914. [PMID: 36948230 DOI: 10.1016/j.arr.2023.101914] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
Protein misfolding is prominent in early cellular pathology of Alzheimer's disease (AD), implicating pathophysiological significance of endoplasmic reticulum stress/unfolded protein response (ER stress/UPR) and highlighting it as a target for drug development. Experimental data from animal AD models and observations on human specimens are, however, inconsistent. ER stress and associated UPR are readily observed in in vitro AD cellular models and in some AD model animals. In the human brain, components and markers of ER stress as well as UPR transducers are observed at Braak stages III-VI associated with severe neuropathology and neuronal death. The picture, however, is further complicated by the brain region- and cell type-specificity of the AD-related pathology. Terms 'disturbed' or 'non-canonical' ER stress/UPR were used to describe the discrepancies between experimental data and the classic ER stress/UPR cascade. Here we discuss possible 'disturbing' or 'interfering' factors which may modify ER stress/UPR in the early AD pathogenesis. We focus on the dysregulation of the ER Ca2+ homeostasis, store-operated Ca2+ entry, and the interaction between the ER and mitochondria. We suggest that a detailed study of the CNS cell type-specific alterations of Ca2+ homeostasis in early AD may deepen our understanding of AD-related dysproteostasis.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy.
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Marco Corazzari
- Department of Health Science (DSS), Center for Translational Research on Autoimmune and Allergic Disease (CAAD) & Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale "Amedeo Avogadro"
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain & Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| |
Collapse
|
5
|
Yoo YM, Joo SS. Melatonin Can Modulate Neurodegenerative Diseases by Regulating Endoplasmic Reticulum Stress. Int J Mol Sci 2023; 24:ijms24032381. [PMID: 36768703 PMCID: PMC9916953 DOI: 10.3390/ijms24032381] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
As people age, their risks of developing degenerative diseases such as cancer, diabetes, Parkinson's Disease (PD), Alzheimer's Disease (AD), rheumatoid arthritis, and osteoporosis are generally increasing. Millions of people worldwide suffer from these diseases as they age. In most countries, neurodegenerative diseases are generally recognized as the number one cause afflicting the elderly. Endoplasmic reticulum (ER) stress has been suggested to be associated with some human neurological diseases, such as PD and AD. Melatonin, a neuroendocrine hormone mainly synthesized in the pineal gland, is involved in pleiotropically biological functions, including the control of the circadian rhythm, immune enhancement, and antioxidant, anti-aging, and anti-tumor effects. Although there are many papers on the prevention or suppression of diseases by melatonin, there are very few papers about the effects of melatonin on ER stress in neurons and neurodegenerative diseases. This paper aims to summarize and present the effects of melatonin reported so far, focusing on its effects on neurons and neurodegenerative diseases related to ER stress. Studies have shown that the primary target molecule of ER stress for melatonin is CHOP, and PERK and GRP78/BiP are the secondary target molecules. Therefore, melatonin is crucial in protecting neurons and treating neurodegeneration against ER stress.
Collapse
Affiliation(s)
- Yeong-Min Yoo
- East Coast Life Sciences Institute, College of Life Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
- Correspondence: (Y.-M.Y.); (S.S.J.); Tel.: +82-10-2494-5309 (Y.-M.Y.); +82-33-640-2856 (S.S.J.); Fax: +82-33-640-2849 (Y.-M.Y. & S.S.J.)
| | - Seong Soo Joo
- Department of Marine Bioscience, College of Life Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
- Correspondence: (Y.-M.Y.); (S.S.J.); Tel.: +82-10-2494-5309 (Y.-M.Y.); +82-33-640-2856 (S.S.J.); Fax: +82-33-640-2849 (Y.-M.Y. & S.S.J.)
| |
Collapse
|
6
|
Ryan KC, Laboy JT, Norman KR. Deregulation of Mitochondrial Calcium Handling Due to Presenilin Loss Disrupts Redox Homeostasis and Promotes Neuronal Dysfunction. Antioxidants (Basel) 2022; 11:antiox11091642. [PMID: 36139715 PMCID: PMC9495597 DOI: 10.3390/antiox11091642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dysfunction and oxidative stress are major contributors to the pathophysiology of neurodegenerative diseases, including Alzheimer’s disease (AD). However, the mechanisms driving mitochondrial dysfunction and oxidative stress are unclear. Familial AD (fAD) is an early onset form of AD caused primarily by mutations in the presenilin-encoding genes. Previously, using Caenorhabditis elegans as a model system to study presenilin function, we found that loss of C. elegans presenilin orthologue SEL-12 results in elevated mitochondrial and cytosolic calcium levels. Here, we provide evidence that elevated neuronal mitochondrial generated reactive oxygen species (ROS) and subsequent neurodegeneration in sel-12 mutants are a consequence of the increase of mitochondrial calcium levels and not cytosolic calcium levels. We also identify mTORC1 signaling as a critical factor in sustaining high ROS in sel-12 mutants in part through its repression of the ROS scavenging system SKN-1/Nrf. Our study reveals that SEL-12/presenilin loss disrupts neuronal ROS homeostasis by increasing mitochondrial ROS generation and elevating mTORC1 signaling, which exacerbates this imbalance by suppressing SKN-1/Nrf antioxidant activity.
Collapse
|
7
|
Kou JJ, Shi JZ, He YY, Hao JJ, Zhang HY, Luo DM, Song JK, Yan Y, Xie XM, Du GH, Pang XB. Luteolin alleviates cognitive impairment in Alzheimer's disease mouse model via inhibiting endoplasmic reticulum stress-dependent neuroinflammation. Acta Pharmacol Sin 2022; 43:840-849. [PMID: 34267346 DOI: 10.1038/s41401-021-00702-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
Luteolin is a flavonoid in a variety of fruits, vegetables, and herbs, which has shown anti-inflammatory, antioxidant, and anti-cancer neuroprotective activities. In this study, we investigated the potential beneficial effects of luteolin on memory deficits and neuroinflammation in a triple-transgenic mouse model of Alzheimer's disease (AD) (3 × Tg-AD). The mice were treated with luteolin (20, 40 mg · kg-1 · d-1, ip) for 3 weeks. We showed that luteolin treatment dose-dependently improved spatial learning, ameliorated memory deficits in 3 × Tg-AD mice, accompanied by inhibiting astrocyte overactivation (GFAP) and neuroinflammation (TNF-α, IL-1β, IL-6, NO, COX-2, and iNOS protein), and decreasing the expression of endoplasmic reticulum (ER) stress markers GRP78 and IRE1α in brain tissues. In rat C6 glioma cells, treatment with luteolin (1, 10 µM) dose-dependently inhibited LPS-induced cell proliferation, excessive release of inflammatory cytokines, and increase of ER stress marker GRP78. In conclusion, luteolin is an effective agent in the treatment of learning and memory deficits in 3 × Tg-AD mice, which may be attributable to the inhibition of ER stress in astrocytes and subsequent neuroinflammation. These results provide the experimental basis for further research and development of luteolin as a therapeutic agent for AD.
Collapse
|
8
|
Jiang H, Jayadev S, Lardelli M, Newman M. A Review of the Familial Alzheimer's Disease Locus PRESENILIN 2 and Its Relationship to PRESENILIN 1. J Alzheimers Dis 2019; 66:1323-1339. [PMID: 30412492 DOI: 10.3233/jad-180656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PRESENILIN 1 (PSEN1) and PRESENILIN 2 (PSEN2) genes are loci for mutations causing familial Alzheimer's disease (fAD). However, the function of these genes and how they contribute to fAD pathogenesis has not been fully determined. This review provides a summary of the overlapping and independent functions of the PRESENILINS with a focus on the lesser studied PSEN2. As a core component of the γ-secretase complex, the PSEN2 protein is involved in many γ-secretase-related physiological activities, including innate immunity, Notch signaling, autophagy, and mitochondrial function. These physiological activities have all been associated with AD progression, indicating that PSEN2 plays a particular role in AD pathogenesis.
Collapse
Affiliation(s)
- Haowei Jiang
- Alzheimer's Disease Genetics Laboratory, Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Morgan Newman
- Alzheimer's Disease Genetics Laboratory, Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
9
|
Abstract
Dementia is a complex clinical syndrome characterised by progressive decline in cognitive function. It usually presents itself as impairment in memory, loss of judgement, abstract thinking and other disturbances that are severe enough to interfere with activities of daily living. It has long been considered as one of the major challenges at present posing an ever-increasing demand on global health and social care systems. Of all the different forms of dementia, Alzheimer's disease (AD) is the most common. The term non-coding RNA (ncRNA) refers to RNA sequences which do not have the ability to be translated into proteins and therefore mainly fall within the realm of the recently acknowledged ‘dark matter’ of the genome. This genomic dark matter encompasses a whole spectrum of differing ncRNA families such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), PIWI-interacting RNAs (piRNAs), transfer RNAs (tRNAs), small nuclear RNAs (snoRNAs) and circular RNAs (circRNAs), to name but a few. Consequently, due to the widespread influences of miRNAs and lncRNAs across all disease pathways, it is of critical importance for researchers in the field of dementia to focus their attention on possible ncRNA-induced pathogeneses, with the ultimate goal of identifying novel diagnostic procedures and drug targets, together with the development of novel therapies to control such a devastating mental condition in the patient population.
Collapse
|
10
|
Presenilins as Drug Targets for Alzheimer's Disease-Recent Insights from Cell Biology and Electrophysiology as Novel Opportunities in Drug Development. Int J Mol Sci 2018; 19:ijms19061621. [PMID: 29857474 PMCID: PMC6032171 DOI: 10.3390/ijms19061621] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/26/2018] [Accepted: 05/28/2018] [Indexed: 01/24/2023] Open
Abstract
A major cause underlying familial Alzheimer's disease (AD) are mutations in presenilin proteins, presenilin 1 (PS1) and presenilin 2 (PS2). Presenilins are components of the γ-secretase complex which, when mutated, can affect amyloid precursor protein (APP) processing to toxic forms of amyloid beta (Aβ). Consequently, presenilins have been the target of numerous and varied research efforts to develop therapeutic strategies for AD. The presenilin 1 gene harbors the largest number of AD-causing mutations resulting in the late onset familial form of AD. As a result, the majority of efforts for drug development focused on PS1 and Aβ. Soon after the discovery of the major involvement of PS1 and PS2 in γ-secretase activity, it became clear that neuronal signaling, particularly calcium ion (Ca2+) signaling, is regulated by presenilins and impacted by mutations in presenilin genes. Intracellular Ca2+ signaling not only controls the activity of neurons, but also gene expression patterns, structural functionality of the cytoskeleton, synaptic connectivity and viability. Here, we will briefly review the role of presenilins in γ-secretase activity, then focus on the regulation of Ca2+ signaling, oxidative stress, and cellular viability by presenilins within the context of AD and discuss the relevance of presenilins in AD drug development efforts.
Collapse
|
11
|
Abstract
The clinical manifestation of neurodegenerative diseases is initiated by the selective alteration in the functionality of distinct neuronal populations. The pathology of many neurodegenerative diseases includes accumulation of misfolded proteins in the brain. In physiological conditions, the proteostasis network maintains normal protein folding, trafficking and degradation; alterations in this network - particularly disturbances to the function of endoplasmic reticulum (ER) - are thought to contribute to abnormal protein aggregation. ER stress triggers a signalling reaction known as the unfolded protein response (UPR), which induces adaptive programmes that improve protein folding and promote quality control mechanisms and degradative pathways or can activate apoptosis when damage is irreversible. In this Review, we discuss the latest advances in defining the functional contribution of ER stress to brain diseases, including novel evidence that relates the UPR to synaptic function, which has implications for cognition and memory. A complex concept is emerging wherein the consequences of ER stress can differ drastically depending on the disease context and the UPR signalling pathway that is altered. Strategies to target specific components of the UPR using small molecules and gene therapy are in development, and promise interesting avenues for future interventions to delay or stop neurodegeneration.
Collapse
|
12
|
Frazier HN, Maimaiti S, Anderson KL, Brewer LD, Gant JC, Porter NM, Thibault O. Calcium's role as nuanced modulator of cellular physiology in the brain. Biochem Biophys Res Commun 2016; 483:981-987. [PMID: 27553276 DOI: 10.1016/j.bbrc.2016.08.105] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/04/2016] [Accepted: 08/18/2016] [Indexed: 12/22/2022]
Abstract
Neuroscientists studying normal brain aging, spinal cord injury, Alzheimer's disease (AD) and other neurodegenerative diseases have focused considerable effort on carefully characterizing intracellular perturbations in calcium dynamics or levels. At the cellular level, calcium is known for controlling life and death and orchestrating most events in between. For many years, intracellular calcium has been recognized as an essential ion associated with nearly all cellular functions from cell growth to degeneration. Often the emphasis is on the negative impact of calcium dysregulation and the typical worse-case-scenario leading inevitably to cell death. However, even high amplitude calcium transients, when executed acutely, can alter neuronal communication and synaptic strength in positive ways, without necessarily killing neurons. Here, we focus on the evidence that calcium has a subtle and distinctive role in shaping and controlling synaptic events that underpin neuronal communication and that these subtle changes in aging or AD may contribute to cognitive decline. We emphasize that calcium imaging in dendritic components is ultimately necessary to directly test for the presence of age- or disease-associated alterations during periods of synaptic activation.
Collapse
Affiliation(s)
- Hilaree N Frazier
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Shaniya Maimaiti
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Katie L Anderson
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Lawrence D Brewer
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - John C Gant
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Nada M Porter
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA
| | - Olivier Thibault
- UKMC, MS-313, Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536, USA.
| |
Collapse
|
13
|
Gama Sosa MA, De Gasperi R, Hof PR, Elder GA. Fibroblast growth factor rescues brain endothelial cells lacking presenilin 1 from apoptotic cell death following serum starvation. Sci Rep 2016; 6:30267. [PMID: 27443835 PMCID: PMC4957214 DOI: 10.1038/srep30267] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/20/2016] [Indexed: 12/05/2022] Open
Abstract
Presenilin 1 (Psen1) is important for vascular brain development and is known to influence cellular stress responses. To understand the role of Psen1 in endothelial stress responses, we investigated the effects of serum withdrawal on wild type (wt) and Psen1−/− embryonic brain endothelial cells. Serum starvation induced apoptosis in Psen1−/− cells but did not affect wt cells. PI3K/AKT signaling was reduced in serum-starved Psen1−/− cells, and this was associated with elevated levels of phospho-p38 consistent with decreased pro-survival AKT signaling in the absence of Psen1. Fibroblast growth factor (FGF1 and FGF2), but not vascular endothelial growth factor (VEGF) rescued Psen1−/− cells from serum starvation induced apoptosis. Inhibition of FGF signaling induced apoptosis in wt cells under serum withdrawal, while blocking γ-secretase activity had no effect. In the absence of serum, FGF2 immunoreactivity was distributed diffusely in cytoplasmic and nuclear vesicles of wt and Psen1−/− cells, as levels of FGF2 in nuclear and cytosolic fractions were not significantly different. Thus, sensitivity of Psen1−/− cells to serum starvation is not due to lack of FGF synthesis but likely to effects of Psen1 on FGF release onto the cell surface and impaired activation of the PI3K/AKT survival pathway.
Collapse
Affiliation(s)
- Miguel A Gama Sosa
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Patrick R Hof
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gregory A Elder
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
14
|
Liang W, Zhao X, Feng J, Song F, Pan Y. Ursolic acid attenuates beta-amyloid-induced memory impairment in mice. ARQUIVOS DE NEURO-PSIQUIATRIA 2016; 74:482-8. [PMID: 27332074 DOI: 10.1590/0004-282x20160065] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/26/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Increasing evidence demonstrates that oxidative stress and inflammatory are involved in amyloid β (Aβ)-induced memory impairments. Ursolic acid (UA), a triterpenoid compound, has potent anti-inflammatory and antioxidant activities. However, it remains unclear whether UA attenuates Aβ-induced neurotoxicity. METHOD The aggregated Aβ25-35 was intracerebroventricularly administered to mice. RESULTS We found that UA significantly reversed the Aβ25-35-induced learning and memory deficits. Our results indicated that one of the potential mechanisms of the neuroprotective effect was attenuating the Aβ25-35-induced accumulation of malondialdehyde (MDA) and depletion of glutathione (GSH) in the hippocampus. Furthermore, UA significantly suppressed the upregulation of IL-1β, IL-6, and tumor necrosis-α factor levels in the hippocampus of Aβ25-35-treated mice. CONCLUSION These findings suggest that UA prevents memory impairment through amelioration of oxidative stress, inflammatory response and may offer a novel therapeutic strategy for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Wenna Liang
- The Third People's Hospital, Liaocheng, Department of Neurology, Shandong , China, The Third People's Hospital of Liaocheng, Department of Neurology, Shandong, China
| | - Xiaoyang Zhao
- The Third People's Hospital, Liaocheng, Department of Neurology, Shandong , China, The Third People's Hospital of Liaocheng, Department of Neurology, Shandong, China
| | - Jinping Feng
- The Third People's Hospital, Liaocheng, Department of Neurology, Shandong , China, The Third People's Hospital of Liaocheng, Department of Neurology, Shandong, China
| | - Fenghua Song
- The Third People's Hospital, Liaocheng, Department of Pharmacy, Shandong , China, The Third People's Hospital of Liaocheng, Department of Pharmacy, Shandong, China
| | - Yunzhi Pan
- The Third Affiliated Hospital, Qiqihar Medical University, Department of Neurology, Heilong Jiang , China, The Third Affiliated Hospital of Qiqihar Medical University, Department of Neurology, Heilong Jiang, China
| |
Collapse
|
15
|
Abstract
Alzheimer disease (AD) is a fatal progressive disease and the most common form of dementia without effective treatments. Previous studies support that the disruption of endoplasmic reticulum Ca through overactivation of ryanodine receptors plays an important role in the pathogenesis of AD. Normalization of intracellular Ca homeostasis could be an effective strategy for AD therapies. Dantrolene, an antagonist of ryanodine receptors and an FDA-approved drug for clinical treatment of malignant hyperthermia and muscle spasms, exhibits neuroprotective effects in multiple models of neurodegenerative disorders. Recent preclinical studies consistently support the therapeutic effects of dantrolene in various types of AD animal models and were summarized in the current review.
Collapse
|
16
|
Yang F, Luo J. Endoplasmic Reticulum Stress and Ethanol Neurotoxicity. Biomolecules 2015; 5:2538-53. [PMID: 26473940 PMCID: PMC4693246 DOI: 10.3390/biom5042538] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/02/2015] [Accepted: 09/21/2015] [Indexed: 12/21/2022] Open
Abstract
Ethanol abuse affects virtually all organ systems and the central nervous system (CNS) is particularly vulnerable to excessive ethanol exposure. Ethanol exposure causes profound damages to both the adult and developing brain. Prenatal ethanol exposure induces fetal alcohol spectrum disorders (FASD) which is associated with mental retardation and other behavioral deficits. A number of potential mechanisms have been proposed for ethanol-induced brain damage; these include the promotion of neuroinflammation, interference with signaling by neurotrophic factors, induction of oxidative stress, modulation of retinoid acid signaling, and thiamine deficiency. The endoplasmic reticulum (ER) regulates posttranslational protein processing and transport. The accumulation of unfolded or misfolded proteins in the ER lumen triggers ER stress and induces unfolded protein response (UPR) which are mediated by three transmembrane ER signaling proteins: pancreatic endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1), and activating transcription factor 6 (ATF6). UPR is initiated to protect cells from overwhelming ER protein loading. However, sustained ER stress may result in cell death. ER stress has been implied in various CNS injuries, including brain ischemia, traumatic brain injury, and aging-associated neurodegeneration, such as Alzheimer's disease (AD), Huntington's disease (HD), Amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD). However, effects of ethanol on ER stress in the CNS receive less attention. In this review, we discuss recent progress in the study of ER stress in ethanol-induced neurotoxicity. We also examine the potential mechanisms underlying ethanol-mediated ER stress and the interaction among ER stress, oxidative stress and autophagy in the context of ethanol neurotoxicity.
Collapse
Affiliation(s)
- Fanmuyi Yang
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 132 Health Sciences Research Building, 1095 Veterans Drive, Lexington, KY 40536, USA.
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 132 Health Sciences Research Building, 1095 Veterans Drive, Lexington, KY 40536, USA.
| |
Collapse
|
17
|
Dong Z, Yan L, Huang G, Zhang L, Mei B, Meng B. Ibuprofen partially attenuates neurodegenerative symptoms in presenilin conditional double-knockout mice. Neuroscience 2014; 270:58-68. [PMID: 24699228 DOI: 10.1016/j.neuroscience.2014.03.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 03/06/2014] [Accepted: 03/23/2014] [Indexed: 01/22/2023]
Abstract
Ibuprofen is a widely used nonsteroidal anti-inflammatory drug that reportedly reduces the risk of Alzheimer's disease (AD) development. The anti-inflammatory effect of ibuprofen occurred via inhibition of cyclooxygenases and anti-amyloidogenesis through modulation of γ-secretase. Presenilin 1 and 2 conditional double-knockout (cDKO) mice exhibited age-dependent memory impairment and forebrain degeneration without elevation of amyloid β deposition. Therefore, cDKO mice can be an ideal animal model on which to independently test the effects of ibuprofen anti-inflammatory properties on the prevention of AD. Three- and six-month-old cDKO mice were fed diet containing 375 ppm ibuprofen for six months. After multiple, well-validated behavioral tests, treatment with ibuprofen improved cognition-related behavioral performance, and drug efficacy was correlated with the timing of administration. Ibuprofen was more effective on six-month-old than on three-month-old cDKO mice. Biochemical analysis demonstrated that the effects of ibuprofen on glial fibrillary acidic protein and CD68 expression levels were uneven in different brain regions of cDKO mice and that age also influenced such effects. Tau hyperphosphorylation and the cleavage of caspase-3 decreased after ibuprofen treatment, and this effect was more significant in the older than the younger group of mice, which was consistent with the results of behavioral tests.
Collapse
Affiliation(s)
- Z Dong
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - L Yan
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - G Huang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - L Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - B Mei
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - B Meng
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
18
|
Gant JC, Blalock EM, Chen KC, Kadish I, Porter NM, Norris CM, Thibault O, Landfield PW. FK506-binding protein 1b/12.6: a key to aging-related hippocampal Ca2+ dysregulation? Eur J Pharmacol 2013; 739:74-82. [PMID: 24291098 DOI: 10.1016/j.ejphar.2013.10.070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/25/2022]
Abstract
It has been recognized for some time that the Ca(2+)-dependent slow afterhyperpolarization (sAHP) is larger in hippocampal neurons of aged compared with young animals. In addition, extensive studies since have shown that other Ca(2+)-mediated electrophysiological responses are increased in hippocampus with aging, including Ca(2+) transients, L-type voltage-gated Ca(2+) channel activity, Ca(2+) spike duration and action potential accommodation. Elevated Ca(2+)-induced Ca(2+) release from ryanodine receptors (RyRs) appears to drive amplification of the Ca(2+) responses. Components of this Ca(2+) dysregulation phenotype correlate with deficits in cognitive function and plasticity, indicating they may play critical roles in aging-related impairment of brain function. However, the molecular mechanisms underlying aging-related Ca(2+) dysregulation are not well understood. FK506-binding proteins 1a and 1b (FKBP1a/1b, also known as FKBP12/12.6) are immunophilin proteins that bind the immunosuppressant drugs FK506 and rapamycin. In muscle cells, FKBP1a/1b also bind RyRs and inhibits Ca(2+)-induced Ca(2+) release, but it is not clear whether FKBPs act similarly in brain cells. Recently, we found that selectively disrupting hippocampal FKBP1b function in young rats, either by microinjecting adeno-associated viral vectors expressing siRNA, or by treatment with rapamycin, increases the sAHP and recapitulates much of the hippocampal Ca(2+) dysregulation phenotype. Moreover, in microarray studies, we found FKBP1b gene expression was downregulated in hippocampus of aging rats and early-stage Alzheimer's disease subjects. These results suggest the novel hypothesis that declining FKBP function is a key factor in aging-related Ca(2+) dysregulation in the brain and point to potential new therapeutic targets for counteracting unhealthy brain aging.
Collapse
Affiliation(s)
- J C Gant
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - E M Blalock
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - K-C Chen
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - I Kadish
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - N M Porter
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - C M Norris
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - O Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States
| | - P W Landfield
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, 800 Rose St., UKMC Lexington, KY 40536, United States.
| |
Collapse
|
19
|
Cornejo VH, Hetz C. The unfolded protein response in Alzheimer’s disease. Semin Immunopathol 2013; 35:277-92. [DOI: 10.1007/s00281-013-0373-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/13/2013] [Indexed: 01/05/2023]
|
20
|
Hooghiemstra AM, Eggermont LHP, Scheltens P, van der Flier WM, Scherder EJA. Exercise and early-onset Alzheimer's disease: theoretical considerations. Dement Geriatr Cogn Dis Extra 2012; 2:132-45. [PMID: 22590474 PMCID: PMC3347875 DOI: 10.1159/000335493] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND/AIMS Although studies show a negative relationship between physical activity and the risk for cognitive impairment and late-onset Alzheimer's disease, studies concerning early-onset Alzheimer's disease (EOAD) are lacking. This review aims to justify the value of exercise interventions in EOAD by providing theoretical considerations that include neurobiological processes. METHODS A literature search on key words related to early-onset dementia, exercise, imaging, neurobiological mechanisms, and cognitive reserve was performed. RESULTS/CONCLUSION Brain regions and neurobiological processes contributing to the positive effects of exercise are affected in EOAD and, thus, provide theoretical support for exercise interventions in EOAD. Finally, we present the design of a randomized controlled trial currently being conducted in early-onset dementia patients.
Collapse
|
21
|
Nonfibrillar Abeta 1-42 inhibits glutamate uptake and phosphorylates p38 in human fibroblasts. Alzheimer Dis Assoc Disord 2011; 25:164-72. [PMID: 20921877 DOI: 10.1097/wad.0b013e3181f9860f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alzheimer disease (AD) is the most prevalent neurodegenerative disease, characterized by an increased deposition of β-amyloid (Abeta) within the central nervous system, leading to neuronal death. The availability of effective models, in which confirming novel pathogenic hypotheses and developing therapeutic targets, represents a very important goal for the field of AD. Fibroblasts from these patients may be relevant models in which addressing these issues, as they display biochemical alterations mirroring SNC ones. In this work, fibroblasts obtained from controls were studied after exposure to nonfibrillar Abeta 1-42, showing decreased glutamate uptake, similar to that observed in AD cells, in absence of transporters modifications. Nonfibrillar Abeta 1-42 was able to induce in control cells mitochondrial alterations and p38-phosphorylation, mirroring similar alterations found in AD fibroblasts. Under our experimental conditions, this treatment induced neither apoptosis nor necrosis. To investigate a putative role of p38-modulation in mediating nonfibrillar Abeta 1-42 toxicity, fibroblasts from controls were pretreated with retinoic-acid, and SB203580, a p38-inhibitor. These pretreatments prevented both p38-phosphorylation and glutamate uptake inhibition. Our results suggest that nonfibrillar Abeta 1-42 downregulates glutamate transporters activity interfering with p38-activation and mitochondrial stress. Thus, modulating complex kinase signaling pathway might represent a future therapeutic target in AD.
Collapse
|
22
|
Abstract
The calcium ion (Ca(2+)) is the main second messenger that helps to transmit depolarization status and synaptic activity to the biochemical machinery of a neuron. These features make Ca(2+) regulation a critical process in neurons, which have developed extensive and intricate Ca(2+) signaling pathways. High intensity Ca(2+) signaling necessitates high ATP consumption to restore basal (low) intracellular Ca(2+) levels after Ca(2+) influx through plasma membrane receptor and voltage-dependent ion channels. Ca(2+) influx may also lead to increased generation of mitochondrial reactive oxygen species (ROS). Impaired abilities of neurons to maintain cellular energy levels and to suppress ROS may impact Ca(2+) signaling during aging and in neurodegenerative disease processes. This review focuses on mitochondrial and endoplasmic reticulum Ca(2+) homeostasis and how they relate to synaptic Ca(2+) signaling processes, neuronal energy metabolism, and ROS generation. Also, the contribution of altered Ca(2+) signaling to neurodegeneration during aging will be considered. Advances in understanding the molecular regulation of Ca(2+) homeostasis and how it is perturbed in neurological disorders may lead to therapeutic strategies that modulate neuronal Ca(2+) signaling to enhance function and counteract disease processes.
Collapse
Affiliation(s)
- Marc Gleichmann
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, USA.
| | | |
Collapse
|
23
|
Rothenberg KG, Siedlak SL, Lee HG, Zhu X, Perry G, Smith MA. Neurodegenerative processes in Alzheimer’s disease: an overview of pathogenesis with strategic biomarker potential. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.10.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Since Alzheimer’s disease (AD) is the leading cause of senile dementia in the USA, affecting 15% of people over the age of 65 years and almost 50% of those aged over 85 years, the need for an adequate and early diagnosis as well as preventative measure against disease onset and progression is increasing. Epidemiological and molecular studies suggest that AD has multiple etiologies, including genetic mutations, genetic variations affecting susceptibility and environmental factors. All these aspects can promote the formation and the accumulation of insoluble amyloid-β and hyperphosphorylated tau. Since the disease is multifactorial and clinical diagnosis is highly exclusive, the need for a sensitive, specific and reliable biomarker for the disease is crucial. While amyloid and amyloid-related compounds may be useful biomarkers in the early diagnosis of AD, the multitude of other characteristic features of AD presented in this article may be similarly appropriate. For example, genetic mutations play a role in a subset of AD patients (often with early disease onset and more severe disease progression), and genetic analysis could thus play a role in disease diagnosis. Similarly, oxidative damage to various proteins, nucleic acids and other cellular compounds, probably arising from mitochondrial abnormalities, is found early in the disease and may provide certain biochemical signatures of disease. Ultimately, specific assays for genetic, protein and oxidative profiles and mitochondrial abnormalities, as well as those for amyloid-β and its immunological response, may serve as a relevant group of biomarkers that could be informative to individuals regarding risk of disease, as well as for indicators of the progression of disease. Correspondingly, new developments in treatment options will probably be available.
Collapse
Affiliation(s)
- Kasia Gustaw Rothenberg
- Department of Psychiatry, University Hospitals Case Medical Center, Cleveland, OH, USA
- Deptartment of Neurodegenerative Diseases, Institute of Agricultural Medicine, 2 Jaczewskiego Street, 20-095, Lublin, Poland
| | - Sandra L Siedlak
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Hyoung-gon Lee
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - George Perry
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
- Neurosciences Institute & Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Mark A Smith
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| |
Collapse
|
24
|
Bose A, Mouton-Liger F, Paquet C, Mazot P, Vigny M, Gray F, Hugon J. Modulation of tau phosphorylation by the kinase PKR: implications in Alzheimer's disease. Brain Pathol 2010; 21:189-200. [PMID: 21029237 DOI: 10.1111/j.1750-3639.2010.00437.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Double-stranded RNA dependent kinase (PKR) is a pro-apoptotic kinase that controls protein translation. Previous studies revealed that activated PKR is increased in brains with Alzheimer's disease (AD). Glycogen Synthase Kinase Aβ (GSK-3β) is responsible for tau phosphorylation and controls several cellular functions also including apoptosis. The goal of this work was to determine if PKR could concurrently trigger GSK-3β activation, tau phosphorylation and apoptosis. In AD brains, both activated kinases co-localize with phosphorylated tau in neurons. In SH-SY5Y cell cultures, tunicamycin and Aβ(1-42) activate PKR, GSK-3β and induce tau phosphorylation and all these processes are attenuated by PKR inhibitors or PKR siRNA. Our results demonstrate that neuronal PKR co-localizes with GSK-3β and tau in AD brains and is able to modulate GSK-3β activation, tau phosphorylation and apoptosis in neuroblastoma cells exposed to tunicamycin or Aβ. PKR could represent a crucial signaling point relaying stress signals to neuronal pathways leading to cellular degeneration in AD.
Collapse
Affiliation(s)
- Anindita Bose
- Inserm UMRS, Institut du Fer à Moulin, Paris, FranceDepartments of Histology Pathology The Memory Clinical Center, Lariboisière Hospital (APHP), University Paris Diderot VII, Paris, France
| | | | | | | | | | | | | |
Collapse
|
25
|
Tsujii H, Eguchi Y, Chenchik A, Mizutani T, Yamada K, Tsujimoto Y. Screening of cell death genes with a mammalian genome-wide RNAi library. J Biochem 2010; 148:157-70. [PMID: 20421362 DOI: 10.1093/jb/mvq042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We report the construction and application of a mammalian genome-wide RNAi library. The oligodeoxynucleotides encoding approximately 200,000 shRNA sequences that targeted 47,400 human transcripts were inserted into a lentivirus vector pFIV-H1-puro, and a pool of pseudovirus particles with a complexity of approximately 200,000 were used to infect target cells. From the cells surviving apoptogenic Fas stimulation, four candidate shRNA sequences were obtained that provided resistance to Fas-induced cell death, including two shRNAs for caspase-8, an shRNA for Bid, and an shRNA for Fas. The reconstructed shRNAs with these sequences were shown to reduce expression of the respective gene products and increase survival after Fas stimulation. When similar selection was performed for tunicamycin-induced apoptosis, no shRNA strongly inhibiting tunicamycin-induced cell death was isolated, although a few reconstructed shRNAs led to a slight increase of survival. Thus, this genome-wide shRNA library proved useful for selection of genes that are involved in cell death, but some limitation was also revealed.
Collapse
Affiliation(s)
- Hisayo Tsujii
- Department of Medical Genetics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Gustaw-Rothenberg K, Lerner A, Bonda DJ, Lee HG, Zhu X, Perry G, Smith MA. Biomarkers in Alzheimer's disease: past, present and future. Biomark Med 2010; 4:15-26. [PMID: 20387301 PMCID: PMC2855161 DOI: 10.2217/bmm.09.86] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Epidemiological and molecular studies suggest that Alzheimer's disease (AD) has multiple etiologies including genetic mutations, genetic variations affecting susceptibility and environmental factors. These aspects can promote the formation and accumulation of insoluble amyloid-beta and hyperphosphorylated tau. Since the disease is multifactorial and clinical diagnosis is highly exclusive, the need for a sensitive, specific and reliable biomarker is crucial. The concept of a biomarker implies sensitivity and specificity relative to the condition being considered. For clinical practice, AD diagnosis has been based on adherence to clinical criteria such as the NINCDS/ADRDA and DSM-IV. A more recent set of diagnostic criteria proposed incorporates imaging findings into the diagnosis of AD. In this article, we consider the most studied candidates or group of candidates for AD biomarkers, including pathological processes and proteins (amyloid-beta, tau, oxidative stress, mitochondrial/metabolic changes and cell-cycle processes), or autoantibodies thereto, as well as genetic factors.
Collapse
Affiliation(s)
- Katarzyna Gustaw-Rothenberg
- University Hospitals, Case Medical Center and University Memory and Cognition Center, Case Western Reserve University, Cleveland, OH, USA
- Department of Neurodegenerative Diseases, Institute of Agricultural Medicine, 2 Jaczewskiego Str, 20-095, Lublin, Poland
| | - Alan Lerner
- University Hospitals, Case Medical Center and University Memory and Cognition Center, Case Western Reserve University, Cleveland, OH, USA
| | - David J Bonda
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Hyoung-gon Lee
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - George Perry
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- UTSA Neurosciences Institute and Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Mark A Smith
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
27
|
van de Hoef DL, Hughes J, Livne-Bar I, Garza D, Konsolaki M, Boulianne GL. Identifying genes that interact with Drosophila presenilin and amyloid precursor protein. Genesis 2009; 47:246-60. [PMID: 19241393 DOI: 10.1002/dvg.20485] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The gamma-secretase complex is involved in cleaving transmembrane proteins such as Notch and one of the genes targeted in Alzheimer's disease known as amyloid precursor protein (APP). Presenilins function within the catalytic core of gamma-secretase, and mutated forms of presenilins were identified as causative factors in familial Alzheimer's disease. Recent studies show that in addition to Notch and APP, numerous signal transduction pathways are modulated by presenilins, including intracellular calcium signaling. Thus, presenilins appear to have diverse roles. To further understand presenilin function, we searched for Presenilin-interacting genes in Drosophila by performing a genetic modifier screen for enhancers and suppressors of Presenilin-dependent Notch-related phenotypes. We identified 177 modifiers, including known members of the Notch pathway and genes involved in intracellular calcium homeostasis. We further demonstrate that 53 of these modifiers genetically interacted with APP. Characterization of these genes may provide valuable insights into Presenilin function in development and disease.
Collapse
Affiliation(s)
- Diana L van de Hoef
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
28
|
Choi SJ, Jeong CH, Choi SG, Chun JY, Kim YJ, Lee J, Shin DH, Heo HJ. Zeatin prevents amyloid beta-induced neurotoxicity and scopolamine-induced cognitive deficits. J Med Food 2009; 12:271-7. [PMID: 19459726 DOI: 10.1089/jmf.2007.0678] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The antioxidative and protective effects of zeatin against amyloid beta-protein (Abeta)-induced neurotoxicity were investigated using PC12 cells. Zeatin showed antioxidative and cell protective effects against Abeta-induced neurotoxicity. In this study, we also evaluated the effect of zeatin on learning and memory capacity in vivo using ICR mice with amnesia induced by scopolamine (1 mg/kg of body weight). Zeatin, when administered to mice at 4.5 mg/kg of body weight, significantly ameliorated scopolamine-induced amnesia as measured in both the passive avoidance test and Y-maze test. Injecting mice with scopolamine impaired performance on the passive avoidance test (48 +/- 4.5% decrease) and on the Y-maze test (12 +/- 1.3% decrease). In contrast, mice treated with zeatin before scopolamine injections were protected from these changes (5-34% decrease in step-through latency; 1-4% decrease in alternation behavior). The present results suggest a possible chemopreventive role of zeatin in Alzheimer's disease.
Collapse
Affiliation(s)
- Soo Jung Choi
- Graduate School of Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Veurink G, Fuller SJ, Atwood CS, Martins RN. ReviewGenetics, lifestyle and the roles of amyloid β and oxidative stress in Alzheimer’s disease. Ann Hum Biol 2009; 30:639-67. [PMID: 14675907 DOI: 10.1080/03014460310001620144] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This paper reviews a wide range of recent studies that have linked AD-associated biochemical and physiological changes with oxidative stress and damage. Some of these changes include disruptions in metal ion homeostasis, mitochondrial damage, reduced glucose metabolism, decreased intracellular pH and inflammation. Although the changes mentioned above are associated with oxidative stress, in most cases, a cause and effect relationship is not clearcut, as many changes are interlinked. Increases in the levels of Abeta peptides, the main protein components of the cerebral amyloid deposits of AD, have been demonstrated to occur in inherited early-onset forms of AD, and as a result of certain environmental and genetic risk factors. Abeta peptides have been shown to exhibit superoxide dismutase activity, producing hydrogen peroxide which may be responsible for the neurotoxicity exhibited by this peptide in vitro. This review also discusses the biochemical aspects of oxidative stress, antioxidant defence mechanisms, and possible antioxidant therapeutic measures which may be effective in counteracting increased levels of oxidative stress. In conclusion, this review provides support for the theory that damage caused by free radicals and oxidative stress is a primary cause of the neurodegeneration seen in AD with Abeta postulated as an initiator of this process.
Collapse
Affiliation(s)
- G Veurink
- The Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Perth, Australia
| | | | | | | |
Collapse
|
30
|
Kawahara M, Negishi-Kato M, Sadakane Y. Calcium dyshomeostasis and neurotoxicity of Alzheimer's beta-amyloid protein. Expert Rev Neurother 2009; 9:681-93. [PMID: 19402778 DOI: 10.1586/ern.09.28] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Neurotoxicity of Alzheimer's beta-amyloid protein (AbetaP) is central to the pathogenesis of Alzheimer's disease (AD). Recent approaches have emphasized the importance of AbetaP oligomerization, which causes synaptic degeneration and neuronal loss, finally leading to the pathogenesis of AD. Although the precise molecular mechanism of AbetaP neurotoxicity remains elusive, our and other numerous findings have demonstrated that AbetaP directly incorporated into neuronal membranes formed calcium-permeable ion channels (amyloid channels) and resulted in an abnormal elevation of the intracellular calcium levels. The formation of amyloid channels and the abnormal increase of intracellular Ca(2+) have also been commonly observed in other neurodegenerative diseases, including conformational diseases such as prion disease or dementia with Lewy bodies. This article reviews the current understanding of the pathology of AD based on the hypothesis that the disruption of calcium homeostasis through amyloid channels may be the molecular basis of AbetaP neurotoxicity. The potential development of preventive agents is also discussed.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Analytical Chemistry, School of Pharmaceutical Sciences, Kyushu University of Health and Welfare, Nobeoka-shi, Miyazaki, Japan.
| | | | | |
Collapse
|
31
|
Varma R, Chai Y, Troncoso J, Gu J, Xing H, Stojilkovic SS, Mattson MP, Haughey NJ. Amyloid-beta induces a caspase-mediated cleavage of P2X4 to promote purinotoxicity. Neuromolecular Med 2009; 11:63-75. [PMID: 19562525 PMCID: PMC2735730 DOI: 10.1007/s12017-009-8073-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 06/16/2009] [Indexed: 11/25/2022]
Abstract
Overproduction of the beta-amyloid fragment 1-42 (A beta(1-42)) is thought to contribute to synaptic dysfunction and neuronal death in Alzheimer's disease. Mounting evidence suggests that purinergic receptors play critical roles in synaptic plasticity and neuronal survival, but the potential involvement of these receptors in A beta(1-42)-induced synaptic dysfunction and neuronal death has not been addressed. Here we report that A beta(1-42) promoted accumulation of the calcium-permeable purinergic receptor P2X4 in neurons. We also report evidence that A beta(1-42) induced a caspase-3-mediated cleavage of the receptor that slowed channel closure times and prevented agonist-induced internalization of the receptor. Molecular interference to reduce the expression of P2X4 in primary rodent neurons attenuated A beta(1-42)-induced neuronal death while induced expression of P2X4 in a neuronal cell line that does not normally express P2-receptors enhanced the toxic effect of A beta(1-42). Together these findings suggest that A beta(1-42)-induced synaptic dysfunction and neuronal death may involve perturbations in P2X4 purinergic receptors.
Collapse
Affiliation(s)
- R Varma
- Department of Neurology, Johns Hopkins, University School of Medicine, Meyer 6-109, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Lu P, Mamiya T, Lu LL, Mouri A, Zou L, Nagai T, Hiramatsu M, Ikejima T, Nabeshima T. Silibinin prevents amyloid beta peptide-induced memory impairment and oxidative stress in mice. Br J Pharmacol 2009; 157:1270-7. [PMID: 19552690 DOI: 10.1111/j.1476-5381.2009.00295.x] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Accumulated evidence suggests that oxidative stress is involved in amyloid beta (Abeta)-induced cognitive dysfunction. Silibinin (silybin), a flavonoid derived from the herb milk thistle (Silybum marianum), has been shown to have antioxidative properties; however, it remains unclear whether silibinin improves Abeta-induced neurotoxicity. In the present study, we examined the effect of silibinin on the memory impairment and accumulation of oxidative stress induced by Abeta(25-35) in mice. EXPERIMENTAL APPROACH Aggregated Abeta(25-35) (3 nmol) was intracerebroventricularly administered to mice. Treatment with silibinin (2, 20 and 200 mg.kg(-1), once a day, p.o.) was started immediately after the injection of Abeta(25-35). Locomotor activity was evaluated 6 days after the Abeta(25-35) treatment, and cognitive function was evaluated in a Y-maze and novel object recognition tests 6-11 days after the Abeta(25-35) treatment. The levels of lipid peroxidation (malondialdehyde) and antioxidant (glutathione) in the hippocampus were measured 7 days after the Abeta(25-35) injection. KEY RESULTS Silibinin prevented the memory impairment induced by Abeta(25-35) in the Y-maze and novel object recognition tests. Repeated treatment with silibinin attenuated the Abeta(25-35)-induced accumulation of malondialdehyde and depletion of glutathione in the hippocampus. CONCLUSIONS AND IMPLICATIONS Silibinin prevents memory impairment and oxidative damage induced by Abeta(25-35) and may be a potential therapeutic agent for Alzheimer's disease.
Collapse
Affiliation(s)
- P Lu
- Department of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lee HG, Casadesus G, Zhu X, Castellani RJ, McShea A, Perry G, Petersen RB, Bajic V, Smith MA. Cell cycle re-entry mediated neurodegeneration and its treatment role in the pathogenesis of Alzheimer's disease. Neurochem Int 2009; 54:84-8. [PMID: 19114068 PMCID: PMC2792898 DOI: 10.1016/j.neuint.2008.10.013] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 10/16/2008] [Accepted: 10/20/2008] [Indexed: 11/23/2022]
Abstract
As one of the earliest pathologic changes, the aberrant re-expression of many cell cycle-related proteins and inappropriate cell cycle control in specific vulnerable neuronal populations in Alzheimer's disease (AD) is emerging as an important component in the pathogenesis leading to AD and other neurodegenerative diseases. These events are clearly representative of a true cell cycle, rather than epiphenomena of other processes since, in AD and other neurodegenerative diseases, there is a true mitotic alteration that leads to DNA replication. While the exact role of cell cycle re-entry is unclear, recent studies using cell culture and animal models strongly support the notion that the dysregulation of cell cycle in neurons leads to the development of AD-related pathology such as hyperphosphorylation of tau and amyloid-beta deposition and ultimately causes neuronal cell death. Importantly, cell cycle re-entry is also evident in mutant amyloid-beta precursor protein and tau transgenic mice and, as in human disease, occurs prior to the development of the pathological hallmarks, neurofibrillary tangles and amyloid-beta plaques. Therefore, the study of aberrant cell cycle regulation in model systems, both cellular and animal, may provide extremely important insights into the pathogenesis of AD and also serve as a means to test novel therapeutic approaches.
Collapse
Affiliation(s)
- Hyoung-Gon Lee
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Greilberger J, Koidl C, Greilberger M, Lamprecht M, Schroecksnadel K, Leblhuber F, Fuchs D, Oettl K. Malondialdehyde, carbonyl proteins and albumin-disulphide as useful oxidative markers in mild cognitive impairment and Alzheimer's disease. Free Radic Res 2008; 42:633-8. [PMID: 18654878 DOI: 10.1080/10715760802255764] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The question arises as to whether oxidative stress has a primary role in neurodegeneration or is a secondary end-stage epiphenomenon. The aim of the present study was to determine oxidative stress parameters like malondialdehyde (MDA), carbonyl proteins (CP) and Albumin-disulphide (Alb-SSR) and relate these parameters to the immune parameter neopterin, folic acid and vitamin B12 as vitamins and homocysteine in patients with neuro-degenerative diseases (NDD), namely mild cognitive impairment (MCI) and Alzheimer's disease (AD) compared to an aged matched control group. MDA, CP and Alb-SSR were significantly increased in the NDD group compared to controls, but not vitamin B12, folic acid and neopterin. Significant correlations were found between CP and Alb-SSR, CP and MDA and between MDA and Alb-SSR including patients with NDD and the control group. These results support the hypothesis that oxidative damage to lipids and proteins is an important early event in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- J Greilberger
- Institute of Physiological Chemistry, Center for Physiological Medicine, Medical University of Graz,8010 Graz, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Popova MS, Stepanichev MY. Cell cycle induction, amyloid-beta, and free radicals in the mechanisms of neurodegenerative process progression in the brain. NEUROCHEM J+ 2008. [DOI: 10.1134/s1819712408030021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Leissring MA, Paul BA, Parker I, Cotman CW, LaFerla FM. Alzheimer's Presenilin-1 Mutation Potentiates Inositol 1,4,5-Trisphosphate-Mediated Calcium Signaling in Xenopus. J Neurochem 2008. [DOI: 10.1111/j.1471-4159.1999.721061.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Human neuroblastoma cells transfected with two Chinese presenilin 1 mutations are sensitized to trophic factor withdrawal and protected by insulin-like growth factor-1. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200805020-00009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
38
|
Pensalfini A, Cecchi C, Zampagni M, Becatti M, Favilli F, Paoli P, Catarzi S, Bagnoli S, Nacmias B, Sorbi S, Liguri G. Protective effect of new S-acylglutathione derivatives against amyloid-induced oxidative stress. Free Radic Biol Med 2008; 44:1624-36. [PMID: 18294463 DOI: 10.1016/j.freeradbiomed.2008.01.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Revised: 12/27/2007] [Accepted: 01/17/2008] [Indexed: 10/22/2022]
Abstract
Recent data support the role of oxidative stress in the pathogenesis of Alzheimer disease (AD). In particular, glutathione (GSH) metabolism is altered and its levels are decreased in affected brain regions and peripheral cells from AD patients and in experimental models of AD. In the past decade, interest in the protective effects of various antioxidants aimed at increasing intracellular GSH content has been growing. Because much experimental evidence suggests a possible protective role of unsaturated fatty acids in age-related diseases, we designed the synthesis of new S-acylglutathione (acyl-SG) thioesters. S-Lauroylglutathione (lauroyl-SG) and S-palmitoleoylglutathione (palmitoleoyl-SG) were easily internalized into the cells and they significantly reduced Abeta42-induced oxidative stress in human neurotypic SH-SY5Y cells. In particular, acyl-SG thioesters can prevent the impairment of intracellular ROS scavengers, intracellular ROS accumulation, lipid peroxidation, and apoptotic pathway activation. Palmitoleoyl-SG seemed more effective in cellular protection against Abeta-induced oxidative damage than lauroyl-SG, suggesting a valuable role for the monounsaturated fatty acid. In this study, we demonstrate that acyl-SG derivatives completely avoid the sharp lipoperoxidation in primary fibroblasts from familial AD patients occurring after exposure to Abeta42 aggregates. Hence, we put forward these derivatives as new antioxidant compounds which could be excellent candidates for therapeutic treatment of AD and other oxidative stress-related diseases.
Collapse
Affiliation(s)
- Anna Pensalfini
- Department of Biochemical Sciences, University of Florence, 50134 Florence, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Differentiation increases the resistance of neuronal cells to amyloid toxicity. Neurochem Res 2008; 33:2516-31. [PMID: 18307032 DOI: 10.1007/s11064-008-9627-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Accepted: 02/06/2008] [Indexed: 10/22/2022]
Abstract
A substantial lack of information is recognized on the features underlying the variable susceptibility to amyloid aggregate toxicity of cells with different phenotypes. Recently, we showed that different cell types are variously affected by early aggregates of a prokaryotic hydrogenase domain (HypF-N). In the present study we investigated whether differentiation affects cell susceptibility to amyloid injury using a human neurotypic SH-SY5Y cell differentiation model. We found that retinoic acid-differentiated cells were significantly more resistant against Abeta1-40, Abeta1-42 and HypF-N prefibrillar aggregate toxicity respect to undifferentiated cells treated similarly. Earlier and sharper increases in cytosolic Ca(2+) and ROS with marked lipid peroxidation and mitochondrial dysfunction were also detected in exposed undifferentiated cells resulting in apoptosis activation. The reduced vulnerability of differentiated cells matched a more efficient Ca(2+)-ATPase equipment and a higher total antioxidant capacity. Finally, increasing the content of membrane cholesterol resulted in a remarkable reduction of vulnerability and ability to bind the aggregates in either undifferentiated and differentiated cells.
Collapse
|
40
|
Cecchi C, Pensalfini A, Stefani M, Baglioni S, Fiorillo C, Cappadona S, Caporale R, Nosi D, Ruggiero M, Liguri G. Replicating neuroblastoma cells in different cell cycle phases display different vulnerability to amyloid toxicity. J Mol Med (Berl) 2007; 86:197-209. [PMID: 17885746 DOI: 10.1007/s00109-007-0265-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 08/29/2007] [Accepted: 09/03/2007] [Indexed: 01/03/2023]
Abstract
A key role of mitotic activation in neuronal cell death in early stages of Alzheimer's disease (AD) has been suggested. Apparently, terminally differentiated neurons are precluded from mitotic division, yet some phenotypic markers of cell cycling are present in AD-vulnerable brain areas. In this paper, we investigated whether dividing human neuroblastoma cells are preferentially vulnerable to amyloid aggregate toxicity in some specific cell cycle stage(s). Our data indicate that Abeta1-40/42 aggregates added to the cell culture media bind to the plasma membrane and are internalized faster in the S than in the G2/M and G1 cells possibly as a result of a lower content in membrane cholesterol in the former. Earlier and sharper increases in reactive oxygen species production triggered a membrane oxidative injury and a significant impairment of antioxidant capacity, eventually culminating with apoptotic activation in S and, to a lesser extent, in G2/M exposed cells. G1 cells appeared more resistant to the amyloid-induced oxidative attack possibly because of their higher antioxidant capacity. The high vulnerability of S cells to aggregate toxicity extends previous data suggesting that neuronal loss in AD could result from mitotic reactivation of terminally differentiated neurons with arrest in the S phase.
Collapse
Affiliation(s)
- Cristina Cecchi
- Department of Biochemical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Proteins synthesized in the endoplasmic reticulum (ER) are properly folded with the assistance of ER chaperones. Malfolded proteins are disposed of by ER-associated protein degradation (ERAD). When the amount of unfolded protein exceeds the folding capacity of the ER, human cells activate a defense mechanism called the ER stress response, which induces expression of ER chaperones and ERAD components and transiently attenuates protein synthesis to decrease the burden on the ER. It has been revealed that three independent response pathways separately regulate induction of the expression of chaperones, ERAD components, and translational attenuation. A malfunction of the ER stress response caused by aging, genetic mutations, or environmental factors can result in various diseases such as diabetes, inflammation, and neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, and bipolar disorder, which are collectively known as 'conformational diseases'. In this review, I will summarize recent progress in this field. Molecules that regulate the ER stress response would be potential candidates for drug targets in various conformational diseases.
Collapse
Affiliation(s)
- Hiderou Yoshida
- Department of Biophysics, Graduate School of Science, Kyoto University, Japan.
| |
Collapse
|
42
|
Wu P, Shen Q, Dong S, Xu Z, Tsien JZ, Hu Y. Calorie restriction ameliorates neurodegenerative phenotypes in forebrain-specific presenilin-1 and presenilin-2 double knockout mice. Neurobiol Aging 2007; 29:1502-11. [PMID: 17499883 DOI: 10.1016/j.neurobiolaging.2007.03.028] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Revised: 03/16/2007] [Accepted: 03/30/2007] [Indexed: 11/15/2022]
Abstract
Conditional double knockout of presenilin-1 and presenilin-2 (cDKO) in forebrain of mice led to brain atrophy, tau hyperphosphorylation, synaptic dysfunction and cognitive deficit. These brain changes recapitulated most of the neurodegenerative phenotypes of Alzheimer's disease (AD). In this report, we have investigated the effects of 4-month calorie restriction (CR) regimen on different phenotypes in cDKO mice. We found that CR improved novel object recognition and contextual fear conditioning memory in the cDKO mice. Histological and biochemical analysis showed that CR attenuated ventricle enlargement, caspase-3 activation and astrogliosis. In addition, the induction of tau hyperphosphorylation in the cDKO mice was reduced by CR, possibly through reduction of p25 accumulation and aberrant CDK5 activation. Finally, DNA microarray analysis demonstrated that CR could increase the expression of neurogenesis related genes and decrease the expression of inflammation related genes in the hippocampus of cDKO mice. The possible molecular mechanisms of the CR effects on alleviating AD pathogenesis have been discussed.
Collapse
Affiliation(s)
- Pu Wu
- Shanghai Institute of Brain Functional Genomics, MOE & STCSM Key Lab of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | | | | | | | | | | |
Collapse
|
43
|
Nelson O, Tu H, Lei T, Bentahir M, de Strooper B, Bezprozvanny I. Familial Alzheimer disease-linked mutations specifically disrupt Ca2+ leak function of presenilin 1. J Clin Invest 2007; 117:1230-9. [PMID: 17431506 PMCID: PMC1847535 DOI: 10.1172/jci30447] [Citation(s) in RCA: 185] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2006] [Accepted: 02/13/2007] [Indexed: 01/19/2023] Open
Abstract
Mutations in presenilins are responsible for approximately 40% of all early-onset familial Alzheimer disease (FAD) cases in which a genetic cause has been identified. In addition, a number of mutations in presenilin-1 (PS1) have been suggested to be associated with the occurrence of frontal temporal dementia (FTD). Presenilins are highly conserved transmembrane proteins that support cleavage of the amyloid precursor protein by gamma-secretase. Recently, we discovered that presenilins also function as passive ER Ca(2+) leak channels. Here we used planar lipid bilayer reconstitution assays and Ca(2+) imaging experiments with presenilin-null mouse embryonic fibroblasts to analyze ER Ca(2+) leak function of 6 FAD-linked PS1 mutants and 3 known FTD-associated PS1 mutants. We discovered that L166P, A246E, E273A, G384A, and P436Q FAD mutations in PS1 abolished ER Ca(2+) leak function of PS1. In contrast, A79V FAD mutation or FTD-associated mutations (L113P, G183V, and Rins352) did not appear to affect ER Ca(2+) leak function of PS1 in our experiments. We validated our findings in Ca(2+) imaging experiments with primary fibroblasts obtained from an FAD patient possessing mutant PS1-A246E. Our results indicate that many FAD mutations in presenilins are loss-of-function mutations affecting ER Ca(2+) leak activity. In contrast, none of the FTD-associated mutations affected ER Ca(2+) leak function of PS1, indicating that the observed effects are disease specific. Our observations are consistent with the potential role of disturbed Ca(2+) homeostasis in Alzheimer disease pathogenesis.
Collapse
Affiliation(s)
- Omar Nelson
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.
Center for Human Genetics, Katholieke Universiteit Leuven, and Department for Molecular and Developmental Genetics, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Huiping Tu
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.
Center for Human Genetics, Katholieke Universiteit Leuven, and Department for Molecular and Developmental Genetics, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Tianhua Lei
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.
Center for Human Genetics, Katholieke Universiteit Leuven, and Department for Molecular and Developmental Genetics, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Mostafa Bentahir
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.
Center for Human Genetics, Katholieke Universiteit Leuven, and Department for Molecular and Developmental Genetics, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Bart de Strooper
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.
Center for Human Genetics, Katholieke Universiteit Leuven, and Department for Molecular and Developmental Genetics, Flanders Institute for Biotechnology, Leuven, Belgium
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.
Center for Human Genetics, Katholieke Universiteit Leuven, and Department for Molecular and Developmental Genetics, Flanders Institute for Biotechnology, Leuven, Belgium
| |
Collapse
|
44
|
Payette DJ, Xie J, Guo Q. Reduction in CHT1-mediated choline uptake in primary neurons from presenilin-1 M146V mutant knock-in mice. Brain Res 2006; 1135:12-21. [PMID: 17196556 PMCID: PMC1805819 DOI: 10.1016/j.brainres.2006.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 11/20/2006] [Accepted: 12/02/2006] [Indexed: 12/31/2022]
Abstract
The memory loss in Alzheimer's disease (AD) has been linked to cholinergic hypoactivity. Mutations in presenilin-1 (PS-1) may regulate cholinergic signaling, although their precise roles in cholinergic neurotransmission in AD are unsettled. Neuronal uptake of choline via the high affinity choline transporter (CHT1) is essential for cholinergic neurotransmission. CHT1 is a Na+-dependent, hemicholinium-3 (HC-3)-sensitive choline transporter. Although cholinergic neurons in the nucleus basalis of Meynert are a major source of cholinergic projections for the cerebral cortex, it is unclear whether cortical neurons exhibit intrinsic CHT1 activity that is altered in AD. We now report that primary cortical neurons express intrinsic and biologically active CHT1, and that, in these neurons, CHT1-mediated choline uptake activity is significantly reduced in PS-1 M146V mutant knock-in mice. Further kinetic studies using HC-3 binding and cell surface biotinylation assays showed that the PS-1 mutation inhibits CHT1 mediated choline uptake by reducing the ligand binding affinity of CHT1 without significantly altering levels of CHT1 expression in the plasma membrane. Since human neocortex has recently been shown to possess intrinsic cholinergic innervation, our results indicate that alterations in CHT1-mediated high affinity choline uptake in cortical neurons may contribute to Alzheimer's dementia.
Collapse
Affiliation(s)
| | | | - Qing Guo
- Correspondence: Qing Guo, Ph.D., Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, Phone: (405) 271-2226 ext. 56268., FAX: (405) 271-3181., E-mail:
| |
Collapse
|
45
|
|
46
|
Nixon RA. Autophagy in neurodegenerative disease: friend, foe or turncoat? Trends Neurosci 2006; 29:528-35. [PMID: 16859759 DOI: 10.1016/j.tins.2006.07.003] [Citation(s) in RCA: 250] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 05/10/2006] [Accepted: 07/10/2006] [Indexed: 02/09/2023]
Abstract
Autophagy, a lysosomal pathway for degrading organelles and long-lived proteins, is becoming recognized as a key adaptive response that can preclude death in stressed or diseased cells. However, during development strong induction of autophagy in specific cell populations mediates a type of programmed cell death that has distinctive 'autophagic' morphology and a requirement for autophagy activity. The recent identification of autophagosomes in neurons in a growing number of neurodegenerative disorders has, therefore, sparked controversy about whether these structures are contributing to neuronal cell death or protecting against it. Emerging evidence supports the view that induction of autophagy is a neuroprotective response and that inadequate or defective autophagy, rather than excessive autophagy, promotes neuronal cell death in most of these disorders. In this review, we consider possible mechanisms underlying autophagy-associated cell death and their relationship to pathways mediating apoptosis and necrosis.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA.
| |
Collapse
|
47
|
Kasri NN, Kocks SL, Verbert L, Hébert SS, Callewaert G, Parys JB, Missiaen L, De Smedt H. Up-regulation of inositol 1,4,5-trisphosphate receptor type 1 is responsible for a decreased endoplasmic-reticulum Ca2+ content in presenilin double knock-out cells. Cell Calcium 2006; 40:41-51. [PMID: 16675011 DOI: 10.1016/j.ceca.2006.03.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 03/12/2006] [Accepted: 03/16/2006] [Indexed: 11/27/2022]
Abstract
Presenilins (PS) are proteins involved in the pathogenesis of autosomal-dominant familial cases of Alzheimer's disease. Mutations in PS are known to induce specific alterations in cellular Ca2+ signaling which might be involved in the pathogenesis of neurodegenerative diseases. Mouse embryonic fibroblasts (MEF) deficient in PS1 and PS2 (PS DKO) as well as the latter rescued with PS1 (Rescue), were used to investigate the underlying mechanism of these alterations in Ca2+ signaling. PS DKO cells were characterized by a decrease in the [Ca2+]ER as measured by ER-targeted aequorin luminescence and an increased level of type 1 inositol 1,4,5-trisphosphate receptor (IP3R1). The lower [Ca2+]ER was associated with an increase in a Ca2+ leak from the ER. The increased IP3R1 expression and the concomitant changes in ER Ca2+ handling were reversed in the Rescue cells. Moreover using RNA-interference mediated reduction of IP3R1 we could demonstrate that the up-regulation of this isoform was responsible for the increased Ca2+ leak and the lowered [Ca2+]ER PS DKO cells. Finally, we show that the decreased [Ca2+]ER in PS DKO cells was protective against apoptosis.
Collapse
Affiliation(s)
- Nael Nadif Kasri
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Brain cells are highly energy dependent for maintaining ion homeostasis during high metabolic activity. During active periods, full mitochondrial function is essential to generate ATP from electrons that originate with the oxidation of NADH. Decreasing brain metabolism is a significant cause of cognitive abnormalities of Alzheimer disease (AD), but it remains uncertain whether this is the cause of further pathology or whether synaptic loss results in a lower energy demand. Synapses are the first to show pathological symptoms in AD before the onset of clinical symptoms. Because synaptic function has high energy demands, interruption in mitochondrial energy supply could be the major factor in synaptic failure in AD. A newly discovered age-related decline in neuronal NADH and redox ratio may jeopardize this function. Mitochondrial dehydrogenases and several mutations affecting energy transfer are frequently altered in aging and AD. Thus, with the accumulation of genetic defects in mitochondria at the level of energy transfer, the issue of neuronal susceptibility to damage as a function of age and age-related disease becomes important. In an aging rat neuron model, mitochondria are both chronically depolarized and produce more reactive oxygen species with age. These concepts suggest that multiple treatment targets may be needed to reverse this multifactorial disease. This review summarizes new insights based on the interaction of mitoenergetic failure, glutamate excitotoxicity, and amyloid toxicity in the exacerbation of AD.
Collapse
Affiliation(s)
- Mordhwaj S Parihar
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794-9626, USA
| | | |
Collapse
|
49
|
Increased susceptibility to amyloid toxicity in familial Alzheimer's fibroblasts. Neurobiol Aging 2006; 28:863-76. [PMID: 16781020 DOI: 10.1016/j.neurobiolaging.2006.05.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2005] [Revised: 04/14/2006] [Accepted: 05/03/2006] [Indexed: 11/18/2022]
Abstract
Much experimental evidence suggests that an imbalance in cellular redox status is a major factor in the pathogenesis of Alzheimer's disease (AD). Our previous data showed a marked increase in membrane lipoperoxidation in primary fibroblasts from familial AD (FAD) patients. In the present study, we demonstrate that when oligomeric structures of Abeta 1-40 and Abeta 1-42 are added to the culture media, they accumulate quicker near the plasma membrane, and are internalized faster and mostly in APPV717I fibroblasts than in age-matched healthy cells; this results in an earlier and sharper increase in the production of reactive oxygen species (ROS). Higher ROS production leads in turn to an increase in membrane oxidative-injury and significant impairment of cellular antioxidant capacity, giving rise to apoptotic cascade activation and finally to a necrotic outcome. In contrast, healthy fibroblasts appear more resistant to amyloid oxidative-attack, possibly as a result of their plasma membrane integrity and powerful antioxidant capacity. Our data are consistent with increasing evidence that prefibrillar aggregates, compared to mature fibrils, are likely the more toxic species of the peptides. These findings provide compelling evidence that cells bearing increased membrane lipoperoxidation are more susceptible to aggregate toxicity as a result of their reduced ability to counteract amyloid oligomeric attack.
Collapse
|
50
|
Yu MS, Suen KC, Kwok NS, So KF, Hugon J, Chang RCC. Beta-amyloid peptides induces neuronal apoptosis via a mechanism independent of unfolded protein responses. Apoptosis 2006; 11:687-700. [PMID: 16532272 DOI: 10.1007/s10495-006-5540-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Accumulation of beta-amyloid (Abeta) peptides in senile plaques is one of the pathological hallmarks in Alzheimer's disease (AD), which can trigger apoptosis. We have previously demonstrated that Abeta triggered calcium release from the ER. Depletion of ER Ca(2+) ions has been reported leading to unfolded protein responses (UPR). While hypothesis has been made about UPR and neurodegeneration in AD, little is known about the effects of extracellular accumulation of Abeta on UPR. We have shown previously that activation of PKR in Abeta-triggered apoptosis. Since UPR can trigger PKR, our study aims to elucidate whether extracellular accumulation of Abeta peptides induce UPR in cultured neurons. Our results showed that Abeta could not trigger UPR signalings including phosphorylation of PERK, alternative cleavage of xbp-1 mRNA and induction of transcription of xbp-1 and Gadd153. Taken together, our results suggest that extracellular accumulation of Abeta peptides induce apoptosis via a mechanism independent of UPR.
Collapse
Affiliation(s)
- Man-Shan Yu
- Laboratory of Neurodegenerative Diseases, Department of Anatomy, Research Centre of Heart, Brain, Hormone and Healthy Aging, Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | | | | | | | | | | |
Collapse
|