51
|
Deng Y, Li W, Niu L, Luo X, Li J, Zhang Y, Liu H, He J, Wan W. Amelioration of Scopolamine-induced Learning and Memory Impairment by the TRPV4 Inhibitor HC067047 in ICR Mice. Neurosci Lett 2021; 767:136209. [PMID: 34480999 DOI: 10.1016/j.neulet.2021.136209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
Alzheimer's disease (AD) is one of the most common causes of neurodegenerative diseases in the elderly. Cholinergic dysfunction is one of the pathological hallmarks of AD and leads to learning and memory impairment. Transient receptor potential vanilloid 4(TRPV4), a nonselective cation channel, is involved in learning and memory functions. HC067047, a TRPV4 specific inhibitor, has been reported to protect neurons against cerebral ischemic injury and amyloid-β -(Aβ) 40-induced hippocampal cell death. However, whether HC067047 could improve scopolamine (SCP)-induced cognitive dysfunction in mice is still unknown. The aims of this study were to verify whether HC067047 could ameliorate the SCP-induced learning and memory impairments in mice and to elucidate its underlying mechanisms of action. In this study, we examined the neuroprotective effect of the HC067047 against cognitive dysfunction induced by SCP (5 mg/kg, i.p.), a muscarinic receptor antagonist. The results showed that administration of HC067047(10 mg/kg, i.p.) significantly ameliorated SCP-induced cognitive dysfunction as assessed by the novel place recognition test (NPRT) and novel object recognition test (NORT). In the Y-maze test, HC067047 significantly enhanced the time spent in the novel arm in SCP mice. To further investigate the molecular mechanisms underlying the neuroprotective effect of HC067047, expression of several proteins involved in apoptosis was examined. The results demonstrated that HC067047 treatment decreased the protein levels of proapoptotic proteins such as Bax and caspase-3 in the hippocampus of SCP mice. In addition, HC067047 enhanced expression of the neurogenesis marker DCX and improved levels of the mature neuronal marker NeuN in SCP mice. These findings suggest the neuroprotective potential of the TRPV4 inhibitor HC067047 for the management of dementia with learning and memory loss.
Collapse
Affiliation(s)
- Yingcheng Deng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Wei Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Lei Niu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China; Liuyang Traditional Chinese Medicine Hospital, 410300, Liuyang, Hunan, China
| | - Xianglin Luo
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Jing Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Yuan Zhang
- Department of Pathology, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Hong Liu
- Department of Orthopedics, 922Hospital of PLA Joint Logistics Support Force
| | - Jie He
- Department of Pathology, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Wei Wan
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, 421001 Hengyang, Hunan, China; China Key Laboratory Of Brain Science Research & Transformation In Tropical Environment Of Hainan Province, Hainan Medical University, 571199, Haikou, Hai nan China.
| |
Collapse
|
52
|
Cellular functions of the protein kinase ATM and their relevance to human disease. Nat Rev Mol Cell Biol 2021; 22:796-814. [PMID: 34429537 DOI: 10.1038/s41580-021-00394-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is a master regulator of double-strand DNA break (DSB) signalling and stress responses. For three decades, ATM has been investigated extensively to elucidate its roles in the DNA damage response (DDR) and in the pathogenesis of ataxia telangiectasia (A-T), a human neurodegenerative disease caused by loss of ATM. Although hundreds of proteins have been identified as ATM phosphorylation targets and many important roles for this kinase have been identified, it is still unclear how ATM deficiency leads to the early-onset cerebellar degeneration that is common in all individuals with A-T. Recent studies suggest the existence of links between ATM deficiency and other cerebellum-specific neurological disorders, as well as the existence of broader similarities with more common neurodegenerative disorders. In this Review, we discuss recent structural insights into ATM regulation, and possible aetiologies of A-T phenotypes, including reactive oxygen species, mitochondrial dysfunction, alterations in transcription, R-loop metabolism and alternative splicing, defects in cellular proteostasis and metabolism, and potential pathogenic roles for hyper-poly(ADP-ribosyl)ation.
Collapse
|
53
|
Patel AG, Nehete PN, Krivoshik SR, Pei X, Cho EL, Nehete BP, Ramani MD, Shao Y, Williams LE, Wisniewski T, Scholtzova H. Innate immunity stimulation via CpG oligodeoxynucleotides ameliorates Alzheimer's disease pathology in aged squirrel monkeys. Brain 2021; 144:2146-2165. [PMID: 34128045 PMCID: PMC8502485 DOI: 10.1093/brain/awab129] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease is the most common cause of dementia and the only illness among the top 10 causes of death for which there is no disease-modifying therapy. The failure rate of clinical trials is very high, in part due to the premature translation of successful results in transgenic mouse models to patients. Extensive evidence suggests that dysregulation of innate immunity and microglia/macrophages plays a key role in Alzheimer's disease pathogenesis. Activated resident microglia and peripheral macrophages can display protective or detrimental phenotypes depending on the stimulus and environment. Toll-like receptors (TLRs) are a family of innate immune regulators known to play an important role in governing the phenotypic status of microglia. We have shown in multiple transgenic Alzheimer's disease mouse models that harnessing innate immunity via TLR9 agonist CpG oligodeoxynucleotides (ODNs) modulates age-related defects associated with immune cells and safely reduces amyloid plaques, oligomeric amyloid-β, tau pathology, and cerebral amyloid angiopathy (CAA) while promoting cognitive benefits. In the current study we have used a non-human primate model of sporadic Alzheimer's disease pathology that develops extensive CAA-elderly squirrel monkeys. The major complications in current immunotherapeutic trials for Alzheimer's disease are amyloid-related imaging abnormalities, which are linked to the presence and extent of CAA; hence, the prominence of CAA in elderly squirrel monkeys makes them a valuable model for studying the safety of the CpG ODN-based concept of immunomodulation. We demonstrate that long-term use of Class B CpG ODN 2006 induces a favourable degree of innate immunity stimulation without producing excessive or sustained inflammation, resulting in efficient amelioration of both CAA and tau Alzheimer's disease-related pathologies in association with behavioural improvements and in the absence of microhaemorrhages in aged elderly squirrel monkeys. CpG ODN 2006 has been well established in numerous human trials for a variety of diseases. The present evidence together with our earlier, extensive preclinical research, validates the beneficial therapeutic outcomes and safety of this innovative immunomodulatory approach, increasing the likelihood of CpG ODN therapeutic efficacy in future clinical trials.
Collapse
Affiliation(s)
- Akash G Patel
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Pramod N Nehete
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Sara R Krivoshik
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Xuewei Pei
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Elizabeth L Cho
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Bharti P Nehete
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Margish D Ramani
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Yongzhao Shao
- Division of Biostatistics, Departments of Population Health and Environmental Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Lawrence E Williams
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Henrieta Scholtzova
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| |
Collapse
|
54
|
Hampel H, Nisticò R, Seyfried NT, Levey AI, Modeste E, Lemercier P, Baldacci F, Toschi N, Garaci F, Perry G, Emanuele E, Valenzuela PL, Lucia A, Urbani A, Sancesario GM, Mapstone M, Corbo M, Vergallo A, Lista S. Omics sciences for systems biology in Alzheimer's disease: State-of-the-art of the evidence. Ageing Res Rev 2021; 69:101346. [PMID: 33915266 DOI: 10.1016/j.arr.2021.101346] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is characterized by non-linear, genetic-driven pathophysiological dynamics with high heterogeneity in biological alterations and disease spatial-temporal progression. Human in-vivo and post-mortem studies point out a failure of multi-level biological networks underlying AD pathophysiology, including proteostasis (amyloid-β and tau), synaptic homeostasis, inflammatory and immune responses, lipid and energy metabolism, oxidative stress. Therefore, a holistic, systems-level approach is needed to fully capture AD multi-faceted pathophysiology. Omics sciences - genomics, epigenomics, transcriptomics, proteomics, metabolomics, lipidomics - embedded in the systems biology (SB) theoretical and computational framework can generate explainable readouts describing the entire biological continuum of a disease. Such path in Neurology is encouraged by the promising results of omics sciences and SB approaches in Oncology, where stage-driven pathway-based therapies have been developed in line with the precision medicine paradigm. Multi-omics data integrated in SB network approaches will help detect and chart AD upstream pathomechanistic alterations and downstream molecular effects occurring in preclinical stages. Finally, integrating omics and neuroimaging data - i.e., neuroimaging-omics - will identify multi-dimensional biological signatures essential to track the clinical-biological trajectories, at the subpopulation or even individual level.
Collapse
|
55
|
Ferrer I, Andrés-Benito P, Ausín K, Pamplona R, Del Rio JA, Fernández-Irigoyen J, Santamaría E. Dysregulated protein phosphorylation: A determining condition in the continuum of brain aging and Alzheimer's disease. Brain Pathol 2021; 31:e12996. [PMID: 34218486 PMCID: PMC8549032 DOI: 10.1111/bpa.12996] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Tau hyperphosphorylation is the first step of neurofibrillary tangle (NFT) formation. In the present study, samples of the entorhinal cortex (EC) and frontal cortex area 8 (FC) of cases with NFT pathology classified as stages I-II, III-IV, and V-VI without comorbidities, and of middle-aged (MA) individuals with no NFT pathology, were analyzed by conventional label-free and SWATH-MS (sequential window acquisition of all theoretical fragment ion spectra mass spectrometry) to assess the (phospho)proteomes. The total number of identified dysregulated phosphoproteins was 214 in the EC, 65 of which were dysregulated at the first stages (I-II) of NFT pathology; 167 phosphoproteins were dysregulated in the FC, 81 of them at stages I-II of NFT pathology. A large percentage of dysregulated phosphoproteins were identified in the two regions and at different stages of NFT progression. The main group of dysregulated phosphoproteins was made up of components of the membranes, cytoskeleton, synapses, proteins linked to membrane transport and ion channels, and kinases. The present results show abnormal phosphorylation of proteins at the first stages of NFT pathology in the elderly (in individuals clinically considered representative of normal aging) and sporadic Alzheimer's disease (sAD). Dysregulated protein phosphorylation in the FC precedes the formation of NFTs and SPs. The most active period of dysregulated phosphorylation is at stages III-IV when a subpopulation of individuals might be clinically categorized as suffering from mild cognitive impairment which is a preceding determinant stage in the progression to dementia. Altered phosphorylation of selected proteins, carried out by activation of several kinases, may alter membrane and cytoskeletal functions, among them synaptic transmission and membrane/cytoskeleton signaling. Besides their implications in sAD, the present observations suggest a molecular substrate for "benign" cognitive deterioration in "normal" brain aging.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain.,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Hospitalet de Llobregat, Spain.,Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL, Hospitalet de Llobregat, Spain
| | - Pol Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain.,CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Hospitalet de Llobregat, Spain.,Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL, Hospitalet de Llobregat, Spain
| | - Karina Ausín
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA, IdiSNA, Pamplona, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (UdL-IRBLleida, Lleida, Spain
| | - José Antonio Del Rio
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology, Science Park Barcelona (PCB, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA, IdiSNA, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA, IdiSNA, Pamplona, Spain
| |
Collapse
|
56
|
Martínez M, Inestrosa NC. The transcriptional landscape of Alzheimer's disease and its association with Wnt signaling pathway. Neurosci Biobehav Rev 2021; 128:454-466. [PMID: 34224789 DOI: 10.1016/j.neubiorev.2021.06.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/31/2021] [Accepted: 06/20/2021] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is a neurological disorder primarily affecting the elderly. The disease manifests as progressive deterioration in cognitive functions, leading to a loss of autonomy. The identification of transcriptional changes in susceptible signaling pathways has provided clues to the origin and progression of AD and has pinpointed synapse loss as the prominent event in early stages of the disease. Synapse failure represents a key pathological correlate of cognitive decline in patients. Genetics and transcriptomics studies have also identified novel genes, processes, and pathways associated with AD. This evidence suggests that a deficiency in Wnt signaling pathway contributes to AD pathogenesis by inducing synaptic dysfunction and neuronal degeneration. In the adult nervous system, Wnt signaling plays a crucial role in synaptic physiology, modulating the synaptic vesicle cycle, trafficking neurotransmitter receptors, and modulating the expression of different genes associated with these processes. In this review, we describe the general transcriptional landscape associated with AD, specifically transcriptional changes associated with the Wnt signaling pathway and their effects in the context of disease.
Collapse
Affiliation(s)
- Milka Martínez
- Centro de Envejecimiento y Regeneración (CARE UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
57
|
Molecular subtyping of Alzheimer's disease with consensus non-negative matrix factorization. PLoS One 2021; 16:e0250278. [PMID: 34014928 PMCID: PMC8136734 DOI: 10.1371/journal.pone.0250278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/01/2021] [Indexed: 11/19/2022] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous disease and exhibits diverse clinical presentations and disease progression. Some pathological and anatomical subtypes have been proposed. However, these subtypes provide a limited mechanistic understanding for AD. Leveraging gene expression data of 222 AD patients from The Religious Orders Study and Memory and Aging Project (ROSMAP) Study, we identified two AD molecular subtypes (synaptic type and inflammatory type) using consensus non-negative matrix factorization (NMF). Synaptic type is characterized by disrupted synaptic vesicle priming and recycling and synaptic plasticity. Inflammatory type is characterized by disrupted IL2, interferon alpha and gamma pathways. The two AD molecular subtypes were validated using independent data from Gene Expression Omnibus. We further demonstrated that the two molecular subtypes are associated with APOE genotypes, with synaptic type more prevalent in AD patients with E3E4 genotype and inflammatory type more prevalent in AD patients with E3E3 genotype (p = 0.031). In addition, two molecular subtypes are differentially represented in male and female AD, with synaptic type more prevalent in male and inflammatory type in female patients (p = 0.051). Identification of AD molecular subtypes has potential in facilitating disease mechanism understanding, clinical trial design, drug discovery, and precision medicine for AD.
Collapse
|
58
|
Ma C, Hunt JB, Kovalenko A, Liang H, Selenica MLB, Orr MB, Zhang B, Gensel JC, Feola DJ, Gordon MN, Morgan D, Bickford PC, Lee DC. Myeloid Arginase 1 Insufficiency Exacerbates Amyloid-β Associated Neurodegenerative Pathways and Glial Signatures in a Mouse Model of Alzheimer's Disease: A Targeted Transcriptome Analysis. Front Immunol 2021; 12:628156. [PMID: 34046031 PMCID: PMC8144303 DOI: 10.3389/fimmu.2021.628156] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
Brain myeloid cells, include infiltrating macrophages and resident microglia, play an essential role in responding to and inducing neurodegenerative diseases, such as Alzheimer's disease (AD). Genome-wide association studies (GWAS) implicate many AD casual and risk genes enriched in brain myeloid cells. Coordinated arginine metabolism through arginase 1 (Arg1) is critical for brain myeloid cells to perform biological functions, whereas dysregulated arginine metabolism disrupts them. Altered arginine metabolism is proposed as a new biomarker pathway for AD. We previously reported Arg1 deficiency in myeloid biased cells using lysozyme M (LysM) promoter-driven deletion worsened amyloidosis-related neuropathology and behavioral impairment. However, it remains unclear how Arg1 deficiency in these cells impacts the whole brain to promote amyloidosis. Herein, we aim to determine how Arg1 deficiency driven by LysM restriction during amyloidosis affects fundamental neurodegenerative pathways at the transcriptome level. By applying several bioinformatic tools and analyses, we found that amyloid-β (Aβ) stimulated transcriptomic signatures in autophagy-related pathways and myeloid cells' inflammatory response. At the same time, myeloid Arg1 deficiency during amyloidosis promoted gene signatures of lipid metabolism, myelination, and migration of myeloid cells. Focusing on Aβ associated glial transcriptomic signatures, we found myeloid Arg1 deficiency up-regulated glial gene transcripts that positively correlated with Aβ plaque burden. We also observed that Aβ preferentially activated disease-associated microglial signatures to increase phagocytic response, whereas myeloid Arg1 deficiency selectively promoted homeostatic microglial signature that is non-phagocytic. These transcriptomic findings suggest a critical role for proper Arg1 function during normal and pathological challenges associated with amyloidosis. Furthermore, understanding pathways that govern Arg1 metabolism may provide new therapeutic opportunities to rebalance immune function and improve microglia/macrophage fitness.
Collapse
Affiliation(s)
- Chao Ma
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Jerry B. Hunt
- Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Andrii Kovalenko
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Huimin Liang
- Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Maj-Linda B. Selenica
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
- Sanders-Brown Center on Aging, Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Michael B. Orr
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Bei Zhang
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - John C. Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - David J. Feola
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Marcia N. Gordon
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Dave Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Paula C. Bickford
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Research Service, James A. Haley Veterans Affairs Hospital, Tampa, FL, United States
| | - Daniel C. Lee
- Sanders-Brown Center on Aging, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, United States
| |
Collapse
|
59
|
Cascella R, Cecchi C. Calcium Dyshomeostasis in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22094914. [PMID: 34066371 PMCID: PMC8124842 DOI: 10.3390/ijms22094914] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 01/12/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disorder that is characterized by amyloid β-protein deposition in senile plaques, neurofibrillary tangles consisting of abnormally phosphorylated tau protein, and neuronal loss leading to cognitive decline and dementia. Despite extensive research, the exact mechanisms underlying AD remain unknown and effective treatment is not available. Many hypotheses have been proposed to explain AD pathophysiology; however, there is general consensus that the abnormal aggregation of the amyloid β peptide (Aβ) is the initial event triggering a pathogenic cascade of degenerating events in cholinergic neurons. The dysregulation of calcium homeostasis has been studied considerably to clarify the mechanisms of neurodegeneration induced by Aβ. Intracellular calcium acts as a second messenger and plays a key role in the regulation of neuronal functions, such as neural growth and differentiation, action potential, and synaptic plasticity. The calcium hypothesis of AD posits that activation of the amyloidogenic pathway affects neuronal Ca2+ homeostasis and the mechanisms responsible for learning and memory. Aβ can disrupt Ca2+ signaling through several mechanisms, by increasing the influx of Ca2+ from the extracellular space and by activating its release from intracellular stores. Here, we review the different molecular mechanisms and receptors involved in calcium dysregulation in AD and possible therapeutic strategies for improving the treatment.
Collapse
|
60
|
Mitochondrial dysfunction: A potential target for Alzheimer's disease intervention and treatment. Drug Discov Today 2021; 26:1991-2002. [PMID: 33962036 DOI: 10.1016/j.drudis.2021.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/05/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative brain disorder which manifests as a progressive decline in cognitive function. Mitochondrial dysfunction plays a critical role in the early stages of AD, and advances the progression of this age-related neurodegenerative disorder. Therefore, it can be a potential target for interventions to treat AD. Several therapeutic strategies to target mitochondrial dysfunction have gained significant attention in the preclinical stage, but the clinical trials performed to date have shown little progress. Thus, we discuss the mechanisms and strategies of different therapeutic agents for targeting mitochondrial dysfunction in AD. We hope that this review will inspire and guide the development of efficient AD drugs in the future.
Collapse
|
61
|
Barthelson K, Pederson SM, Newman M, Jiang H, Lardelli M. In-Frame and Frameshift Mutations in Zebrafish Presenilin 2 Affect Different Cellular Functions in Young Adult Brains. J Alzheimers Dis Rep 2021; 5:395-404. [PMID: 34189411 PMCID: PMC8203281 DOI: 10.3233/adr-200279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mutations in PRESENILIN 2 (PSEN2) cause early onset familial Alzheimer's disease (EOfAD) but their mode of action remains elusive. One consistent observation for all PRESENILIN gene mutations causing EOfAD is that a transcript is produced with a reading frame terminated by the normal stop codon-the "reading frame preservation rule". Mutations that do not obey this rule do not cause the disease. The reasons for this are debated. OBJECTIVE To predict cellular functions affected by heterozygosity for a frameshift, or a reading frame-preserving mutation in zebrafish psen2 using bioinformatic techniques. METHODS A frameshift mutation (psen2 N140fs ) and a reading frame-preserving (in-frame) mutation (psen2 T141 _ L142delinsMISLISV ) were previously isolated during genome editing directed at the N140 codon of zebrafish psen2 (equivalent to N141 of human PSEN2). We mated a pair of fish heterozygous for each mutation to generate a family of siblings including wild type and heterozygous mutant genotypes. Transcriptomes from young adult (6 months) brains of these genotypes were analyzed. RESULTS The in-frame mutation uniquely caused subtle, but statistically significant, changes to expression of genes involved in oxidative phosphorylation, long-term potentiation and the cell cycle. The frameshift mutation uniquely affected genes involved in Notch and MAPK signaling, extracellular matrix receptor interactions and focal adhesion. Both mutations affected ribosomal protein gene expression but in opposite directions. CONCLUSION A frameshift and an in-frame mutation at the same position in zebrafish psen2 cause discrete effects. Changes in oxidative phosphorylation, long-term potentiation and the cell cycle may promote EOfAD pathogenesis in humans.
Collapse
Affiliation(s)
- Karissa Barthelson
- Alzheimer’s Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Stephen Martin Pederson
- Bioinformatics Hub, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Morgan Newman
- Alzheimer’s Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Haowei Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Michael Lardelli
- Alzheimer’s Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| |
Collapse
|
62
|
Sharma C, Kim S, Nam Y, Jung UJ, Kim SR. Mitochondrial Dysfunction as a Driver of Cognitive Impairment in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22094850. [PMID: 34063708 PMCID: PMC8125007 DOI: 10.3390/ijms22094850] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most frequent cause of age-related neurodegeneration and cognitive impairment, and there are currently no broadly effective therapies. The underlying pathogenesis is complex, but a growing body of evidence implicates mitochondrial dysfunction as a common pathomechanism involved in many of the hallmark features of the AD brain, such as formation of amyloid-beta (Aβ) aggregates (amyloid plaques), neurofibrillary tangles, cholinergic system dysfunction, impaired synaptic transmission and plasticity, oxidative stress, and neuroinflammation, that lead to neurodegeneration and cognitive dysfunction. Indeed, mitochondrial dysfunction concomitant with progressive accumulation of mitochondrial Aβ is an early event in AD pathogenesis. Healthy mitochondria are critical for providing sufficient energy to maintain endogenous neuroprotective and reparative mechanisms, while disturbances in mitochondrial function, motility, fission, and fusion lead to neuronal malfunction and degeneration associated with excess free radical production and reduced intracellular calcium buffering. In addition, mitochondrial dysfunction can contribute to amyloid-β precursor protein (APP) expression and misprocessing to produce pathogenic fragments (e.g., Aβ1-40). Given this background, we present an overview of the importance of mitochondria for maintenance of neuronal function and how mitochondrial dysfunction acts as a driver of cognitive impairment in AD. Additionally, we provide a brief summary of possible treatments targeting mitochondrial dysfunction as therapeutic approaches for AD.
Collapse
Affiliation(s)
- Chanchal Sharma
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Sehwan Kim
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Korea; (S.K.); (Y.N.)
| | - Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Korea; (S.K.); (Y.N.)
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Korea;
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Korea; (S.K.); (Y.N.)
- Correspondence: ; Tel.: +82-53-950-7362; Fax: +82-53-943-2762
| |
Collapse
|
63
|
Lawingco T, Chaudhury S, Brookes KJ, Guetta-Baranes T, Guerreiro R, Bras J, Hardy J, Francis P, Thomas A, Belbin O, Morgan K. Genetic variants in glutamate-, Aβ-, and tau-related pathways determine polygenic risk for Alzheimer's disease. Neurobiol Aging 2021; 101:299.e13-299.e21. [PMID: 33303219 DOI: 10.1016/j.neurobiolaging.2020.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/29/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022]
Abstract
Synapse loss is an early event in late-onset Alzheimer's disease (LOAD). In this study, we have assessed the capacity of a polygenic risk score (PRS) restricted to synapse-encoding loci to predict LOAD. We used summary statistics from the International Genetics of Alzheimer's Project genome-wide association meta-analysis of 74,046 patients for model construction and tested the "synaptic PRS" in 2 independent data sets of controls and pathologically confirmed LOAD. The mean synaptic PRS was 2.3-fold higher in LOAD than that in controls (p < 0.0001) with a predictive accuracy of 72% in the target data set (n = 439) and 73% in the validation data set (n = 136), a 5%-6% improvement compared with the APOE locus (p < 0.00001). The model comprises 8 variants from 4 previously identified (BIN1, PTK2B, PICALM, APOE) and 2 novel (DLG2, MINK1) LOAD loci involved in glutamate signaling (p = 0.01) or APP catabolism or tau binding (p = 0.005). As the simplest PRS model with good predictive accuracy to predict LOAD, we conclude that synapse-encoding genes are enriched for LOAD risk-modifying loci. The synaptic PRS could be used to identify individuals at risk of LOAD before symptom onset.
Collapse
Affiliation(s)
- Ted Lawingco
- Human Genetics Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Sultan Chaudhury
- Human Genetics Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Keeley J Brookes
- Human Genetics Group, School of Life Sciences, University of Nottingham, Nottingham, UK; Biosciences School of Science & Technology, Nottingham Trent University, Nottingham, UK
| | - Tamar Guetta-Baranes
- Human Genetics Group, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Rita Guerreiro
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA; Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Jose Bras
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA; Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - John Hardy
- UK Dementia Research Institute and Department of Neurodegenerative Disease and Reta Lila Weston Institute, UCL Institute of Neurology and UCL Movement Disorders Centre, University College London, London, UK; Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | | | - Alan Thomas
- Brains for Dementia Research Resource, Newcastle, UK
| | - Olivia Belbin
- Sant Pau Memory Unit and Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Kevin Morgan
- Human Genetics Group, School of Life Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
64
|
Hadjidemetriou M, Rivers-Auty J, Papafilippou L, Eales J, Kellett KAB, Hooper NM, Lawrence CB, Kostarelos K. Nanoparticle-Enabled Enrichment of Longitudinal Blood Proteomic Fingerprints in Alzheimer's Disease. ACS NANO 2021; 15:7357-7369. [PMID: 33730479 PMCID: PMC8155389 DOI: 10.1021/acsnano.1c00658] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Blood-circulating biomarkers have the potential to detect Alzheimer's disease (AD) pathology before clinical symptoms emerge and to improve the outcomes of clinical trials for disease-modifying therapies. Despite recent advances in understanding concomitant systemic abnormalities, there are currently no validated or clinically used blood-based biomarkers for AD. The extremely low concentration of neurodegeneration-associated proteins in blood necessitates the development of analytical platforms to address the "signal-to-noise" issue and to allow an in-depth analysis of the plasma proteome. Here, we aimed to discover and longitudinally track alterations of the blood proteome in a transgenic mouse model of AD, using a nanoparticle-based proteomics enrichment approach. We employed blood-circulating, lipid-based nanoparticles to extract, analyze and monitor AD-specific protein signatures and to systemically uncover molecular pathways associated with AD progression. Our data revealed the existence of multiple proteomic signals in blood, indicative of the asymptomatic stages of AD. Comprehensive analysis of the nanoparticle-recovered blood proteome by label-free liquid chromatography-tandem mass spectrometry resulted in the discovery of AD-monitoring signatures that could discriminate the asymptomatic phase from amyloidopathy and cognitive deterioration. While the majority of differentially abundant plasma proteins were found to be upregulated at the initial asymptomatic stages, the abundance of these molecules was significantly reduced as a result of amyloidosis, suggesting a disease-stage-dependent fluctuation of the AD-specific blood proteome. The potential use of the proposed nano-omics approach to uncover information in the blood that is directly associated with brain neurodegeneration was further exemplified by the recovery of focal adhesion cascade proteins. We herein propose the integration of nanotechnology with already existing proteomic analytical tools in order to enrich the identification of blood-circulating signals of neurodegeneration, reinvigorating the potential clinical utility of the blood proteome at predicting the onset and kinetics of the AD progression trajectory.
Collapse
Affiliation(s)
- Marilena Hadjidemetriou
- Nanomedicine
Lab, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
- (M.H.)
| | - Jack Rivers-Auty
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Lana Papafilippou
- Nanomedicine
Lab, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - James Eales
- Division
of Cardiovascular Sciences, School of Medical Sciences, Faculty of
Biology, Medicine and Health, The University
of Manchester M13 9PT, Manchester, United Kingdom
| | - Katherine A. B. Kellett
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Nigel M. Hooper
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Catherine B. Lawrence
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science
Centre, Manchester M13 9PT, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine
Lab, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
- (K.K.)
| |
Collapse
|
65
|
Ma Y, Ye J, Zhao L, Pan D. MicroRNA-146a inhibition promotes total neurite outgrowth and suppresses cell apoptosis, inflammation, and STAT1/MYC pathway in PC12 and cortical neuron cellular Alzheimer's disease models. ACTA ACUST UNITED AC 2021; 54:e9665. [PMID: 33729395 PMCID: PMC7959174 DOI: 10.1590/1414-431x20209665] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/30/2020] [Indexed: 01/20/2023]
Abstract
This study aimed to explore the effect of microRNA (miR)-146a inhibition on regulating cell apoptosis, total neurite outgrowth, inflammation, and STAT1/MYC pathway in Alzheimer's disease (AD). PC12 and cortical neuron cellular AD models were constructed by Aβ1-42 insult. For the former model, nerve growth factor (NGF) stimulation was previously conducted. miR-146a inhibitor and negative-control (NC) inhibitor were transfected into the two cellular AD models, and then cells were named miR-inhibitor group and NC-inhibitor group, respectively. After transfection, cell apoptosis, total neurite outgrowth, supernatant inflammation cytokines, and STAT1/MYC pathway were detected. miR-146a expression was similar between PC12 cellular AD model and control cells (NGF-stimulated PC12 cells), while miR-146a expression was increased in cortical neuron cellular AD model compared with control cells (rat embryo primary cortical neurons). In both PC12 and cortical neuron cellular AD models, miR-146a expression was reduced in miR-inhibitor group compared with NC-inhibitor group after transfection. Furthermore, cell apoptosis was attenuated, while total neurite outgrowth was elevated in miR-inhibitor group compared with NC-inhibitor group. As for supernatant inflammatory cytokines, tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and IL-17 levels were lower in miR-inhibitor group than in NC-inhibitor group. Additionally, STAT1 and c-Myc mRNA and protein expressions were attenuated in miR-inhibitor group compared with NC-inhibitor group. In conclusion, miR-146a potentially represented a viable therapeutic target for AD.
Collapse
Affiliation(s)
- Yinghui Ma
- Department of Neurosurgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Jiye Ye
- Department of Neurosurgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | | | - Dongmei Pan
- Department of Gerontology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| |
Collapse
|
66
|
Is γ-secretase a beneficial inactivating enzyme of the toxic APP C-terminal fragment C99? J Biol Chem 2021; 296:100489. [PMID: 33662398 PMCID: PMC8027268 DOI: 10.1016/j.jbc.2021.100489] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic, biochemical, and anatomical grounds led to the proposal of the amyloid cascade hypothesis centered on the accumulation of amyloid beta peptides (Aβ) to explain Alzheimer's disease (AD) etiology. In this context, a bulk of efforts have aimed at developing therapeutic strategies seeking to reduce Aβ levels, either by blocking its production (γ- and β-secretase inhibitors) or by neutralizing it once formed (Aβ-directed immunotherapies). However, so far the vast majority of, if not all, clinical trials based on these strategies have failed, since they have not been able to restore cognitive function in AD patients, and even in many cases, they have worsened the clinical picture. We here propose that AD could be more complex than a simple Aβ-linked pathology and discuss the possibility that a way to reconcile undoubted genetic evidences linking processing of APP to AD and a consistent failure of Aβ-based clinical trials could be to envision the pathological contribution of the direct precursor of Aβ, the β-secretase-derived C-terminal fragment of APP, βCTF, also referred to as C99. In this review, we summarize scientific evidences pointing to C99 as an early contributor to AD and postulate that γ-secretase should be considered as not only an Aβ-generating protease, but also a beneficial C99-inactivating enzyme. In that sense, we discuss the limitations of molecules targeting γ-secretase and propose alternative strategies seeking to reduce C99 levels by other means and notably by enhancing its lysosomal degradation.
Collapse
|
67
|
Defining early changes in Alzheimer's disease from RNA sequencing of brain regions differentially affected by pathology. Sci Rep 2021; 11:4865. [PMID: 33649380 PMCID: PMC7921390 DOI: 10.1038/s41598-021-83872-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 02/03/2021] [Indexed: 01/01/2023] Open
Abstract
Tau pathology in Alzheimer’s disease (AD) spreads in a predictable pattern that corresponds with disease symptoms and severity. At post-mortem there are cortical regions that range from mildly to severely affected by tau pathology and neuronal loss. A comparison of the molecular signatures of these differentially affected areas within cases and between cases and controls may allow the temporal modelling of disease progression. Here we used RNA sequencing to explore differential gene expression in the mildly affected primary visual cortex and moderately affected precuneus of ten age-, gender- and RNA quality-matched post-mortem brains from AD patients and healthy controls. The two regions in AD cases had similar transcriptomic signatures but there were broader abnormalities in the precuneus consistent with the greater tau load. Both regions were characterised by upregulation of immune-related genes such as those encoding triggering receptor expressed on myeloid cells 2 and membrane spanning 4-domains A6A and milder changes in insulin/IGF1 signalling. The precuneus in AD was also characterised by changes in vesicle secretion and downregulation of the interneuronal subtype marker, somatostatin. The ‘early’ AD transcriptome is characterised by perturbations in synaptic vesicle secretion on a background of neuroimmune dysfunction. In particular, the synaptic deficits that characterise AD may begin with the somatostatin division of inhibitory neurotransmission.
Collapse
|
68
|
Goult BT. The Mechanical Basis of Memory - the MeshCODE Theory. Front Mol Neurosci 2021; 14:592951. [PMID: 33716664 PMCID: PMC7947202 DOI: 10.3389/fnmol.2021.592951] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
One of the major unsolved mysteries of biological science concerns the question of where and in what form information is stored in the brain. I propose that memory is stored in the brain in a mechanically encoded binary format written into the conformations of proteins found in the cell-extracellular matrix (ECM) adhesions that organise each and every synapse. The MeshCODE framework outlined here represents a unifying theory of data storage in animals, providing read-write storage of both dynamic and persistent information in a binary format. Mechanosensitive proteins that contain force-dependent switches can store information persistently, which can be written or updated using small changes in mechanical force. These mechanosensitive proteins, such as talin, scaffold each synapse, creating a meshwork of switches that together form a code, the so-called MeshCODE. Large signalling complexes assemble on these scaffolds as a function of the switch patterns and these complexes would both stabilise the patterns and coordinate synaptic regulators to dynamically tune synaptic activity. Synaptic transmission and action potential spike trains would operate the cytoskeletal machinery to write and update the synaptic MeshCODEs, thereby propagating this coding throughout the organism. Based on established biophysical principles, such a mechanical basis for memory would provide a physical location for data storage in the brain, with the binary patterns, encoded in the information-storing mechanosensitive molecules in the synaptic scaffolds, and the complexes that form on them, representing the physical location of engrams. Furthermore, the conversion and storage of sensory and temporal inputs into a binary format would constitute an addressable read-write memory system, supporting the view of the mind as an organic supercomputer.
Collapse
Affiliation(s)
- Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
69
|
Tong TM, Dao TTH, Doan LP, Nguyen DT, Nguyen QTT, Do TTT, Truong KD, Phan MD, Nguyen HN, Tran TC, Giang H. Genetic analysis of Vietnamese patients with early-onset Alzheimer's disease. Int J Neurosci 2021; 132:1190-1197. [PMID: 33397166 DOI: 10.1080/00207454.2020.1870974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Purpose of the study: Alzheimer's disease (AD) is the most common type of dementia and its prevalence is rapidly increasing worldwide. Early-onset Alzheimer's disease (EOAD) constitutes of patients with age of onset earlier than 65 year-old and is known to be associated with genetic mutations. In this study, we reported the first genetic analysis of Vietnamese patients with EOAD.Materials and methods: We analyzed targeted sequencing data obtained from a cohort of 51 Vietnamese EOAD patients to identify pathogenic variants in twenty nine well-characterized neurodengerative genes.Results: We identified four missense mutations in APP/PSEN1 genes from six individuals, which accounts for 11.8% of all tested cases. Three of these mutations were previously reported as pathogenic and one mutation in the APP gene was newly identified and might be specific for Vietnamese patients. Our study also found eight individuals carrying homozygous APOE ε4 allele, the main risk factor gene for late-onset AD.Conclusions: Our findings showed that mutation rate in APP/PSEN genes in Vietnamese EOAD patients is consistent with that in other ethnic groups. Although further functional studies are required to validate the pathogenesis of the new mutations, our study demonstrated the necessity of genetic screening for EOAD patients as well as additional genetic data collection in Vietnamese population.
Collapse
Affiliation(s)
- Trang Mai Tong
- Department of Neurology, University Medical Center, Ho Chi Minh City, Vietnam
| | | | | | | | | | | | | | - Minh-Duy Phan
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | - Hoai-Nghia Nguyen
- Center for Molecular Biomedicine, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Thang Cong Tran
- Department of Neurology, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| |
Collapse
|
70
|
Yan M, Zheng T. Role of the endolysosomal pathway and exosome release in tau propagation. Neurochem Int 2021; 145:104988. [PMID: 33582164 DOI: 10.1016/j.neuint.2021.104988] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 02/08/2023]
Abstract
The progressive deposition of misfolded and aggregated forms of Tau protein in the brain is a pathological hallmark of tauopathies, such as Alzheimer's disease (AD) and frontotemporal degeneration (FTD). The misfolded Tau can be released into the extracellular space and internalized by neighboring cells, acting as seeds to trigger the robust conversion of soluble Tau into insoluble filamentous aggregates in a prion-like manner, ultimately contributing to the progression of the disease. However, molecular mechanisms accountable for the propagation of Tau pathology are poorly defined. We reviewed the Tau processing imbalance in endosomal, lysosomal, and exosomal pathways in AD. Increased exosome release counteracts the endosomal-lysosomal dysfunction of Tau processing but increases the number of aggregates and the propagation of Tau. This review summarizes our current understanding of the underlying tauopathy mechanisms with an emphasis on the emerging role of the endosomal-lysosomal-exosome pathways in this process. The components CHMP6, TSG101, and other components of the ESCRT complex, as well as Rab GTPase such as Rab35 and Rab7A, regulate vesicle cargoes routing from endosome to lysosome and affect Tau traffic, degradation, or secretion. Thus, the significant molecular pathways that should be potential therapeutic targets for treating tauopathies are determined.
Collapse
Affiliation(s)
- Minli Yan
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou, 310009, China
| | - Tingting Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou, 310009, China.
| |
Collapse
|
71
|
Zhou L, Shi X, Yin H, Huang Y, Wang R, Ma L. Design, Synthesis and Biological Evaluation of Nobiletin Derivatives as Multifunctional Agents for the Treatment of Alzheimer's Disease. ChemistrySelect 2021. [DOI: 10.1002/slct.202004239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Licheng Zhou
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Ximeng Shi
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Huanhuan Yin
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Yi Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| |
Collapse
|
72
|
Ando K, Houben S, Homa M, de Fisenne MA, Potier MC, Erneux C, Brion JP, Leroy K. Alzheimer's Disease: Tau Pathology and Dysfunction of Endocytosis. Front Mol Neurosci 2021; 13:583755. [PMID: 33551742 PMCID: PMC7862548 DOI: 10.3389/fnmol.2020.583755] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/22/2020] [Indexed: 11/21/2022] Open
Affiliation(s)
- Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Sarah Houben
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Mégane Homa
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Marie-Ange de Fisenne
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Marie-Claude Potier
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christophe Erneux
- Institut de Recherche Interdisciplinaire en Biologie Humaine et moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, Brussels, Belgium
| |
Collapse
|
73
|
Chan JW, Chan NCY, Sadun AA. Glaucoma as Neurodegeneration in the Brain. Eye Brain 2021; 13:21-28. [PMID: 33500674 PMCID: PMC7822087 DOI: 10.2147/eb.s293765] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/07/2021] [Indexed: 12/31/2022] Open
Abstract
Glaucoma, a group of diseases characterized by progressive optic nerve degeneration that results in irreversible blindness, can be considered a neurodegenerative disorder of both the eye and the brain. Increasing evidence from human and animal studies have shown that glaucoma shares some common neurodegenerative pathways with Alzheimer’s disease (AD) and other tauopathies, such as chronic traumatic encephalopathy (CTE) and frontotemporal dementia. This hypothesis is based on the focal adhesion pathway hypothesis and the spreading hypothesis of tau. Not only has the Apolipoprotein E (APOE) gene been shown to be associated with AD, but also with primary open angle glaucoma (POAG). This review will highlight the relevant literature in the past 20 years from PubMed that show the pathogenic overlap between POAG and AD. Neurodegenerative pathways that contribute to transsynaptic neurodegeneration in AD and other tauopathies might also be similar to those in glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Jane W Chan
- Department of Ophthalmology, Doheny Eye Institute, Pasadena, CA, USA
| | - Noel C Y Chan
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T., Hong Kong, People's Republic of China
| | - Alfredo A Sadun
- Department of Ophthalmology, Doheny Eye Institute, Pasadena, CA, USA.,Department of Ophthalmology, University of California, Los Angeles, CA, USA
| |
Collapse
|
74
|
Avramopoulos D, Kapogiannis D, Leoutsakos JM, Lyketsos CG, Mahairaki V, Nowrangi M, Oishi K, Oh ES, Rosenberg PB, Samus Q, Smith GS, Witwer K, Yasar S, Zandi PP. Developing Treatments for Alzheimer's and Related Disorders with Precision Medicine: A Vision. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:395-402. [PMID: 35023131 PMCID: PMC9358929 DOI: 10.1007/978-3-030-78787-5_49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Precision medicine, also known as personalized medicine, is concerned with finding the right treatment for the right patient at the right time. It is a way of thinking focused on parsing heterogeneity ultimately down to the level of the individual. Its main mission is to identify characteristics of heterogeneous clinical conditions so as to target tailored therapies to individuals. Precision Medicine however is not an agnostic collection of all manner of clinical, genetic and other biologic data in select cohorts. This is an important point. Simply collecting as much information as possible on individuals without applying this way of thinking should not be considered Precision Medicine.
Collapse
Affiliation(s)
- Dimitrios Avramopoulos
- Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Intramural Research Program, National Institute on Aging (NIA/NIH), Baltimore, MD, USA
| | - Jeannie-Marie Leoutsakos
- Departments of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Constantine G Lyketsos
- Departments of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Vasiliki Mahairaki
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Milap Nowrangi
- Departments of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kenichi Oishi
- Departments of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Esther S Oh
- Departments of Medicine (Geriatric Medicine and Gerontology), School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Paul B Rosenberg
- Departments of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Quincy Samus
- Departments of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Gwenn S Smith
- Departments of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kenneth Witwer
- Departments of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sevil Yasar
- Departments of Medicine (Geriatric Medicine and Gerontology), School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Peter P Zandi
- Departments of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
75
|
Podleśny-Drabiniok A, Marcora E, Goate AM. Microglial Phagocytosis: A Disease-Associated Process Emerging from Alzheimer’s Disease Genetics. Trends Neurosci 2020; 43:965-979. [DOI: 10.1016/j.tins.2020.10.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 10/05/2020] [Indexed: 01/02/2023]
|
76
|
Fani L, Ahmad S, Ikram MK, Ghanbari M, Ikram MA. Immunity and amyloid beta, total tau and neurofilament light chain: Findings from a community-based cohort study. Alzheimers Dement 2020; 17:446-456. [PMID: 33215849 PMCID: PMC8048997 DOI: 10.1002/alz.12212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 12/24/2022]
Abstract
Introduction We investigated how components of immunity relate to biomarkers of Alzheimer's disease (AD) in plasma and explored the influence of AD genetic risk factors in the population‐based Rotterdam Study. Methods In 7397 persons, we calculated the granulocyte‐to‐lymphocyte ratio (GLR), platelet‐to‐lymphocyte ratio (PLR), and systemic immune‐inflammation index (SII). In 3615 of these persons, plasma amyloid‐beta (Aβ)42 and Aβ40 were measured. Next, we constructed an overall genetic risk score (GRS) based on genome‐wide significant variants, both including and excluding APOE ε4. Results All innate immunity phenotypes were related to higher Aβ, most strongly with a doubling in GLR leading to a 1.9% higher Aβ42 (95% confidence interval [95% CI] 0.4 to 3.3%) and 3.2% higher Aβ40 (95% CI 2.0 to 4.3%). Higher AD GRS including APOE ε4 was associated with higher immunity markers. Discussion Higher levels of immunity markers were associated with higher Aβ in plasma. Participants with a higher genetic predisposition to AD had higher immunity markers, where these effects were mainly driven by APOE ε4.
Collapse
Affiliation(s)
- Lana Fani
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Erasmus MC, Department of Neurology, the Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
77
|
Agüero P, Sainz MJ, García-Ayllón MS, Sáez-Valero J, Téllez R, Guerrero-López R, Pérez-Pérez J, Jiménez-Escrig A, Gómez-Tortosa E. α-Secretase nonsense mutation (ADAM10 Tyr167*) in familial Alzheimer's disease. Alzheimers Res Ther 2020; 12:139. [PMID: 33129344 PMCID: PMC7603780 DOI: 10.1186/s13195-020-00708-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The disintegrin metalloproteinase 10 (ADAM10) is the main α-secretase acting in the non-amyloidogenic processing of APP. Some ADAM10 gene variants have been associated with higher susceptibility to develop late-onset AD, though clear clinical-genetic correlates remain elusive. METHODS Clinical-genetic and biomarker study of a first family with early- and late-onset AD associated with a nonsense ADAM10 mutation (p.Tyr167*). CSF analysis included AD core biomarkers, as well as Western blot of ADAM10 species and sAPPα and sAPPβ peptides. We evaluate variant's pathogenicity, pattern of segregation, and further screened for the p.Tyr167* mutation in 197 familial AD cases from the same cohort, 200 controls from the same background, and 274 AD cases from an independent Spanish cohort. RESULTS The mutation was absent from public databases and segregated with the disease. CSF Aβ42, total tau, and phosphorylated tau of affected siblings were consistent with AD. The predicted haploinsufficiency effect of the nonsense mutation was supported by (a) ADAM10 isoforms in CSF decreased around 50% and (b) 70% reduction of CSF sAPPα peptide, both compared to controls, while sAPPβ levels remained unchanged. Interestingly, sporadic AD cases had a similar decrease in CSF ADAM10 levels to that of mutants, though their sAPPα and sAPPβ levels resembled those of controls. Therefore, a decreased sAPPα/sAPPβ ratio was an exclusive feature of mutant ADAM10 siblings. The p.Tyr167* mutation was not found in any of the other AD cases or controls screened. CONCLUSIONS This family illustrates the role of ADAM10 in the amyloidogenic process and the clinical development of the disease. Similarities between clinical and biomarker findings suggest that this family could represent a genetic model for sporadic late-onset AD due to age-related downregulation of α-secretase. This report encourages future research on ADAM10 enhancers.
Collapse
Affiliation(s)
- Pablo Agüero
- Department of Neurology, Fundación Jiménez Díaz, Avenida de los Reyes Católicos 2, 28040, Madrid, Spain
| | - María José Sainz
- Department of Neurology, Fundación Jiménez Díaz, Avenida de los Reyes Católicos 2, 28040, Madrid, Spain
| | - María-Salud García-Ayllón
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d'Alacant, Spain
- Unidad de Investigación, Hospital General Universitario de Elche, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Elche, Spain
| | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sant Joan d'Alacant, Spain
| | - Raquel Téllez
- Department of Immunology, Fundación Jiménez Díaz, Madrid, Spain
| | - Rosa Guerrero-López
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD) and CIBERER, Madrid, Spain
| | | | | | - Estrella Gómez-Tortosa
- Department of Neurology, Fundación Jiménez Díaz, Avenida de los Reyes Católicos 2, 28040, Madrid, Spain.
| |
Collapse
|
78
|
Soheili-Nezhad S, van der Linden RJ, Olde Rikkert M, Sprooten E, Poelmans G. Long genes are more frequently affected by somatic mutations and show reduced expression in Alzheimer's disease: Implications for disease etiology. Alzheimers Dement 2020; 17:489-499. [PMID: 33075204 PMCID: PMC8048495 DOI: 10.1002/alz.12211] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/11/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022]
Abstract
Aging, the greatest risk factor for Alzheimer's disease (AD), may lead to the accumulation of somatic mutations in neurons. We investigated whether somatic mutations, specifically in longer genes, are implicated in AD etiology. First, we modeled the theoretical likelihood of genes being affected by aging‐induced somatic mutations, dependent on their length. We then tested this model and found that long genes are indeed more affected by somatic mutations and that their expression is more frequently reduced in AD brains. Furthermore, using gene‐set enrichment analysis, we investigated the potential consequences of such long gene disruption. We found that long genes are involved in synaptic adhesion and other synaptic pathways that are predicted to be inhibited in the brains of AD patients. Taken together, our findings indicate that long gene–dependent synaptic impairment may contribute to AD pathogenesis.
Collapse
Affiliation(s)
- Sourena Soheili-Nezhad
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Marcel Olde Rikkert
- Department of Geriatric Medicine, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emma Sprooten
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
79
|
Weninger S, Sperling B, Alexander R, Ivarsson M, Menzies FM, Powchik P, Weber CJ, Altar CA, Crystal RG, Haggarty SJ, Loring J, Bain LJ, Carrillo MC. Active immunotherapy and alternative therapeutic modalities for Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2020; 6:e12090. [PMID: 33083513 PMCID: PMC7550557 DOI: 10.1002/trc2.12090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 12/17/2022]
Abstract
As knowledge of Alzheimer's disease (AD) progression improves, the field has recognized the need to diversify the pipeline, broaden strategies and approaches to therapies, as well as delivery mechanisms. A better understanding of the earliest biological processes of AD/dementia would help inform drug target selection. Currently there are a number of programs exploring these alternate avenues. This meeting will allow experts in the field (academia, industry, government) to provide perspectives and experiences that can help elucidate what the pipeline looks like today and what avenues hold promise in developing new therapies across the stages of AD. The focus here is on Active Immunotherapies and Alternative Therapeutic Modalities. This topic includes active vaccines, antisense oligomers, and cell-based therapy among others, and highlights new clinical developments that utilize these modalities.
Collapse
Affiliation(s)
| | | | - Robert Alexander
- Takeda Pharmaceuticals International Co. Cambridge Massachusetts USA
| | - Magnus Ivarsson
- Rodin Therapeutics 300 Technology Square Cambridge Massachusetts USA
| | | | - Peter Powchik
- United Neuroscience 9 Exchange Place, I. F. S. C Dublin Ireland
| | | | | | - Ronald G Crystal
- Department of Genetic Medicine Weill Cornell Medicine New York New York USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory Center for Genomic Medicine Department of Neurology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts USA
| | | | - Lisa J Bain
- Independent Science Writer Elverson Pennsylvania USA
| | | |
Collapse
|
80
|
Gadhave K, Gehi BR, Kumar P, Xue B, Uversky VN, Giri R. The dark side of Alzheimer's disease: unstructured biology of proteins from the amyloid cascade signaling pathway. Cell Mol Life Sci 2020; 77:4163-4208. [PMID: 31894361 PMCID: PMC11104979 DOI: 10.1007/s00018-019-03414-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/17/2019] [Accepted: 12/04/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a leading cause of age-related dementia worldwide. Despite more than a century of intensive research, we are not anywhere near the discovery of a cure for this disease or a way to prevent its progression. Among the various molecular mechanisms proposed for the description of the pathogenesis and progression of AD, the amyloid cascade hypothesis, according to which accumulation of a product of amyloid precursor protein (APP) cleavage, amyloid β (Aβ) peptide, induces pathological changes in the brain observed in AD, occupies a unique niche. Although multiple proteins have been implicated in this amyloid cascade signaling pathway, their structure-function relationships are mostly unexplored. However, it is known that two major proteins related to AD pathology, Aβ peptide, and microtubule-associated protein tau belong to the category of intrinsically disordered proteins (IDPs), which are the functionally important proteins characterized by a lack of fixed, ordered three-dimensional structure. IDPs and intrinsically disordered protein regions (IDPRs) play numerous vital roles in various cellular processes, such as signaling, cell cycle regulation, macromolecular recognition, and promiscuous binding. However, the deregulation and misfolding of IDPs may lead to disturbed signaling, interactions, and disease pathogenesis. Often, molecular recognition-related IDPs/IDPRs undergo disorder-to-order transition upon binding to their biological partners and contain specific disorder-based binding motifs, known as molecular recognition features (MoRFs). Knowing the intrinsic disorder status and disorder-based functionality of proteins associated with amyloid cascade signaling pathway may help to untangle the mechanisms of AD pathogenesis and help identify therapeutic targets. In this paper, we have used multiple computational tools to evaluate the presence of intrinsic disorder and MoRFs in 27 proteins potentially relevant to the amyloid cascade signaling pathway. Among these, BIN1, APP, APOE, PICALM, PSEN1 and CD33 were found to be highly disordered. Furthermore, their disorder-based binding regions and associated short linear motifs have also been identified. These findings represent important foundation for the future research, and experimental characterization of disordered regions in these proteins is required to better understand their roles in AD pathogenesis.
Collapse
Affiliation(s)
- Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | | | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Bin Xue
- Department of Cell Biology, Microbiology and Molecular Biology, School of Natural Sciences and Mathematics, College of Arts and Sciences, University of South Florida, Tampa, FL, 33620, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, 33620, USA.
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia.
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India.
| |
Collapse
|
81
|
Mo D, Li X, Raabe CA, Rozhdestvensky TS, Skryabin BV, Brosius J. Circular RNA Encoded Amyloid Beta peptides-A Novel Putative Player in Alzheimer's Disease. Cells 2020; 9:E2196. [PMID: 33003364 PMCID: PMC7650678 DOI: 10.3390/cells9102196] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 02/05/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related detrimental dementia. Amyloid beta peptides (Aβ) play a crucial role in the pathology of AD. In familial AD, Aβ are generated from the full-length amyloid beta precursor protein (APP) via dysregulated proteolytic processing; however, in the case of sporadic AD, the mechanism of Aβ biogenesis remains elusive. circRNAs are a class of transcripts preferentially expressed in brain. We identified a circRNA harboring the Aβ-coding region of the APP gene termed circAβ-a. This circular RNA was detected in the brains of AD patients and non-dementia controls. With the aid of our recently established approach for analysis of circRNA functions, we demonstrated that circAβ-a is efficiently translated into a novel Aβ-containing Aβ175 polypeptide (19.2 KDa) in both cultured cells and human brain. Furthermore, Aβ175 was shown to be processed into Aβ peptides-a hallmark of AD. In summary, our analysis revealed an alternative pathway of Aβ biogenesis. Consequently, circAβ-a and its corresponding translation product could potentially represent novel therapeutic targets for AD treatment. Importantly, our data point to yet another evolutionary route for potentially increasing proteome complexity by generating additional polypeptide variants using back-splicing of primary transcripts that yield circular RNA templates.
Collapse
Affiliation(s)
- Dingding Mo
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9b, 50931 Cologne, Germany;
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, O&N IV Herestraat 49—box 602, 3000 Leuven, Belgium
- Medical Faculty, Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany; (T.S.R.); (B.V.S.)
| | - Xinping Li
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9b, 50931 Cologne, Germany;
| | - Carsten A. Raabe
- Institute of Experimental Pathology, Centre for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany; (C.A.R.); (J.B.)
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Strasse 56, D-48149 Münster, Germany
| | - Timofey S. Rozhdestvensky
- Medical Faculty, Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany; (T.S.R.); (B.V.S.)
| | - Boris V. Skryabin
- Medical Faculty, Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany; (T.S.R.); (B.V.S.)
| | - Juergen Brosius
- Institute of Experimental Pathology, Centre for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany; (C.A.R.); (J.B.)
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610212, China
| |
Collapse
|
82
|
Kilinc D, Vreulx AC, Mendes T, Flaig A, Marques-Coelho D, Verschoore M, Demiautte F, Amouyel P, Eysert F, Dourlen P, Chapuis J, Costa MR, Malmanche N, Checler F, Lambert JC. Pyk2 overexpression in postsynaptic neurons blocks amyloid β 1-42-induced synaptotoxicity in microfluidic co-cultures. Brain Commun 2020; 2:fcaa139. [PMID: 33718872 PMCID: PMC7941669 DOI: 10.1093/braincomms/fcaa139] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/12/2020] [Accepted: 08/03/2020] [Indexed: 01/06/2023] Open
Abstract
Recent meta-analyses of genome-wide association studies identified a number of genetic risk factors of Alzheimer's disease; however, little is known about the mechanisms by which they contribute to the pathological process. As synapse loss is observed at the earliest stage of Alzheimer's disease, deciphering the impact of Alzheimer's risk genes on synapse formation and maintenance is of great interest. In this article, we report a microfluidic co-culture device that physically isolates synapses from pre- and postsynaptic neurons and chronically exposes them to toxic amyloid β peptides secreted by model cell lines overexpressing wild-type or mutated (V717I) amyloid precursor protein. Co-culture with cells overexpressing mutated amyloid precursor protein exposed the synapses of primary hippocampal neurons to amyloid β1-42 molecules at nanomolar concentrations and induced a significant decrease in synaptic connectivity, as evidenced by distance-based assignment of postsynaptic puncta to presynaptic puncta. Treating the cells with antibodies that target different forms of amyloid β suggested that low molecular weight oligomers are the likely culprit. As proof of concept, we demonstrate that overexpression of protein tyrosine kinase 2 beta-an Alzheimer's disease genetic risk factor involved in synaptic plasticity and shown to decrease in Alzheimer's disease brains at gene expression and protein levels-selectively in postsynaptic neurons is protective against amyloid β1-42-induced synaptotoxicity. In summary, our lab-on-a-chip device provides a physiologically relevant model of Alzheimer's disease-related synaptotoxicity, optimal for assessing the impact of risk genes in pre- and postsynaptic compartments.
Collapse
Affiliation(s)
- Devrim Kilinc
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Anaïs-Camille Vreulx
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Tiago Mendes
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Amandine Flaig
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Diego Marques-Coelho
- Brain Institute, Federal University of Rio Grande do Norte, Natal 59056-450, Brazil
| | - Maxime Verschoore
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Florie Demiautte
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Philippe Amouyel
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | | | - Fanny Eysert
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Pierre Dourlen
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Julien Chapuis
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Marcos R Costa
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Nicolas Malmanche
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| | - Frédéric Checler
- CNRS UMR7275 Laboratory of Excellence "Distalz", IPMC, Université Côte d'Azur, Inserm, Valbonne 06560, France
| | - Jean-Charles Lambert
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, Lille 59019, France
| |
Collapse
|
83
|
Gerardo B, Cabral Pinto M, Nogueira J, Pinto P, Almeida A, Pinto E, Marinho-Reis P, Diniz L, Moreira PI, Simões MR, Freitas S. Associations between Trace Elements and Cognitive Decline: An Exploratory 5-Year Follow-Up Study of an Elderly Cohort. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E6051. [PMID: 32825289 PMCID: PMC7503463 DOI: 10.3390/ijerph17176051] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/12/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022]
Abstract
Trace elements (TE) homeostasis is crucial in normal brain functioning. Although imbalances have the potential to exacerbate events leading neurodegenerative diseases, few studies have directly addressed the eventual relationships between TE levels in the human body and future cognitive status. The present study aimed to assess how different TE body-levels relate to cognitive decline. This exploratory research included a study-group (RES) of 20 elderly individuals living in two Portuguese geographical areas of interest (Estarreja; Mértola), as well as a 20 subjects neuropsychological control-group (CTR). Participants were neuropsychologically assessed through the Mini Mental State Examination (MMSE) and the Montreal Cognitive Assessment (MoCA) and the RES group was biomonitored for TE through fingernail analysis. After 5 years, the cognitive assessments were repeated. Analyses of the RES neuropsychological data showed an average decrease of 6.5 and 5.27 points in MMSE and MoCA, respectively, but TE contents in fingernails were generally within the referenced values for non-exposed individuals. Higher levels of Nickel and Selenium significantly predicted lesser cognitive decline within 5 years. Such preliminary results evidence an association between higher contents of these TE and higher cognitive scores at follow-up, suggesting their contribution to the maintenance of cognitive abilities. Future expansion of the present study is needed in order to comprehensively assess the potential benefits of these TE.
Collapse
Affiliation(s)
- Bianca Gerardo
- Center for Research in Neuropsychology and Cognitive and Behavioral Intervention (CINEICC), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal; (J.N.); (M.R.S.); (S.F.)
- Psychological Assessment and Psychometrics Laboratory (PsyAssessmentLab), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal;
| | - Marina Cabral Pinto
- Geobiotec Research Centre, Department of Geosciences, University of Aveiro, 3810-193 Aveiro, Portugal; (M.C.P.); (P.M.-R.)
| | - Joana Nogueira
- Center for Research in Neuropsychology and Cognitive and Behavioral Intervention (CINEICC), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal; (J.N.); (M.R.S.); (S.F.)
- Psychological Assessment and Psychometrics Laboratory (PsyAssessmentLab), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal;
| | - Paula Pinto
- Psychological Assessment and Psychometrics Laboratory (PsyAssessmentLab), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal;
| | - Agostinho Almeida
- LAQV/REQUIMTE, Laboratory of Applied Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (A.A.); (E.P.); (L.D.)
| | - Edgar Pinto
- LAQV/REQUIMTE, Laboratory of Applied Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (A.A.); (E.P.); (L.D.)
- Department of Environmental Health, School of Health, P.Porto, CISA/Research Center in Environment and Health, 4200-072 Porto, Portugal
| | - Paula Marinho-Reis
- Geobiotec Research Centre, Department of Geosciences, University of Aveiro, 3810-193 Aveiro, Portugal; (M.C.P.); (P.M.-R.)
- Departamento de Ciências da Terra, Instituto de Ciências da Terra, Polo da Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Luísa Diniz
- LAQV/REQUIMTE, Laboratory of Applied Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (A.A.); (E.P.); (L.D.)
| | - Paula I. Moreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal;
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mário R. Simões
- Center for Research in Neuropsychology and Cognitive and Behavioral Intervention (CINEICC), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal; (J.N.); (M.R.S.); (S.F.)
- Psychological Assessment and Psychometrics Laboratory (PsyAssessmentLab), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal;
| | - Sandra Freitas
- Center for Research in Neuropsychology and Cognitive and Behavioral Intervention (CINEICC), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal; (J.N.); (M.R.S.); (S.F.)
- Psychological Assessment and Psychometrics Laboratory (PsyAssessmentLab), Faculty of Psychology and Educational Sciences (FPCEUC), Univ Coimbra, 3000-115 Coimbra, Portugal;
| |
Collapse
|
84
|
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease and the leading cause of dementia in the elderly. Recent decades have been marked by considerable advances in our understanding of genetic and environmental risk factors and also of the AD mechanism(s) of action. Nonetheless, there is still no cure and the myriad ways AD affects the brain is overwhelmingly complex. Such complexity is manifest in part by the fact that genetic background interacts with the environment, including traffic-derived particulate air pollution, to greatly exacerbate AD risk. Determining the mechanisms by which particulate air pollution acts as an AD risk factor has the potential to reveal yet unknown aspects of AD pathology. This review carefully peels back the layers of complexity to discern whether a unifying disease model, one with proteostasis imbalance at its core, holds up to scrutiny in light of the recent literature. While the data are compelling, it is now time for carefully designed studies to definitively determine whether particulate air pollution acts with ageing, genetic background and other sources of proteotoxic stress to disrupt the delicate proteostasis balance.
Collapse
Affiliation(s)
- Elise A Kikis
- Biology Department, the University of the South, 735 University Avenue, Sewanee, TN 37383, USA
| |
Collapse
|
85
|
Liang JJH, McKinnon IA, Rankin CH. The contribution of C. elegans neurogenetics to understanding neurodegenerative diseases. J Neurogenet 2020; 34:527-548. [DOI: 10.1080/01677063.2020.1803302] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Joseph J. H. Liang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Issa A. McKinnon
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Catharine H. Rankin
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Psychology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
86
|
Katsumata Y, Fardo DW, Bachstetter AD, Artiushin SC, Wang WX, Wei A, Brzezinski LJ, Nelson BG, Huang Q, Abner EL, Anderson S, Patel I, Shaw BC, Price DA, Niedowicz DM, Wilcock DW, Jicha GA, Neltner JH, Van Eldik LJ, Estus S, Nelson PT. Alzheimer Disease Pathology-Associated Polymorphism in a Complex Variable Number of Tandem Repeat Region Within the MUC6 Gene, Near the AP2A2 Gene. J Neuropathol Exp Neurol 2020; 79:3-21. [PMID: 31748784 PMCID: PMC8204704 DOI: 10.1093/jnen/nlz116] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/18/2019] [Accepted: 10/27/2019] [Indexed: 02/06/2023] Open
Abstract
We found evidence of late-onset Alzheimer disease (LOAD)-associated genetic polymorphism within an exon of Mucin 6 (MUC6) and immediately downstream from another gene: Adaptor Related Protein Complex 2 Subunit Alpha 2 (AP2A2). PCR analyses on genomic DNA samples confirmed that the size of the MUC6 variable number tandem repeat (VNTR) region was highly polymorphic. In a cohort of autopsied subjects with quantitative digital pathology data (n = 119), the size of the polymorphic region was associated with the severity of pTau pathology in neocortex. In a separate replication cohort of autopsied subjects (n = 173), more pTau pathology was again observed in subjects with longer VNTR regions (p = 0.031). Unlike MUC6, AP2A2 is highly expressed in human brain. AP2A2 expression was lower in a subset analysis of brain samples from persons with longer versus shorter VNTR regions (p = 0.014 normalizing with AP2B1 expression). Double-label immunofluorescence studies showed that AP2A2 protein often colocalized with neurofibrillary tangles in LOAD but was not colocalized with pTau proteinopathy in progressive supranuclear palsy, or with TDP-43 proteinopathy. In summary, polymorphism in a repeat-rich region near AP2A2 was associated with neocortical pTau proteinopathy (because of the unique repeats, prior genome-wide association studies were probably unable to detect this association), and AP2A2 was often colocalized with neurofibrillary tangles in LOAD.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - David W Fardo
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Sergey C Artiushin
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Wang-Xia Wang
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Angela Wei
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Lena J Brzezinski
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Bela G Nelson
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Qingwei Huang
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Erin L Abner
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Sonya Anderson
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Indumati Patel
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Benjamin C Shaw
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Douglas A Price
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Dana M Niedowicz
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Donna W Wilcock
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Gregory A Jicha
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Janna H Neltner
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Steven Estus
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Peter T Nelson
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| |
Collapse
|
87
|
Endosomal-lysosomal dysfunction in metabolic diseases and Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:303-324. [PMID: 32739009 DOI: 10.1016/bs.irn.2020.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The endosomal-lysosomal pathways and related autophagic processes are responsible for proteostasis, involving complexes between lysosomes and autophagosomes. Lysosomes are a key component of homeostasis, involved in cell signaling, metabolism, and quality control, and they experience functional compromise in metabolic diseases, aging, and neurodegenerative diseases. Many genetic mutations and risk factor genes associated with proteinopathies, as well as with metabolic diseases like diabetes, negatively influence endocytic trafficking and autophagic clearance. In contrast, health-improving exercise induces autophagy-lysosomal degradation, perhaps promoting efficient digestion of injured organelles so that undamaged organelles ensure cellular healthiness. Reductions in lysosomal hydrolases are implicated in Alzheimer's, Parkinson's, and lysosomal storage diseases, as well as obesity-related pathology, and members of the cathepsin enzyme family are involved in clearing both Aβ42 and α-synuclein. Upregulation of cathepsin hydrolases improves synaptic and memory functions in models of dementia and in exercising humans, thus identifying lysosomal-related systems as vital for healthy cognitive aging.
Collapse
|
88
|
Li X, Zhou H, Yang P, Shi HX, Xiong Y, Nie ZY, Yu JQ, Wang YA, Zhou R, Wang LY. Cyclin-dependent Kinase 5 Regulates Cortical Neurotransmission and Neural Circuits Associated with Motor Control in the Secondary Motor Cortex in the Mouse. Neuroscience 2020; 438:9-24. [PMID: 32353462 DOI: 10.1016/j.neuroscience.2020.04.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 11/17/2022]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a regulator of axon growth and radial neuronal migration in the developing mouse brain, and it plays critical roles in cortical structure formation and brain function. However, the function of Cdk5 in cortico-cortical and cortico-sensorimotor networks in the adult remains largely unknown. In this study, we investigated the function of Cdk5 in the rostral secondary motor cortex (M2) in the male mouse using CRISPR/Cas9 gene editing and somatic brain transgenesis, to produce M2-specific knockdown of Cdk5 in neurons in the male mouse. Mouse deficient in Cdk5 in the M2 exhibited a reduction in both the number of functional synapses and the total basal dendritic length, as well as motor dysfunction. Furthermore, whole-cell patch-clamp recordings in layer V green fluorescent protein (GFP)-tag pyramidal neurons revealed a decrease in the frequency and amplitude of miniature EPSCs and miniature IPSCs, as well as a reduction in the population synaptic responses (fEPSPs) in these mice. Specifically, retrograde labeling showed that Cdk5 knockdown in the M2 caused a reduction in long-range projections to the M2 from the thalamus/prefrontal cortex and claustrum. Collectively, our findings show a new regulatory role of Cdk5 in neural circuit maintenance, and that the changes in neural transmission and circuits in the mice with Cdk5 knockdown in the M2 likely contribute to the motor dysfunction in these animals.
Collapse
Affiliation(s)
- Xing Li
- Department of Pharmacology, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Hu Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Pei Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hua-Xiang Shi
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ying Xiong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zhi-Yong Nie
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jian-Qiang Yu
- Department of Pharmacology, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Yong-An Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ru Zhou
- Department of Pharmacology, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China.
| | - Li-Yun Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
89
|
Park J, Kim H, Kim J, Cheon M. A practical application of generative adversarial networks for RNA-seq analysis to predict the molecular progress of Alzheimer's disease. PLoS Comput Biol 2020; 16:e1008099. [PMID: 32706788 PMCID: PMC7406107 DOI: 10.1371/journal.pcbi.1008099] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 08/05/2020] [Accepted: 06/28/2020] [Indexed: 12/15/2022] Open
Abstract
Next-generation sequencing (NGS) technology has become a powerful tool for dissecting the molecular and pathological signatures of a variety of human diseases. However, the limited availability of biological samples from different disease stages is a major hurdle in studying disease progressions and identifying early pathological changes. Deep learning techniques have recently begun to be applied to analyze NGS data and thereby predict the progression of biological processes. In this study, we applied a deep learning technique called generative adversarial networks (GANs) to predict the molecular progress of Alzheimer's disease (AD). We successfully applied GANs to analyze RNA-seq data from a 5xFAD mouse model of AD, which recapitulates major AD features of massive amyloid-β (Aβ) accumulation in the brain. We examined how the generator is featured to have specific-sample generation and biological gene association. Based on the above observations, we suggested virtual disease progress by latent space interpolation to yield the transition curves of various genes with pathological changes from normal to AD state. By performing pathway analysis based on the transition curve patterns, we identified several pathological processes with progressive changes, such as inflammatory systems and synapse functions, which have previously been demonstrated to be involved in the pathogenesis of AD. Interestingly, our analysis indicates that alteration of cholesterol biosynthesis begins at a very early stage of AD, suggesting that it is the first effect to mediate the cholesterol metabolism of AD downstream of Aβ accumulation. Here, we suggest that GANs are a useful tool to study disease progression, leading to the identification of early pathological signatures.
Collapse
Affiliation(s)
- Jinhee Park
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- School of Electronics Engineering, Kyungpook National University, Daegu, Korea
| | - Hyerin Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| | - Jaekwang Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| | - Mookyung Cheon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| |
Collapse
|
90
|
Soheili-Nezhad S, Jahanshad N, Guelfi S, Khosrowabadi R, Saykin AJ, Thompson PM, Beckmann CF, Sprooten E, Zarei M. Imaging genomics discovery of a new risk variant for Alzheimer's disease in the postsynaptic SHARPIN gene. Hum Brain Mapp 2020; 41:3737-3748. [PMID: 32558014 PMCID: PMC7416020 DOI: 10.1002/hbm.25083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/29/2020] [Accepted: 05/14/2020] [Indexed: 12/26/2022] Open
Abstract
Molecular mechanisms underlying Alzheimer's disease (AD) are difficult to investigate, partly because diagnosis lags behind the insidious pathological processes. Therefore, identifying AD neuroimaging markers and their genetic modifiers may help study early mechanisms of neurodegeneration. We aimed to identify brain regions of the highest vulnerability to AD using a data-driven search in the AD Neuroimaging Initiative (ADNI, n = 1,100 subjects), and further explored genetic variants affecting this critical brain trait using both ADNI and the younger UK Biobank cohort (n = 8,428 subjects). Tensor-Based Morphometry (TBM) and Independent Component Analysis (ICA) identified the limbic system and its interconnecting white-matter as the most AD-vulnerable brain feature. Whole-genome analysis revealed a common variant in SHARPIN that was associated with this imaging feature (rs34173062, p = 2.1 × 10-10 ). This genetic association was validated in the UK Biobank, where it was correlated with entorhinal cortical thickness bilaterally (p = .002 left and p = 8.6 × 10-4 right), and with parental history of AD (p = 2.3 × 10-6 ). Our findings suggest that neuroanatomical variation in the limbic system and AD risk are associated with a novel variant in SHARPIN. The role of this postsynaptic density gene product in β1-integrin adhesion is in line with the amyloid precursor protein (APP) intracellular signaling pathway and the recent genome-wide evidence.
Collapse
Affiliation(s)
- Sourena Soheili-Nezhad
- Institute of Medical Science and Technology, Shahid Beheshti University, Tehran, Iran.,Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands.,Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, The Netherlands
| | - Neda Jahanshad
- Imaging Genetics Center, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA
| | - Sebastian Guelfi
- Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, University College London (UCL) Institute of Neurology, London, UK
| | - Reza Khosrowabadi
- Institute for Cognitive and Brain Science, Shahid Beheshti University, Tehran, Iran
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul M Thompson
- Imaging Genetics Center, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, California, USA
| | - Christian F Beckmann
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands.,Donders Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, The Netherlands.,Oxford Centre for Functional Magnetic Resonance Imaging of the Brain (FMRIB), Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Emma Sprooten
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mojtaba Zarei
- Institute of Medical Science and Technology, Shahid Beheshti University, Tehran, Iran
| | | |
Collapse
|
91
|
Ubeda-Bañon I, Saiz-Sanchez D, Flores-Cuadrado A, Rioja-Corroto E, Gonzalez-Rodriguez M, Villar-Conde S, Astillero-Lopez V, Cabello-de la Rosa JP, Gallardo-Alcañiz MJ, Vaamonde-Gamo J, Relea-Calatayud F, Gonzalez-Lopez L, Mohedano-Moriano A, Rabano A, Martinez-Marcos A. The human olfactory system in two proteinopathies: Alzheimer's and Parkinson's diseases. Transl Neurodegener 2020; 9:22. [PMID: 32493457 PMCID: PMC7271529 DOI: 10.1186/s40035-020-00200-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/20/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's and Parkinson's diseases are the most prevalent neurodegenerative disorders. Their etiologies are idiopathic, and treatments are symptomatic and orientated towards cognitive or motor deficits. Neuropathologically, both are proteinopathies with pathological aggregates (plaques of amyloid-β peptide and neurofibrillary tangles of tau protein in Alzheimer's disease, and Lewy bodies mostly composed of α-synuclein in Parkinson's disease). These deposits appear in the nervous system in a predictable and accumulative sequence with six neuropathological stages. Both disorders present a long prodromal period, characterized by preclinical signs including hyposmia. Interestingly, the olfactory system, particularly the anterior olfactory nucleus, is initially and preferentially affected by the pathology. Cerebral atrophy revealed by magnetic resonance imaging must be complemented by histological analyses to ascertain whether neuronal and/or glial loss or neuropil remodeling are responsible for volumetric changes. It has been proposed that these proteinopathies could act in a prion-like manner in which a misfolded protein would be able to force native proteins into pathogenic folding (seeding), which then propagates through neurons and glia (spreading). Existing data have been examined to establish why some neuronal populations are vulnerable while others are resistant to pathology and to what extent glia prevent and/or facilitate proteinopathy spreading. Connectomic approaches reveal a number of hubs in the olfactory system (anterior olfactory nucleus, olfactory entorhinal cortex and cortical amygdala) that are key interconnectors with the main hubs (the entorhinal-hippocampal-cortical and amygdala-dorsal motor vagal nucleus) of network dysfunction in Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Isabel Ubeda-Bañon
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Daniel Saiz-Sanchez
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Alicia Flores-Cuadrado
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Ernesto Rioja-Corroto
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Melania Gonzalez-Rodriguez
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Sandra Villar-Conde
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | - Veronica Astillero-Lopez
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| | | | | | - Julia Vaamonde-Gamo
- Neurology Service, Ciudad Real General University Hospital, 13005 Ciudad Real, Spain
| | | | - Lucia Gonzalez-Lopez
- Pathology Service, Ciudad Real General University Hospital, 13005 Ciudad Real, Spain
| | | | - Alberto Rabano
- Neuropathology Department and Tissue Bank, CIEN Foundation, Carlos III Health Institute, Madrid, Spain
| | - Alino Martinez-Marcos
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha, 13005 Ciudad Real, Spain
| |
Collapse
|
92
|
Li Y, Fan H, Sun J, Ni M, Zhang L, Chen C, Hong X, Fang F, Zhang W, Ma P. Circular RNA expression profile of Alzheimer's disease and its clinical significance as biomarkers for the disease risk and progression. Int J Biochem Cell Biol 2020; 123:105747. [PMID: 32315771 DOI: 10.1016/j.biocel.2020.105747] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/01/2020] [Accepted: 04/13/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To investigate circular RNA (circRNA) expression profile via microarray, and further assess the potential of candidate circRNAs as biomarkers in Alzheimer's disease (AD). METHODS CircRNA expression profile in cerebrospinal fluid from 8 AD patients and 8 control (Ctrl) subjects was assessed by microarray. Subsequently, 10 candidate circRNAs from microarray were validated by reverse transcription quantitative polymerase chain reaction (RT-qPCR) in cerebrospinal fluid from 80 AD patients and 40 Ctrl subjects. RESULTS By microarray, 112 circRNAs were upregulated and 51 circRNAs were downregulated in AD patients compared with Ctrl subjects, and these circRNAs were enriched in AD related pathways such as neurotrophin signaling pathway, natural killer cell mediated cytotoxicity and cholinergic synapse. By RT-qPCR, circ-LPAR1, circ-AXL and circ-GPHN were increased, whereas circ-PCCA, circ-HAUS4, circ-KIF18B and circ-TTC39C were decreased in AD patients compared with Ctrl subjects, and these circRNAs were disclosed to predict AD risk by receiver operating characteristics curve analysis. Further forward-stepwise multivariate logistic regression revealed that circ-AXL, circ-GPHN, circ-ITPR3, circ-PCCA and cic-TTC39C were independent predictive factors for AD risk. Besides, in AD patients, circ-AXL and circ-GPHN negatively correlated, while circ-PCCA and circ-HAUS4 positively correlated with mini-mental state examination score; Circ-AXL negatively correlated, while circ-PCCA, circ-HAUS4 and circ-KIF18B positively correlated with Aβ42; Circ-AXL and circ-GPHN positively correlated, whereas circ-HAUS4 negatively correlated with t-tau; Circ-AXL positively correlated with p-tau. CONCLUSION Our study provides an overview of circRNA expression profile in AD, and identifies that circ-AXL, circ-GPHN and circ-PCCA hold clinical implications for guiding disease management in AD patients.
Collapse
Affiliation(s)
- Yuanlong Li
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Hua Fan
- The First Affiliated Hospital of Henan University of Science and Technology, School of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| | - Jun Sun
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Ming Ni
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Clinical Pharmacy, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Lei Zhang
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Ci Chen
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Xuejiao Hong
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Fengqin Fang
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Wei Zhang
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Peizhi Ma
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, China; Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China; Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, China.
| |
Collapse
|
93
|
Bellenguez C, Grenier-Boley B, Lambert JC. Genetics of Alzheimer’s disease: where we are, and where we are going. Curr Opin Neurobiol 2020; 61:40-48. [DOI: 10.1016/j.conb.2019.11.024] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022]
|
94
|
Baldacci F, Mazzucchi S, Della Vecchia A, Giampietri L, Giannini N, Koronyo-Hamaoui M, Ceravolo R, Siciliano G, Bonuccelli U, Elahi FM, Vergallo A, Lista S, Giorgi FS. The path to biomarker-based diagnostic criteria for the spectrum of neurodegenerative diseases. Expert Rev Mol Diagn 2020; 20:421-441. [PMID: 32066283 PMCID: PMC7445079 DOI: 10.1080/14737159.2020.1731306] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
Abstract
Introduction: The postmortem examination still represents the reference standard for detecting the pathological nature of chronic neurodegenerative diseases (NDD). This approach displays intrinsic conceptual limitations since NDD represent a dynamic spectrum of partially overlapping phenotypes, shared pathomechanistic alterations that often give rise to mixed pathologies.Areas covered: We scrutinized the international clinical diagnostic criteria of NDD and the literature to provide a roadmap toward a biomarker-based classification of the NDD spectrum. A few pathophysiological biomarkers have been established for NDD. These are time-consuming, invasive, and not suitable for preclinical detection. Candidate screening biomarkers are gaining momentum. Blood neurofilament light-chain represents a robust first-line tool to detect neurodegeneration tout court and serum progranulin helps detect genetic frontotemporal dementia. Ultrasensitive assays and retinal scans may identify Aβ pathology early, in blood and the eye, respectively. Ultrasound also represents a minimally invasive option to investigate the substantia nigra. Protein misfolding amplification assays may accurately detect α-synuclein in biofluids.Expert opinion: Data-driven strategies using quantitative rather than categorical variables may be more reliable for quantification of contributions from pathophysiological mechanisms and their spatial-temporal evolution. A systems biology approach is suitable to untangle the dynamics triggering loss of proteostasis, driving neurodegeneration and clinical evolution.
Collapse
Affiliation(s)
- Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
| | - Sonia Mazzucchi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Linda Giampietri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicola Giannini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fanny M. Elahi
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière Hospital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière Hospital, Paris, France
| | - Filippo Sean Giorgi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
95
|
Zhao H, Li S, Li Z, Yang S, Li D, Zheng J, Gao H, Yun L, Gu Y, Li L, Zhao J, Fu Y. Intranasal delivery of 9-cis retinoic acid reduces beta-amyloid deposition via inhibiting astrocyte-mediated inflammation. Aging (Albany NY) 2020; 12:5469-5478. [PMID: 32209731 PMCID: PMC7138573 DOI: 10.18632/aging.102970] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is associated with the accumulation and deposition of a beta-amyloid (Αβ) peptide in the brain, resulting in increased neuroinflammation and synaptic dysfunction. Intranasal delivery of targeted drugs to the brain represents a noninvasive pathway that bypasses the blood-brain barrier and minimizes systemic exposure. The aim of this study was to evaluate the therapeutic effect of intranasally delivered 9-cis retinoic acid (RA) on the neuropathology of an AD mouse model. Herein, we observed dramatically decreased Αβ deposition in the brains of amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic mice (APP/PS1) treated intranasally with 9-cis RA for 4 weeks compared to that in the brains of vehicle-treated mice. Importantly, intranasal delivery of 9-cis RA suppressed Αβ-associated astrocyte activation and neuroinflammation and ultimately restored synaptic deficits in APP/PS1 transgenic mice. These results support the critical roles of Αβ-associated neuroinflammation responses to synaptic deficits, particularly during the deposition of Αβ. Our findings provide strong evidence that intranasally delivered 9-cis RA attenuates neuronal dysfunction in an AD mouse model and is a promising therapeutic strategy for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Shuo Li
- Department of Ultrasonography, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Zhuo Li
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, China
| | - Shuo Yang
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Dandan Li
- Department of Neurology, The Second Hospital of Heilongjiang Province, Harbin, China
| | - Jiaolin Zheng
- Department of Neurology, The Second Hospital of Harbin Medical University, Harbin, China
| | - Hongmei Gao
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Ling Yun
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - YingLi Gu
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Longxuan Li
- Department of Neurology, Gongli Hospital of The Second Military Medical University, Shanghai, China
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuan Fu
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
96
|
Wang Y, Zhang X, Song Q, Hou Y, Liu J, Sun Y, Wang P. Characterization of the chromatin accessibility in an Alzheimer's disease (AD) mouse model. ALZHEIMERS RESEARCH & THERAPY 2020; 12:29. [PMID: 32293531 PMCID: PMC7092509 DOI: 10.1186/s13195-020-00598-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
Background The pathological hallmarks of Alzheimer’s disease (AD) involve alterations in the expression of numerous genes associated with transcriptional levels, which are determined by chromatin accessibility. Here, the landscape of chromatin accessibility was studied to understand the outline of the transcription and expression of AD-associated metabolism genes in an AD mouse model. Methods The assay for transposase-accessible chromatin by sequencing (ATAC-seq) was used to investigate the AD-associated chromatin reshaping in the APPswe/PS1dE9 (APP/PS1) mouse model. ATAC-seq data in the hippocampus of 8-month-old APP/PS1 mice were generated, and the relationship between chromatin accessibility and gene expression was analyzed in combination with RNA sequencing. Gene ontology (GO) analysis was applied to elucidate biological processes and signaling pathways altered in APP/PS1 mice. Critical transcription factors were identified; alterations in chromatin accessibility were further confirmed using chromatin immunoprecipitation assays. Results We identified 1690 increased AD-associated chromatin-accessible regions in the hippocampal tissues of APP/PS1 mice. These regions were enriched in genes related to diverse signaling pathways, including the PI3K-Akt, Hippo, TGF-β, and Jak-Stat signaling pathways, which play essential roles in regulating cell proliferation, apoptosis, and inflammatory responses. A total of 1003 decreased chromatin-accessible regions were considered to be related with declined AD-associated biological processes including cellular response to hyperoxia and insulin stimulus, synaptic transmission, and positive regulation of autophagy. In the APP/PS1 hippocampus, 1090 genes were found to be upregulated and 1081 downregulated. Interestingly, enhanced ATAC-seq signal was found in approximately 740 genes, with 43 exhibiting upregulated mRNA levels. Several genes involved in AD development were found to have a significantly increased expression in APP/PS1 mice compared to controls, including Sele, Clec7a, Cst7, and Ccr6. The signatures of numerous transcription factors, including Olig2, NeuroD1, TCF4, and NeuroG2, were found enriched in the AD-associated accessible chromatin regions. The transcription-activating marks of H3K4me3 and H3K27ac were also found increased in the promoters of these genes. These results indicate that the mechanism for the upregulation of genes could be attributed to the enrichment of open chromatin regions with transcription factors motifs and the histone marks H3K4me3 and H3K27ac. Conclusion Our study reveals that alterations in chromatin accessibility may be an initial mechanism in AD pathogenesis. Supplementary information Supplementary information accompanies this paper at 10.1186/s13195-020-00598-2.
Collapse
Affiliation(s)
- Yaqi Wang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Xiaomin Zhang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Qiao Song
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Yuli Hou
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Jing Liu
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China
| | - Yu Sun
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China.
| | - Peichang Wang
- Clinical Laboratory of Xuanwu Hospital, Capital Medical University, Beijing, 100053, People's Republic of China.
| |
Collapse
|
97
|
Glennon EB, Lau DHW, Gabriele RMC, Taylor MF, Troakes C, Opie-Martin S, Elliott C, Killick R, Hanger DP, Perez-Nievas BG, Noble W. Bridging Integrator-1 protein loss in Alzheimer's disease promotes synaptic tau accumulation and disrupts tau release. Brain Commun 2020; 2. [PMID: 32500121 PMCID: PMC7272218 DOI: 10.1093/braincomms/fcaa011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Polymorphisms associated with BIN1 (bridging integrator 1) confer the second greatest risk for developing late-onset Alzheimer’s disease. The biological consequences of this genetic variation are not fully understood; however, BIN1 is a binding partner for tau. Tau is normally a highly soluble cytoplasmic protein, but in Alzheimer’s disease, tau is abnormally phosphorylated and accumulates at synapses to exert synaptotoxicity. The purpose of this study was to determine whether alterations in BIN1 and tau in Alzheimer’s disease promote the damaging redistribution of tau to synapses, as a mechanism by which BIN1 polymorphisms may increase the risk of developing Alzheimer’s disease. We show that BIN1 is lost from the cytoplasmic fraction of Alzheimer’s disease cortex, and this is accompanied by the progressive mislocalization of phosphorylated tau to synapses. We confirmed proline 216 in tau as critical for tau interaction with the BIN1-SH3 domain and showed that the phosphorylation of tau disrupts this binding, suggesting that tau phosphorylation in Alzheimer’s disease disrupts tau–BIN1 associations. Moreover, we show that BIN1 knockdown in rat primary neurons to mimic BIN1 loss in Alzheimer’s disease brain causes the damaging accumulation of phosphorylated tau at synapses and alterations in dendritic spine morphology. We also observed reduced release of tau from neurons upon BIN1 silencing, suggesting that BIN1 loss disrupts the function of extracellular tau. Together, these data indicate that polymorphisms associated with BIN1 that reduce BIN1 protein levels in the brain likely act synergistically with increased tau phosphorylation to increase the risk of Alzheimer’s disease by disrupting cytoplasmic tau–BIN1 interactions, promoting the damaging mis-sorting of phosphorylated tau to synapses to alter synapse structure and reducing the release of physiological forms of tau to disrupt tau function.
Collapse
Affiliation(s)
- Elizabeth B Glennon
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Dawn H-W Lau
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Rebecca M C Gabriele
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Matthew F Taylor
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Claire Troakes
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK.,King's College London, MRC London Neurodegenerative Diseases Brain Bank, London, UK
| | - Sarah Opie-Martin
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Christina Elliott
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Old Age Psychiatry, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Richard Killick
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Old Age Psychiatry, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Diane P Hanger
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Beatriz G Perez-Nievas
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| | - Wendy Noble
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, 5 Cutcombe Road, London, SE5 9RX. UK
| |
Collapse
|
98
|
Alzheimer’s Disease Genetics: Review of Novel Loci Associated with Disease. CURRENT GENETIC MEDICINE REPORTS 2020. [DOI: 10.1007/s40142-020-00182-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
99
|
Yang GX, Huang Y, Zheng LL, Zhang L, Su L, Wu YH, Li J, Zhou LC, Huang J, Tang Y, Wang R, Ma L. Design, synthesis and evaluation of diosgenin carbamate derivatives as multitarget anti-Alzheimer’s disease agents. Eur J Med Chem 2020; 187:111913. [DOI: 10.1016/j.ejmech.2019.111913] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022]
|
100
|
Fossel M. A unified model of dementias and age-related neurodegeneration. Alzheimers Dement 2020; 16:365-383. [PMID: 31943780 DOI: 10.1002/alz.12012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/09/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022]
|