1
|
Turton SM, Padgett S, Maisel MT, Johnson CE, Buzinova VA, Barth SE, Kohler K, Spearman HM, Macheda T, Manauis EC, Guo LZ, Whitlock HR, Bachstetter AD, Sunderam S, O'Hara BF, Duncan MJ, Murphy MP. Interactions between daily sleep-wake rhythms, γ-secretase, and amyloid-β peptide pathology point to complex underlying relationships. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167840. [PMID: 40222459 DOI: 10.1016/j.bbadis.2025.167840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Disrupted or insufficient sleep is a well-documented risk factor for Alzheimer's disease (AD) and related dementias. Previous studies in our lab and others have shown that chronic fragmentation of the daily sleep-wake rhythm in mice can accelerate the development of AD-related neuropathology in the brain, including increases in the levels of amyloid-β (Aβ). Although sleep is known to increase clearance of Aβ via the glymphatic system, little is known about the effect of sleep on Aβ production and the role this might play in amyloid deposition. To examine the relationship of Aβ production and its interaction with sleep and sleep dysfunction, we treated mice from an APP × PS1 mutant knock-in line (APPΔNLh/ΔNLh × PS1P264L/P264L) with an inhibitor of γ-secretase (LY-450,139; Semagacestat®) during a protocol of mild sleep fragmentation (SF). Compared to the male mice, the female mice slept less, and had more Aβ pathology. Semagacestat treatment reduced Aβ, but only in the most soluble extractable fraction. Although the female mice showed an increase in the amount of Aβ following SF, this effect was blocked by Semagacestat, an effect that was not seen in the male mice. SF also led to a significant, sex-dependent changes in the relative amounts of C-terminal fragments of the amyloid precursor protein, the immediate substrate of the γ-secretase enzyme. These findings indicate that the relationship between disruption of the daily sleep-wake rhythm and the development of AD-related pathology is complex, and may involve unappreciated interactions with biological sex. Consideration of these factors is necessary for a better understanding of AD risk, especially the elevated risk in women.
Collapse
Affiliation(s)
| | | | | | - Carrie E Johnson
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA
| | - Valeria A Buzinova
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA
| | | | | | | | | | | | | | | | - Adam D Bachstetter
- The Sanders-Brown Center on Aging, USA; The Spinal Cord and Brain Injury Research Center, USA; Department of Neuroscience, USA
| | | | | | | | - M Paul Murphy
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA.
| |
Collapse
|
2
|
Peng L, Zhang Z, Li Q, Song Z, Yan C, Ling H. Unveiling the multifaceted pathogenesis and therapeutic drugs of Alzheimer's disease: A comprehensive review. Heliyon 2024; 10:e39217. [PMID: 39629139 PMCID: PMC11612466 DOI: 10.1016/j.heliyon.2024.e39217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/02/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder characterized by the accumulation of β-amyloid (Aβ) plaques and tau phosphorylation-induced neurofibrillary tangles. This review comprehensively summarizes AD pathogenesis and related factors, drawing on a wealth of authoritative reports and research findings. Specifically, we delve into the intricate mechanisms underlying AD pathology, including Aβ deposition, tau protein phosphorylation, cholinergic dysfunction, neuroinflammation, mitochondrial oxidative stress, ferroptosis, imbalance in the gut microbiota, and microRNA dysregulation. We also explored the effects of these factors on the brain, including synaptic damage and cognitive impairment. Moreover, our review highlights the associations between the pathogenesis of AD and inflammatory cytokines in the peripheral blood and cerebrospinal fluid, dysbiosis of the gut microbiota, and changes in microRNA expression. Overall, we provided a systematic and illustrative overview of the pathogenesis and therapeutic drugs for AD, offering help in the prevention and treatment of this condition.
Collapse
Affiliation(s)
- Liting Peng
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Zhiming Zhang
- Department of Anesthesiology, The First People's Hospital of Chenzhou, The Chenzhou Affiliated Hospital, Hengyang Medical School, University of South China, Chenzhou, 423000, Hunan, China
| | - Qi Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Zhenjiang Song
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Canqun Yan
- The Health Management Center, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hongyan Ling
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| |
Collapse
|
3
|
Pei J, Kinch LN, Cong Q. Computational analysis of propeptide-containing proteins and prediction of their post-cleavage conformation changes. Proteins 2024; 92:1206-1219. [PMID: 38775337 DOI: 10.1002/prot.26702] [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: 01/23/2024] [Revised: 04/10/2024] [Accepted: 04/29/2024] [Indexed: 10/26/2024]
Abstract
A propeptide is removed from a precursor protein to generate its active or mature form. Propeptides play essential roles in protein folding, transportation, and activation and are present in about 2.3% of reviewed proteins in the UniProt database. They are often found in secreted or membrane-bound proteins including proteolytic enzymes, hormones, and toxins. We identified a variety of globular and nonglobular Pfam domains in protein sequences designated as propeptides, some of which form intramolecular interactions with other domains in the mature proteins. Propeptide-containing enzymes mostly function as proteases, as they are depleted in other enzyme classes such as hydrolases acting on DNA and RNA, isomerases, and lyases. We applied AlphaFold to generate structural models for over 7000 proteins with propeptides having no less than 20 residues. Analysis of residue contacts in these models revealed conformational changes for over 300 proteins before and after the cleavage of the propeptide. Examples of conformation change occur in several classes of proteolytic enzymes in the families of subtilisins, trypsins, aspartyl proteases, and thermolysin-like metalloproteases. In most of the observed cases, cleavage of the propeptide releases the constraints imposed by the covalent bond between the propeptide and the mature protein, and cleavage enables stronger interactions between the propeptide and the mature protein. These findings suggest that post-cleavage propeptides could play critical roles in regulating the activity of mature proteins.
Collapse
Affiliation(s)
- Jimin Pei
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lisa N Kinch
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
4
|
Murray A, Muñiz-García A, Alić I, Nižetić D. It's good to know what to BACE the specificity of your inhibitors on. J Clin Invest 2024; 134:e183677. [PMID: 39145447 PMCID: PMC11324289 DOI: 10.1172/jci183677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024] Open
Abstract
Production, aggregation, and clearance of the amyloid β peptide (Aβ) are important processes governing the initial pathogenesis of Alzheimer's disease (AD). Inhibition of β-site amyloid precursor protein (APP) cleaving enzyme (BACE1) (one of two key proteases responsible for Aβ production) as an AD-therapeutic approach so far has failed to yield a successful drug. BACE1 and its homologue BACE2 are frequently inhibited by the same inhibitors. Several genetic and cerebral organoid modeling studies suggest that BACE2 has dose-dependent AD-suppressing activity, which makes its unwanted inhibition potentially counterproductive for AD treatment. The in vivo effects of an unwanted cross inhibition of BACE2 have so far been impossible to monitor because of the lack of an easily accessible pharmacodynamic marker specific for BACE2 cleavage. In this issue of the JCI, work led by Stefan F. Lichtenthaler identifies soluble VEGFR3 (sVEGFR3) as a pharmacodynamic plasma marker for BACE2 activity not shared with BACE1.
Collapse
Affiliation(s)
- Aoife Murray
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ana Muñiz-García
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ivan Alić
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Dean Nižetić
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
5
|
Alldred MJ, Pidikiti H, Ibrahim KW, Lee SH, Heguy A, Hoffman GE, Roussos P, Wisniewski T, Wegiel J, Stutzmann GE, Mufson EJ, Ginsberg SD. Analysis of microisolated frontal cortex excitatory layer III and V pyramidal neurons reveals a neurodegenerative phenotype in individuals with Down syndrome. Acta Neuropathol 2024; 148:16. [PMID: 39105932 PMCID: PMC11578391 DOI: 10.1007/s00401-024-02768-0] [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: 03/04/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 08/07/2024]
Abstract
We elucidated the molecular fingerprint of vulnerable excitatory neurons within select cortical lamina of individuals with Down syndrome (DS) for mechanistic understanding and therapeutic potential that also informs Alzheimer's disease (AD) pathophysiology. Frontal cortex (BA9) layer III (L3) and layer V (L5) pyramidal neurons were microisolated from postmortem human DS and age- and sex-matched controls (CTR) to interrogate differentially expressed genes (DEGs) and key biological pathways relevant to neurodegenerative programs. We identified > 2300 DEGs exhibiting convergent dysregulation of gene expression in both L3 and L5 pyramidal neurons in individuals with DS versus CTR subjects. DEGs included over 100 triplicated human chromosome 21 genes in L3 and L5 neurons, demonstrating a trisomic neuronal karyotype in both laminae. In addition, thousands of other DEGs were identified, indicating gene dysregulation is not limited to trisomic genes in the aged DS brain, which we postulate is relevant to AD pathobiology. Convergent L3 and L5 DEGs highlighted pertinent biological pathways and identified key pathway-associated targets likely underlying corticocortical neurodegeneration and related cognitive decline in individuals with DS. Select key DEGs were interrogated as potential hub genes driving dysregulation, namely the triplicated DEGs amyloid precursor protein (APP) and superoxide dismutase 1 (SOD1), along with key signaling DEGs including mitogen activated protein kinase 1 and 3 (MAPK1, MAPK3) and calcium calmodulin dependent protein kinase II alpha (CAMK2A), among others. Hub DEGs determined from multiple pathway analyses identified potential therapeutic candidates for amelioration of cortical neuron dysfunction and cognitive decline in DS with translational relevance to AD.
Collapse
Affiliation(s)
- Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Harshitha Pidikiti
- Center for Dementia Research, Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Kyrillos W Ibrahim
- Center for Dementia Research, Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
| | - Sang Han Lee
- Center for Dementia Research, Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Adriana Heguy
- Genome Technology Center, New York University Grossman School of Medicine, New York, NY, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry and the Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry and the Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Wisniewski
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Jerzy Wegiel
- Department of Developmental Neurobiology, Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Grace E Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University/The Chicago Medical School, North Chicago, IL, USA
| | - Elliott J Mufson
- Department of Translational Neuroscience and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, 10962, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Murphy MP, Buzinova VA, Johnson CE. The amyloid-β peptide: Guilty as charged? Biochim Biophys Acta Mol Basis Dis 2024; 1870:166945. [PMID: 37935338 PMCID: PMC10842071 DOI: 10.1016/j.bbadis.2023.166945] [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: 07/27/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023]
Abstract
Recent years have seen both considerable progress and controversy in the Alzheimer's disease (AD) field. After decades of slow to negligible movement towards the development of disease modifying therapies, promising outcomes in recent clinical trials with several monoclonal antibodies targeting various forms of the amyloid-β (Aβ) peptide have at last opened a possible way forward. In fact, at this point multiple anti-Aβ therapeutics are close to receiving (or have already received) regulatory approval. Although these outcomes are not without some degree of divisiveness, the fact remains that targeting amyloid for removal has finally shown at least modest efficacy in slowing the otherwise relentless progression of the disease. Although the validation of the long standing amyloid cascade hypothesis would seem to be at hand, what remains is the puzzling issue of why - if Aβ indeed causes AD - does removing it from the brain not stop the disease entirely.
Collapse
Affiliation(s)
- M Paul Murphy
- Department of Molecular and Cellular Biochemistry and the Sanders-Brown Center on Aging University of Kentucky, 789 S. Limestone Street, Lexington, KY 40536, USA.
| | - Valeria A Buzinova
- Department of Molecular and Cellular Biochemistry and the Sanders-Brown Center on Aging University of Kentucky, 789 S. Limestone Street, Lexington, KY 40536, USA
| | - Carrie E Johnson
- Department of Molecular and Cellular Biochemistry and the Sanders-Brown Center on Aging University of Kentucky, 789 S. Limestone Street, Lexington, KY 40536, USA
| |
Collapse
|
7
|
Rudisch DM, Krasko MN, Barnett DGS, Mueller KD, Russell JA, Connor NP, Ciucci MR. Early ultrasonic vocalization deficits and related thyroarytenoid muscle pathology in the transgenic TgF344-AD rat model of Alzheimer's disease. Front Behav Neurosci 2024; 17:1294648. [PMID: 38322496 PMCID: PMC10844490 DOI: 10.3389/fnbeh.2023.1294648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/01/2023] [Indexed: 02/08/2024] Open
Abstract
Background Alzheimer's disease (AD) is a progressive neurologic disease and the most common cause of dementia. Classic pathology in AD is characterized by inflammation, abnormal presence of tau protein, and aggregation of β-amyloid that disrupt normal neuronal function and lead to cell death. Deficits in communication also occur during disease progression and significantly reduce health, well-being, and quality of life. Because clinical diagnosis occurs in the mid-stage of the disease, characterizing the prodrome and early stages in humans is currently challenging. To overcome these challenges, we use the validated TgF344-AD (F344-Tg(Prp-APP, Prp-PS1)19/Rrrc) transgenic rat model that manifests cognitive, behavioral, and neuropathological dysfunction akin to AD in humans. Objectives The overarching goal of our work is to test the central hypothesis that pathology and related behavioral deficits such as communication dysfunction in part manifest in the peripheral nervous system and corresponding target tissues already in the early stages. The primary aims of this study are to test the hypotheses that: (1) changes in ultrasonic vocalizations (USV) occur in the prodromal stage at 6 months of age and worsen at 9 months of age, (2) inflammation as well as AD-related pathology can be found in the thyroarytenoid muscle (TA) at 12 months of age (experimental endpoint tissue harvest), and to (3) demonstrate that the TgF344-AD rat model is an appropriate model for preclinical investigations of early AD-related vocal deficits. Methods USVs were collected from male TgF344-AD (N = 19) and wildtype (WT) Fischer-344 rats (N = 19) at 6 months (N = 38; WT: n = 19; TgF344-AD: n = 19) and 9 months of age (N = 18; WT: n = 10; TgF344-AD: n = 8) and acoustically analyzed for duration, mean power, principal frequency, low frequency, high frequency, peak frequency, and call type. RT-qPCR was used to assay peripheral inflammation and AD-related pathology via gene expressions in the TA muscle of male TgF344-AD rats (n = 6) and WT rats (n = 6) at 12 months of age. Results This study revealed a significant reduction in mean power of ultrasonic calls from 6 to 9 months of age and increased peak frequency levels over time in TgF344-AD rats compared to WT controls. Additionally, significant downregulation of AD-related genes Uqcrc2, Bace2, Serpina3n, and Igf2, as well as downregulation of pro-inflammatory gene Myd88 was found in the TA muscle of TgF344-AD rats at 12 months of age. Discussion Our findings demonstrate early and progressive vocal deficits in the TgF344-AD rat model. We further provide evidence of dysregulation of AD-pathology-related genes as well as inflammatory genes in the TA muscles of TgF344-AD rats in the early stage of the disease, confirming this rat model for early-stage investigations of voice deficits and related pathology.
Collapse
Affiliation(s)
- Denis Michael Rudisch
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- UW Institute for Clinical and Translational Research, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Maryann N Krasko
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - David G S Barnett
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Kimberly D Mueller
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - John A Russell
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Nadine P Connor
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Michelle R Ciucci
- Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, WI, United States
- Department of Surgery, Division of Otolaryngology - Head and Neck Surgery, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
8
|
Kohler K, Macheda T, Hobbs MM, Maisel MT, Rodriguez A, Farris L, Wessel CR, Infantino C, Niedowicz DM, Helman AM, Beckett TL, Unrine JM, Murphy MP. Exposure to Lead in Drinking Water Causes Cognitive Impairment via an Alzheimer's Disease Gene-Dependent Mechanism in Adult Mice. J Alzheimers Dis 2024; 100:S291-S304. [PMID: 39121129 PMCID: PMC11616619 DOI: 10.3233/jad-240640] [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] [Indexed: 08/11/2024]
Abstract
Background Exposure to lead (Pb) is a major public health problem that could occur through contaminated soil, air, food, or water, either during the course of everyday life, or while working in hazardous occupations. Although Pb has long been known as a neurodevelopmental toxicant in children, a recent and growing body of epidemiological research indicates that cumulative, low-level Pb exposure likely drives age-related neurologic dysfunction in adults. Environmental Pb exposure in adulthood has been linked to risk of late-onset Alzheimer's disease (AD) and dementia. Objective Although the biological mechanism underlying this link is unknown, it has been proposed that Pb exposure may increase the risk of AD via altering the expression of AD-related genes and, possibly, by activating the molecular pathways underlying AD-related pathology. Methods We investigated Pb exposure using a line of genetically modified mice with AD-causing knock-in mutations in the amyloid precursor protein and presenilin 1 (APPΔNL/ΔNL x PS1P264L/P264L) that had been crossed with Leprdb/db mice to impart vulnerability to vascular pathology. Results Our data show that although Pb exposure in adult mice impairs cognitive function, this effect is not related to either an increase in amyloid pathology or to changes in the expression of common AD-related genes. Pb exposure also caused a significant increase in blood pressure, a well known effect of Pb. Interestingly, although the increase in blood pressure was unrelated to genotype, only mice that carried AD-related mutations developed cognitive dysfunction, in spite of showing no significant change in cerebrovascular pathology. Conclusions These results raise the possibility that the increased risk of dementia associated with Pb exposure in adults may be tied to its subsequent interaction with either pre-existing or developing AD-related neuropathology.
Collapse
Affiliation(s)
- Katharina Kohler
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Teresa Macheda
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Misty M. Hobbs
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - M. Tyler Maisel
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Antonela Rodriguez
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Lindsey Farris
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Caitlin R. Wessel
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | | | - Dana M. Niedowicz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Alex M. Helman
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Tina L. Beckett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
| | - Jason M. Unrine
- Department of Plant and Soil Sciences, University of Kentucky Martin-Gatton College of Agriculture, Food, and Environment, Lexington, KY, U.S.A
- Kentucky Water Research Institute, University of Kentucky, Lexington, Kentucky, USA
| | - M. Paul Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, U.S.A
- Kentucky Water Research Institute, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
9
|
Ohno M. A Strategy for Allowing Earlier Diagnosis and Rigorous Evaluation of BACE1 Inhibitors in Preclinical Alzheimer's Disease. J Alzheimers Dis 2024; 99:431-445. [PMID: 38701146 DOI: 10.3233/jad-231451] [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] [Indexed: 05/05/2024]
Abstract
Given continued failure of BACE1 inhibitor programs at symptomatic and prodromal stages of Alzheimer's disease (AD), clinical trials need to target the earlier preclinical stage. However, trial design is complex in this population with negative diagnosis of classical hippocampal amnesia on standard memory tests. Besides recent advances in brain imaging, electroencephalogram, and fluid-based biomarkers, new cognitive markers should be established for earlier diagnosis that can optimize recruitment to BACE1 inhibitor trials in presymptomatic AD. Notably, accelerated long-term forgetting (ALF) is emerging as a sensitive cognitive measure that can discriminate between asymptomatic individuals with high risks for developing AD and healthy controls. ALF is a form of declarative memory impairment characterized by increased forgetting rates over longer delays (days to months) despite normal storage within the standard delays of testing (20-60 min). Therefore, ALF may represent a harbinger of preclinical dementia and the impairment of systems memory consolidation, during which memory traces temporarily stored in the hippocampus become gradually integrated into cortical networks. This review provides an overview of the utility of ALF in a rational design of next-generation BACE1 inhibitor trials in preclinical AD. I explore potential mechanisms underlying ALF and relevant early-stage biomarkers useful for BACE1 inhibitor evaluation, including synaptic protein alterations, astrocytic dysregulation and neuron hyperactivity in the hippocampal-cortical network. Furthermore, given the physiological role of the isoform BACE2 as an AD-suppressor gene, I also discuss the possible association between the poor selectivity of BACE1 inhibitors and their side effects (e.g., cognitive worsening) in prior clinical trials.
Collapse
Affiliation(s)
- Masuo Ohno
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
10
|
Yeap YJ, Kandiah N, Nizetic D, Lim KL. BACE2: A Promising Neuroprotective Candidate for Alzheimer's Disease. J Alzheimers Dis 2023; 94:S159-S171. [PMID: 36463454 PMCID: PMC10473127 DOI: 10.3233/jad-220867] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia that affects millions of predominantly elderly individuals worldwide. Despite intensive research over several decades, controversies still surround the etiology of AD and the disease remains incurable. Meanwhile, new molecular players of the central amyloid cascade hypothesis have emerged and among these is a protease known as β-site APP cleavage enzyme 2 (BACE2). Unlike BACE1, BACE2 cleaves the amyloid-β protein precursor within the Aβ domain that accordingly prevents the generation of Aβ42 peptides, the aggregation of which is commonly regarded as the toxic entity that drives neurodegeneration in AD. Given this non-amyloidogenic role of BACE2, it is attractive to position BACE2 as a therapeutic target for AD. Indeed, several groups including ours have demonstrated a neuroprotective role for BACE2 in AD. In this review, we discuss emerging evidence supporting the ability of BACE2 in mitigating AD-associated pathology in various experimental systems including human pluripotent stem cell-derived cerebral organoid disease models. Alongside this, we also provide an update on the identification of single nucleotide polymorphisms occurring in the BACE2 gene that are linked to increased risk and earlier disease onset in the general population. In particular, we highlight a recently identified point mutation on BACE2 that apparently leads to sporadic early-onset AD. We believe that a better understanding of the role of BACE2 in AD would provide new insights for the development of viable therapeutic strategies for individuals with dementia.
Collapse
Affiliation(s)
- Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nagaendran Kandiah
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Dean Nizetic
- Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kah-Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Brain Sciences, Imperial College London, London, UK
- Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
11
|
Hampel H, Caruso G, Nisticò R, Piccioni G, Mercuri NB, Giorgi FS, Ferrarelli F, Lemercier P, Caraci F, Lista S, Vergallo A. Biological Mechanism-based Neurology and Psychiatry: A BACE1/2 and Downstream Pathway Model. Curr Neuropharmacol 2023; 21:31-53. [PMID: 34852743 PMCID: PMC10193755 DOI: 10.2174/1570159x19666211201095701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 02/04/2023] Open
Abstract
In oncology, comprehensive omics and functional enrichment studies have led to an extensive profiling of (epi)genetic and neurobiological alterations that can be mapped onto a single tumor's clinical phenotype and divergent clinical phenotypes expressing common pathophysiological pathways. Consequently, molecular pathway-based therapeutic interventions for different cancer typologies, namely tumor type- and site-agnostic treatments, have been developed, encouraging the real-world implementation of a paradigm shift in medicine. Given the breakthrough nature of the new-generation translational research and drug development in oncology, there is an increasing rationale to transfertilize this blueprint to other medical fields, including psychiatry and neurology. In order to illustrate the emerging paradigm shift in neuroscience, we provide a state-of-the-art review of translational studies on the β-site amyloid precursor protein cleaving enzyme (BACE) and its most studied downstream effector, neuregulin, which are molecular orchestrators of distinct biological pathways involved in several neurological and psychiatric diseases. This body of data aligns with the evidence of a shared genetic/biological architecture among Alzheimer's disease, schizoaffective disorder, and autism spectrum disorders. To facilitate a forward-looking discussion about a potential first step towards the adoption of biological pathway-based, clinical symptom-agnostic, categorization models in clinical neurology and psychiatry for precision medicine solutions, we engage in a speculative intellectual exercise gravitating around BACE-related science, which is used as a paradigmatic case here. We draw a perspective whereby pathway-based therapeutic strategies could be catalyzed by highthroughput techniques embedded in systems-scaled biology, neuroscience, and pharmacology approaches that will help overcome the constraints of traditional descriptive clinical symptom and syndrome-focused constructs in neurology and psychiatry.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | | | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- School of Pharmacy, University of Rome “Tor Vergata”, Rome, Italy
| | - Gaia Piccioni
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- Department of Physiology and Pharmacology “V.Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Nicola B. Mercuri
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- IRCCS Santa Lucia Foundation, Rome, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Pablo Lemercier
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, Troina, Italy
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Simone Lista
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
- Memory Resources and Research Center (CMRR), Neurology Department, Gui de Chauliac University Hospital, Montpellier, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| |
Collapse
|
12
|
Bergstedt J, Azzou SAK, Tsuo K, Jaquaniello A, Urrutia A, Rotival M, Lin DTS, MacIsaac JL, Kobor MS, Albert ML, Duffy D, Patin E, Quintana-Murci L. The immune factors driving DNA methylation variation in human blood. Nat Commun 2022; 13:5895. [PMID: 36202838 PMCID: PMC9537159 DOI: 10.1038/s41467-022-33511-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/21/2022] [Indexed: 11/08/2022] Open
Abstract
Epigenetic changes are required for normal development, yet the nature and respective contribution of factors that drive epigenetic variation in humans remain to be fully characterized. Here, we assessed how the blood DNA methylome of 884 adults is affected by DNA sequence variation, age, sex and 139 factors relating to life habits and immunity. Furthermore, we investigated whether these effects are mediated or not by changes in cellular composition, measured by deep immunophenotyping. We show that DNA methylation differs substantially between naïve and memory T cells, supporting the need for adjustment on these cell-types. By doing so, we find that latent cytomegalovirus infection drives DNA methylation variation and provide further support that the increased dispersion of DNA methylation with aging is due to epigenetic drift. Finally, our results indicate that cellular composition and DNA sequence variation are the strongest predictors of DNA methylation, highlighting critical factors for medical epigenomics studies.
Collapse
Affiliation(s)
- Jacob Bergstedt
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France.
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Sadoune Ait Kaci Azzou
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
| | - Kristin Tsuo
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
| | - Anthony Jaquaniello
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
| | | | - Maxime Rotival
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
| | - David T S Lin
- Edwin S.H. Leong Healthy Aging Program, Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Julia L MacIsaac
- Edwin S.H. Leong Healthy Aging Program, Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Michael S Kobor
- Edwin S.H. Leong Healthy Aging Program, Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | | | - Darragh Duffy
- Institut Pasteur, Université Paris Cité, Translational Immunology Unit, Institut Pasteur, Paris, France
| | - Etienne Patin
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France.
| | - Lluís Quintana-Murci
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France.
- Chair of Human Genomics and Evolution, Collège de France, Paris, France.
| |
Collapse
|
13
|
Mumford P, Tosh J, Anderle S, Gkanatsiou Wikberg E, Lau G, Noy S, Cleverley K, Saito T, Saido TC, Yu E, Brinkmalm G, Portelius E, Blennow K, Zetterberg H, Tybulewicz V, Fisher EMC, Wiseman FK. Genetic Mapping of APP and Amyloid-β Biology Modulation by Trisomy 21. J Neurosci 2022; 42:6453-6468. [PMID: 35835549 PMCID: PMC9398545 DOI: 10.1523/jneurosci.0521-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/03/2022] [Accepted: 06/18/2022] [Indexed: 12/04/2022] Open
Abstract
Individuals who have Down syndrome (DS) frequently develop early onset Alzheimer's disease (AD), a neurodegenerative condition caused by the buildup of aggregated amyloid-β (Aβ) and tau proteins in the brain. Aβ is produced by amyloid precursor protein (APP), a gene located on chromosome 21. People who have DS have three copies of chromosome 21 and thus also an additional copy of APP; this genetic change drives the early development of AD in these individuals. Here we use a combination of next-generation mouse models of DS (Tc1, Dp3Tyb, Dp(10)2Yey and Dp(17)3Yey) and a knockin mouse model of Aβ accumulation (AppNL-F ) to determine how chromosome 21 genes, other than APP, modulate APP/Aβ in the brain when in three copies. Using both male and female mice, we demonstrate that three copies of other chromosome 21 genes are sufficient to partially ameliorate Aβ accumulation in the brain. We go on to identify a subregion of chromosome 21 that contains the gene(s) causing this decrease in Aβ accumulation and investigate the role of two lead candidate genes, Dyrk1a and Bace2 Thus, an additional copy of chromosome 21 genes, other than APP, can modulate APP/Aβ in the brain under physiological conditions. This work provides critical mechanistic insight into the development of disease and an explanation for the typically later age of onset of dementia in people who have AD in DS, compared with those who have familial AD caused by triplication of APP SIGNIFICANCE STATEMENT Trisomy of chromosome 21 is a commonly occurring genetic risk factor for early-onset Alzheimer's disease (AD), which has been previously attributed to people with Down syndrome having three copies of the amyloid precursor protein (APP) gene, which is encoded on chromosome 21. However, we have shown that an extra copy of other chromosome 21 genes modifies AD-like phenotypes independently of APP copy number (Wiseman et al., 2018; Tosh et al., 2021). Here, we use a mapping approach to narrow down the genetic cause of the modulation of pathology, demonstrating that gene(s) on chromosome 21 decrease Aβ accumulation in the brain, independently of alterations to full-length APP or C-terminal fragment abundance and that just 38 genes are sufficient to cause this.
Collapse
Affiliation(s)
- Paige Mumford
- The UK Dementia Research Institute, University College London, London, WC1N 3BG, United Kingdom
| | - Justin Tosh
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, United Kingdom
| | - Silvia Anderle
- The UK Dementia Research Institute, University College London, London, WC1N 3BG, United Kingdom
| | - Eleni Gkanatsiou Wikberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg S-431 80, Sweden
| | - Gloria Lau
- The UK Dementia Research Institute, University College London, London, WC1N 3BG, United Kingdom
| | - Sue Noy
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, United Kingdom
| | - Karen Cleverley
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, United Kingdom
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama Japan, 351-0198
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama Japan, 351-0198
| | - Eugene Yu
- Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Children's Guild Foundation Down Syndrome Research Program, Buffalo, New York NY 14263
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg S-431 80, Sweden
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg S-431 80, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal S-43180, Sweden
| | - Henrik Zetterberg
- The UK Dementia Research Institute, University College London, London, WC1N 3BG, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal S-43180, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Victor Tybulewicz
- The Francis Crick Institute, London, NW1 1AT, United Kingdom
- Department of Immunology and Inflammation, Imperial College, London, W12 0NN, United Kingdom
- LonDownS: London Down Syndrome Consortium
| | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, United Kingdom
- LonDownS: London Down Syndrome Consortium
| | - Frances K Wiseman
- The UK Dementia Research Institute, University College London, London, WC1N 3BG, United Kingdom
- LonDownS: London Down Syndrome Consortium
| |
Collapse
|
14
|
Sawa M, Overk C, Becker A, Derse D, Albay R, Weldy K, Salehi A, Beach TG, Doran E, Head E, Yu YE, Mobley WC. Impact of increased APP gene dose in Down syndrome and the Dp16 mouse model. Alzheimers Dement 2022; 18:1203-1234. [PMID: 34757693 PMCID: PMC9085977 DOI: 10.1002/alz.12463] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/21/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION People with Down syndrome (DS) are predisposed to Alzheimer's disease (AD). The amyloid hypothesis informs studies of AD. In AD-DS, but not sporadic AD, increased APP copy number is necessary, defining the APP gene dose hypothesis. Which amyloid precursor protein (APP) products contribute needs to be determined. METHODS Brain levels of full-length protein (fl-hAPP), C-terminal fragments (hCTFs), and amyloid beta (Aβ) peptides were measured in DS, AD-DS, non-demented controls (ND), and sporadic AD cases. The APP gene-dose hypothesis was evaluated in the Dp16 model. RESULTS DS and AD-DS differed from ND and AD for all APP products. In AD-DS, Aβ42 and Aβ40 levels exceeded AD. APP products were increased in the Dp16 model; increased APP gene dose was necessary for loss of vulnerable neurons, tau pathology, and activation of astrocytes and microglia. DISCUSSION Increases in APP products other than Aβ distinguished AD-DS from AD. Deciphering AD-DS pathogenesis necessitates deciphering which APP products contribute and how.
Collapse
Affiliation(s)
- Mariko Sawa
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Cassia Overk
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ann Becker
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Dominique Derse
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ricardo Albay
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Kim Weldy
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624
| | - Ahmad Salehi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305
| | - Thomas G. Beach
- Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ 85351
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, CA, 92697
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, 92697
| | - Y. Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093-0624,Correspondence to: William Mobley M.D., Department of Neurosciences, UCSD School of Medicine, 9500 Gilman Drive, GPL 355, La Jolla, CA 92093-0624;
| |
Collapse
|
15
|
Luo J, Zou H, Guo Y, Huang K, Ngan ESW, Li P. BACE2 variant identified from HSCR patient causes AD-like phenotypes in hPSC-derived brain organoids. Cell Death Discov 2022; 8:47. [PMID: 35110536 PMCID: PMC8811022 DOI: 10.1038/s41420-022-00845-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/30/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
β-site APP-cleaving enzyme 2 (BACE2) is a homolog of BACE1, which is considered as the most promising therapeutic target for Alzheimer's disease (AD). However, the expression and functional role of BACE2 in central nervous system (CNS) remain obscured. Previously, we identified several BACE2 rare variants in Hirschsprung disease (HSCR) patients and proved that BACE2-mediated APP cleavage might represent a novel HSCR pathogenesis mechanism in enteric nervous system. Here, we validated that these HSCR-associated BACE2 variants were loss-of-function mutations. Using the human pluripotent stem cell (hPSC)-derived brain organoids (BOs), we further demonstrated that BACE2 was mainly expressed in the ventricular zone and cortical plate of BOs, and its expression level was gradually increased along with the BO maturation. Functionally, we found that the BOs carrying the BACE2 loss-of-function mutation (BACE2G446R) showed greater apoptosis and increased levels of Aβ oligomers compared to the control BOs, resembling with the AD-associated phenotypes. All these phenotypes could be rescued via the removal of APP protein in BACE2G446R BOs. Furthermore, rather than BACE2G446R, BACE2WT overexpression in BOs carrying the APP Swedish/Indiana mutations attenuated the AD-associated phenotypes, including Aβ accumulation and neuronal cell death. Taken together, our results unravel that BACE2 can protect the neuronal cell from apoptosis caused by Aβ accumulation, and the deficiency of BACE2-mediated APP cleavage may represent a common pathological mechanism for both HSCR and AD.
Collapse
Affiliation(s)
- Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Yibo Guo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Ke Huang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Elly Sau-Wai Ngan
- Department of Surgery, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, People's Republic of China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, People's Republic of China.
| |
Collapse
|
16
|
Vitória JJM, Trigo D, da Cruz E Silva OAB. Revisiting APP secretases: an overview on the holistic effects of retinoic acid receptor stimulation in APP processing. Cell Mol Life Sci 2022; 79:101. [PMID: 35089425 PMCID: PMC11073327 DOI: 10.1007/s00018-021-04090-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide and is characterized by the accumulation of the β-amyloid peptide (Aβ) in the brain, along with profound alterations in phosphorylation-related events and regulatory pathways. The production of the neurotoxic Aβ peptide via amyloid precursor protein (APP) proteolysis is a crucial step in AD development. APP is highly expressed in the brain and is complexly metabolized by a series of sequential secretases, commonly denoted the α-, β-, and γ-cleavages. The toxicity of resulting fragments is a direct consequence of the first cleaving event. β-secretase (BACE1) induces amyloidogenic cleavages, while α-secretases (ADAM10 and ADAM17) result in less pathological peptides. Hence this first cleavage event is a prime therapeutic target for preventing or reverting initial biochemical events involved in AD. The subsequent cleavage by γ-secretase has a reduced impact on Aβ formation but affects the peptides' aggregating capacity. An array of therapeutic strategies are being explored, among them targeting Retinoic Acid (RA) signalling, which has long been associated with neuronal health. Additionally, several studies have described altered RA levels in AD patients, reinforcing RA Receptor (RAR) signalling as a promising therapeutic strategy. In this review we provide a holistic approach focussing on the effects of isoform-specific RAR modulation with respect to APP secretases and discuss its advantages and drawbacks in subcellular AD related events.
Collapse
Affiliation(s)
- José J M Vitória
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diogo Trigo
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Odete A B da Cruz E Silva
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
17
|
Alić I, Goh PA, Murray A, Portelius E, Gkanatsiou E, Gough G, Mok KY, Koschut D, Brunmeir R, Yeap YJ, O'Brien NL, Groet J, Shao X, Havlicek S, Dunn NR, Kvartsberg H, Brinkmalm G, Hithersay R, Startin C, Hamburg S, Phillips M, Pervushin K, Turmaine M, Wallon D, Rovelet-Lecrux A, Soininen H, Volpi E, Martin JE, Foo JN, Becker DL, Rostagno A, Ghiso J, Krsnik Ž, Šimić G, Kostović I, Mitrečić D, Francis PT, Blennow K, Strydom A, Hardy J, Zetterberg H, Nižetić D. Patient-specific Alzheimer-like pathology in trisomy 21 cerebral organoids reveals BACE2 as a gene dose-sensitive AD suppressor in human brain. Mol Psychiatry 2021; 26:5766-5788. [PMID: 32647257 PMCID: PMC8190957 DOI: 10.1038/s41380-020-0806-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/18/2020] [Accepted: 05/29/2020] [Indexed: 11/15/2022]
Abstract
A population of more than six million people worldwide at high risk of Alzheimer's disease (AD) are those with Down Syndrome (DS, caused by trisomy 21 (T21)), 70% of whom develop dementia during lifetime, caused by an extra copy of β-amyloid-(Aβ)-precursor-protein gene. We report AD-like pathology in cerebral organoids grown in vitro from non-invasively sampled strands of hair from 71% of DS donors. The pathology consisted of extracellular diffuse and fibrillar Aβ deposits, hyperphosphorylated/pathologically conformed Tau, and premature neuronal loss. Presence/absence of AD-like pathology was donor-specific (reproducible between individual organoids/iPSC lines/experiments). Pathology could be triggered in pathology-negative T21 organoids by CRISPR/Cas9-mediated elimination of the third copy of chromosome 21 gene BACE2, but prevented by combined chemical β and γ-secretase inhibition. We found that T21 organoids secrete increased proportions of Aβ-preventing (Aβ1-19) and Aβ-degradation products (Aβ1-20 and Aβ1-34). We show these profiles mirror in cerebrospinal fluid of people with DS. We demonstrate that this protective mechanism is mediated by BACE2-trisomy and cross-inhibited by clinically trialled BACE1 inhibitors. Combined, our data prove the physiological role of BACE2 as a dose-sensitive AD-suppressor gene, potentially explaining the dementia delay in ~30% of people with DS. We also show that DS cerebral organoids could be explored as pre-morbid AD-risk population detector and a system for hypothesis-free drug screens as well as identification of natural suppressor genes for neurodegenerative diseases.
Collapse
Grants
- MR/S011277/1 Medical Research Council
- MR/L501542/1 Medical Research Council
- RF1 AG059695 NIA NIH HHS
- G-0907 Parkinson's UK
- MR/N026004/1 Medical Research Council
- MR/R024901/1 Medical Research Council
- Wellcome Trust
- 217199 Wellcome Trust
- G0901254 Medical Research Council
- G0701075 Medical Research Council
- 098330 Wellcome Trust
- William Harvey Academy Fellowship, co-funded by the People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme (FP7/2007-2013) under REA grant agreement n° 608765
- Fondation pour la Recherche Médicale (Foundation for Medical Research in France)
- National Research Foundation Singapore (National Research Foundation-Prime Minister’s office, Republic of Singapore)
- BrightFocus Foundation (BrightFocus)
- Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- Svenska Forskningsrådet Formas (Swedish Research Council Formas)
- KB holds the Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences, and is supported by the Swedish Alzheimer Foundation (#AF-742881), Hjärnfonden, Sweden (#FO2017-0243), and the Swedish State Support for Clinical Research (#ALFGBG-715986).
- Wellcome Trust (Wellcome)
- JH received funding from the Dementia Research Institute, an anonymous foundation and the Dolby foundation
- HZ is a Wallenberg Academy Fellow supported by grants from the Swedish Research Council, the European Research Council, Swedish State Support for Clinical Research (ALFGBG-720931) the UK Dementia Research Institute at UCL
Collapse
Affiliation(s)
- Ivan Alić
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- Department of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Pollyanna A Goh
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Aoife Murray
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Eleni Gkanatsiou
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Gillian Gough
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Kin Y Mok
- LonDownS Consortium, London, UK
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - David Koschut
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Reinhard Brunmeir
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Niamh L O'Brien
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Jürgen Groet
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
- LonDownS Consortium, London, UK
| | - Xiaowei Shao
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Steven Havlicek
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - N Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Rosalyn Hithersay
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - Carla Startin
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
| | - Sarah Hamburg
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
| | - Margaret Phillips
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Konstantin Pervushin
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Mark Turmaine
- Division of Biosciences, University College London, Gower Street, London, WC1E 6BT, UK
| | - David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Anne Rovelet-Lecrux
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Neurology and CNR-MAJ, F 76000, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Hilkka Soininen
- University of Eastern Finland, Institute of Clinical Medicine/Neurology, Kuopio, FI-70211, Finland
| | - Emanuela Volpi
- School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Joanne E Martin
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Agueda Rostagno
- Department of Pathology & Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA
| | - Jorge Ghiso
- Department of Pathology & Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Goran Šimić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Dinko Mitrečić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Paul T Francis
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Andre Strydom
- LonDownS Consortium, London, UK
- Division of Psychiatry, University College London, London, WC1E 6BT, UK
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - John Hardy
- LonDownS Consortium, London, UK
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, S-405 30, Sweden
- Dementia Research Institute & Reta Lila Weston Institute, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Dean Nižetić
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK.
- LonDownS Consortium, London, UK.
| |
Collapse
|
18
|
Farris F, Matafora V, Bachi A. The emerging role of β-secretases in cancer. J Exp Clin Cancer Res 2021; 40:147. [PMID: 33926496 PMCID: PMC8082908 DOI: 10.1186/s13046-021-01953-3] [Citation(s) in RCA: 20] [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: 03/17/2021] [Accepted: 04/19/2021] [Indexed: 01/08/2023] Open
Abstract
BACE1 and BACE2 belong to a class of proteases called β-secretases involved in ectodomain shedding of different transmembrane substrates. These enzymes have been extensively studied in Alzheimer's disease as they are responsible for the processing of APP in neurotoxic Aβ peptides. These proteases, especially BACE2, are overexpressed in tumors and correlate with poor prognosis. Recently, different research groups tried to address the role of BACE1 and 2 in cancer development and progression. In this review, we summarize the latest findings on β-secretases in cancer, highlighting the mechanisms that build the rationale to propose inhibitors of these proteins as a new line of treatment for different tumor types.
Collapse
Affiliation(s)
| | | | - Angela Bachi
- IFOM- FIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
19
|
Hampel H, Vassar R, De Strooper B, Hardy J, Willem M, Singh N, Zhou J, Yan R, Vanmechelen E, De Vos A, Nisticò R, Corbo M, Imbimbo BP, Streffer J, Voytyuk I, Timmers M, Tahami Monfared AA, Irizarry M, Albala B, Koyama A, Watanabe N, Kimura T, Yarenis L, Lista S, Kramer L, Vergallo A. The β-Secretase BACE1 in Alzheimer's Disease. Biol Psychiatry 2021; 89:745-756. [PMID: 32223911 PMCID: PMC7533042 DOI: 10.1016/j.biopsych.2020.02.001] [Citation(s) in RCA: 410] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 01/18/2023]
Abstract
BACE1 (beta-site amyloid precursor protein cleaving enzyme 1) was initially cloned and characterized in 1999. It is required for the generation of all monomeric forms of amyloid-β (Aβ), including Aβ42, which aggregates into bioactive conformational species and likely initiates toxicity in Alzheimer's disease (AD). BACE1 concentrations and rates of activity are increased in AD brains and body fluids, thereby supporting the hypothesis that BACE1 plays a critical role in AD pathophysiology. Therefore, BACE1 is a prime drug target for slowing down Aβ production in early AD. Besides the amyloidogenic pathway, BACE1 has other substrates that may be important for synaptic plasticity and synaptic homeostasis. Indeed, germline and adult conditional BACE1 knockout mice display complex neurological phenotypes. Despite BACE1 inhibitor clinical trials conducted so far being discontinued for futility or safety reasons, BACE1 remains a well-validated therapeutic target for AD. A safe and efficacious compound with high substrate selectivity as well as a more accurate dose regimen, patient population, and disease stage may yet be found. Further research should focus on the role of Aβ and BACE1 in physiological processes and key pathophysiological mechanisms of AD. The functions of BACE1 and the homologue BACE2, as well as the biology of Aβ in neurons and glia, deserve further investigation. Cellular and molecular studies of BACE1 and BACE2 knockout mice coupled with biomarker-based human research will help elucidate the biological functions of these important enzymes and identify their substrates and downstream effects. Such studies will have critical implications for BACE1 inhibition as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Harald Hampel
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey; Sorbonne University, GRC No. 21, Alzheimer Precision Medicine, Pitié-Salpêtrière Hospital, Paris, France.
| | - Robert Vassar
- Department of Neurology, Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Bart De Strooper
- Department of Neurosciences, KU Leuven, Leuven, Belgium; Centre for Brain and Disease Research, VIB (Flanders Institute for Biotechnology), Leuven, Belgium; Dementia Research Institute, University College London, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience and Reta Lilla Weston Laboratories, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Michael Willem
- Chair of Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Neeraj Singh
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut
| | - John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut
| | | | | | - Robert Nisticò
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy; School of Pharmacy, Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | | | - Johannes Streffer
- Reference Center for Biological Markers of Dementia, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; UCB Biopharma SPRL, Braine-l'Alleud, Belgium
| | - Iryna Voytyuk
- Department of Neurosciences, KU Leuven, Leuven, Belgium; Centre for Brain and Disease Research, VIB (Flanders Institute for Biotechnology), Leuven, Belgium; ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge, United Kingdom
| | - Maarten Timmers
- Reference Center for Biological Markers of Dementia, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Janssen Research and Development, a division of Janssen Pharmaceutica, Beerse, Belgium
| | - Amir Abbas Tahami Monfared
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Michael Irizarry
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Bruce Albala
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Akihiko Koyama
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | | | | | - Lisa Yarenis
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Simone Lista
- Sorbonne University, GRC No. 21, Alzheimer Precision Medicine, Pitié-Salpêtrière Hospital, Paris, France; Institute of Memory and Alzheimer's Disease, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute, INSERM U 1127, CNRS UMR 7225, Paris, France
| | - Lynn Kramer
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Andrea Vergallo
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey; Sorbonne University, GRC No. 21, Alzheimer Precision Medicine, Pitié-Salpêtrière Hospital, Paris, France; Institute of Memory and Alzheimer's Disease, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute, INSERM U 1127, CNRS UMR 7225, Paris, France.
| |
Collapse
|
20
|
RCAN1 Inhibits BACE2 Turnover by Attenuating Proteasome-Mediated BACE2 Degradation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1920789. [PMID: 32566665 PMCID: PMC7293731 DOI: 10.1155/2020/1920789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 05/22/2020] [Indexed: 11/23/2022]
Abstract
Amyloid-β protein (Aβ) is the main component of neuritic plaques, the pathological hallmark of Alzheimer's disease (AD). β-site APP cleaving enzyme 1 (BACE1) is a major β-secretase contributing to Aβ generation. β-site APP cleaving enzyme 2 (BACE2), the homolog of BACE1, is not only a θ-secretase but also a conditional β-secretase. Previous studies showed that regulator of calcineurin 1 (RCAN1) is markedly increased by AD and promotes BACE1 expression. However, the role of RCAN1 in BACE2 regulation remains elusive. Here, we showed that RCAN1 increases BACE2 protein levels. Moreover, RCAN1 inhibits the turnover of BACE2 protein. Furthermore, RCAN1 attenuates proteasome-mediated BACE2 degradation, but not lysosome-mediated BACE2 degradation. Taken together, our work indicates that RCAN1 inhibits BACE2 turnover by attenuating proteasome-mediated BACE2 degradation. It advances our understanding of BACE2 regulation and provides a potential mechanism of BACE2 dysregulation in AD.
Collapse
|
21
|
Cairns DM, Rouleau N, Parker RN, Walsh KG, Gehrke L, Kaplan DL. A 3D human brain-like tissue model of herpes-induced Alzheimer's disease. SCIENCE ADVANCES 2020; 6:eaay8828. [PMID: 32494701 PMCID: PMC7202879 DOI: 10.1126/sciadv.aay8828] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/18/2020] [Indexed: 05/23/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that causes cognitive decline, memory loss, and inability to perform everyday functions. Hallmark features of AD-including generation of amyloid plaques, neurofibrillary tangles, gliosis, and inflammation in the brain-are well defined; however, the cause of the disease remains elusive. Growing evidence implicates pathogens in AD development, with herpes simplex virus type I (HSV-1) gaining increasing attention as a potential causative agent. Here, we describe a multidisciplinary approach to produce physiologically relevant human tissues to study AD using human-induced neural stem cells (hiNSCs) and HSV-1 infection in a 3D bioengineered brain model. We report a herpes-induced tissue model of AD that mimics human disease with multicellular amyloid plaque-like formations, gliosis, neuroinflammation, and decreased functionality, completely in the absence of any exogenous mediators of AD. This model will allow for future studies to identify potential downstream drug targets for treating this devastating disease.
Collapse
Affiliation(s)
- Dana M. Cairns
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Nicolas Rouleau
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Rachael N. Parker
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | | | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
22
|
Qiu K, Liang W, Wang S, Kong T, Wang X, Li C, Wang Z, Wu Y. BACE2 degradation is mediated by both the proteasome and lysosome pathways. BMC Mol Cell Biol 2020; 21:13. [PMID: 32160867 PMCID: PMC7066761 DOI: 10.1186/s12860-020-00260-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/05/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease is the most common neurodegenerative disease in the elderly. Amyloid-β protein (Aβ) is the major component of neuritic plaques which are the hallmark of AD pathology. β-site APP cleaving enzyme 1 (BACE1) is the major β-secretase contributing to Aβ generation. β-site APP-cleaving enzyme 2 (BACE2), the homolog of BACE1, might play a complex role in the pathogenesis of Alzheimer's disease as it is not only a θ-secretase but also a conditional β-secretase. Dysregulation of BACE2 is observed in Alzheimer's disease. However, the regulation of BACE2 is less studied compared with BACE1, including its degradation pathways. In this study, we investigated the turnover rates and degradation pathways of BACE2 in both neuronal cells and non-neuronal cells. RESULTS Both lysosomal inhibition and proteasomal inhibition cause a time- and dose-dependent increase of transiently overexpressed BACE2 in HEK293 cells. The half-life of transiently overexpressed BACE2 protein is approximately 6 h. Moreover, the half-life of endogenous BACE2 protein is approximately 4 h in both HEK293 cells and mouse primary cortical neurons. Furthermore, both lysosomal inhibition and proteasomal inhibition markedly increases endogenous BACE2 in HEK293 cells and mouse primary cortical neurons. CONCLUSIONS This study demonstrates that BACE2 is degraded by both the proteasome and lysosome pathways in both neuronal and non-neuronal cells at endogenous level and in transient overexpression system. It indicates that BACE2 dysregulation might be mediated by the proteasomal and lysosomal impairment in Alzheimer's disease. This study advances our understanding of the regulation of BACE2 and provides a potential mechanism of its dysregulation in Alzheimer's disease.
Collapse
Affiliation(s)
- Kaixin Qiu
- Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, LixiaDistrict, Jinan, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Wenping Liang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Shuai Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Tingting Kong
- Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, LixiaDistrict, Jinan, Shandong, China
| | - Xin Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Chunyan Li
- Cheeloo College of Medicine, Shandong University, 44 Wenhua West Road, LixiaDistrict, Jinan, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yili Wu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of mental disorders, Institute of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, 272067, Shandong, China.
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong, China.
| |
Collapse
|
23
|
Gong M, Wang Z, Liu Y, Li W, Ye S, Zhu J, Zhang H, Wang J, He K. A transcriptomic analysis of Nsmce1 overexpression in mouse hippocampal neuronal cell by RNA sequencing. Funct Integr Genomics 2019; 20:459-470. [PMID: 31792732 DOI: 10.1007/s10142-019-00728-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/10/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
Mouse Nsmce1 gene is the homolog of non-structural maintenance of chromosomes element 1 (NSE1) that is mainly involved in maintenance of genome integrity, DNA damage response, and DNA repair. Defective DNA repair may cause neurological disorders such as Alzheimer's disease (AD). So far, there is no direct evidence for the correlation between Nsmce1 and AD. In order to explore the function of Nsmce1 in the regulation of nervous system, we have overexpressed or knocked down Nsmce1 in the mouse hippocampal neuronal cells (MHNCs) HT-22 and detected its regulation of AD marker genes as well as cell proliferation. The results showed that the expression of App, Bace2, and Mapt could be inhibited by Nsmce1 overexpression and activated by the knockdown of Nsmce1. Moreover, the HT-22 cell proliferation ability could be promoted by Nsmce1 overexpression and inhibited by knockdown of Nsmce1. Furthermore, we performed a transcriptomics study by RNA sequencing (RNA-seq) to evaluate and quantify the gene expression profiles in response to the overexpression of Nsmce1 in HT-22 cells. As a result, 224 significantly dysregulated genes including 83 upregulated and 141 downregulated genes were identified by the comparison of Nsmce1 /+ to WT cells, which were significantly enriched in several Gene Ontology (GO) terms and pathways. In addition, the complexity of the alternative splicing (AS) landscape was increased by Nsmce1 overexpression in HT-22 cells. Thousands of AS events were identified to be mainly involved in the pathway of ubiquitin-mediated proteolysis (UMP) as well as 3 neurodegenerative diseases including AD. The protein-protein interaction network was reconstructed to show top 10 essential genes regulated by Nsmce1. Our sequencing data is available in the Gene Expression Omnibus (GEO) database with accession number as GSE113436. These results may provide some evidence of molecular and cellular functions of Nsmce1 in MHNCs.
Collapse
Affiliation(s)
- Mengting Gong
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, 230601, Anhui, China
| | - Zhen Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yanjun Liu
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
| | - Wenxing Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, Yunnan, China
| | - Shoudong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, 230601, Anhui, China
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
| | - Jie Zhu
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
| | - Hui Zhang
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
| | - Jing Wang
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China
| | - Kan He
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, 230601, Anhui, China.
- Department of Biostatistics, School of Life Sciences, Anhui University, Hefei, 230601, Anhui, China.
| |
Collapse
|
24
|
Egan MF, Kost J, Voss T, Mukai Y, Aisen PS, Cummings JL, Tariot PN, Vellas B, van Dyck CH, Boada M, Zhang Y, Li W, Furtek C, Mahoney E, Harper Mozley L, Mo Y, Sur C, Michelson D. Randomized Trial of Verubecestat for Prodromal Alzheimer's Disease. N Engl J Med 2019; 380:1408-1420. [PMID: 30970186 PMCID: PMC6776078 DOI: 10.1056/nejmoa1812840] [Citation(s) in RCA: 382] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Prodromal Alzheimer's disease offers an opportunity to test the effect of drugs that modify the deposition of amyloid in the brain before the onset of dementia. Verubecestat is an orally administered β-site amyloid precursor protein-cleaving enzyme 1 (BACE-1) inhibitor that blocks production of amyloid-beta (Aβ). The drug did not prevent clinical progression in a trial involving patients with mild-to-moderate dementia due to Alzheimer's disease. METHODS We conducted a randomized, double-blind, placebo-controlled, 104-week trial to evaluate verubecestat at doses of 12 mg and 40 mg per day, as compared with placebo, in patients who had memory impairment and elevated brain amyloid levels but whose condition did not meet the case definition of dementia. The primary outcome was the change from baseline to week 104 in the score on the Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB; scores range from 0 to 18, with higher scores indicating worse cognition and daily function). Secondary outcomes included other assessments of cognition and daily function. RESULTS The trial was terminated for futility after 1454 patients had been enrolled; 485 had been assigned to receive verubecestat at a dose of 12 mg per day (the 12-mg group), 484 to receive verubecestat at a dose of 40 mg per day (the 40-mg group), and 485 to receive placebo. A total of 234 patients, 231 patients, and 239 patients per group, respectively, completed 104 weeks of the trial regimen. The estimated mean change from baseline to week 104 in the CDR-SB score was 1.65 in the 12-mg group, 2.02 in the 40-mg group, and 1.58 in the placebo group (P = 0.67 for the comparison between the 12-mg group and the placebo group and P = 0.01 for the comparison between the 40-mg group and the placebo group), suggesting a worse outcome in the higher-dose group than in the placebo group. The estimated rate of progression to dementia due to Alzheimer's disease was 24.5, 25.5, and 19.3 events per 100 patient-years in the 12-mg group, the 40-mg group, and the placebo group, respectively (hazard ratio for 40 mg vs. placebo, 1.38; 97.51% confidence interval, 1.07 to 1.79, not adjusted for multiple comparisons), favoring placebo. Adverse events were more common in the verubecestat groups than in the placebo group. CONCLUSIONS Verubecestat did not improve clinical ratings of dementia among patients with prodromal Alzheimer's disease, and some measures suggested that cognition and daily function were worse among patients who received verubecestat than among those who received placebo. (Funded by Merck Sharp & Dohme; ClinicalTrials.gov number, NCT01953601.).
Collapse
Affiliation(s)
- Michael F Egan
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - James Kost
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Tiffini Voss
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Yuki Mukai
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Paul S Aisen
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Jeffrey L Cummings
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Pierre N Tariot
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Bruno Vellas
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Christopher H van Dyck
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Merce Boada
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Ying Zhang
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Wen Li
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Christine Furtek
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Erin Mahoney
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Lyn Harper Mozley
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Yi Mo
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - Cyrille Sur
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| | - David Michelson
- From Merck, Kenilworth, NJ (M.F.E., J.K., T.V., Y. Mukai, Y.Z., W.L., C.F., E.M., L.H.M., Y. Mo, C.S., D.M.); the University of Southern California, San Diego (P.S.A.); Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas (J.L.C.); Banner Alzheimer's Institute, Phoenix, AZ (P.N.T.); Gerontopole, INSERM Unité 1027, Alzheimer's Disease Research and Clinical Center, Toulouse University Hospital, Toulouse, France (B.V.); Yale University School of Medicine, New Haven, CT (C.H.D.); and the Research Center and Memory Clínic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociènces Aplicades-Universitat Internacional de Catalunya, Barcelona, and the Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid - both in Spain (M.B.)
| |
Collapse
|
25
|
Abstract
Virtually all adults with Down syndrome (DS) show the neuropathological changes of Alzheimer disease (AD) by the age of 40 years. This association is partially due to overexpression of amyloid precursor protein, encoded by APP, as a result of the location of this gene on chromosome 21. Amyloid-β accumulates in the brain across the lifespan of people with DS, which provides a unique opportunity to understand the temporal progression of AD and the epigenetic factors that contribute to the age of dementia onset. This age dependency in the development of AD in DS can inform research into the presentation of AD in the general population, in whom a longitudinal perspective of the disease is not often available. Comparison of the risk profiles, biomarker profiles and genetic profiles of adults with DS with those of individuals with AD in the general population can help to determine common and distinct pathways as well as mechanisms underlying increased risk of dementia. This Review evaluates the similarities and differences between the pathological cascades and genetics underpinning DS and AD with the aim of providing a platform for common exploration of these disorders.
Collapse
Affiliation(s)
- Ira T Lott
- Department of Pediatrics and Neurology, School of Medicine, University of California, Irvine, CA, USA.
| | - Elizabeth Head
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
26
|
Rubin LH, Sundermann EE, Moore DJ. The current understanding of overlap between characteristics of HIV-associated neurocognitive disorders and Alzheimer's disease. J Neurovirol 2019; 25:661-672. [PMID: 30671777 DOI: 10.1007/s13365-018-0702-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/04/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022]
Abstract
The advent of effective antiretroviral medications (ARVs) has led to an aging of the HIV population with approximately 50% of people with HIV (PWH) being over the age of 50 years. Neurocognitive complications, typically known as HIV-associated neurocognitive disorders (HAND), persist in the era of ARVs and, in addition to risk of HAND, older PWH are also at risk for age-associated, neurodegenerative disorders including Alzheimer's disease (AD). It has been postulated that risk for AD may be greater among PWH due to potential compounding effects of HIV and aging on mechanisms of neural insult. We are now faced with the challenge of disentangling AD from HAND, which has important prognostic and treatment implications given the more rapidly debilitating trajectory of AD. Herein, we review the evidence to date demonstrating both parallels and differences in the profiles of HAND and AD. We specifically address similarities and difference of AD and HAND as it relates to (1) neuropsychological profiles (cross-sectional/longitudinal), (2) AD-associated neuropathological features as evidenced from neuropathological, cerebrospinal fluid and neuroimaging assessments, (3) biological mechanisms underlying cortical amyloid deposition, (4) parallels in mechanisms of neural insult, and (5) common risk factors. Our current understanding of the similarities and dissimilarities of AD and HAND should be further delineated and leveraged in the development of differential diagnostic methods that will allow for the early identification of AD and more suitable and effective treatment interventions among graying PWH.
Collapse
Affiliation(s)
- Leah H Rubin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Erin E Sundermann
- Department of Psychiatry, University of California, San Diego (UCSD) School of Medicine, La Jolla, CA, USA.
| | - David J Moore
- Department of Psychiatry, University of California, San Diego (UCSD) School of Medicine, La Jolla, CA, USA
| |
Collapse
|
27
|
Wang Z, Xu Q, Cai F, Liu X, Wu Y, Song W. BACE2, a conditional β-secretase, contributes to Alzheimer's disease pathogenesis. JCI Insight 2019; 4:123431. [PMID: 30626751 DOI: 10.1172/jci.insight.123431] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/05/2018] [Indexed: 12/21/2022] Open
Abstract
Deposition of amyloid-β protein (Aβ) to form neuritic plaques is the characteristic neuropathology of Alzheimer's disease (AD). Aβ is generated from amyloid precursor protein (APP) by β- and γ-secretase cleavages. BACE1 is the β-secretase and its inhibition induces severe side effects, whereas its homolog BACE2 normally suppresses Aβ by cleaving APP/Aβ at the θ-site (Phe20) within the Aβ domain. Here, we report that BACE2 also processes APP at the β site, and the juxtamembrane helix (JH) of APP inhibits its β-secretase activity, enabling BACE2 to cleave nascent APP and aggravate AD symptoms. JH-disrupting mutations and clusterin binding to JH triggered BACE2-mediated β-cleavage. Both BACE2 and clusterin were elevated in aged mouse brains, and enhanced β-cleavage during aging. Therefore, BACE2 contributes to AD pathogenesis as a conditional β-secretase and could be a preventive and therapeutic target for AD without the side effects of BACE1 inhibition.
Collapse
Affiliation(s)
- Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China.,Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qin Xu
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Xi Liu
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Yili Wu
- Department of Psychiatry, Jining Medical University, Jining, Shandong, China
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China.,Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Cleavage of potassium channel Kv2.1 by BACE2 reduces neuronal apoptosis. Mol Psychiatry 2018; 23:1542-1554. [PMID: 29703946 DOI: 10.1038/s41380-018-0060-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/24/2018] [Accepted: 03/13/2018] [Indexed: 12/20/2022]
Abstract
Potassium channel Kv2.1 regulates potassium current in cortical neurons and potassium efflux is necessary for cell apoptosis. As a major component of delayed rectifier current potassium channels, Kv2.1 forms clusters in the membrane of hippocampal neurons. BACE2 is an aspartyl protease to cleave APP to prevent the generation of Aβ, a central component of neuritic plaques in Alzheimer's brain. We now identified Kv2.1 as a novel substrate of BACE2. We found that BACE2 cleaved Kv2.1 at Thr376, Ala717, and Ser769 sites and disrupted Kv2.1 clustering on cell membrane, resulting in decreased Ik of Kv2.1 and a hyperpolarizing shift in primary neurons. Furthermore, we discovered that the BACE2-cleaved Kv2.1 forms, Kv2.1-1-375, Kv2.1-1-716, and Kv2.1-1-768, depressed the delayed rectifier Ik surge and reduced neuronal apoptosis. Our study suggests that BACE2 plays a neuroprotective role by cleavage of Kv2.1 to prevent the outward potassium currents, a potential new target for Alzheimer's treatment.
Collapse
|
29
|
Zhang C, Li J, Wang H, Song SW. Identification of a five B cell-associated gene prognostic and predictive signature for advanced glioma patients harboring immunosuppressive subtype preference. Oncotarget 2018; 7:73971-73983. [PMID: 27738332 PMCID: PMC5342028 DOI: 10.18632/oncotarget.12605] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/27/2016] [Indexed: 12/29/2022] Open
Abstract
High grade gliomas contribute to most brain tumor mortality. A few studies reported that the immune system affected glioma development, and immune biomarkers helped understand the disease and formulate effective immunotherapy for patients. Currently, no B lymphocyte-based prognostic signature was reported in gliomas. By applying 78 B cell lineage-specific genes, we conducted a whole-genome gene expression analysis in 782 high grade gliomas derived from three independent datasets by Cox regression analysis and risk score method for signature identification, and then used Gene Ontology, Gene Set Enrichment Analysis, and other statistical methods for functional annotations of the signature-defined differences. We developed a five B cell-associated gene signature for prognosis of high grade glioma patients, which is independent of clinicopathological and genetic features. The signature identified high risk patients suitable for chemoradiotherapy, whereas low risk patients should rule out chemotherapy with radiotherapy only. We found that tumors of TCGA Mesenchymal subtype and wild type IDH1 were preferentially stratified to the high risk group, which bore strong immunosuppressive microenvironment, while tumors of TCGA Proneural subtype and mutated IDH1 were significantly accumulated to the low risk group, which exhibited less immunosuppressive state. The five B cell-associated gene signature predicts poor survival of high risk patients bearing strong immunosuppression and helps select optimal therapeutic regimens for glioma patients.
Collapse
Affiliation(s)
- Chuanbao Zhang
- Beijing Neurosurgical Institute, Capital Medical University, TiantanXili, Dongcheng District, Beijing 100050, China
| | - Jiye Li
- Beijing Neurosurgical Institute, Capital Medical University, TiantanXili, Dongcheng District, Beijing 100050, China.,Beijing Institute for Brain Disorders, Youanmen, Beijing, 100069, China.,Center for Brain Disorders Research, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Haoyuan Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Sonya Wei Song
- Beijing Neurosurgical Institute, Capital Medical University, TiantanXili, Dongcheng District, Beijing 100050, China.,Beijing Institute for Brain Disorders, Youanmen, Beijing, 100069, China.,Center for Brain Disorders Research, Capital Medical University, Youanmen, Beijing, 100069, China
| |
Collapse
|
30
|
Lovell MA, Lynn BC, Fister S, Bradley-Whitman M, Murphy MP, Beckett TL, Norris CM. A Novel Small Molecule Modulator of Amyloid Pathology. J Alzheimers Dis 2018; 53:273-87. [PMID: 27163808 DOI: 10.3233/jad-151160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Because traditional approaches to drug development for Alzheimer's disease are becoming increasingly expensive and in many cases disappointingly unsuccessful, alternative approaches are required to shift the paradigm. Following leads from investigations of dihydropyridine calcium channel blockers, we observed unique properties from a class of functionalized naphthyridines and sought to develop these as novel therapeutics that minimize amyloid pathology without the adverse effects associated with current therapeutics. Our data show methyl 2,4-dimethyl-5-oxo-5,6-dihydrobenzo[c][2,7]naphthyridine-1-carboxylate (BNC-1) significantly decreases amyloid burden in a well-established mouse model of amyloid pathology through a unique mechanism mediated by Elk-1, a transcriptional repressor of presenilin-1. Additionally, BNC-1 treatment leads to increased levels of synaptophysin and synapsin, markers of synaptic integrity, but does not adversely impact presenilin-2 or processing of Notch-1, thus avoiding negative off target effects associated with pan-gamma secretase inhibition. Overall, our data show BNC-1 significantly decreases amyloid burden and improves markers of synaptic integrity in a well-established mouse model of amyloid deposition by promoting phosphorylation and activation of Elk-1, a transcriptional repressor of presenilin-1 but not presenilin-2. These data suggest BNC-1 might be a novel, disease-modifying therapeutic that will alter the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Mark A Lovell
- Department of Chemistry, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Bert C Lynn
- Department of Chemistry, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Universisty of Kentucky Mass Spectrometry Center, Lexington, KY, USA
| | - Shuling Fister
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | | - M Paul Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Department of Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Tina L Beckett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Department of Pharmacology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
31
|
Head E, Helman AM, Powell D, Schmitt FA. Down syndrome, beta-amyloid and neuroimaging. Free Radic Biol Med 2018; 114:102-109. [PMID: 28935420 PMCID: PMC5748259 DOI: 10.1016/j.freeradbiomed.2017.09.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 12/20/2022]
Abstract
This review focuses on the role of Aβ in AD pathogenesis in Down syndrome and current approaches for imaging Aβ in vivo. We will describe how Aβ deposits with age, the posttranslational modifications that can occur, and detection in biofluids. Three unique case studies describing partial trisomy 21 cases without APP triplication, and the occurrences of low level mosaic trisomy 21 in an early onset AD patient are presented. Brain imaging for Aβ includes those by positron emission tomography and ligands (Pittsburgh Compound B, Florbetapir, and FDDNP) that bind Aβ have been published and are summarized here. In combination, we have learned a great deal about Aβ in DS in terms of characterizing age of onset of this pathology and it is exciting to note that there is a clinical trial in DS targeting Aβ that may lead to clinical benefits.
Collapse
Affiliation(s)
- Elizabeth Head
- University of Kentucky, Sanders-Brown Center on Aging, 800 South Limestone Street, Lexington, KY 40536, United States; University of Kentucky, Department of Pharmacology & Nutritional Sciences, Lexington, KY 40536, United States.
| | - Alex M Helman
- University of Kentucky, Sanders-Brown Center on Aging, 800 South Limestone Street, Lexington, KY 40536, United States; University of Kentucky, Department of Pharmacology & Nutritional Sciences, Lexington, KY 40536, United States; University of Kentucky, Magnetic Resonance Imaging and Spectroscopy Center, Lexington, KY 40536, United States; University of Kentucky, Department of Neurology, Lexington, KY 40536, United States
| | - David Powell
- University of Kentucky, Magnetic Resonance Imaging and Spectroscopy Center, Lexington, KY 40536, United States
| | - Frederick A Schmitt
- University of Kentucky, Sanders-Brown Center on Aging, 800 South Limestone Street, Lexington, KY 40536, United States; University of Kentucky, Department of Neurology, Lexington, KY 40536, United States
| |
Collapse
|
32
|
Karelina T, Demin O, Nicholas T, Lu Y, Duvvuri S, Barton HA. A Translational Systems Pharmacology Model for Aβ Kinetics in Mouse, Monkey, and Human. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:666-675. [PMID: 28571112 PMCID: PMC5658289 DOI: 10.1002/psp4.12211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 03/13/2017] [Accepted: 05/18/2017] [Indexed: 01/06/2023]
Abstract
A mechanistic model of amyloid beta production, degradation, and distribution was constructed for mouse, monkey, and human, calibrated and externally verified across multiple datasets. Simulations of single‐dose avagacestat treatment demonstrate that the Aβ42 brain inhibition may exceed that in cerebrospinal fluid (CSF). The dose that achieves 50% CSF Aβ40 inhibition for humans (both healthy and with Alzheimer's disease (AD)) is about 1 mpk, one order of magnitude lower than for mouse (10 mpk), mainly because of differences in pharmacokinetics. The predicted maximal percent of brain Aβ42 inhibition after single‐dose avagacestat is higher for AD subjects (about 60%) than for healthy individuals (about 45%). The probability of achieving a normal physiological level for Aβ42 in brain (1 nM) during multiple avagacestat dosing can be increased by using a dosing regimen that achieves higher exposure. The proposed model allows prediction of brain pharmacodynamics for different species given differing dosing regimens.
Collapse
Affiliation(s)
- T Karelina
- Institute for Systems Biology, Moscow, Russia
| | - O Demin
- Institute for Systems Biology, Moscow, Russia
| | | | | | | | | |
Collapse
|
33
|
Andreeva TV, Lukiw WJ, Rogaev EI. Biological Basis for Amyloidogenesis in Alzheimer's Disease. BIOCHEMISTRY (MOSCOW) 2017; 82:122-139. [PMID: 28320296 DOI: 10.1134/s0006297917020043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Certain cellular proteins normally soluble in the living organism under certain conditions form aggregates with a specific cross-β sheet structure called amyloid. These intra- or extracellular insoluble aggregates (fibers or plaques) are hallmarks of many neurodegenerative pathologies including Alzheimer's disease (AD), Huntington's disease, Parkinson's disease, prion disease, and other progressive neurological diseases that develop in the aging human central nervous system. Amyloid diseases (amyloidoses) are widespread in the elderly human population, a rapidly expanding demographic in many global populations. Increasing age is the most significant risk factor for neurodegenerative diseases associated with amyloid plaques. To date, nearly three dozen different misfolded proteins targeting brain and other organs have been identified in amyloid diseases and AD, the most prevalent neurodegenerative amyloid disease affecting over 15 million people worldwide. Here we (i) highlight the latest data on mechanisms of amyloid formation and further discuss a hypothesis on the amyloid cascade as a primary mechanism of AD pathogenesis and (ii) review the evolutionary aspects of amyloidosis, which allow new insight on human-specific mechanisms of dementia development.
Collapse
Affiliation(s)
- T V Andreeva
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, 119991, Russia.
| | | | | |
Collapse
|
34
|
Head E, Lott IT, Wilcock DM, Lemere CA. Aging in Down Syndrome and the Development of Alzheimer's Disease Neuropathology. Curr Alzheimer Res 2016; 13:18-29. [PMID: 26651341 PMCID: PMC4948181 DOI: 10.2174/1567205012666151020114607] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/18/2015] [Accepted: 09/01/2015] [Indexed: 02/04/2023]
Abstract
Chromosome 21, triplicated in Down Syndrome, contains several genes that are thought to play a critical role in the development of AD neuropathology. The overexpression of the gene for the amyloid precursor protein (APP), on chromosome 21, leads to early onset beta-amyloid (Aβ) plaques in DS. In addition to Aβ accumulation, middle-aged people with DS develop neurofibrillary tangles, cerebrovascular pathology, white matter pathology, oxidative damage, neuroinflammation and neuron loss. There is also evidence of potential compensatory responses in DS that benefit the brain and delay the onset of dementia after there is sufficient neuropathology for a diagnosis of AD. This review describes some of the existing literature and also highlights gaps in our knowledge regarding AD neuropathology in DS. It will be critical in the future to develop networked brain banks with standardized collection procedures to fully characterize the regional and temporal pathological events associated with aging in DS. As more information is acquired regarding AD evolution in DS, there will be opportunities to develop interventions that are age-appropriate to delay AD in DS.
Collapse
Affiliation(s)
- Elizabeth Head
- Sanders Brown Center on Aging, University of Kentucky, 800 South Limestone Street, Lexington, KY, 40536, USA.
| | | | | | | |
Collapse
|
35
|
Wiseman FK, Al-Janabi T, Hardy J, Karmiloff-Smith A, Nizetic D, Tybulewicz VLJ, Fisher EMC, Strydom A. A genetic cause of Alzheimer disease: mechanistic insights from Down syndrome. Nat Rev Neurosci 2015; 16:564-74. [PMID: 26243569 PMCID: PMC4678594 DOI: 10.1038/nrn3983] [Citation(s) in RCA: 375] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Down syndrome, which arises in individuals carrying an extra copy of chromosome 21, is associated with a greatly increased risk of early-onset Alzheimer disease. It is thought that this risk is conferred by the presence of three copies of the gene encoding amyloid precursor protein (APP)--an Alzheimer disease risk factor--although the possession of extra copies of other chromosome 21 genes may also play a part. Further study of the mechanisms underlying the development of Alzheimer disease in people with Down syndrome could provide insights into the mechanisms that cause dementia in the general population.
Collapse
Affiliation(s)
- Frances K Wiseman
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Tamara Al-Janabi
- Division of Psychiatry, University College London, Maple House, 149 Tottenham Court Road, London W1T 7NF, UK
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Annette Karmiloff-Smith
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Malet Street, London WC1E 7HX, UK
| | - Dean Nizetic
- Lee Kong Chian School of Medicine, Nanyang Technological University, Novena Campus, 11 Mandalay Road, Singapore 308232; and the Blizard Institute, Barts and the London School of Medicine, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | | | - Elizabeth M C Fisher
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - André Strydom
- Division of Psychiatry, University College London, Maple House, 149 Tottenham Court Road, London W1T 7NF, UK
| |
Collapse
|
36
|
Holler CJ, Davis PR, Beckett TL, Platt TL, Webb RL, Head E, Murphy MP. Bridging integrator 1 (BIN1) protein expression increases in the Alzheimer's disease brain and correlates with neurofibrillary tangle pathology. J Alzheimers Dis 2015; 42:1221-7. [PMID: 25024306 DOI: 10.3233/jad-132450] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Recent genome wide association studies have implicated bridging integrator 1 (BIN1) as a late-onset Alzheimer's disease (AD) susceptibility gene. There are at least 15 different known isoforms of BIN1, with many being expressed in the brain including the longest isoform (iso1), which is brain-specific and localizes to axon initial segments and nodes of Ranvier. It is currently unknown what role BIN1 plays in AD. We analyzed BIN1 protein expression from a large number (n = 71) of AD cases and controls from five different brain regions (hippocampus, inferior parietal cortex, inferior temporal cortex, frontal cortex (BA9), and superior and middle temporal gyri). We found that the amount of the largest isoform of BIN1 was significantly reduced in the AD brain compared to age-matched controls, and smaller BIN1 isoforms were significantly increased. Further, BIN1 was significantly correlated with the amount of neurofibrillary tangle (NFT) pathology but not with either diffuse or neuritic plaques, or with the amount of amyloid-β peptide. BIN1 is known to be abnormally expressed in another human disease, myotonic dystrophy, which also features prominent NFT pathology. These data suggest that BIN1 is likely involved in AD as a modulator of NFT pathology, and that this role may extend to other human diseases that feature tau pathology.
Collapse
Affiliation(s)
- Christopher J Holler
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Paulina R Davis
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - Tina L Beckett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Thomas L Platt
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Robin L Webb
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - M Paul Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA University of Kentucky Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
37
|
Opportunities and Limitations of Modelling Alzheimer's Disease with Induced Pluripotent Stem Cells. J Clin Med 2014; 3:1357-72. [PMID: 26237606 PMCID: PMC4470188 DOI: 10.3390/jcm3041357] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/31/2014] [Accepted: 11/12/2014] [Indexed: 01/16/2023] Open
Abstract
Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) has opened the way for patient-specific disease modelling. Following their differentiation into neuronal cell types, iPSC have enabled the investigation of human neurodegenerative diseases, such as Alzheimer's disease (AD). While human iPSCs certainly provide great opportunities to repeatedly interrogate specific human brain cell types of individuals with familial and sporadic forms of the disease, the complex aetiology and timescale over which AD develops in humans poses particular challenges to iPSC-based AD models. Here, we discuss the current state-of-play in the context of these and other iPSC model-related challenges and elaborate on likely future developments in this field of research.
Collapse
|
38
|
Alcarraz‐Vizán G, Casini P, Cadavez L, Visa M, Montane J, Servitja J, Novials A. Inhibition of BACE2 counteracts hIAPP‐induced insulin secretory defects in pancreatic β‐cells. FASEB J 2014; 29:95-104. [DOI: 10.1096/fj.14-255489] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Gema Alcarraz‐Vizán
- Diabetes and Obesity Research LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)BarcelonaSpain
| | - Paola Casini
- Diabetes and Obesity Research LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)BarcelonaSpain
| | - Lisa Cadavez
- Diabetes and Obesity Research LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)BarcelonaSpain
| | - Montse Visa
- Diabetes and Obesity Research LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)BarcelonaSpain
| | - Joel Montane
- Diabetes and Obesity Research LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)BarcelonaSpain
| | - Joan‐Marc Servitja
- Diabetes and Obesity Research LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)BarcelonaSpain
| | - Anna Novials
- Diabetes and Obesity Research LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)BarcelonaSpain
| |
Collapse
|
39
|
Mok KY, Jones EL, Hanney M, Harold D, Sims R, Williams J, Ballard C, Hardy J. Polymorphisms in BACE2 may affect the age of onset Alzheimer's dementia in Down syndrome. Neurobiol Aging 2013; 35:1513.e1-5. [PMID: 24462566 PMCID: PMC3969241 DOI: 10.1016/j.neurobiolaging.2013.12.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/03/2013] [Accepted: 12/19/2013] [Indexed: 02/01/2023]
Abstract
It is known that Alzheimer's disease (AD) presents at an early age in people with Down syndrome (DS). The trisomy 21 in DS provides an opportunity to study the effect of duplicated genes in AD. APP and BACE2 are 2 genes located in chromosome 21 and related to AD. We looked into our cohort of 67 DS cases with dementia for the effect of BACE2 variants in age of onset of dementia. Of the 83 single-nucleotide polymorphisms (SNPs), 6 were associated with age of onset and another 8 SNPs were borderline associated. Our finding also replicated a previous study showing association of rs2252576 with AD.
Collapse
Affiliation(s)
- Kin Y Mok
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK; Reta Lila Weston Institute, UCL Institute of Neurology, London, UK.
| | - Emma L Jones
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | | | - Denise Harold
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Henry Wellcome Building for Biomedical Research, Heath Park, Cardiff, UK
| | - Rebecca Sims
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Henry Wellcome Building for Biomedical Research, Heath Park, Cardiff, UK
| | - Julie Williams
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Henry Wellcome Building for Biomedical Research, Heath Park, Cardiff, UK
| | - Clive Ballard
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK; Reta Lila Weston Institute, UCL Institute of Neurology, London, UK; The London Down Syndrome (LonDownS) Consortium, UK
| |
Collapse
|
40
|
Xu Y, Li W, Liu X, Ma H, Tu Z, Dai Y. Analysis of microRNA expression profile by small RNA sequencing in Down syndrome fetuses. Int J Mol Med 2013; 32:1115-25. [PMID: 24071828 DOI: 10.3892/ijmm.2013.1499] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/22/2013] [Indexed: 11/06/2022] Open
Abstract
Down syndrome (DS) is caused by trisomy of human chromosome 21 (Hsa21) and is associated with numerous deleterious phenotypes, including cognitive impairment, childhood leukemia and immune defects. Five Hsa21‑derived microRNAs (i.e., hsa-miR-99a, let-7c, miR-125b-2, miR-155 and miR-802) are involved in variable phenotypes of DS. However, the changes involved in the genome-wide microRNA expression of DS fetuses under the influence of trisomy 21 have yet to be determined. To investigate the expression characteristic of microRNAs during the development of DS fetuses and identify whether another microRNA gene resides in the Hsa21, Illumina high-throughput sequencing technology was employed to comprehensively characterize the microRNA expression profiles of the DS and normal fetal cord blood mononuclear cells (CBMCs). In total, 149 of 395 identified microRNAs were significantly differentially expressed (fold change >2.0 and P<0.001) and 2 of 181 candidate novel microRNAs were identified as residing within the ̔DS critical region̓ of human chromosome 21 (chr21q22.2‑22.3). Additionally, 7 of 14 Hsa21-derived microRNAs were detected, although not all seven were overexpressed in DS CBMCs compared with the control. Gene ontology enrichment analyses revealed that a set of abnormally expressed microRNAs were involved in the regulation of transcription, gene expression, cellular biosynthetic process and nucleic acid metabolic process. Significantly, most of the mRNA targets in these categories were associated with immune modulation (i.e., SOD1, MXD4, PBX1, BCLAF1 and FOXO1). Findings of the present study provided a considerable insight into understanding the expression characteristic of microRNAs in the DS fetal CBMCs. To the best of our knowledge, this is the first study to examine genome-wide microRNA expression profiles in the DS fetus. Differentially expressed microRNAs may be involved in hemopoietic abnormalities and the immune defects of DS fetuses and newborns.
Collapse
Affiliation(s)
- Yong Xu
- Clinical Medical Research Center, Pingshan People's Hospital, Shenzhen, Guangdong 518118, P.R. China
| | | | | | | | | | | |
Collapse
|
41
|
Platt TL, Reeves VL, Murphy MP. Transgenic models of Alzheimer's disease: better utilization of existing models through viral transgenesis. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1437-48. [PMID: 23619198 DOI: 10.1016/j.bbadis.2013.04.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/09/2013] [Accepted: 04/11/2013] [Indexed: 12/30/2022]
Abstract
Animal models have been used for decades in the Alzheimer's disease (AD) research field and have been crucial for the advancement of our understanding of the disease. Most models are based on familial AD mutations of genes involved in the amyloidogenic process, such as the amyloid precursor protein (APP) and presenilin 1 (PS1). Some models also incorporate mutations in tau (MAPT) known to cause frontotemporal dementia, a neurodegenerative disease that shares some elements of neuropathology with AD. While these models are complex, they fail to display pathology that perfectly recapitulates that of the human disease. Unfortunately, this level of pre-existing complexity creates a barrier to the further modification and improvement of these models. However, as the efficacy and safety of viral vectors improves, their use as an alternative to germline genetic modification is becoming a widely used research tool. In this review we discuss how this approach can be used to better utilize common mouse models in AD research. This article is part of a Special Issue entitled: Animal Models of Disease.
Collapse
Affiliation(s)
- Thomas L Platt
- Department of Cellular and Molecular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | | | | |
Collapse
|
42
|
Qi Q, Zhou X, Jiang Y, Hao N, Zhou J, Zhang L. A rare de novo duplication of chromosome 21q22.12 → q22.3 with other concomitant deletion and duplication of small fragments in 21q associated with Down syndrome: Prenatal diagnosis, molecular cytogenetic characterization. Mol Cytogenet 2013; 6:11. [PMID: 23497671 PMCID: PMC3599797 DOI: 10.1186/1755-8166-6-11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/12/2013] [Indexed: 11/30/2022] Open
Abstract
Background Karyotyping is considered the gold standard for the genome-wide detection of genomic imbalances in prenatal diagnosis, but it has a number of inherent limitations, namely the time required to culture cell and the limited resolution(5 ~ 10 Mb). Although fluorescence in situ hybridization (FISH) can also be used as a rapid prenatal diagnosis for common aneuploidies, it is labor intensive, requires prior knowledge of the regions of interest, and can only be used to diagnose one or a few genomic regions simultaneously. Array comparative genomic hybridization (aCGH) can overcome the resolution, the locus-specific, and the time limitations of the karyotyping and FISH techniques and is currently the most powerful method for detecting chromosomal alterations in pre and postnatal clinical cases. Several investigations have suggested that the aCGH testing should be considered a first-tier test for the diagnosis of cytogenetic aberrations in the fetus. Results This study used karyotyping, FISH, sequence-tagged site (STS) analysis and aCGH to diagnose a case of de novo duplication of chromosome 21q22.12 → q22.3 with other concomitant deletion and duplication of small fragments in 21q associated with Down syndrome prenatally. Conclusions FISH, aCGH and STS analysis are useful in prenatal investigation of the nature of de novo alterations of small fragments of the chromosome.
Collapse
Affiliation(s)
- Qingwei Qi
- Department of Obstetrics & Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Peking Union Medical College, Dongdan, Dongcheng District, Beijing, 100730, P, R, China.
| | | | | | | | | | | |
Collapse
|
43
|
Wang H, Li R, Shen Y. β-Secretase: its biology as a therapeutic target in diseases. Trends Pharmacol Sci 2013; 34:215-25. [PMID: 23452816 DOI: 10.1016/j.tips.2013.01.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/21/2013] [Accepted: 01/29/2013] [Indexed: 11/24/2022]
Abstract
β-Secretase (BACE1, β-site APP cleaving enzyme 1) is an aspartic proteinase that has multiple functions in various physiological processes, such as cell differentiation, immunoregulation, and cell death. There is increasing evidence that changes in BACE1 activity are involved in many diseases, such as Alzheimer's disease (AD), schizophrenia, epileptic behavior, and others. However, a deeper understanding of the molecular biology of BACE1 is necessary for further exploration of cell development, immunological regulation, and disease pathogenesis. Here, we review the molecular and cellular biology of BACE1, including its enzymatic properties, structure, biosynthesis, and physiological functions to provide a new perspective and rational assessment of drugability. Lastly, we discuss proposed strategies to control BACE1 activity for possible therapeutic application.
Collapse
Affiliation(s)
- Haibo Wang
- Center for Advanced Therapeutic Strategies for Brain Disorders, Roskamp Institute, Sarasota, FL 34203, USA
| | | | | |
Collapse
|
44
|
Beckett TL, Webb RL, Niedowicz DM, Holler CJ, Matveev S, Baig I, LeVine H, Keller JN, Murphy MP. Postmortem Pittsburgh Compound B (PiB) binding increases with Alzheimer's disease progression. J Alzheimers Dis 2013; 32:127-38. [PMID: 22766739 DOI: 10.3233/jad-2012-120655] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The development of imaging reagents is of considerable interest in the Alzheimer's disease (AD) field. Some of these, such as Pittsburgh Compound B (PiB), were designed to bind to the amyloid-β peptide (Aβ), the major component of amyloid deposits in the AD brain. Although these agents were designed for imaging amyloid deposits in vivo, a major avenue of evaluation relies on postmortem cross validation with established indices of AD pathology. In this study, we evaluated changes in the postmortem binding of PiB and its relationship to other aspects of Aβ-related pathology in a series of AD cases and age-matched controls. We also examined cases of preclinical AD (PCAD) and amnestic mild cognitive impairment (MCI), both considered early points in the AD continuum. PiB binding was found to increase with the progression of the disease and paralleled increases in the less soluble forms of Aβ, including SDS-stable Aβ oligomers. Increased PiB binding and its relationship to Aβ was only significant in a brain region vulnerable to the development of AD pathology (the superior and middle temporal gyri) but not in an unaffected region (cerebellum). This implies that the amyloid deposited in disease-affected regions may possess fundamental, brain region specific characteristics that may not as yet be fully appreciated. These data support the idea that PiB is a useful diagnostic tool for AD, particularly in the early stage of the disease, and also show that PiB could be a useful agent for the discovery of novel disease-related properties of amyloid.
Collapse
Affiliation(s)
- Tina L Beckett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536-0230, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Beckett TL, Studzinski CM, Keller JN, Paul Murphy M, Niedowicz DM. A ketogenic diet improves motor performance but does not affect β-amyloid levels in a mouse model of Alzheimer's disease. Brain Res 2013; 1505:61-7. [PMID: 23415649 DOI: 10.1016/j.brainres.2013.01.046] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/14/2013] [Accepted: 01/29/2013] [Indexed: 11/25/2022]
Abstract
β-Amyloid (Aβ), a small, fibrillogenic peptide, is known to play an important role in the pathogenesis of Alzheimer's disease (AD) in the brain. In addition, Aβ accumulates in skeletal muscle cells in individuals with sporadic inclusion body myositis (sIBM), an age-related muscle disease. Because of the socioeconomic burden associated with age-related diseases, particularly AD, there has been considerable emphasis on studying potential therapeutic strategies. The high-fat, low carbohydrate ketogenic diet has been used extensively to treat refractory childhood epilepsy and has been studied as a potential treatment for other neurological diseases, including Parkinson's disease and AD. In this study, we fed young APP/PS1 knock-in mice, which have a whole body knock-in of AD-related genes, a ketogenic diet and determined the effect on Aβ levels in the brain and skeletal muscle, as well motor performance and oxidative stress. Aβ and its precursor, the β-C-terminal fragment of amyloid precursor protein (CTFβ), were unchanged overall in both the brain and quadriceps after 1 month on the ketogenic diet, and there was no effect on nitrotyrosine, a product of oxidative stress. The ketogenic diet improved performance on the Rota-rod apparatus (p=0.007), however. These data indicate that the ketogenic diet may have some efficacy in the treatment of both neurologic and muscle diseases though the underlying mechanisms do not involve amelioration of Aβ pathology.
Collapse
Affiliation(s)
- Tina L Beckett
- Sanders Brown Center on Aging, University of Kentucky, 800S. Limestone, Lexington, KY 40536-0230, United States
| | | | | | | | | |
Collapse
|
46
|
Borjabad A, Volsky DJ. Common transcriptional signatures in brain tissue from patients with HIV-associated neurocognitive disorders, Alzheimer's disease, and Multiple Sclerosis. J Neuroimmune Pharmacol 2012; 7:914-26. [PMID: 23065460 PMCID: PMC3515772 DOI: 10.1007/s11481-012-9409-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 09/26/2012] [Indexed: 01/01/2023]
Abstract
HIV-Associated Neurocognitive Disorders (HAND) is a common manifestation of HIV infection that afflicts about 50 % of HIV-positive individuals. As people with access to antiretroviral treatments live longer, HAND can be found in increasing segments of populations at risk for other chronic, neurodegenerative conditions such as Alzheimer's disease (AD) and Multiple Sclerosis (MS). If brain diseases of diverse etiologies utilize similar biological pathways in the brain, they may coexist in a patient and possibly exacerbate neuropathogenesis and morbidity. To test this proposition, we conducted comparative meta-analysis of selected publicly available microarray datasets from brain tissues of patients with HAND, AD, and MS. In pair-wise and three-way analyses, we found a large number of dysregulated genes and biological processes common to either HAND and AD or HAND and MS, or to all three diseases. The common characteristic of all three diseases was up-regulation of broadly ranging immune responses in the brain. In addition, HAND and AD share down-modulation of processes involved, among others, in synaptic transmission and cell-cell signaling while HAND and MS share defective processes of neurogenesis and calcium/calmodulin-dependent protein kinase activity. Our approach could provide insight into the identification of common disease mechanisms and better intervention strategies for complex neurocognitive disorders.
Collapse
Affiliation(s)
- Alejandra Borjabad
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center, Columbia University, New York, NY 10019, USA.
| | | |
Collapse
|
47
|
Abdul-Hay SO, Sahara T, McBride M, Kang D, Leissring MA. Identification of BACE2 as an avid ß-amyloid-degrading protease. Mol Neurodegener 2012; 7:46. [PMID: 22986058 PMCID: PMC3470943 DOI: 10.1186/1750-1326-7-46] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 08/16/2012] [Indexed: 11/10/2022] Open
Abstract
Background Proteases that degrade the amyloid ß-protein (Aß) have emerged as key players in the etiology and potential treatment of Alzheimer’s disease (AD), but it is unlikely that all such proteases have been identified. To discover new Aß-degrading proteases (AßDPs), we conducted an unbiased, genome-scale, functional cDNA screen designed to identify proteases capable of lowering net Aß levels produced by cells, which were subsequently characterized for Aß-degrading activity using an array of downstream assays. Results The top hit emerging from the screen was ß-site amyloid precursor protein-cleaving enzyme 2 (BACE2), a rather unexpected finding given the well-established role of its close homolog, BACE1, in the production of Aß. BACE2 is known to be capable of lowering Aß levels via non-amyloidogenic processing of APP. However, in vitro, BACE2 was also found to be a particularly avid AßDP, with a catalytic efficiency exceeding all known AßDPs except insulin-degrading enzyme (IDE). BACE1 was also found to degrade Aß, albeit ~150-fold less efficiently than BACE2. Aß is cleaved by BACE2 at three peptide bonds—Phe19-Phe20, Phe20-Ala21, and Leu34-Met35—with the latter cleavage site being the initial and principal one. BACE2 overexpression in cultured cells was found to lower net Aß levels to a greater extent than multiple, well-established AßDPs, including neprilysin (NEP) and endothelin-converting enzyme-1 (ECE1), while showing comparable effectiveness to IDE. Conclusions This study identifies a new functional role for BACE2 as a potent AßDP. Based on its high catalytic efficiency, its ability to degrade Aß intracellularly, and other characteristics, BACE2 represents a particulary strong therapeutic candidate for the treatment or prevention of AD.
Collapse
Affiliation(s)
- Samer O Abdul-Hay
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Birdsall Bldg,, Rm, 117, Jacksonville, FL 32224, USA
| | | | | | | | | |
Collapse
|
48
|
|
49
|
β-Secretases, Alzheimer's Disease, and Down Syndrome. Curr Gerontol Geriatr Res 2012; 2012:362839. [PMID: 22481915 PMCID: PMC3299320 DOI: 10.1155/2012/362839] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 12/05/2011] [Indexed: 01/01/2023] Open
Abstract
Individuals with Down Syndrome (DS), or trisomy 21, develop Alzheimer's disease (AD) pathology by approximately 40 years of age. Chromosome 21 harbors several genes implicated in AD, including the amyloid precursor protein and one homologue of the β-site APP cleaving enzyme, BACE2. Processing of the amyloid precursor protein by β-secretase (BACE) is the rate-limiting step in the production of the pathogenic Aβ peptide. Increased amounts of APP in the DS brain result in increased amounts of Aβ and extracellular plaque formation beginning early in life. BACE dysregulation potentially represents an overlapping biological mechanism with sporadic AD and a common therapeutic target. As the lifespan for those with DS continues to increase, age-related concerns such as obesity, depression, and AD are of growing concern. The ability to prevent or delay the progression of neurodegenerative diseases will promote healthy aging and improve quality of life for those with DS.
Collapse
|