1
|
Dai L, Shen Y. Insights into T-cell dysfunction in Alzheimer's disease. Aging Cell 2021; 20:e13511. [PMID: 34725916 PMCID: PMC8672785 DOI: 10.1111/acel.13511] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/22/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
T cells, the critical immune cells of the adaptive immune system, are often dysfunctional in Alzheimer's disease (AD) and are involved in AD pathology. Reports highlight neuroinflammation as a crucial modulator of AD pathogenesis, and aberrant T cells indirectly contribute to neuroinflammation by secreting proinflammatory mediators via direct crosstalk with glial cells infiltrating the brain. However, the mechanisms underlying T‐cell abnormalities in AD appear multifactorial. Risk factors for AD and pathological hallmarks of AD have been tightly linked with immune responses, implying the potential regulatory effects of these factors on T cells. In this review, we discuss how the risk factors for AD, particularly Apolipoprotein E (ApoE), Aβ, α‐secretase, β‐secretase, γ‐secretase, Tau, and neuroinflammation, modulate T‐cell activation and the association between T cells and pathological AD hallmarks. Understanding these associations is critical to provide a comprehensive view of appropriate therapeutic strategies for AD.
Collapse
Affiliation(s)
- Linbin Dai
- Institute on Aging and Brain Disorders The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Sciences and Technology of China Hefei China
- Neurodegenerative Disease Research Center University of Science and Technology of China Hefei China
- Hefei National Laboratory for Physical Sciences at the Microscale University of Science and Technology of China Hefei China
| | - Yong Shen
- Institute on Aging and Brain Disorders The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Sciences and Technology of China Hefei China
- Neurodegenerative Disease Research Center University of Science and Technology of China Hefei China
- Hefei National Laboratory for Physical Sciences at the Microscale University of Science and Technology of China Hefei China
| |
Collapse
|
2
|
Ayton S, Bush AI. β-amyloid: The known unknowns. Ageing Res Rev 2021; 65:101212. [PMID: 33188924 DOI: 10.1016/j.arr.2020.101212] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) stands out as a major disease without any form of preventative or disease modifying therapy. This is not for lack of trying. 33 phase 3 clinical trials of drugs targeting amyloid beta (Aβ) have failed to slow cognitive decline in AD. The field is at a cross-roads about whether to continue anti-Aβ therapy or more actively pursue alternative targets. With the burden of this disease to patients, families, and healthcare budgets growing yearly, the need for disease modifying AD therapies has become one of the highest priorities in all of medicine. While pathology, genetic and biochemical data offer a popular narrative for the causative role of Aβ, there are alternative explanations, and dissenting findings that, now more than ever, warrant thorough reanalysis. This review questions the major assumptions about Aβ on which therapies for AD were premised, and invites renewed interrogation into AD pathogenesis.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
3
|
Feng W, Zhang Y, Wang Z, Xu H, Wu T, Marshall C, Gao J, Xiao M. Microglia prevent beta-amyloid plaque formation in the early stage of an Alzheimer's disease mouse model with suppression of glymphatic clearance. ALZHEIMERS RESEARCH & THERAPY 2020; 12:125. [PMID: 33008458 PMCID: PMC7532614 DOI: 10.1186/s13195-020-00688-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/15/2020] [Indexed: 12/13/2022]
Abstract
Background Soluble beta-amyloid (Aβ) can be cleared from the brain through various mechanisms including enzymatic degradation, glial cell phagocytosis, transport across the blood-brain barrier, and glymphatic clearance. However, the relative contribution of each clearance system and their compensatory effects in delaying the pathological process of Alzheimer’s disease (AD) are currently unknown. Methods Fluorescent trace, immunofluorescence, and Western blot analyses were performed to compare glymphatic clearance ability and Aβ accumulation among 3-month-old APP695/PS1-dE9 transgenic (APP/PS1) mice, wild-type mice, aquaporin 4 knock out (AQP4−/−) mice, and AQP4−/−/APP/PS1 mice. The consequence of selectively eliminating microglial cells, or downregulating apolipoprotein E (apoE) expression, on Aβ burden, was also investigated in the frontal cortex of AQP4−/−/APP/PS1 mice and APP/PS1 mice. Results AQP4 deletion in APP/PS1 mice significantly exaggerated glymphatic clearance dysfunction, and intraneuronal accumulation of Aβ and apoE, although it did not lead to Aβ plaque deposition. Notably, microglia, but not astrocytes, increased activation and phagocytosis of Aβ in the cerebral cortex of AQP4−/−/APP/PS1 mice, compared with APP/PS1 mice. Selectively eliminating microglia in the frontal cortex via local injection of clodronate liposomes resulted in deposition of Aβ plaques in AQP4−/−/APP/PS1 mice, but not APP/PS1 mice. Moreover, knockdown of apoE reduced intraneuronal Aβ levels in both APP/PS1 mice and AQP4−/−/APP/PS1 mice, indicating an inhibitory effect of apoE on Aβ clearance. Conclusion The above results suggest that the glymphatic system mediated Aβ and apoE clearance and microglia mediated Aβ degradation synergistically prevent Aβ plague formation in the early stages of the AD mouse model. Protecting one or both of them might be beneficial to delaying the onset of AD.
Collapse
Affiliation(s)
- Weixi Feng
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China.,Brain Institute, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yanli Zhang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China.,Brain Institute, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Ze Wang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China.,Brain Institute, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Hanrong Xu
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China.,Brain Institute, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Wu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Charles Marshall
- Department of Physical Therapy, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, USA
| | - Junying Gao
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China. .,Brain Institute, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China.
| | - Ming Xiao
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, China. .,Brain Institute, The Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Long JM, Holtzman DM. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019; 179:312-339. [PMID: 31564456 PMCID: PMC6778042 DOI: 10.1016/j.cell.2019.09.001] [Citation(s) in RCA: 1608] [Impact Index Per Article: 321.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer disease (AD) is a heterogeneous disease with a complex pathobiology. The presence of extracellular β-amyloid deposition as neuritic plaques and intracellular accumulation of hyperphosphorylated tau as neurofibrillary tangles remains the primary neuropathologic criteria for AD diagnosis. However, a number of recent fundamental discoveries highlight important pathological roles for other critical cellular and molecular processes. Despite this, no disease-modifying treatment currently exists, and numerous phase 3 clinical trials have failed to demonstrate benefits. Here, we review recent advances in our understanding of AD pathobiology and discuss current treatment strategies, highlighting recent clinical trials and opportunities for developing future disease-modifying therapies.
Collapse
Affiliation(s)
- Justin M Long
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
5
|
Transcriptional Effects of ApoE4: Relevance to Alzheimer's Disease. Mol Neurobiol 2017; 55:5243-5254. [PMID: 28879423 DOI: 10.1007/s12035-017-0757-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/24/2017] [Indexed: 10/18/2022]
Abstract
The major genetic risk factor for sporadic Alzheimer's disease (AD) is the lipid binding and transporting carrier protein apolipoprotein E, epsilon 4 allele (ApoE4). One of the unsolved mysteries of AD is how the presence of ApoE4 elicits this age-associated, currently incurable neurodegenerative disease. Recently, we showed that ApoE4 acts as a transcription factor and binds to the promoters of genes involved in a range of processes linked to aging and AD disease pathogenesis. These findings point to novel therapeutic strategies for AD and aging, resulting in an extension of human healthspan, the disease-free and functional period of life. Here, we review the effects and implications of the putative transcriptional role of ApoE4 and propose a model of Alzheimer's disease that focuses on the transcriptional nature of ApoE4 and its downstream effects, with the aim that this knowledge will help to define the role ApoE4 plays as a risk factor for AD, aging, and other processes such as inflammation and cardiovascular disease.
Collapse
|
6
|
Lodeiro M, Puerta E, Ismail MAM, Rodriguez-Rodriguez P, Rönnbäck A, Codita A, Parrado-Fernandez C, Maioli S, Gil-Bea F, Merino-Serrais P, Cedazo-Minguez A. Aggregation of the Inflammatory S100A8 Precedes Aβ Plaque Formation in Transgenic APP Mice: Positive Feedback for S100A8 and Aβ Productions. J Gerontol A Biol Sci Med Sci 2017; 72:319-328. [PMID: 27131040 DOI: 10.1093/gerona/glw073] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 04/07/2016] [Indexed: 11/12/2022] Open
Abstract
Inflammation plays an important role in Alzheimer's disease (AD) and other neurodegenerative disorders. Although chronic inflammation in later stages of AD is well described, little is known about the inflammatory processes in preclinical or early stages of the disease prior to plaque deposition. In this study, we report that the inflammatory mediator S100A8 is increased with aging in the mouse brain. It is observed as extracellular aggregates, which do not correspond to corpora amylacea. S100A8 aggregation is enhanced in the hippocampi of two different mouse models for amyloid-β (Aβ) overproduction (Tg2576 and TgAPParctic mice). S100A8 aggregates are seen prior the formation of Aβ plaques and do not colocalize. In vitro treatment of glial cells from primary cultures with Aβ42 resulted in an increased production of S100A8. In parallel, treatment of a neuronal cell line with recombinant S100A8 protein resulted in enhanced Aβ42 and decreased Aβ40 production. Our results suggest that important inflammatory processes are occurring prior to Aβ deposition and the existence of a positive feedback between S100A8 and Aβ productions. The possible relevance of aging- or AD-dependent formation of S100A8 aggregates in the hippocampus thus affecting learning and memory processes is discussed.
Collapse
Affiliation(s)
- Maria Lodeiro
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Elena Puerta
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Muhammad-Al-Mustafa Ismail
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Patricia Rodriguez-Rodriguez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Annica Rönnbäck
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Alina Codita
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Cristina Parrado-Fernandez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Silvia Maioli
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Francisco Gil-Bea
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Division of Neurosciences, Department of Cellular and Molecular Neuropharmacology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Paula Merino-Serrais
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Huynh TPV, Davis AA, Ulrich JD, Holtzman DM. Apolipoprotein E and Alzheimer's disease: the influence of apolipoprotein E on amyloid-β and other amyloidogenic proteins. J Lipid Res 2017; 58:824-836. [PMID: 28246336 DOI: 10.1194/jlr.r075481] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 02/25/2017] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is one of the fastest-growing causes of death and disability in persons 65 years of age or older, affecting more than 5 million Americans alone. Clinical manifestations of AD include progressive decline in memory, executive function, language, and other cognitive domains. Research efforts within the last three decades have identified APOE as the most significant genetic risk factor for late-onset AD, which accounts for >99% of cases. The apoE protein is hypothesized to affect AD pathogenesis through a variety of mechanisms, from its effects on the blood-brain barrier, the innate immune system, and synaptic function to the accumulation of amyloid-β (Aβ). Here, we discuss the role of apoE on the biophysical properties and metabolism of the Aβ peptide, the principal component of amyloid plaques and cerebral amyloid angiopathy (CAA). CAA is characterized by the deposition of amyloid proteins (including Aβ) in the leptomeningeal medium and small arteries, which is found in most AD cases but sometimes occurs as an independent entity. Accumulation of these pathologies in the brain is one of the pathological hallmarks of AD. Beyond Aβ, we will extend the discussion to the potential role of apoE on other amyloidogenic proteins found in AD, and also a number of diverse neurodegenerative diseases.
Collapse
Affiliation(s)
- Tien-Phat V Huynh
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110
| | - Albert A Davis
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110
| | - Jason D Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110
| |
Collapse
|
8
|
Habib A, Sawmiller D, Tan J. Restoring Soluble Amyloid Precursor Protein α Functions as a Potential Treatment for Alzheimer's Disease. J Neurosci Res 2016; 95:973-991. [PMID: 27531392 DOI: 10.1002/jnr.23823] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/13/2016] [Accepted: 06/15/2016] [Indexed: 12/14/2022]
Abstract
Soluble amyloid precursor protein α (sAPPα), a secreted proteolytic fragment of nonamyloidogenic amyloid precursor protein (APP) processing, is known for numerous neuroprotective functions. These functions include but are not limited to proliferation, neuroprotection, synaptic plasticity, memory formation, neurogenesis, and neuritogenesis in cell culture and animal models. In addition, sAPPα influences amyloid-β (Aβ) production by direct modulation of APP β-secretase proteolysis as well as Aβ-related or unrelated tau pathology, hallmark pathologies of Alzheimer's disease (AD). Thus, the restoration of sAPPα levels and functions in the brain by increasing nonamyloidogenic APP processing and/or manipulation of its signaling could reduce AD pathology and cognitive impairment. It is likely that identification and characterization of sAPPα receptors in the brain, downstream effectors, and signaling pathways will pave the way for an attractive therapeutic target for AD prevention or intervention. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ahsan Habib
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Darrell Sawmiller
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jun Tan
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
9
|
Han SH, Park JC, Mook-Jung I. Amyloid β-interacting partners in Alzheimer's disease: From accomplices to possible therapeutic targets. Prog Neurobiol 2016; 137:17-38. [DOI: 10.1016/j.pneurobio.2015.12.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022]
|
10
|
Wong BX, Hung YH, Bush AI, Duce JA. Metals and cholesterol: two sides of the same coin in Alzheimer's disease pathology. Front Aging Neurosci 2014; 6:91. [PMID: 24860500 PMCID: PMC4030154 DOI: 10.3389/fnagi.2014.00091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/28/2014] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease. It begins years prior to the onset of clinical symptoms, such as memory loss and cognitive decline. Pathological hallmarks of AD include the accumulation of β-amyloid in plaques and hyperphosphorylated tau in neurofibrillary tangles. Copper, iron, and zinc are abnormally accumulated and distributed in the aging brain. These metal ions can adversely contribute to the progression of AD. Dysregulation of cholesterol metabolism has also been implicated in the development of AD pathology. To date, large bodies of research have been carried out independently to elucidate the role of metals or cholesterol on AD pathology. Interestingly, metals and cholesterol affect parallel molecular and biochemical pathways involved in AD pathology. The possible links between metal dyshomeostasis and altered brain cholesterol metabolism in AD are reviewed.
Collapse
Affiliation(s)
- Bruce X Wong
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Ya Hui Hung
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Ashley I Bush
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - James A Duce
- Oxidation Biology Unit, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia ; School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds Leeds, North Yorkshire, UK
| |
Collapse
|
11
|
Apolipoprotein E, amyloid-beta, and neuroinflammation in Alzheimer's disease. Neurosci Bull 2014; 30:317-30. [PMID: 24652457 DOI: 10.1007/s12264-013-1422-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 01/23/2014] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation and deposition of amyloid-beta (Aβ) peptides in the brain. Neuroinflammation occurs in the AD brain and plays a critical role in the neurodegenerative pathology. Particularly, Aβ evokes an inflammatory response that leads to synaptic dysfunction, neuronal death, and neurodegeneration. Apolipoprotein E (ApoE) proteins are involved in cholesterol transport, Aβ binding and clearance, and synaptic functions in the brain. The ApoE4 isoform is a key risk factor for AD, while the ApoE2 isoform has a neuroprotective effect. However, studies have reached different conclusions about the roles of the isoforms; some show that both ApoE3 and ApoE4 have anti-inflammatory effects, while others show that ApoE4 causes a predisposition to inflammation or promotes an inflammatory response following lipopolysaccharide treatment. These discrepancies may result from the differences in models, cell types, experimental conditions, and inflammatory stimuli used. Further, little was known about the role of ApoE isoforms in the Aβ-induced inflammatory response and in the neuroinflammation of AD. Our recent work showed that ApoE isoforms differentially regulate and modify the Aβ-induced inflammatory response in neural cells, with ApoE2 suppressing and ApoE4 promoting the response. In this article, we review the roles, mechanisms, and interrelations among Aβ, ApoE, and neuroinflammation in AD.
Collapse
|
12
|
Abstract
The canonical pathogenesis of Alzheimer's disease links the expression of apolipoprotein E ε4 allele (ApoE) to amyloid precursor protein (APP) processing and Aβ peptide accumulation by a set of mechanisms that is incompletely defined. The development of a simple system that focuses not on a single variable but on multiple factors and pathways would be valuable both for dissecting the underlying mechanisms and for identifying candidate therapeutics. Here we show that, although both ApoE3 and ApoE4 associate with APP with nanomolar affinities, only ApoE4 significantly (i) reduces the ratio of soluble amyloid precursor protein alpha (sAPPα) to Aβ; (ii) reduces Sirtuin T1 (SirT1) expression, resulting in markedly differing ratios of neuroprotective SirT1 to neurotoxic SirT2; (iii) triggers Tau phosphorylation and APP phosphorylation; and (iv) induces programmed cell death. We describe a subset of drug candidates that interferes with the APP-ApoE interaction and returns the parameters noted above to normal. Our data support the hypothesis that neuronal connectivity, as reflected in the ratios of critical mediators such as sAPPα:Aβ, SirT1:SirT2, APP:phosphorylated (p)-APP, and Tau:p-Tau, is programmatically altered by ApoE4 and offer a simple system for the identification of program mediators and therapeutic candidates.
Collapse
|
13
|
Gil-Bea F, Akterin S, Persson T, Mateos L, Sandebring A, Avila-Cariño J, Gutierrez-Rodriguez A, Sundström E, Holmgren A, Winblad B, Cedazo-Minguez A. Thioredoxin-80 is a product of alpha-secretase cleavage that inhibits amyloid-beta aggregation and is decreased in Alzheimer's disease brain. EMBO Mol Med 2012; 4:1097-111. [PMID: 22933306 PMCID: PMC3491839 DOI: 10.1002/emmm.201201462] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/14/2012] [Accepted: 07/17/2012] [Indexed: 11/07/2022] Open
Abstract
Thioredoxin-1 (Trx1) is an endogenous dithiol reductant and antioxidant that was shown to be decreased in Alzheimer's disease (AD) neurons. A truncated form of Trx1, thioredoxin 80 (Trx80), was reported to be secreted from monocytes having cytokine activity. Here, we show that Trx80 is present in human brain in an aggregated form. Trx80 localizes mainly to neurons and is dramatically decreased in AD brains. Trx80 levels in cerebrospinal fluid (CSF) correlate with those of the classical AD biomarkers amyloid-β (Aβ) 1-42 and total tau. Moreover, Trx80 measurements in CSF discriminate between patients with stable mild cognitive impairment, prodomal AD and mild AD. We report that ADAM10 and 17, two α-secretases processing the Aβ precursor protein, are responsible for Trx80 generation. In contrast to the periphery, Trx80 has no pro-inflammatory effects in glia, either by itself or in combination with Aβ or apolipoprotein E. Instead, Trx80 inhibits Aβ(1-42) aggregation and protects against its toxicity. Thus, a reduction in Trx80 production would result in increased Aβ polymerization and enhanced neuronal vulnerability. Our data suggest that a deficit in Trx80 could participate in AD pathogenesis.
Collapse
Affiliation(s)
- Francisco Gil-Bea
- Department of Neurobiology, KI-Alzheimer's Disease Research Center, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Spencer JPE, Vafeiadou K, Williams RJ, Vauzour D. Neuroinflammation: modulation by flavonoids and mechanisms of action. Mol Aspects Med 2012; 33:83-97. [PMID: 22107709 DOI: 10.1016/j.mam.2011.10.016] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 10/14/2011] [Indexed: 01/01/2023]
Abstract
Neuroinflammatory processes are known to contribute to the cascade of events culminating in the neuronal damage that underpins neurodegenerative disorders such as Parkinson's and Alzheimer's disease. Recently, there has been much interest in the potential neuroprotective effects of flavonoids, a group of plant secondary metabolites known to have diverse biological activity in vivo. With respect to the brain, flavonoids, such as those found in cocoa, tea, berries and citrus, have been shown to be highly effective in preventing age-related cognitive decline and neurodegeneration in both animals and humans. Evidence suggests that flavonoids may express such ability through a multitude of physiological functions, including an ability to modulate the brains immune system. This review will highlight the evidence for their potential to inhibit neuroinflammation through an attenuation of microglial activation and associated cytokine release, iNOS expression, nitric oxide production and NADPH oxidase activity. We will also detail the current evidence indicting that their regulation of these immune events appear to be mediated by their actions on intracellular signaling pathways, including the nuclear factor-κB (NF-κB) cascade and mitogen-activated protein kinase (MAPK) pathway. As such, flavonoids represent important precursor molecules in the quest to develop of a new generation of drugs capable of counteracting neuroinflammation and neurodegenerative disease.
Collapse
Affiliation(s)
- Jeremy P E Spencer
- Molecular Nutrition Group, Centre for Integrative Neuroscience and Neurodynamics, School of Chemistry, Food and Pharmacy, University of Reading, Reading RG6 6AP, UK.
| | | | | | | |
Collapse
|
15
|
Abstract
Sir Harold Himsworth's prescient observations 75 years ago have recently been expanded to include a clear relationship between insulin resistance and central nervous system function. Insulin is a master regulator of corporeal ageing in all known species, determining the rate and expression of ageing in multiple body systems. Thus, it is not surprising that insulin also plays an important role in brain ageing and cognitive decline that is associated with pathological brain ageing. Brain ageing is accompanied by reduced insulin effectiveness, either by an inadequate cellular response to insulin or by insulin deficiency attributable to reduced insulin transport across the blood-brain barrier. Age-associated brain insulin abnormalities may contribute to cognitive decline in ageing, as have been documented in older adults with Type 2 diabetes mellitus and hypertension. With more extreme pathology, brain insulin resistance may be associated with neurogenerative diseases such as Alzheimer's disease, and the condition which precedes Alzheimer's disease, known as amnestic mild cognitive impairment. In the following review, we discuss the mechanisms through which insulin resistance may induce or potentiate pathological brain ageing and thereby create a neurobiological environment that promotes neurodegeneration and associated cognitive decline. This topic is timely, given that insulin resistance-associated conditions such as diabetes and obesity have reached epidemic proportions. The prevalence of such chronic conditions, in combination with a rapidly ageing population, may result in a corresponding increase in the prevalence of Alzheimer's disease and other cognitive disorders. Fortunately, insulin resistance-associated conditions are amenable to both pharmacologic and lifestyle interventions that may reduce the deleterious impact of insulin resistance on the ageing brain.
Collapse
Affiliation(s)
- B Cholerton
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98108, USA
| | | | | |
Collapse
|
16
|
Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in Alzheimer's disease. Neuron 2009; 63:287-303. [PMID: 19679070 PMCID: PMC3044446 DOI: 10.1016/j.neuron.2009.06.026] [Citation(s) in RCA: 1070] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Revised: 06/22/2009] [Accepted: 06/25/2009] [Indexed: 01/17/2023]
Abstract
The epsilon4 allele of apolipoprotein E (APOE) is the major genetic risk factor for Alzheimer's disease (AD). Although there have been numerous studies attempting to elucidate the underlying mechanism for this increased risk, how apoE4 influences AD onset and progression has yet to be proven. However, prevailing evidence suggests that the differential effects of apoE isoforms on Abeta aggregation and clearance play the major role in AD pathogenesis. Other potential mechanisms, such as the differential modulation of neurotoxicity and tau phosphorylation by apoE isoforms as well as its role in synaptic plasticity and neuroinflammation, have not been ruled out. Inconsistent results among studies have made it difficult to define whether the APOE epsilon4 allele represents a gain of toxic function, a loss of neuroprotective function, or both. Therapeutic strategies based on apoE propose to reduce the toxic effects of apoE4 or to restore the physiological, protective functions of apoE.
Collapse
Affiliation(s)
- Jungsu Kim
- Department of Neurology, Developmental Biology, Hope Center for Neurological Disorders, Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
17
|
Abstract
Neuronal models for Alzheimer's disease research frequently have limitations as a result of their nonhuman origin and/or transformed state. Here we examined the potential of readily accessible neural crest-derived human epidermal melanocytes isolated from elderly individuals as a model system for Alzheimer's disease research. The amyloidogenic isoforms of amyloid precursor protein (APP; isoforms APP751/770) and amyloid beta (Abeta)1-40 were detected in epidermal melanocytes using immunocytochemistry and western blotting. Incubation of epidermal melanocytes with aggregated Abeta1-40 peptide caused a concentration-dependent reduction in cell viability, whereas age-matched dermal fibroblasts remained unaffected. These findings suggest that epidermal melanocytes from elderly donors are capable of amyloidogenesis and are sensitive to Abeta1-40 cytotoxicity. Thus, these cells may provide a readily accessible human cell model for Alzheimer's disease research.
Collapse
|
18
|
Cedazo-Mínguez A. Apolipoprotein E and Alzheimer's disease: molecular mechanisms and therapeutic opportunities. J Cell Mol Med 2008; 11:1227-38. [PMID: 18205697 PMCID: PMC4401287 DOI: 10.1111/j.1582-4934.2007.00130.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Multiple genetic and environmental factors are likely to contribute to the development of Alzheimer's disease (AD). The most important known risk factor for AD is presence of the E4 isoform of apolipoprotein E (apoE). Epidemiological studies demonstrated that apoE4 carriers have a higher risk and develop the disease and an early onset. Moreover, apoE4 is the only molecule that has been associated with all the biochemical disturbances characteristic of the disease: amyloid-beta (Abeta) deposition, tangle formation, oxidative stress, lipid homeostasis deregulation, synaptic plasticity loss and cholinergic dysfunction. This large body of evidence suggest that apoE is a key player in the pathogenesis of AD. This short review examines the current facts and hypotheses of the association between apoE4 and AD, as well as the therapeutic possibilities that apoE might offer for the treatment of this disease.
Collapse
Affiliation(s)
- Angel Cedazo-Mínguez
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, KI-Alzheimer's Disease Research Center, NOVUM, Stockholm, Sweden.
| |
Collapse
|
19
|
|
20
|
Parsons RB, Subramaniam D, Austen BM. A specific inhibitor of cholesterol biosynthesis, BM15.766, reduces the expression of ?-secretase and the production of amyloid-?in vitro. J Neurochem 2007; 102:1276-91. [PMID: 17472704 DOI: 10.1111/j.1471-4159.2007.04619.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have previously shown that statins reduce the production of amyloid-beta (Abeta) by both isoprenoid- and cholesterol-dependent mechanisms. These pathways contribute to the regulation of the dimerisation of BACE into its physiologically active form. Statins reduce cellular cholesterol levels by 20-40%; therefore, it is possible that the remaining cholesterol within the cell may play a significant role in the production of Abeta. Incubation of cells with the specific cholesterol biosynthesis inhibitor BM15.766 together with 50 micromol/L simvastatin and 400 micromol/L mevalonate reduced cellular cholesterol levels in a dose-dependent manner with increasing BM15.766 concentration (r = -0.9736, p = 0.0264). Furthermore, decreases in cellular cholesterol levels correlated with reductions in total Abeta production (r = 0.9683, p = 0.0317). A total of 2.5 micromol/L BM15.766 inhibited the dimerisation of BACE, whilst the expression of BACE monomer was reduced by 5 micromol/L BM15.766. BM15.766 treatment localised BACE predominantly within the Golgi, and reduced total BACE expression per cell. Similar changes were observed in the expression of the Golgi marker golgin-97, suggesting that reduced BACE expression may arise from a decrease in protein trafficking and an increase in degradation. By targeting cholesterol synthesis using specific cholesterol biosynthesis inhibitors, it is possible to reduce Abeta production without reducing protein isoprenylation.
Collapse
Affiliation(s)
- Richard B Parsons
- Department of Basic Medical Sciences, St. Georges, University of London, Cranmer Terrace, London, UK.
| | | | | |
Collapse
|
21
|
Grimm MOW, Grimm HS, Hartmann T. Amyloid beta as a regulator of lipid homeostasis. Trends Mol Med 2007; 13:337-44. [PMID: 17644432 DOI: 10.1016/j.molmed.2007.06.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 05/25/2007] [Accepted: 06/28/2007] [Indexed: 11/18/2022]
Abstract
The beta-amyloid peptide (A beta) is widely considered to be the molecule that causes Alzheimer's disease (AD). Besides this pathological function of A beta, recently published data reveal that A beta also has an essential physiological role in lipid homeostasis. Cholesterol increases A beta production, and conversely A beta production causes a decrease in cholesterol synthesis. The latter appears to be mediated by the inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR), a key enzyme in cholesterol synthesis, in an action similar to that of statins. Moreover, A beta regulates sphingolipid metabolism by directly activating sphingomyelinases (SMases). This review summarizes the molecular basis for the known physiological functions of A beta and amyloid precursor protein (APP), the roles of A beta and APP in lipid homeostasis and the medical implications of addressing lipid homeostasis in respect to AD. This knowledge might provide new insights for current and future therapeutic approaches to AD.
Collapse
Affiliation(s)
- Marcus O W Grimm
- Universität des Saarlandes, Kirrberger Str. 61.4, D-66421 Homburg, Germany
| | | | | |
Collapse
|
22
|
Zinser EG, Hartmann T, Grimm MOW. Amyloid beta-protein and lipid metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:1991-2001. [PMID: 17418089 DOI: 10.1016/j.bbamem.2007.02.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 02/14/2007] [Accepted: 02/15/2007] [Indexed: 01/16/2023]
Abstract
Lipids play an important part as risk or protective factors for Alzheimer's disease. This review summarizes the current findings in which lipids influence Alzheimer's disease and introduces the molecular mechanism how these lipids are linked to amyloid production. Besides the pathological impact of amyloid in Alzheimer's disease, amyloid has a physiological function in regulating lipid homeostasis in return. The understanding of the resulting regulatory cycles between amyloid precursor protein processing and lipids provides a platform for the development of new causal therapeutic approaches for Alzheimer's disease.
Collapse
Affiliation(s)
- Eva G Zinser
- Universität des Saarlandes, Uniklinikum Homburg, Neurobiologie, Neurologie, Gebäude 90, 66421 Homburg/Saar, Germany
| | | | | |
Collapse
|
23
|
Devi L, Prabhu BM, Galati DF, Avadhani NG, Anandatheerthavarada HK. Accumulation of amyloid precursor protein in the mitochondrial import channels of human Alzheimer's disease brain is associated with mitochondrial dysfunction. J Neurosci 2006; 26:9057-68. [PMID: 16943564 PMCID: PMC6675337 DOI: 10.1523/jneurosci.1469-06.2006] [Citation(s) in RCA: 613] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction is one of the major intracellular lesions of Alzheimer's disease (AD). However, the causative factors involved in the mitochondrial dysfunction in human AD are not well understood. Here we report that nonglycosylated full-length and C-terminal truncated amyloid precursor protein (APP) accumulates exclusively in the protein import channels of mitochondria of human AD brains but not in age-matched controls. Furthermore, in AD brains, mitochondrially associated APP formed stable approximately 480 kDa complexes with the translocase of the outer mitochondrial membrane 40 (TOM40) import channel and a super complex of approximately 620 kDa with both mitochondrial TOM40 and the translocase of the inner mitochondrial membrane 23 (TIM23) import channel TIM23 in an "N(in mitochondria)-C(out cytoplasm)" orientation. Accumulation of APP across mitochondrial import channels, which varied with the severity of AD, inhibited the entry of nuclear-encoded cytochrome c oxidase subunits IV and Vb proteins, which was associated with decreased cytochrome c oxidase activity and increased levels of H2O2. Regional distribution of mitochondrial APP showed higher levels in AD-vulnerable brain regions, such as the frontal cortex, hippocampus, and amygdala. Mitochondrial accumulation of APP was also observed in the cholinergic, dopaminergic, GABAergic, and glutamatergic neuronal types in the category III AD brains. The levels of translocationally arrested mitochondrial APP directly correlated with mitochondrial dysfunction. Moreover, apolipoprotein genotype analysis revealed that AD subjects with the E3/E4 alleles had the highest content of mitochondrial APP. Collectively, these results suggest that abnormal accumulation of APP across mitochondrial import channels, causing mitochondrial dysfunction, is a hallmark of human AD pathology.
Collapse
Affiliation(s)
- Latha Devi
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Badanavalu M. Prabhu
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Domenico F. Galati
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Narayan G. Avadhani
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | |
Collapse
|
24
|
Abstract
Tremendous progress has been made in understanding the processes of the Alzheimer's disease (AD) cascade, laying the groundwork for improvements in diagnosis and treatment. Advancement has been made in understanding the genetic basis of AD, with identification of causative genes for early-onset familial AD, and the role of the polymorphism of the APOE gene in the late-onset form of the disease. Understanding cerebral degeneration and accumulation of beta-amyloid has generated hopes for discovery of disease-modifying treatments. Progress is needed in understanding the mechanisms that link beta-amyloid accumulation and neuronal death. The next 5 years will be crucial in this respect.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Research and Development Department, Chiesi Farmaceutici, Parma, Italy
| | | | | |
Collapse
|
25
|
Nicotra A, Lupo G, Giurdanella G, Anfuso CD, Ragusa N, Tirolo C, Marchetti B, Alberghina M. MAPKs mediate the activation of cytosolic phospholipase A2 by amyloid β(25–35) peptide in bovine retina pericytes. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1733:172-86. [PMID: 15863364 DOI: 10.1016/j.bbalip.2004.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 10/15/2004] [Accepted: 12/28/2004] [Indexed: 10/25/2022]
Abstract
We have previously shown that, in bovine retina pericytes, amyloid beta(1-42) and its truncated form containing amino acids 25-35, after 24 h treatment, stimulate arachidonic acid (AA) release and phosphatidylcholine hydrolysis, by activation of both cytosolic (cPLA(2)) and Ca(2+)-independent (iPLA(2)) phospholipase A(2). A putative role for MAP kinases in this process emerged. Here we studied the role of the MAP-kinase family as well as both cPLA(2) and iPLA(2) mRNA expression by a semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) in the same sublethal model of amyloid-beta (Abeta) damage to pericytes in vitro. Abeta(25-35) peptide evoked AA release as well as stimulated phosphorylation of ERK1/2, p38 MAPKs and cPLA(2), but not c-Jun N-terminal kinase (JNK/SAPK). PD98059, an inhibitor of ERK-activating kinase MEK-1, and SB203580, an inhibitor of p38 protein kinase, abolished the stimulation of AA release and MAPK activities. In cells stimulated by Abeta(25-35) peptide, Western blotting and confocal microscopy analyses confirmed either an increase in the phosphorylated form of ERKs and p38 or their nuclear translocation. A complete inhibition of MAPK activation and AA release was also observed when pericytes were treated with GF109203X, a general PKC inhibitor, indicating the important role of both PKC and the two MAPKs in mediating the Abeta peptide response. Compared with samples untreated or treated with reverse Abeta(35-25) peptide, pretreatment with 50 microM Abeta(25-35) for 24 h significantly increased the level of constitutively expressed iPLA(2) mRNA by 25%, which seems to depend on the activation of kinases. By contrast, the level of cPLA(2) mRNA remained unchanged. Together, these data link either the stimulation of PKC-ERK-p38 cascades or PLA(2) activity by Abeta peptide to prooxidant mechanism induced by amyloid, which may initially stimulate the cell reaction as well as metabolic repair, such as during inflammation.
Collapse
Affiliation(s)
- Ambra Nicotra
- Department of Biochemistry, University of Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Zha Q, Ruan Y, Hartmann T, Beyreuther K, Zhang D. GM1 ganglioside regulates the proteolysis of amyloid precursor protein. Mol Psychiatry 2004; 9:946-52. [PMID: 15052275 DOI: 10.1038/sj.mp.4001509] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Plaques containing amyloid beta-peptides (Abeta) are a major feature in Alzheimer's disease (AD), and GM1 ganglioside is an important component of cellular plasma membranes and especially enriched in lipid raft. GM1-bound Abeta (GM1/Abeta), found in brains exhibiting early pathological changes of AD including diffuse plaques, has been suggested to be involved in the initiation of amyloid fibril formation in vivo by acting as a seed. However, the role of GM1 in amyloid beta-protein precursor (APP) processing is not yet defined. In this study, we report that exogenous GM1 ganglioside promotes Abeta biogenesis and decreases sAPPalpha secretion in SH-SY5Y and COS7 cells stably transfected with human APP695 cDNA without affecting full-length APP and the sAPPbeta levels. We also observe that GM1 increases extracellular levels of Abeta in primary cultures of mixed rat cortical neurons transiently transfected with human APP695 cDNA. These findings suggest a regulatory role for GM1 in APP processing pathways.
Collapse
Affiliation(s)
- Q Zha
- 1Department of Biochemistry, Institute of Mental Health, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
27
|
Palotás A, Pákáski M, Palotás M, Hugyecz M, Molnár J, Penke B, Janka Z, Kálmán J. Effect of haloperidol and risperidone on amyloid precursor protein levels in vivo. Brain Res Bull 2003; 62:93-9. [PMID: 14638382 DOI: 10.1016/j.brainresbull.2003.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The neurotoxic beta-amyloid peptide of Alzheimer's disease is formed from the amyloid precursor protein (APP), which is a member of an evolutionarily highly conserved gene family with significant functional importance. Because behavioral and psychiatric symptoms treated with antipsychotics may influence the course of the disease, we have investigated traditional and atypical antipsychotic drugs, administered through the intraperitoneal route, for their effects on rat cortical APP. Western-immunoblotting was utilized for semi-quantitative evaluation of APP levels. Treatment with haloperidol resulted in an acute elevation of cortical APP both in therapeutic and toxic doses, however, it had no significant chronic impact on APP. Atypical antipsychotic risperidone did not change cortical APP concentration. These results indicate that both haloperidol and risperidone are considered to be relatively safe with respect to APP metabolism. Possible mechanisms, including involvement of calcium and APP itself as a receptor, are discussed.
Collapse
Affiliation(s)
- András Palotás
- Department of Psychiatry, University of Szeged, Pécsi u. 4, Szeged H-6720, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Cedazo-Mínguez A, Popescu BO, Blanco-Millán JM, Akterin S, Pei JJ, Winblad B, Cowburn RF. Apolipoprotein E and β-amyloid (1-42) regulation of glycogen synthase kinase-3β. J Neurochem 2003; 87:1152-64. [PMID: 14622095 DOI: 10.1046/j.1471-4159.2003.02088.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glycogen synthase kinase-3beta (GSK-3beta) is implicated in regulating apoptosis and tau protein hyperphosphorylation in Alzheimer's disease (AD). We investigated the effects of two key AD molecules, namely apoE (E3 and E4 isoforms) and beta-amyloid (Abeta) 1-42 on GSK-3beta and its major upstream regulators, intracellular calcium and protein kinases C and B (PKC and PKB) in human SH-SY5Y neuroblastoma cells. ApoE3 induced a mild, transient, Ca2+-independent and early activation of GSK-3beta. ApoE4 effects were biphasic, with an early strong GSK-3beta activation that was partially dependent on extracellular Ca2+, followed by a GSK-3beta inactivation. ApoE4 also activated PKC-alpha and PKB possibly giving the subsequent GSK-3beta inhibition. Abeta(1-42) effects were also biphasic with a strong activation dependent partially on extracellular Ca2+ followed by an inactivation. Abeta(1-42) induced an early and potent activation of PKC-alpha and a late decrease of PKB activity. ApoE4 and Abeta(1-42) were more toxic than apoE3 as shown by MTT reduction assays and generation of activated caspase-3. ApoE4 and Abeta(1-42)-induced early activation of GSK-3beta could lead to apoptosis and tau hyperphosphorylation. A late inhibition of GSK-3beta through activation of upstream kinases likely compensates the effects of apoE4 and Abeta(1-42) on GSK-3beta, the unbalanced regulation of which may contribute to AD pathology.
Collapse
Affiliation(s)
- A Cedazo-Mínguez
- Neurotec, Section for Experimental Geriatrics, Karolinska Institutet, NOVUM, KFC, plan 4, S-141 86 Huddinge, Sweden.
| | | | | | | | | | | | | |
Collapse
|
29
|
Zhu Y, Hui DY. Apolipoprotein E binding to low density lipoprotein receptor-related protein-1 inhibits cell migration via activation of cAMP-dependent protein kinase A. J Biol Chem 2003; 278:36257-63. [PMID: 12857755 DOI: 10.1074/jbc.m303171200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Smooth muscle cell migration and proliferation contribute to neointimal hyperplasia and vascular stenosis after endothelial denudation. Previous studies revealed that apolipoprotein E (apoE) is an effective inhibitor of platelet-derived growth factor-directed smooth muscle cell migration and proliferation and that the anti-migratory function is mediated via apoE binding to low density lipoprotein receptor-related protein-1 (LRP-1). This study was undertaken to identify the intracellular pathway by which apoE binding to LRP-1 results in inhibition of smooth muscle cell migration. The results showed that apoE increased intracellular cAMP levels 3-fold after 5 min, and the increase was sustained for more than 1 h. As a consequence, apoE also increased protein kinase A (PKA) activity in smooth muscle cells. Importantly, suppression of PKA activity with a cell-permeable peptide inhibitor of PKA abolished the inhibitory effect of apoE on smooth muscle cell migration. These results indicated that apoE inhibition of smooth muscle cell migration is mediated via the activation of cAMP-dependent PKA. Additional experiments revealed that apoE also inhibited fibroblasts migration toward platelet-derived growth factor by a similar mechanism of cAMP-dependent PKA activation. It is noteworthy that apoE failed to increase cAMP levels or inhibit migration of LRP-1-negative mouse embryonic fibroblasts and LRP-1-deficient smooth muscle cells. Taken together, these findings established the mechanism by which apoE inhibits cell migration, i.e. via cAMP-dependent protein kinase A activation as a consequence of its binding to LRP-1.
Collapse
Affiliation(s)
- Yanjuan Zhu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267-0529, USA
| | | |
Collapse
|
30
|
Olsson A, Höglund K, Sjögren M, Andreasen N, Minthon L, Lannfelt L, Buerger K, Möller HJ, Hampel H, Davidsson P, Blennow K. Measurement of alpha- and beta-secretase cleaved amyloid precursor protein in cerebrospinal fluid from Alzheimer patients. Exp Neurol 2003; 183:74-80. [PMID: 12957490 DOI: 10.1016/s0014-4886(03)00027-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
One of the major histopathological hallmarks of Alzheimer's disease (AD) is redundant senile plaques mainly composed of beta-amyloid (Abeta) aggregates. Alternative cleavage of the amyloid precursor protein (APP), occurring in both normal and AD subjects, results in the generation and secretion of soluble APP (sAPP) and Abeta. We examined the cerebrospinal fluid (CSF) for alpha- and beta-secretase cleaved sAPP (alpha-sAPP and beta-sAPP) in 81 sporadic AD patients, 19 patients with mild cognitive impairment, and 42 healthy controls by using newly developed sandwich enzyme-linked immunosorbent assay methods. We found that neither the level of CSF-alpha-sAPP nor CSF-beta-sAPP differed between sporadic AD patients and healthy controls. These findings further support the conclusion that there is no change in APP expression in sporadic AD. However, the level of CSF-beta-sAPP was significantly increased in patients with mild cognitive impairment compared to controls. We also investigated the relationship between the CSF level of alpha/beta-sAPP and Abeta(42) and the apoE epsilon 4 (apoE4) allele. Significantly lower levels of CSF-alpha-sAPP were found in AD patients possessing one or two apoE4 alleles than in those not possessing the apoE4 allele. Neither the levels of CSF-beta-sAPP nor CSF-Abeta(42) differed when comparing ApoE4 allele-positive with allele-negative individuals.
Collapse
Affiliation(s)
- Annika Olsson
- Institute of Clinical Neuroscience, Experimental Neuroscience Section, Göteborg University, Sahlgrenska University Hospital/, Mölndal, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Pakaski M, Balaspiri L, Checler F, Kasa P. Human amyloid-beta causes changes in the levels of endothelial protein kinase C and its alpha isoform in vitro. Neurochem Int 2002; 41:409-14. [PMID: 12213228 DOI: 10.1016/s0197-0186(02)00026-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Amyloid-beta (A(beta)) deposits and neurofibrillary pathology are characteristic features of Alzheimer's disease (AD). The association of A(beta) with cerebral vessels is an intriguing feature of AD. While there is considerable evidence of altered activities of the major isoforms of protein kinase C (PKC) in the vasculature and neurons of AD brains, little is known about the relationship between the Abeta toxicity and the altered PKC levels in cerebral endothelial cells. In this study, cultured brain endothelial cells exposed to A(beta)1-40 revealed a translocation of PKC from the membrane fraction to the cytosol. The content of the isoform PKC(alpha), involved in the regulation of amyloid precursor protein (APP) secretion, was decreased in the membrane-bound fraction of rat endothelial cells and increased in the cytosol after A(beta)1-40 treatment. These data suggest that the accumulation of A(beta) peptide in the cerebral vasculature may play a significant role in the down-regulation of PKC seen in the AD cerebral vasculature.
Collapse
Affiliation(s)
- Magdolna Pakaski
- Department of Psychiatry, Alzheimer's Disease Research Centre, University of Szeged, Somogyi 4, H-6720 Szeged, Hungary.
| | | | | | | |
Collapse
|
32
|
Villa A, Santiago J, García-Silva S, Ruiz-León Y, Pascual A. Serum is required for release of Alzheimer's amyloid precursor protein in neuroblastoma cells. Neurochem Int 2002; 41:261-9. [PMID: 12106777 DOI: 10.1016/s0197-0186(02)00019-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The beta-amyloid peptide, the major component of the senile plaques that characterize Alzheimer's disease, is generated from a set of alternatively spliced beta-amyloid precursor proteins (APPs), which are proteolytically cleaved by the action of a set of enzymes referred to generically as secretases. The major processing pathway involves the proteolytic cleavage of APP by alpha-secretase and results in the release of soluble non-amyloidogenic full-length amino terminal fragments (sAPP), which appear to be involved in neurotrophic events. A reduced production of these neuroprotective sAPP would contribute, together with deposition of the beta-amyloid peptide, to the neurodegenerative processes that lead to the cellular death in Alzheimer's disease. In the present work, we describe a dramatic reduction of sAPP content in medium conditioned by neuronal cells grown under low-serum conditions, when compared with the levels released in the presence of 10% serum. The inhibitory effect on sAPP release appears to be quite specific since that reduction occurs without major changes in cell proliferation, expression of APP-mRNA or intracellular APP levels. Under low-serum conditions, cells showed a more differentiated morphology and no apoptotic signs were observed. Since the alpha-secretase has been described as a membrane anchored protein, our results suggest that the serum contains an essential factor(s) involved in the alpha-secretase activity.
Collapse
Affiliation(s)
- Ana Villa
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas, Arturo Duperier 4, 28029 Madrid, Spain
| | | | | | | | | |
Collapse
|
33
|
Bisaglia M, Venezia V, Piccioli P, Stanzione S, Porcile C, Russo C, Mancini F, Milanese C, Schettini G. Acetaminophen protects hippocampal neurons and PC12 cultures from amyloid beta-peptides induced oxidative stress and reduces NF-kappaB activation. Neurochem Int 2002; 41:43-54. [PMID: 11918971 DOI: 10.1016/s0197-0186(01)00136-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present findings show that an atypical non-steroidal anti-inflammatory drug, such as acetaminophen, retains the ability to recover amyloid beta-peptides driven neuronal apoptosis through the impairment of oxidative stress. Moreover, this compound reduces the increased NF-kappaB binding activity, which occurs in these degenerative conditions. Therapeutic interventions aimed at reducing the inflammatory response in Alzheimer's disease (AD) recently suggested the application of non-steroidal anti-inflammatory drugs. Although the anti-inflammatory properties of acetaminophen are controversial, it emerged that in an amyloid-driven astrocytoma cell degeneration model acetaminophen proved to be effective. On these bases, we analyzed the role of acetaminophen against the toxicity exerted by different Abeta-peptides on rat primary hippocampal neurons and on a rat pheochromocytoma cell line. We found a consistent protection from amyloid beta-fragments 1-40 and 1-42-induced impairment of mitochondrial redox activity on both cell cultures, associated with a marked reduction of apoptotic nuclear fragmentation. An antioxidant component of the protective activity emerged from the analysis of the reduction of phospholipid peroxidation, and also from a significant reduction of cytoplasmic accumulation of peroxides in the pheochromocytoma cell line. Moreover, activation of NF-kappaB by amyloid-derived peptides was greatly impaired by acetaminophen pre-treatment in hippocampal cells. This evidence points out antioxidant and anti-transcriptional properties of acetaminophen besides the known capability to interfere with inflammation within the central nervous system, and suggests that it can be exploited as a possible therapeutic approach against AD.
Collapse
Affiliation(s)
- M Bisaglia
- Pharmacology and Neuroscience, National Cancer Research Institute c/o Advanced Biotechnology Centre, L.go R. Benzi 10, 16132 Genova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cedazo-Mínguez A, Cowburn RF. Apolipoprotein E isoform-specific disruption of phosphoinositide hydrolysis: protection by estrogen and glutathione. FEBS Lett 2001; 504:45-9. [PMID: 11522294 DOI: 10.1016/s0014-5793(01)02761-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The mechanism(s) by which the E4 isoform of apolipoprotein E (apoE4) influences Alzheimer's disease (AD) are not fully known. We report that apoE4, but not apoE3, disrupts carbachol-stimulated phosphoinositide (PI) hydrolysis in SH-SY5Y neuroblastoma cells. Carbachol responses were also disrupted by beta-amyloid (Abeta) (1-42) and apoE4/Abeta(1-42) complexes, but not by apoE3/Abeta(1-42). Glutathione and estrogen protected against apoE4 and Abeta(1-42) effects, as well as those of H(2)O(2). Estrogen protection was partially blocked by wortmannin, suggesting the involvement of phosphatidylinositol 3-kinase. An apoE4-induced disruption of acetylcholine muscarinic receptor-mediated signalling may explain the lower effectiveness of cholinergic replacement treatments in apoE4 AD patients. Also, the beneficial effect of estrogen in AD may be partially due to its ability to protect against apoE4- and Abeta(1-42)-mediated disruption of PI hydrolysis.
Collapse
Affiliation(s)
- A Cedazo-Mínguez
- Karolinska Institutet, NEUROTEC, Division of Experimental Geriatrics, NOVUM, KFC, 141 86, Huddinge, Sweden.
| | | |
Collapse
|
35
|
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder with multiple etiologies. The presence of the E4 isoform of apolipoprotein E (apoE) has been shown to increase the risk and to decrease the age of onset for AD and is the major susceptibility factor known for the disease. ApoE4 has been shown to intensify all the biochemical disturbances characteristic of AD, including beta amyloid (Abeta) deposition, tangle formation, neuronal cell death, oxidative stress, synaptic plasticity and dysfunctions of lipid homeostasis and cholinergic signalling. In contrast, other apoE isoforms are protective. Here we review and discuss these major hypotheses of the apoE4-AD association.
Collapse
Affiliation(s)
- A Cedazo-Mínguez
- Karolinska Institutet, NEUROTEC, Division of Experimental Geriatrics, Novum, KFC, Huddinge, Stockholm, Sweden.
| | | |
Collapse
|